a LANGE medical book
Harper’s
Illustrated
Biochemistry
twenty-sixth edition
Robert K. Murray, MD, PhD
Professor (Emeritus) of Biochemistry
University of Toronto
Toronto, Ontario
Daryl K. Granner, MD
Joe C. Davis Professor of Biomedical Science
Director, Vanderbilt Diabetes Center
Professor of Molecular Physiology and Biophysics
and of Medicine
Vanderbilt University
Nashville, Tennessee
Peter A. Mayes, PhD, DSc
Emeritus Professor of Veterinary Biochemistry
Royal Veterinary College
University of London
London
Victor W. Rodwell, PhD
Professor of Biochemistry
Purdue University
West Lafayette, Indiana
Lange Medical Books/McGraw-Hill
Medical Publishing Division
New York Chicago San Francisco Lisbon London Madrid Mexico City
Milan New Delhi San Juan Seoul Singapore Sydney Toronto
Harper’s Illustrated Biochemistry, Twenty-Sixth Edition
Copyright © 2003 by The McGraw-Hill Companies, Inc. All rights reserved. Printed in the United States of America. Except as
permitted under the United States Copyright Act of 1976, no part of this publication may be reproduced or distributed in any
form or by any means, or stored in a data base or retrieval system, without the prior written permission of the publisher.
Previous editions copyright © 2000, 1996, 1993, 1990 by Appleton & Lange; copyright © 1988 by Lange Medical Publications.
2 3 4 5 6 7 8 9 0 DOC/DOC 0 9 8 7 6 5 4 3
ISBN 0-07-138901-6
ISSN 1043-9811
Notice
Medicine is an ever-changing science. As new research and clinical experience broaden our knowledge, changes in treat-
ment and drug therapy are required. The authors and the publisher of this work have checked with sources believed to be
reliable in their efforts to provide information that is complete and generally in accord with the standards accepted at the
time of publication. However, in view of the possibility of human error or changes in medical sciences, neither the authors
nor the publisher nor any other party who has been involved in the preparation or publication of this work warrants that
the information contained herein is in every respect accurate or complete, and they disclaim all responsibility for any er-
rors or omissions or for the results obtained from use of the information contained in this work. Readers are encouraged
to confirm the information contained herein with other sources. For example and in particular, readers are advised to
check the product information sheet included in the package of each drug they plan to administer to be certain that the
information contained in this work is accurate and that changes have not been made in the recommended dose or in the
contraindications for administration. This recommendation is of particular importance in connection with new or infre-
quently used drugs.
This book was set in Garamond by Pine Tree Composition
The editors were Janet Foltin, Jim Ransom, and Janene Matragrano Oransky.
The production supervisor was Phil Galea.
The illustration manager was Charissa Baker.
The text designer was Eve Siegel.
The cover designer was Mary McKeon.
The index was prepared by Kathy Pitcoff.
RR Donnelley was printer and binder.
This book is printed on acid-free paper.
ISBN-0-07-121766-5 (International Edition)
Copyright © 2003. Exclusive rights by the McGraw-Hill Companies, Inc., for
manufacture and export. This book cannot be re-exported from the country to which it
is consigned by McGraw-Hill. The International Edition is not available in North America.
Authors
David A. Bender, PhD
Peter A. Mayes, PhD, DSc
Sub-Dean Royal Free and University College Medical
Emeritus Professor of Veterinary Biochemistry, Royal
School, Assistant Faculty Tutor and Tutor to Med-
Veterinary College, University of London
ical Students, Senior Lecturer in Biochemistry, De-
partment of Biochemistry and Molecular Biology,
University College London
Robert K. Murray, MD, PhD
Professor
(Emeritus) of Biochemistry, University of
Toronto
Kathleen M. Botham, PhD, DSc
Reader in Biochemistry, Royal Veterinary College,
University of London
Margaret L. Rand, PhD
Scientist, Research Institute, Hospital for Sick Chil-
Daryl K. Granner, MD
dren, Toronto, and Associate Professor, Depart-
ments of Laboratory Medicine and Pathobiology
Joe C. Davis Professor of Biomedical Science, Director,
and Department of Biochemistry, University of
Vanderbilt Diabetes Center, Professor of Molecular
Toronto
Physiology and Biophysics and of Medicine, Vander-
bilt University, Nashville, Tennessee
Victor W. Rodwell, PhD
Professor of Biochemistry, Purdue University, West
Frederick W. Keeley, PhD
Lafayette, Indiana
Associate Director and Senior Scientist, Research Insti-
tute, Hospital for Sick Children, Toronto, and Pro-
fessor, Department of Biochemistry, University of
P. Anthony Weil, PhD
Toronto
Professor of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nash-
Peter J. Kennelly, PhD
ville, Tennessee
Professor of Biochemistry, Virginia Polytechnic Insti-
tute and State University, Blacksburg, Virginia
vii
Preface
The authors and publisher are pleased to present the twenty-sixth edition of Harper’s Illustrated Biochemistry. Review
of Physiological Chemistry was first published in 1939 and revised in 1944, and it quickly gained a wide readership. In
1951, the third edition appeared with Harold A. Harper, University of California School of Medicine at San Fran-
cisco, as author. Dr. Harper remained the sole author until the ninth edition and co-authored eight subsequent edi-
tions. Peter Mayes and Victor Rodwell have been authors since the tenth edition, Daryl Granner since the twentieth
edition, and Rob Murray since the twenty-first edition. Because of the increasing complexity of biochemical knowl-
edge, they have added co-authors in recent editions.
Fred Keeley and Margaret Rand have each co-authored one chapter with Rob Murray for this and previous edi-
tions. Peter Kennelly joined as a co-author in the twenty-fifth edition, and in the present edition has co-authored
with Victor Rodwell all of the chapters dealing with the structure and function of proteins and enzymes. The follow-
ing additional co-authors are very warmly welcomed in this edition: Kathleen Botham has co-authored, with Peter
Mayes, the chapters on bioenergetics, biologic oxidation, oxidative phosphorylation, and lipid metabolism. David
Bender has co-authored, also with Peter Mayes, the chapters dealing with carbohydrate metabolism, nutrition, diges-
tion, and vitamins and minerals. P. Anthony Weil has co-authored chapters dealing with various aspects of DNA, of
RNA, and of gene expression with Daryl Granner. We are all very grateful to our co-authors for bringing their ex-
pertise and fresh perspectives to the text.
CHANGES IN THE TWENTY-SIXTH EDITION
A major goal of the authors continues to be to provide both medical and other students of the health sciences with a
book that both describes the basics of biochemistry and is user-friendly and interesting. A second major ongoing
goal is to reflect the most significant advances in biochemistry that are important to medicine. However, a third
major goal of this edition was to achieve a substantial reduction in size, as feedback indicated that many readers pre-
fer shorter texts.
To achieve this goal, all of the chapters were rigorously edited, involving their amalgamation, division, or dele-
tion, and many were reduced to approximately one-half to two-thirds of their previous size. This has been effected
without loss of crucial information but with gain in conciseness and clarity.
Despite the reduction in size, there are many new features in the twenty-sixth edition. These include:
• A new chapter on amino acids and peptides, which emphasizes the manner in which the properties of biologic
peptides derive from the individual amino acids of which they are comprised.
• A new chapter on the primary structure of proteins, which provides coverage of both classic and newly emerging
“proteomic” and “genomic” methods for identifying proteins. A new section on the application of mass spectrometry
to the analysis of protein structure has been added, including comments on the identification of covalent modifica-
tions.
• The chapter on the mechanisms of action of enzymes has been revised to provide a comprehensive description of
the various physical mechanisms by which enzymes carry out their catalytic functions.
• The chapters on integration of metabolism, nutrition, digestion and absorption, and vitamins and minerals have
been completely re-written.
• Among important additions to the various chapters on metabolism are the following: update of the information
on oxidative phosphorylation, including a description of the rotary ATP synthase; new insights into the role of
GTP in gluconeogenesis; additional information on the regulation of acetyl-CoA carboxylase; new information on
receptors involved in lipoprotein metabolism and reverse cholesterol transport; discussion of the role of leptin in
fat storage; and new information on bile acid regulation, including the role of the farnesoid X receptor (FXR).
• The chapter on membrane biochemistry in the previous edition has been split into two, yielding two new chapters
on the structure and function of membranes and intracellular traffic and sorting of proteins.
• Considerable new material has been added on RNA synthesis, protein synthesis, gene regulation, and various as-
pects of molecular genetics.
• Much of the material on individual endocrine glands present in the twenty-fifth edition has been replaced with
new chapters dealing with the diversity of the endocrine system, with molecular mechanisms of hormone action,
and with signal transduction.
ix
x
/
PREFACE
• The chapter on plasma proteins, immunoglobulins, and blood coagulation in the previous edition has been split
into two new chapters on plasma proteins and immunoglobulins and on hemostasis and thrombosis.
• New information has been added in appropriate chapters on lipid rafts and caveolae, aquaporins, connexins, dis-
orders due to mutations in genes encoding proteins involved in intracellular membrane transport, absorption of
iron, and conformational diseases and pharmacogenomics.
• A new and final chapter on “The Human Genome Project” (HGP) has been added, which builds on the material
covered in Chapters 35 through 40. Because of the impact of the results of the HGP on the future of biology and
medicine, it appeared appropriate to conclude the text with a summary of its major findings and their implica-
tions for future work.
• As initiated in the previous edition, references to useful Web sites have been included in a brief Appendix at the
end of the text.
ORGANIZATION OF THE BOOK
The text is divided into two introductory chapters (“Biochemistry & Medicine” and “Water & pH”) followed by six
main sections.
Section I deals with the structures and functions of proteins and enzymes, the workhorses of the body. Because
almost all of the reactions in cells are catalyzed by enzymes, it is vital to understand the properties of enzymes before
considering other topics.
Section II explains how various cellular reactions either utilize or release energy, and it traces the pathways by
which carbohydrates and lipids are synthesized and degraded. It also describes the many functions of these two
classes of molecules.
Section III deals with the amino acids and their many fates and also describes certain key features of protein ca-
tabolism.
Section IV describes the structures and functions of the nucleotides and nucleic acids, and covers many major
topics such as DNA replication and repair, RNA synthesis and modification, and protein synthesis. It also discusses
new findings on how genes are regulated and presents the principles of recombinant DNA technology.
Section V deals with aspects of extracellular and intracellular communication. Topics covered include membrane
structure and function, the molecular bases of the actions of hormones, and the key field of signal transduction.
Section VI consists of discussions of eleven special topics: nutrition, digestion, and absorption; vitamins and
minerals; intracellular traffic and sorting of proteins; glycoproteins; the extracellular matrix; muscle and the cy-
toskeleton; plasma proteins and immunoglobulins; hemostasis and thrombosis; red and white blood cells; the me-
tabolism of xenobiotics; and the Human Genome Project.
ACKNOWLEDGMENTS
The authors thank Janet Foltin for her thoroughly professional approach. Her constant interest and input have had a
significant impact on the final structure of this text. We are again immensely grateful to Jim Ransom for his excel-
lent editorial work; it has been a pleasure to work with an individual who constantly offered wise and informed alter-
natives to the sometimes primitive text transmitted by the authors. The superb editorial skills of Janene Matragrano
Oransky and Harriet Lebowitz are warmly acknowledged, as is the excellent artwork of Charissa Baker and her col-
leagues. The authors are very grateful to Kathy Pitcoff for her thoughtful and meticulous work in preparing the
Index. Suggestions from students and colleagues around the world have been most helpful in the formulation of this
edition. We look forward to receiving similar input in the future.
Robert K. Murray, MD, PhD
Daryl K. Granner, MD
Peter A. Mayes, PhD, DSc
Victor W. Rodwell, PhD
Toronto, Ontario
Nashville, Tennessee
London
West Lafayette, Indiana
March 2003
Contents
Authors
vii
Preface
ix
1. Biochemistry & Medicine
Robert K. Murray, MD, PhD
1
2. Water & pH
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
5
SECTION I. STRUCTURES & FUNCTIONS OF PROTEINS & ENZYMES
14
3. Amino Acids & Peptides
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
14
4. Proteins: Determination of Primary Structure
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
21
5. Proteins: Higher Orders of Structure
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
30
6. Proteins: Myoglobin & Hemoglobin
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
40
7. Enzymes: Mechanism of Action
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
49
8. Enzymes: Kinetics
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
60
9. Enzymes: Regulation of Activities
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
72
SECTION II. BIOENERGETICS & THE METABOLISM OF CARBOHYDRATES
& LIPIDS
80
10. Bioenergetics: The Role of ATP
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
80
11. Biologic Oxidation
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
86
12. The Respiratory Chain & Oxidative Phosphorylation
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
92
13. Carbohydrates of Physiologic Significance
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
102
iii
iv
/
CONTENTS
14. Lipids of Physiologic Significance
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
111
15. Overview of Metabolism
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
122
16. The Citric Acid Cycle: The Catabolism of Acetyl-CoA
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
130
17. Glycolysis & the Oxidation of Pyruvate
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
136
18. Metabolism of Glycogen
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
145
19. Gluconeogenesis & Control of the Blood Glucose
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
153
20. The Pentose Phosphate Pathway & Other Pathways of Hexose Metabolism
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
163
21. Biosynthesis of Fatty Acids
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
173
22. Oxidation of Fatty Acids: Ketogenesis
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
180
23. Metabolism of Unsaturated Fatty Acids & Eicosanoids
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
190
24. Metabolism of Acylglycerols & Sphingolipids
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
197
25. Lipid Transport & Storage
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
205
26. Cholesterol Synthesis, Transport, & Excretion
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
219
27. Integration of Metabolism—the Provision of Metabolic Fuels
David A. Bender, PhD, & Peter A. Mayes, PhD, DSc
231
SECTION III. METABOLISM OF PROTEINS & AMINO ACIDS
237
28. Biosynthesis of the Nutritionally Nonessential Amino Acids
Victor W. Rodwell, PhD
237
29. Catabolism of Proteins & of Amino Acid Nitrogen
Victor W. Rodwell, PhD
242
CONTENTS
/
v
30. Catabolism of the Carbon Skeletons of Amino Acids
Victor W. Rodwell, PhD
249
31. Conversion of Amino Acids to Specialized Products
Victor W. Rodwell, PhD
264
32. Porphyrins & Bile Pigments
Robert K. Murray, MD, PhD
270
SECTION IV. STRUCTURE, FUNCTION, & REPLICATION
OF INFORMATIONAL MACROMOLECULES
286
33. Nucleotides
Victor W. Rodwell, PhD
286
34. Metabolism of Purine & Pyrimidine Nucleotides
Victor W. Rodwell, PhD
293
35. Nucleic Acid Structure & Function
Daryl K. Granner, MD
303
36. DNA Organization, Replication, & Repair
Daryl K. Granner, MD, & P. Anthony Weil, PhD
314
37. RNA Synthesis, Processing, & Modification
Daryl K. Granner, MD, & P. Anthony Weil, PhD
341
38. Protein Synthesis & the Genetic Code
Daryl K. Granner, MD
358
39. Regulation of Gene Expression
Daryl K. Granner, MD, & P. Anthony Weil, PhD
374
40. Molecular Genetics, Recombinant DNA, & Genomic Technology
Daryl K. Granner, MD, & P. Anthony Weil, PhD
396
SECTION V. BIOCHEMISTRY OF EXTRACELLULAR
& INTRACELLULAR COMMUNICATION
415
41. Membranes: Structure & Function
Robert K. Murray, MD, PhD, & Daryl K. Granner, MD
415
42. The Diversity of the Endocrine System
Daryl K. Granner, MD
434
43. Hormone Action & Signal Transduction
Daryl K. Granner, MD
456
vi
/
CONTENTS
SECTION VI. SPECIAL TOPICS
474
44. Nutrition, Digestion, & Absorption
David A. Bender, PhD, & Peter A. Mayes, PhD, DSc
474
45. Vitamins & Minerals
David A. Bender, PhD, & Peter A. Mayes, PhD, DSc
481
46. Intracellular Traffic & Sorting of Proteins
Robert K. Murray, MD, PhD
498
47. Glycoproteins
Robert K. Murray, MD, PhD
514
48. The Extracellular Matrix
Robert K. Murray, MD, PhD, & Frederick W. Keeley, PhD
535
49. Muscle & the Cytoskeleton
Robert K. Murray, MD, PhD
556
50. Plasma Proteins & Immunoglobulins
Robert K. Murray, MD, PhD
580
51. Hemostasis & Thrombosis
Margaret L. Rand, PhD, & Robert K. Murray, MD, PhD
598
52. Red & White Blood Cells
Robert K. Murray, MD, PhD
609
53. Metabolism of Xenobiotics
Robert K. Murray, MD, PhD
626
54. The Human Genome Project
Robert K. Murray, MD, PhD
633
Appendix
639
Index
643
Biochemistry & Medicine
1
Robert K. Murray, MD, PhD
INTRODUCTION
biochemistry is increasingly becoming their common
language.
Biochemistry can be defined as the science concerned
with the chemical basis of life (Gk bios “life”). The cell is
the structural unit of living systems. Thus, biochem-
A Reciprocal Relationship Between
istry can also be described as the science concerned with
Biochemistry & Medicine Has Stimulated
the chemical constituents of living cells and with the reac-
Mutual Advances
tions and processes they undergo. By this definition, bio-
The two major concerns for workers in the health sci-
chemistry encompasses large areas of cell biology, of
molecular biology, and of molecular genetics.
ences—and particularly physicians—are the understand-
ing and maintenance of health and the understanding
and effective treatment of diseases. Biochemistry im-
The Aim of Biochemistry Is to Describe &
pacts enormously on both of these fundamental con-
Explain, in Molecular Terms, All Chemical
cerns of medicine. In fact, the interrelationship of bio-
Processes of Living Cells
chemistry and medicine is a wide, two-way street.
The major objective of biochemistry is the complete
Biochemical studies have illuminated many aspects of
understanding, at the molecular level, of all of the
health and disease, and conversely, the study of various
chemical processes associated with living cells. To
aspects of health and disease has opened up new areas
achieve this objective, biochemists have sought to iso-
of biochemistry. Some examples of this two-way street
late the numerous molecules found in cells, determine
are shown in Figure 1-1. For instance, a knowledge of
their structures, and analyze how they function. Many
protein structure and function was necessary to eluci-
techniques have been used for these purposes; some of
date the single biochemical difference between normal
them are summarized in Table 1-1.
hemoglobin and sickle cell hemoglobin. On the other
hand, analysis of sickle cell hemoglobin has contributed
significantly to our understanding of the structure and
A Knowledge of Biochemistry Is Essential
function of both normal hemoglobin and other pro-
to All Life Sciences
teins. Analogous examples of reciprocal benefit between
The biochemistry of the nucleic acids lies at the heart of
biochemistry and medicine could be cited for the other
genetics; in turn, the use of genetic approaches has been
paired items shown in Figure 1-1. Another example is
critical for elucidating many areas of biochemistry.
the pioneering work of Archibald Garrod, a physician
Physiology, the study of body function, overlaps with
in England during the early 1900s. He studied patients
biochemistry almost completely. Immunology employs
with a number of relatively rare disorders
(alkap-
numerous biochemical techniques, and many immuno-
tonuria, albinism, cystinuria, and pentosuria; these are
logic approaches have found wide use by biochemists.
described in later chapters) and established that these
Pharmacology and pharmacy rest on a sound knowl-
conditions were genetically determined. Garrod desig-
edge of biochemistry and physiology; in particular,
nated these conditions as inborn errors of metabo-
most drugs are metabolized by enzyme-catalyzed reac-
lism. His insights provided a major foundation for the
tions. Poisons act on biochemical reactions or processes;
development of the field of human biochemical genet-
this is the subject matter of toxicology. Biochemical ap-
ics. More recent efforts to understand the basis of the
proaches are being used increasingly to study basic as-
genetic disease known as familial hypercholesterol-
pects of pathology (the study of disease), such as in-
emia, which results in severe atherosclerosis at an early
flammation, cell injury, and cancer. Many workers in
age, have led to dramatic progress in understanding of
microbiology, zoology, and botany employ biochemical
cell receptors and of mechanisms of uptake of choles-
approaches almost exclusively. These relationships are
terol into cells. Studies of oncogenes in cancer cells
not surprising, because life as we know it depends on
have directed attention to the molecular mechanisms
biochemical reactions and processes. In fact, the old
involved in the control of normal cell growth. These
barriers among the life sciences are breaking down, and
and many other examples emphasize how the study of
1
2
/
CHAPTER 1
Table 1-1. The principal methods and
NORMAL BIOCHEMICAL PROCESSES ARE
preparations used in biochemical laboratories.
THE BASIS OF HEALTH
The World Health Organization
(WHO) defines
Methods for Separating and Purifying Biomolecules1
health as a state of “complete physical, mental and so-
Salt fractionation (eg, precipitation of proteins with ammo-
cial well-being and not merely the absence of disease
nium sulfate)
and infirmity.” From a strictly biochemical viewpoint,
Chromatography: Paper; ion exchange; affinity; thin-layer;
health may be considered that situation in which all of
gas-liquid; high-pressure liquid; gel filtration
the many thousands of intra- and extracellular reactions
Electrophoresis: Paper; high-voltage; agarose; cellulose
that occur in the body are proceeding at rates commen-
acetate; starch gel; polyacrylamide gel; SDS-polyacryl-
surate with the organism’s maximal survival in the
amide gel
physiologic state. However, this is an extremely reduc-
Ultracentrifugation
Methods for Determining Biomolecular Structures
tionist view, and it should be apparent that caring for
Elemental analysis
the health of patients requires not only a wide knowl-
UV, visible, infrared, and NMR spectroscopy
edge of biologic principles but also of psychologic and
Use of acid or alkaline hydrolysis to degrade the biomole-
social principles.
cule under study into its basic constituents
Use of a battery of enzymes of known specificity to de-
Biochemical Research Has Impact on
grade the biomolecule under study (eg, proteases, nucle-
Nutrition & Preventive Medicine
ases, glycosidases)
Mass spectrometry
One major prerequisite for the maintenance of health is
Specific sequencing methods (eg, for proteins and nucleic
that there be optimal dietary intake of a number of
acids)
chemicals; the chief of these are vitamins, certain
X-ray crystallography
amino acids, certain fatty acids, various minerals, and
Preparations for Studying Biochemical Processes
water. Because much of the subject matter of both bio-
Whole animal (includes transgenic animals and animals
chemistry and nutrition is concerned with the study of
with gene knockouts)
various aspects of these chemicals, there is a close rela-
Isolated perfused organ
tionship between these two sciences. Moreover, more
Tissue slice
emphasis is being placed on systematic attempts to
Whole cells
maintain health and forestall disease, ie, on preventive
Homogenate
Isolated cell organelles
medicine. Thus, nutritional approaches to—for exam-
Subfractionation of organelles
ple—the prevention of atherosclerosis and cancer are
Purified metabolites and enzymes
receiving increased emphasis. Understanding nutrition
Isolated genes (including polymerase chain reaction and
depends to a great extent on a knowledge of biochem-
site-directed mutagenesis)
istry.
1Most of these methods are suitable for analyzing the compo-
nents present in cell homogenates and other biochemical prepa-
Most & Perhaps All Disease Has
rations. The sequential use of several techniques will generally
a Biochemical Basis
permit purification of most biomolecules. The reader is referred
to texts on methods of biochemical research for details.
We believe that most if not all diseases are manifesta-
tions of abnormalities of molecules, chemical reactions,
or biochemical processes. The major factors responsible
disease can open up areas of cell function for basic bio-
for causing diseases in animals and humans are listed in
chemical research.
Table 1-2. All of them affect one or more critical
The relationship between medicine and biochem-
chemical reactions or molecules in the body. Numerous
istry has important implications for the former. As long
examples of the biochemical bases of diseases will be en-
as medical treatment is firmly grounded in a knowledge
countered in this text; the majority of them are due to
of biochemistry and other basic sciences, the practice of
causes 5, 7, and 8. In most of these conditions, bio-
medicine will have a rational basis that can be adapted
chemical studies contribute to both the diagnosis and
to accommodate new knowledge. This contrasts with
treatment. Some major uses of biochemical investiga-
unorthodox health cults and at least some “alternative
tions and of laboratory tests in relation to diseases are
medicine” practices, which are often founded on little
summarized in Table 1-3.
more than myth and wishful thinking and generally
Additional examples of many of these uses are pre-
lack any intellectual basis.
sented in various sections of this text.
BIOCHEMISTRY & MEDICINE
/
3
BIOCHEMISTRY
Nucleic
acids
Proteins
Lipids
Carbohydrates
Genetic
Sickle cell
Athero-
Diabetes
diseases
anemia
sclerosis
mellitus
MEDICINE
Figure 1-1. Examples of the two-way street connecting biochemistry and
medicine. Knowledge of the biochemical molecules shown in the top part of the
diagram has clarified our understanding of the diseases shown in the bottom
half—and conversely, analyses of the diseases shown below have cast light on
many areas of biochemistry. Note that sickle cell anemia is a genetic disease and
that both atherosclerosis and diabetes mellitus have genetic components.
Impact of the Human Genome Project
(HGP) on Biochemistry & Medicine
Table 1-3. Some uses of biochemical
Remarkable progress was made in the late 1990s in se-
investigations and laboratory tests in
quencing the human genome. This culminated in July
relation to diseases.
2000, when leaders of the two groups involved in this
effort (the International Human Genome Sequencing
Consortium and Celera Genomics, a private company)
Use
Example
announced that over 90% of the genome had been se-
1.
To reveal the funda-
Demonstration of the na-
quenced. Draft versions of the sequence were published
mental causes and
ture of the genetic de-
mechanisms of diseases
fects in cystic fibrosis.
2.
To suggest rational treat-
A diet low in phenylalanine
Table 1-2. The major causes of diseases. All of
ments of diseases based
for treatment of phenyl-
on (1) above
ketonuria.
the causes listed act by influencing the various
3.
To assist in the diagnosis
Use of the plasma enzyme
biochemical mechanisms in the cell or in the
of specific diseases
creatine kinase MB
body.1
(CK-MB) in the diagnosis
of myocardial infarction.
1. Physical agents: Mechanical trauma, extremes of temper-
4.
To act as screening tests
Use of measurement of
ature, sudden changes in atmospheric pressure, radia-
for the early diagnosis
blood thyroxine or
tion, electric shock.
of certain diseases
thyroid-stimulating hor-
2. Chemical agents, including drugs: Certain toxic com-
mone (TSH) in the neo-
pounds, therapeutic drugs, etc.
natal diagnosis of con-
3. Biologic agents: Viruses, bacteria, fungi, higher forms of
genital hypothyroidism.
parasites.
5.
To assist in monitoring
Use of the plasma enzyme
4. Oxygen lack: Loss of blood supply, depletion of the
the progress (eg, re-
alanine aminotransferase
oxygen-carrying capacity of the blood, poisoning of
covery, worsening, re-
(ALT) in monitoring the
the oxidative enzymes.
mission, or relapse) of
progress of infectious
5. Genetic disorders: Congenital, molecular.
certain diseases
hepatitis.
6. Immunologic reactions: Anaphylaxis, autoimmune
6.
To assist in assessing
Use of measurement of
disease.
the response of dis-
blood carcinoembryonic
7. Nutritional imbalances: Deficiencies, excesses.
eases to therapy
antigen (CEA) in certain
8. Endocrine imbalances: Hormonal deficiencies, excesses.
patients who have been
treated for cancer of the
1Adapted, with permission, from Robbins SL, Cotram RS, Kumar V:
colon.
The Pathologic Basis of Disease, 3rd ed. Saunders, 1984.
4
/
CHAPTER 1
in early 2001. It is anticipated that the entire sequence
• The judicious use of various biochemical laboratory
will be completed by 2003. The implications of this
tests is an integral component of diagnosis and moni-
work for biochemistry, all of biology, and for medicine
toring of treatment.
are tremendous, and only a few points are mentioned
• A sound knowledge of biochemistry and of other re-
here. Many previously unknown genes have been re-
lated basic disciplines is essential for the rational
vealed; their protein products await characterization.
practice of medical and related health sciences.
New light has been thrown on human evolution, and
procedures for tracking disease genes have been greatly
refined. The results are having major effects on areas
REFERENCES
such as proteomics, bioinformatics, biotechnology, and
Fruton JS: Proteins, Enzymes, Genes: The Interplay of Chemistry and
pharmacogenomics. Reference to the human genome
Biology. Yale Univ Press, 1999. (Provides the historical back-
will be made in various sections of this text. The
ground for much of today’s biochemical research.)
Human Genome Project is discussed in more detail in
Garrod AE: Inborn errors of metabolism. (Croonian Lectures.)
Chapter 54.
Lancet 1908;2:1, 73, 142, 214.
International Human Genome Sequencing Consortium. Initial se-
SUMMARY
quencing and analysis of the human genome. Nature
2001:409;860. (The issue [15 February] consists of articles
• Biochemistry is the science concerned with studying
dedicated to analyses of the human genome.)
the various molecules that occur in living cells and
Kornberg A: Basic research: The lifeline of medicine. FASEB J
organisms and with their chemical reactions. Because
1992;6:3143.
life depends on biochemical reactions, biochemistry
Kornberg A: Centenary of the birth of modern biochemistry.
has become the basic language of all biologic sci-
FASEB J 1997;11:1209.
ences.
McKusick VA: Mendelian Inheritance in Man. Catalogs of Human
Genes and Genetic Disorders, 12th ed. Johns Hopkins Univ
• Biochemistry is concerned with the entire spectrum
Press, 1998. [Abbreviated MIM]
of life forms, from relatively simple viruses and bacte-
Online Mendelian Inheritance in Man (OMIM): Center for Med-
ria to complex human beings.
ical Genetics, Johns Hopkins University and National Center
• Biochemistry and medicine are intimately related.
for Biotechnology Information, National Library of Medi-
Health depends on a harmonious balance of bio-
chemical reactions occurring in the body, and disease
(The numbers assigned to the entries in MIM and OMIM will be
cited in selected chapters of this work. Consulting this exten-
reflects abnormalities in biomolecules, biochemical
sive collection of diseases and other relevant entries—specific
reactions, or biochemical processes.
proteins, enzymes, etc—will greatly expand the reader’s
• Advances in biochemical knowledge have illumi-
knowledge and understanding of various topics referred to
nated many areas of medicine. Conversely, the study
and discussed in this text. The online version is updated al-
of diseases has often revealed previously unsuspected
most daily.)
aspects of biochemistry. The determination of the se-
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
herited Disease, 8th ed. McGraw-Hill, 2001.
quence of the human genome, nearly complete, will
Venter JC et al: The Sequence of the Human Genome. Science
have a great impact on all areas of biology, including
2001;291:1304. (The issue [16 February] contains the Celera
biochemistry, bioinformatics, and biotechnology.
draft version and other articles dedicated to analyses of the
• Biochemical approaches are often fundamental in il-
human genome.)
luminating the causes of diseases and in designing
Williams DL, Marks V: Scientific Foundations of Biochemistry in
appropriate therapies.
Clinical Practice, 2nd ed. Butterworth-Heinemann, 1994.
Water & pH
2
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
oxygen atom pulls electrons away from the hydrogen
nuclei, leaving them with a partial positive charge,
Water is the predominant chemical component of liv-
while its two unshared electron pairs constitute a region
ing organisms. Its unique physical properties, which in-
of local negative charge.
clude the ability to solvate a wide range of organic and
Water, a strong dipole, has a high dielectric con-
inorganic molecules, derive from water’s dipolar struc-
stant. As described quantitatively by Coulomb’s law,
ture and exceptional capacity for forming hydrogen
the strength of interaction F between oppositely
bonds. The manner in which water interacts with a sol-
charged particles is inversely proportionate to the di-
vated biomolecule influences the structure of each. An
electric constant ε of the surrounding medium. The di-
excellent nucleophile, water is a reactant or product in
electric constant for a vacuum is unity; for hexane it is
many metabolic reactions. Water has a slight propensity
1.9; for ethanol it is 24.3; and for water it is 78.5.
to dissociate into hydroxide ions and protons. The
Water therefore greatly decreases the force of attraction
acidity of aqueous solutions is generally reported using
between charged and polar species relative to water-free
the logarithmic pH scale. Bicarbonate and other buffers
environments with lower dielectric constants. Its strong
normally maintain the pH of extracellular fluid be-
dipole and high dielectric constant enable water to dis-
tween 7.35 and 7.45. Suspected disturbances of acid-
solve large quantities of charged compounds such as
base balance are verified by measuring the pH of arter-
salts.
ial blood and the CO2 content of venous blood. Causes
of acidosis (blood pH < 7.35) include diabetic ketosis
and lactic acidosis. Alkalosis (pH > 7.45) may, for ex-
Water Molecules Form Hydrogen Bonds
ample, follow vomiting of acidic gastric contents. Regu-
An unshielded hydrogen nucleus covalently bound to
lation of water balance depends upon hypothalamic
an electron-withdrawing oxygen or nitrogen atom can
mechanisms that control thirst, on antidiuretic hor-
interact with an unshared electron pair on another oxy-
mone (ADH), on retention or excretion of water by the
gen or nitrogen atom to form a hydrogen bond. Since
kidneys, and on evaporative loss. Nephrogenic diabetes
water molecules contain both of these features, hydro-
insipidus, which involves the inability to concentrate
gen bonding favors the self-association of water mole-
urine or adjust to subtle changes in extracellular fluid
cules into ordered arrays (Figure 2-2). Hydrogen bond-
osmolarity, results from the unresponsiveness of renal
ing profoundly influences the physical properties of
tubular osmoreceptors to ADH.
water and accounts for its exceptionally high viscosity,
surface tension, and boiling point. On average, each
molecule in liquid water associates through hydrogen
WATER IS AN IDEAL BIOLOGIC SOLVENT
bonds with 3.5 others. These bonds are both relatively
weak and transient, with a half-life of about one mi-
Water Molecules Form Dipoles
crosecond. Rupture of a hydrogen bond in liquid water
A water molecule is an irregular, slightly skewed tetra-
requires only about 4.5 kcal/mol, less than 5% of the
hedron with oxygen at its center (Figure 2-1). The two
energy required to rupture a covalent O H bond.
hydrogens and the unshared electrons of the remaining
Hydrogen bonding enables water to dissolve many
two sp3-hybridized orbitals occupy the corners of the
organic biomolecules that contain functional groups
tetrahedron. The 105-degree angle between the hydro-
which can participate in hydrogen bonding. The oxy-
gens differs slightly from the ideal tetrahedral angle,
gen atoms of aldehydes, ketones, and amides provide
109.5 degrees. Ammonia is also tetrahedral, with a 107-
pairs of electrons that can serve as hydrogen acceptors.
degree angle between its hydrogens. Water is a dipole,
Alcohols and amines can serve both as hydrogen accep-
a molecule with electrical charge distributed asymmetri-
tors and as donors of unshielded hydrogen atoms for
cally about its structure. The strongly electronegative
formation of hydrogen bonds (Figure 2-3).
5
6
/
CHAPTER 2
H
CH3
CH
2
O
H
O
2e
H
2e
H
H
105°
3
CH CH
2
O
H
O
H
CH2
CH3
Figure 2-1. The water molecule has tetrahedral
R
RII
geometry.
C
O
H
N
RI
RIII
INTERACTION WITH WATER INFLUENCES
Figure 2-3. Additional polar groups participate in
THE STRUCTURE OF BIOMOLECULES
hydrogen bonding. Shown are hydrogen bonds formed
Covalent & Noncovalent Bonds Stabilize
between an alcohol and water, between two molecules
Biologic Molecules
of ethanol, and between the peptide carbonyl oxygen
and the peptide nitrogen hydrogen of an adjacent
The covalent bond is the strongest force that holds
amino acid.
molecules together (Table 2-1). Noncovalent forces,
while of lesser magnitude, make significant contribu-
tions to the structure, stability, and functional compe-
phosphatidyl serine or phosphatidyl ethanolamine con-
tence of macromolecules in living cells. These forces,
tact water while their hydrophobic fatty acyl side chains
which can be either attractive or repulsive, involve in-
cluster together, excluding water. This pattern maxi-
teractions both within the biomolecule and between it
mizes the opportunities for the formation of energeti-
and the water that forms the principal component of
cally favorable charge-dipole, dipole-dipole, and hydro-
the surrounding environment.
gen bonding interactions between polar groups on the
biomolecule and water. It also minimizes energetically
unfavorable contact between water and hydrophobic
Biomolecules Fold to Position Polar &
groups.
Charged Groups on Their Surfaces
Most biomolecules are amphipathic; that is, they pos-
Hydrophobic Interactions
sess regions rich in charged or polar functional groups
as well as regions with hydrophobic character. Proteins
Hydrophobic interaction refers to the tendency of non-
tend to fold with the R-groups of amino acids with hy-
polar compounds to self-associate in an aqueous envi-
drophobic side chains in the interior. Amino acids with
ronment. This self-association is driven neither by mu-
charged or polar amino acid side chains (eg, arginine,
tual attraction nor by what are sometimes incorrectly
glutamate, serine) generally are present on the surface
referred to as
“hydrophobic bonds.” Self-association
in contact with water. A similar pattern prevails in a
arises from the need to minimize energetically unfavor-
phospholipid bilayer, where the charged head groups of
able interactions between nonpolar groups and water.
Table 2-1. Bond energies for atoms of biologic
H H
H H
O
O
significance.
H
H
H
H O
O
H
O
Bond
Energy
Bond
Energy
H
O
H H
Type
(kcal/mol)
Type
(kcal/mol)
H
O
H
O—O
34
O==O
96
S—S
51
C—H
99
C—N
70
C==S
108
Figure 2-2. Left: Association of two dipolar water
S—H
81
O—H
110
molecules by a hydrogen bond (dotted line). Right:
C—C
82
C==C
147
Hydrogen-bonded cluster of four water molecules.
C—O
84
C==N
147
Note that water can serve simultaneously both as a hy-
N—H
94
C==O
164
drogen donor and as a hydrogen acceptor.
WATER & pH
/
7
While the hydrogens of nonpolar groups such as the
the backbone to water while burying the relatively hy-
methylene groups of hydrocarbons do not form hydro-
drophobic nucleotide bases inside. The extended back-
gen bonds, they do affect the structure of the water that
bone maximizes the distance between negatively
surrounds them. Water molecules adjacent to a hy-
charged backbone phosphates, minimizing unfavorable
drophobic group are restricted in the number of orien-
electrostatic interactions.
tations (degrees of freedom) that permit them to par-
ticipate in the maximum number of energetically
WATER IS AN EXCELLENT NUCLEOPHILE
favorable hydrogen bonds. Maximal formation of mul-
Metabolic reactions often involve the attack by lone
tiple hydrogen bonds can be maintained only by in-
pairs of electrons on electron-rich molecules termed
creasing the order of the adjacent water molecules, with
nucleophiles on electron-poor atoms called elec-
a corresponding decrease in entropy.
trophiles. Nucleophiles and electrophiles do not neces-
It follows from the second law of thermodynamics
sarily possess a formal negative or positive charge.
that the optimal free energy of a hydrocarbon-water
Water, whose two lone pairs of sp3 electrons bear a par-
mixture is a function of both maximal enthalpy (from
tial negative charge, is an excellent nucleophile. Other
hydrogen bonding) and minimum entropy (maximum
nucleophiles of biologic importance include the oxygen
degrees of freedom). Thus, nonpolar molecules tend to
atoms of phosphates, alcohols, and carboxylic acids; the
form droplets with minimal exposed surface area, re-
sulfur of thiols; the nitrogen of amines; and the imid-
ducing the number of water molecules affected. For the
azole ring of histidine. Common electrophiles include
same reason, in the aqueous environment of the living
the carbonyl carbons in amides, esters, aldehydes, and
cell the hydrophobic portions of biopolymers tend to
ketones and the phosphorus atoms of phosphoesters.
be buried inside the structure of the molecule, or within
Nucleophilic attack by water generally results in the
a lipid bilayer, minimizing contact with water.
cleavage of the amide, glycoside, or ester bonds that
hold biopolymers together. This process is termed hy-
Electrostatic Interactions
drolysis. Conversely, when monomer units are joined
together to form biopolymers such as proteins or glyco-
Interactions between charged groups shape biomolecu-
gen, water is a product, as shown below for the forma-
lar structure. Electrostatic interactions between oppo-
tion of a peptide bond between two amino acids.
sitely charged groups within or between biomolecules
are termed salt bridges. Salt bridges are comparable in
strength to hydrogen bonds but act over larger dis-
O
tances. They thus often facilitate the binding of charged
+H3N
OH + H
NH
molecules and ions to proteins and nucleic acids.
O-
Alanine
Van der Waals Forces
O
Van der Waals forces arise from attractions between
Valine
transient dipoles generated by the rapid movement of
electrons on all neutral atoms. Significantly weaker
than hydrogen bonds but potentially extremely numer-
H2O
ous, van der Waals forces decrease as the sixth power of
the distance separating atoms. Thus, they act over very
O
short distances, typically 2-4 Å.
+H3N
NH
O-
Multiple Forces Stabilize Biomolecules
O
The DNA double helix illustrates the contribution of
multiple forces to the structure of biomolecules. While
each individual DNA strand is held together by cova-
While hydrolysis is a thermodynamically favored re-
lent bonds, the two strands of the helix are held to-
action, the amide and phosphoester bonds of polypep-
gether exclusively by noncovalent interactions. These
tides and oligonucleotides are stable in the aqueous en-
noncovalent interactions include hydrogen bonds be-
vironment of the cell. This seemingly paradoxic
tween nucleotide bases
(Watson-Crick base pairing)
behavior reflects the fact that the thermodynamics gov-
and van der Waals interactions between the stacked
erning the equilibrium of a reaction do not determine
purine and pyrimidine bases. The helix presents the
the rate at which it will take place. In the cell, protein
charged phosphate groups and polar ribose sugars of
catalysts called enzymes are used to accelerate the rate
8
/
CHAPTER 2
of hydrolytic reactions when needed. Proteases catalyze
H7O3+. The proton is nevertheless routinely repre-
the hydrolysis of proteins into their component amino
sented as H+, even though it is in fact highly hydrated.
acids, while nucleases catalyze the hydrolysis of the
Since hydronium and hydroxide ions continuously
phosphoester bonds in DNA and RNA. Careful control
recombine to form water molecules, an individual hy-
of the activities of these enzymes is required to ensure
drogen or oxygen cannot be stated to be present as an
that they act only on appropriate target molecules.
ion or as part of a water molecule. At one instant it is
an ion. An instant later it is part of a molecule. Individ-
Many Metabolic Reactions Involve
ual ions or molecules are therefore not considered. We
Group Transfer
refer instead to the probability that at any instant in
time a hydrogen will be present as an ion or as part of a
In group transfer reactions, a group G is transferred
water molecule. Since 1 g of water contains 3.46 × 1022
from a donor D to an acceptor A, forming an acceptor
molecules, the ionization of water can be described sta-
group complex A-G:
tistically. To state that the probability that a hydrogen
exists as an ion is 0.01 means that a hydrogen atom has
D−G + A = A−G + D
one chance in 100 of being an ion and 99 chances out
of 100 of being part of a water molecule. The actual
The hydrolysis and phosphorolysis of glycogen repre-
probability of a hydrogen atom in pure water existing as
sent group transfer reactions in which glucosyl groups
a hydrogen ion is approximately 1.8 × 10−9. The proba-
are transferred to water or to orthophosphate. The
bility of its being part of a molecule thus is almost
equilibrium constant for the hydrolysis of covalent
unity. Stated another way, for every hydrogen ion and
bonds strongly favors the formation of split products.
hydroxyl ion in pure water there are 1.8 billion or 1.8 ×
The biosynthesis of macromolecules also involves group
109 water molecules. Hydrogen ions and hydroxyl ions
transfer reactions in which the thermodynamically un-
nevertheless contribute significantly to the properties of
favored synthesis of covalent bonds is coupled to fa-
water.
vored reactions so that the overall change in free energy
For dissociation of water,
favors biopolymer synthesis. Given the nucleophilic
character of water and its high concentration in cells,
+
why are biopolymers such as proteins and DNA rela-
[H
][OH− ]
K=
tively stable? And how can synthesis of biopolymers
[H O
]
2
occur in an apparently aqueous environment? Central
to both questions are the properties of enzymes. In the
where brackets represent molar concentrations (strictly
absence of enzymic catalysis, even thermodynamically
speaking, molar activities) and K is the dissociation
highly favored reactions do not necessarily take place
constant. Since one mole (mol) of water weighs 18 g,
rapidly. Precise and differential control of enzyme ac-
one liter (L) (1000 g) of water contains 1000 × 18 =
tivity and the sequestration of enzymes in specific or-
55.56 mol. Pure water thus is 55.56 molar. Since the
ganelles determine under what physiologic conditions a
probability that a hydrogen in pure water will exist as a
given biopolymer will be synthesized or degraded.
hydrogen ion is 1.8 × 10−9, the molar concentration of
Newly synthesized polymers are not immediately hy-
H+ ions (or of OH− ions) in pure water is the product
drolyzed, in part because the active sites of biosynthetic
of the probability, 1.8 × 10−9, times the molar concen-
enzymes sequester substrates in an environment from
tration of water, 55.56 mol/L. The result is 1.0 × 10−7
which water can be excluded.
mol/L.
We can now calculate K for water:
Water Molecules Exhibit a Slight but
Important Tendency to Dissociate
+
−
−7
−7
[H
][OH
]
[10
][10
]
K =
=
The ability of water to ionize, while slight, is of central
[H O]
[5556]
2
importance for life. Since water can act both as an acid
−14
−16
=
0 018×10
=18×10
mol
/
L
and as a base, its ionization may be represented as an
intermolecular proton transfer that forms a hydronium
The molar concentration of water, 55.56 mol/L, is
ion (H3O+) and a hydroxide ion (OH−):
too great to be significantly affected by dissociation. It
+
−
H O
+
H O
=
H O
+
OH
therefore is considered to be essentially constant. This
2
2
3
constant may then be incorporated into the dissociation
The transferred proton is actually associated with a
constant K to provide a useful new constant Kw termed
cluster of water molecules. Protons exist in solution not
the ion product for water. The relationship between
only as H3O+, but also as multimers such as H5O2+ and
Kw and K is shown below:
WATER & pH
/
9
+
−
termediates, whose phosphoryl group contains two dis-
[H
][OH
]
−16
K
=
=18×10
mol/L
sociable protons, the first of which is strongly acidic.
[
H O2
]
The following examples illustrate how to calculate
+
−
the pH of acidic and basic solutions.
K
w
=
(K )[
H O2
]
=
[H
][OH
]
Example 1: What is the pH of a solution whose hy-
−16
=
(18 ×10
mol/L)(5556
mol/L)
drogen ion concentration is 3.2 × 10−4 mol/L?
−14
2
=1 00×10
(mol
/
L
)
+
pH =
−log [H
]
−4
Note that the dimensions of K are moles per liter and
=
−log (3.2×10
)
those of Kw are moles2 per liter2. As its name suggests,
−4
=
−log (3.2)−log (10
)
the ion product Kw is numerically equal to the product
of the molar concentrations of H+ and OH−:
=
−0.5+40
=35
−
Kw
=[H+
[
]
Example 2: What is the pH of a solution whose hy-
At 25 °C, Kw = (10−7)2, or 10−14 (mol/L)2. At tempera-
droxide ion concentration is 4.0 × 10−4 mol/L? We first
tures below 25 °C, Kw is somewhat less than 10−14; and
define a quantity pOH that is equal to −log [OH−] and
at temperatures above 25 °C it is somewhat greater than
that may be derived from the definition of Kw:
10−14. Within the stated limitations of the effect of tem-
perature, Kw equals 10-14 (mol/L)2 for all aqueous so-
+
−
−14
Kw
=[H
][OH
]
=10
lutions, even solutions of acids or bases. We shall use
Kw to calculate the pH of acidic and basic solutions.
Therefore:
+
−
pH IS THE NEGATIVE LOG OF THE
log [H
]+log [OH
]= log
10−14
HYDROGEN ION CONCENTRATION
or
The term pH was introduced in 1909 by Sörensen,
who defined pH as the negative log of the hydrogen ion
pH + pOH = 14
concentration:
To solve the problem by this approach:
pH =
− log [H+
]
−
−4
[OH
] =
40×10
This definition, while not rigorous, suffices for many
biochemical purposes. To calculate the pH of a solution:
pOH=
−log [OH−]
−4
1. Calculate hydrogen ion concentration [H+].
=
−log (40×10
)
2. Calculate the base 10 logarithm of [H+].
−4
=
−log (40)−log
(10
)
3. pH is the negative of the value found in step 2.
=
−0.60+4.0
For example, for pure water at 25°C,
=34
+
−7
pH =
−log [H
]
=
−log 10
=
−(−7) = 7.0
Now:
Low pH values correspond to high concentrations of
pH =14 −pOH = 14
− .
H+ and high pH values correspond to low concentra-
= 10.6
tions of H+.
−2
Acids are proton donors and bases are proton ac-
Example 3: What are the pH values of (a) 2.0 × 10
ceptors. Strong acids (eg, HCl or H2SO4) completely
mol/L KOH and of (b) 2.0 × 10−6 mol/L KOH? The
dissociate into anions and cations even in strongly acidic
OH− arises from two sources, KOH and water. Since
solutions (low pH). Weak acids dissociate only partially
pH is determined by the total [H+] (and pOH by the
in acidic solutions. Similarly, strong bases (eg, KOH or
total [OH−]), both sources must be considered. In the
NaOH)—but not weak bases (eg, Ca[OH]2)—are
first case
(a), the contribution of water to the total
completely dissociated at high pH. Many biochemicals
[OH−] is negligible. The same cannot be said for the
are weak acids. Exceptions include phosphorylated in-
second case (b):
10
/
CHAPTER 2
below are the expressions for the dissociation constant
Concentration (mol/L)
(Ka ) for two representative weak acids, RCOOH and
(a)
(b)
RNH3+.
Molarity of KOH
2.0 × 10−2
2.0 × 10−6
+
[OH−] from KOH
2.0 × 10−2
2.0 × 10−6
R—COOH =R—COO−+H
[OH−] from water
1.0 × 10−7
1.0 × 10−7
−
+
[R—COO
][H
]
Total [OH−]
2.00001 × 10−2
2.1 × 10−6
K
a
=
[R—COOH]
+
+
3
R—NH =R—NH +H
2
Once a decision has been reached about the significance
+
[R—NH ][H
2
]
of the contribution by water, pH may be calculated as
K
=
a
above.
+
[R
—
NH
3
]
The above examples assume that the strong base
KOH is completely dissociated in solution and that the
Since the numeric values of Ka for weak acids are nega-
concentration of OH− ions was thus equal to that of the
tive exponential numbers, we express Ka as pKa, where
KOH. This assumption is valid for dilute solutions of
strong bases or acids but not for weak bases or acids.
p
a
K
= − log K
Since weak electrolytes dissociate only slightly in solu-
tion, we must use the dissociation constant to calcu-
Note that pKa is related to Ka as pH is to [H+]. The
late the concentration of [H+] (or [OH−]) produced by
stronger the acid, the lower its pKa value.
a given molarity of a weak acid (or base) before calcu-
pKa is used to express the relative strengths of both
lating total [H+] (or total [OH−]) and subsequently pH.
acids and bases. For any weak acid, its conjugate is a
strong base. Similarly, the conjugate of a strong base is
a weak acid. The relative strengths of bases are ex-
Functional Groups That Are Weak Acids
pressed in terms of the pKa of their conjugate acids. For
Have Great Physiologic Significance
polyproteic compounds containing more than one dis-
Many biochemicals possess functional groups that are
sociable proton, a numerical subscript is assigned to
weak acids or bases. Carboxyl groups, amino groups,
each in order of relative acidity. For a dissociation of
and the second phosphate dissociation of phosphate es-
the type
ters are present in proteins and nucleic acids, most
coenzymes, and most intermediary metabolites. Knowl-
R—NH3+
→R—NH2
edge of the dissociation of weak acids and bases thus is
basic to understanding the influence of intracellular pH
the pKa is the pH at which the concentration of the
on structure and biologic activity. Charge-based separa-
acid RNH3+ equals that of the base RNH2.
tions such as electrophoresis and ion exchange chro-
From the above equations that relate Ka to [H+] and
matography also are best understood in terms of the
to the concentrations of undissociated acid and its con-
dissociation behavior of functional groups.
jugate base, when
We term the protonated species
(eg, HA or
RNH3+) the acid and the unprotonated species (eg,
−
[R
—
COO
] = [R—COOH]
A− or RNH2) its conjugate base. Similarly, we may
refer to a base (eg, A− or RNH2) and its conjugate
or when
acid (eg, HA or RNH3+). Representative weak acids
(left), their conjugate bases (center), and the pKa values
+
(right) include the following:
[R
—
NH
] = [R
—
NH
]
2
3
−
2
R—CH —COOH
R—CH
2
—COO
pK
a
=
4−5
then
+
R—CH
—NH
R—CH
—NH
pK
=9−10
2
3
2
2
a
+
Ka
=
[H
]
−
2
H CO
3
HCO
3
pK
a
=
64
−
−2
Thus, when the associated (protonated) and dissociated
2
H PO
4
HPO
4
pK
a
=72
(conjugate base) species are present at equal concentra-
tions, the prevailing hydrogen ion concentration [H+]
We express the relative strengths of weak acids and
is numerically equal to the dissociation constant, Ka. If
bases in terms of their dissociation constants. Shown
the logarithms of both sides of the above equation are
WATER & pH
/
11
taken and both sides are multiplied by −1, the expres-
Substitute pH and pKa for −log [H+] and −log Ka, re-
sions would be as follows:
spectively; then:
Ka =[H+]
[HA]
pH=pK
−log
+
a
−
−log
Ka
=
−log [H
]
[A
]
Since −log Ka is defined as pKa, and −log [H+] de-
Inversion of the last term removes the minus sign
fines pH, the equation may be rewritten as
and gives the Henderson-Hasselbalch equation:
pK
=
pH
−
a
[A
]
pH=pK
a
+ log
[HA]
ie, the pKa of an acid group is the pH at which the pro-
tonated and unprotonated species are present at equal
The Henderson-Hasselbalch equation has great pre-
concentrations. The pKa for an acid may be determined
dictive value in protonic equilibria. For example,
by adding 0.5 equivalent of alkali per equivalent of
acid. The resulting pH will be the pKa of the acid.
(1) When an acid is exactly half-neutralized, [A−] =
[HA]. Under these conditions,
The Henderson-Hasselbalch Equation
−
[A
]
1
Describes the Behavior
pH=pK
a
+ log
=
pK
a
+log
=
pK
a
+
0
[HA]
1
of Weak Acids & Buffers
The Henderson-Hasselbalch equation is derived below.
Therefore, at half-neutralization, pH = pKa.
A weak acid, HA, ionizes as follows:
(2) When the ratio [A−]/[HA] = 100:1,
HA = H+ +A−
−
[A
]
pH=pK
a
+
log
The equilibrium constant for this dissociation is
[HA]
pH
=
pK
a
+
log 100 / 1=pK
a
+
2
−
[H+][A
]
K
a
=
[HA]
(3) When the ratio [A−]/[HA] = 1:10,
Cross-multiplication gives
pH=pK
a
+ log 1/ 10 =pK
a
+(−1)
+
−
[H
][A
] =
K
a
[HA
]
If the equation is evaluated at ratios of [A−]/[HA]
ranging from 103 to 10−3 and the calculated pH values
Divide both sides by [A−]:
are plotted, the resulting graph describes the titration
curve for a weak acid (Figure 2-4).
+
[HA]
[H
]
=K
a
−
[A
]
Solutions of Weak Acids & Their Salts
Take the log of both sides:
Buffer Changes in pH
Solutions of weak acids or bases and their conjugates
[HA]
+
log [H
]= log
K
exhibit buffering, the ability to resist a change in pH
−
a [A
]
following addition of strong acid or base. Since many
[HA]
metabolic reactions are accompanied by the release or
=
log
K
a
+log
−
uptake of protons, most intracellular reactions are
[A
]
buffered. Oxidative metabolism produces CO2, the an-
hydride of carbonic acid, which if not buffered would
Multiply through by −1:
produce severe acidosis. Maintenance of a constant pH
involves buffering by phosphate, bicarbonate, and pro-
+
[HA]
−log [H
] =
−log
K
−
log
teins, which accept or release protons to resist a change
a
−
[A
]
12
/
CHAPTER 2
1.0
1.0
to the pKa. A solution of a weak acid and its conjugate
base buffers most effectively in the pH range pKa ± 1.0
0.8
0.8
pH unit.
Figure 2-4 also illustrates the net charge on one
0.6
0.6
molecule of the acid as a function of pH. A fractional
charge of −0.5 does not mean that an individual mole-
0.4
0.4
cule bears a fractional charge, but the probability that a
given molecule has a unit negative charge is 0.5. Con-
0.2
0.2
sideration of the net charge on macromolecules as a
function of pH provides the basis for separatory tech-
0
0
niques such as ion exchange chromatography and elec-
2
3
4
5
6
7
8
trophoresis.
pH
Figure 2-4. Titration curve for an acid of the type
Acid Strength Depends on
HA. The heavy dot in the center of the curve indicates
Molecular Structure
the pKa 5.0.
Many acids of biologic interest possess more than one
dissociating group. The presence of adjacent negative
charge hinders the release of a proton from a nearby
in pH. For experiments using tissue extracts or en-
group, raising its pKa. This is apparent from the pKa
zymes, constant pH is maintained by the addition of
values for the three dissociating groups of phosphoric
buffers such as MES ([2-N-morpholino]ethanesulfonic
acid and citric acid (Table 2-2). The effect of adjacent
acid, pKa 6.1), inorganic orthophosphate (pKa2 7.2),
charge decreases with distance. The second pKa for suc-
HEPES (N-hydroxyethylpiperazine-N9-2-ethanesulfonic
cinic acid, which has two methylene groups between its
acid, pKa
6.8), or Tris
(tris[hydroxymethyl] amino-
carboxyl groups, is 5.6, whereas the second pKa for glu-
methane, pKa 8.3). The value of pKa relative to the de-
sired pH is the major determinant of which buffer is se-
lected.
Buffering can be observed by using a pH meter
Table 2-2. Relative strengths of selected acids of
while titrating a weak acid or base (Figure 2-4). We
can also calculate the pH shift that accompanies addi-
biologic significance. Tabulated values are the pKa
tion of acid or base to a buffered solution. In the exam-
values (−log of the dissociation constant) of
ple, the buffered solution (a weak acid, pKa
= 5.0, and
selected monoprotic, diprotic, and triprotic acids.
its conjugate base) is initially at one of four pH values.
We will calculate the pH shift that results when 0.1
Monoprotic Acids
meq of KOH is added to 1 meq of each solution:
Formic
pK
3.75
Lactic
pK
3.86
Acetic
pK
4.76
Initial pH
5.00
5.37
5.60
5.86
Ammonium ion
pK
9.25
[A−]initial
0.50
0.70
0.80
0.88
[HA]initial
0.50
0.30
0.20
0.12
Diprotic Acids
([A−]/[HA])initial
1.00
2.33
4.00
7.33
Carbonic
pK1
6.37
Addition of 0.1 meq of KOH produces
pK2
10.25
[A−]final
0.60
0.80
0.90
0.98
Succinic
pK1
4.21
[HA]final
0.40
0.20
0.10
0.02
pK2
5.64
([A−]/[HA])final
1.50
4.00
9.00
49.0
Glutaric
pK1
4.34
log ([A−]/[HA])final
0.176
0.602
0.95
1.69
pK2
5.41
Final pH
5.18
5.60
5.95
6.69
Triprotic Acids
∆pH
0.18
0.60
0.95
1.69
Phosphoric
pK1
2.15
pK2
6.82
pK3
12.38
Citric
pK1
3.08
Notice that the change in pH per milliequivalent of
pK2
4.74
OH− added depends on the initial pH. The solution re-
pK3
5.40
sists changes in pH most effectively at pH values close
WATER & pH
/
13
taric acid, which has one additional methylene group,
• Macromolecules exchange internal surface hydrogen
is 5.4.
bonds for hydrogen bonds to water. Entropic forces
dictate that macromolecules expose polar regions to
pKa Values Depend on the Properties
an aqueous interface and bury nonpolar regions.
of the Medium
• Salt bonds, hydrophobic interactions, and van der
Waals forces participate in maintaining molecular
The pKa of a functional group is also profoundly influ-
structure.
enced by the surrounding medium. The medium may
• pH is the negative log of [H+]. A low pH character-
either raise or lower the pKa depending on whether the
izes an acidic solution, and a high pH denotes a basic
undissociated acid or its conjugate base is the charged
solution.
species. The effect of dielectric constant on pKa may be
observed by adding ethanol to water. The pKa of a car-
• The strength of weak acids is expressed by pKa, the
negative log of the acid dissociation constant. Strong
boxylic acid increases, whereas that of an amine decreases
because ethanol decreases the ability of water to solvate
acids have low pKa values and weak acids have high
pKa values.
a charged species. The pKa values of dissociating groups
in the interiors of proteins thus are profoundly affected
• Buffers resist a change in pH when protons are pro-
by their local environment, including the presence or
duced or consumed. Maximum buffering capacity
absence of water.
occurs ± 1 pH unit on either side of pKa. Physiologic
buffers include bicarbonate, orthophosphate, and
proteins.
SUMMARY
REFERENCES
• Water forms hydrogen-bonded clusters with itself and
with other proton donors or acceptors. Hydrogen
Segel IM: Biochemical Calculations. Wiley, 1968.
bonds account for the surface tension, viscosity, liquid
Wiggins PM: Role of water in some biological processes. Microbiol
state at room temperature, and solvent power of water.
Rev 1990;54:432.
• Compounds that contain O, N, or S can serve as hy-
drogen bond donors or acceptors.
SECTION I
Structures & Functions
of Proteins & Enzymes
Amino Acids & Peptides
3
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
more than 20 amino acids, its redundancy limits the
available codons to the 20 L-α-amino acids listed in
In addition to providing the monomer units from which
Table 3-1, classified according to the polarity of their R
the long polypeptide chains of proteins are synthesized,
groups. Both one- and three-letter abbreviations for each
the L-α-amino acids and their derivatives participate in
amino acid can be used to represent the amino acids in
cellular functions as diverse as nerve transmission and
peptides (Table 3-1). Some proteins contain additional
the biosynthesis of porphyrins, purines, pyrimidines,
amino acids that arise by modification of an amino acid
and urea. Short polymers of amino acids called peptides
already present in a peptide. Examples include conver-
perform prominent roles in the neuroendocrine system
sion of peptidyl proline and lysine to 4-hydroxyproline
as hormones, hormone-releasing factors, neuromodula-
and 5-hydroxylysine; the conversion of peptidyl gluta-
tors, or neurotransmitters. While proteins contain only
mate to γ-carboxyglutamate; and the methylation,
L-α-amino acids, microorganisms elaborate peptides
formylation, acetylation, prenylation, and phosphoryla-
that contain both D- and L-α-amino acids. Several of
tion of certain aminoacyl residues. These modifications
these peptides are of therapeutic value, including the an-
extend the biologic diversity of proteins by altering their
tibiotics bacitracin and gramicidin A and the antitumor
solubility, stability, and interaction with other proteins.
agent bleomycin. Certain other microbial peptides are
toxic. The cyanobacterial peptides microcystin and
nodularin are lethal in large doses, while small quantities
Only L-
-Amino Acids Occur in Proteins
promote the formation of hepatic tumors. Neither hu-
With the sole exception of glycine, the α-carbon of
mans nor any other higher animals can synthesize 10 of
amino acids is chiral. Although some protein amino
the 20 common L-α-amino acids in amounts adequate
acids are dextrorotatory and some levorotatory, all share
to support infant growth or to maintain health in adults.
the absolute configuration of L-glyceraldehyde and thus
Consequently, the human diet must contain adequate
are L-α-amino acids. Several free L-α-amino acids fulfill
quantities of these nutritionally essential amino acids.
important roles in metabolic processes. Examples in-
clude ornithine, citrulline, and argininosuccinate that
PROPERTIES OF AMINO ACIDS
participate in urea synthesis; tyrosine in formation of
thyroid hormones; and glutamate in neurotransmitter
The Genetic Code Specifies
biosynthesis. D-Amino acids that occur naturally in-
20 L-
-Amino Acids
clude free D-serine and D-aspartate in brain tissue,
Of the over 300 naturally occurring amino acids, 20 con-
D-alanine and D-glutamate in the cell walls of gram-
stitute the monomer units of proteins. While a nonre-
positive bacteria, and D-amino acids in some nonmam-
dundant three-letter genetic code could accommodate
malian peptides and certain antibiotics.
14
Table 3-1. L- α-Amino acids present in proteins.
Name
Symbol
Structural Formula
pK1
pK2
pK3
With Aliphatic Side Chains
-COOH
-NH3+
R Group
Glycine
Gly [G]
2.4
9.8
H CH COO-
+
NH
3
Alanine
Ala [A]
2.4
9.9
CH3
CH COO-
+
NH
3
H3C
CH
CH COO-
Valine
Val [V]
2.2
9.7
+
H3C
NH3
H3C
CH
CH2
CH
COO-
Leucine
Leu [L]
2.3
9.7
H3C
NH3
+
CH3
CH2
Isoleucine
Ile [I]
CH CH COO-
2.3
9.8
CH3
NH
+
3
With Side Chains Containing Hydroxylic (OH) Groups
Serine
Ser [S]
2.2
9.2
about 13
CH
2
CH
COO-
+
OH
NH3
Threonine
Thr [T]
2.1
9.1
about 13
CH
3
CH
CH
COO-
+
OH
NH3
Tyrosine
Tyr [Y]
See below.
With Side Chains Containing Sulfur Atoms
Cysteine
Cys [C]
CH2
CH
COO-
1.9
10.8
8.3
+
SH
NH3
Methionine
Met [M]
CH2
CH2
CH
COO-
2.1
9.3
+
S CH
3
NH3
With Side Chains Containing Acidic Groups or Their Amides
Aspartic acid
Asp [D]
-
2.0
9.9
3.9
-OOC
CH2
CH COO
+
NH
3
Asparagine
Asn [N]
2.1
8.8
H2N
C
CH2
CH
COO-
+
O
NH3
–OOC
CH2
CH2
CH COO-
Glutamic acid
Glu [E]
2.1
9.5
4.1
+
NH
3
H2N
C
CH2
CH2
CH
COO-
Glutamine
Gln [Q]
2.2
9.1
+
O
NH3
(continued)
15
16
/
CHAPTER 3
Table 3-1. L-α-Amino acids present in proteins. (continued)
Name
Symbol
Structural Formula
pK1
pK2
pK3
With Side Chains Containing Basic Groups
-COOH
-NH3+
R Group
Arginine
Arg [R]
1.8
9.0
12.5
H
N
CH
2
CH2
CH
2
CH COO-
+
+
C
NH2
NH3
NH2
-
CH2
CH2
CH2
CH2
CH COO
Lysine
Lys [K]
2.2
9.2
10.8
+
+
NH3
NH
3
CH2
CH COO-
Histidine
His [H]
1.8
9.3
6.0
HN
N
+
NH
3
Containing Aromatic Rings
Histidine
His [H]
See above.
CH2
CH COO-
Phenylalanine
Phe [F]
2.2
9.2
+
NH
3
Tyrosine
Tyr [Y]
2.2
9.1
10.1
HO
CH2
CH
COO-
+
NH
3
Tryptophan
Trp [W]
2.4
9.4
CH2
CH
COO-
+
NH
3
N
H
Imino Acid
Proline
Pro [P]
2.0
10.6
+
N
COO-
H2
Amino Acids May Have Positive, Negative,
Molecules that contain an equal number of ioniz-
or Zero Net Charge
able groups of opposite charge and that therefore bear
no net charge are termed zwitterions. Amino acids in
Charged and uncharged forms of the ionizable
blood and most tissues thus should be represented as in
COOH and NH3+ weak acid groups exist in solu-
A, below.
tion in protonic equilibrium:
+
NH3
NH2
+
R—COOH =R—COO−+H
-
O
OH
+
+
R—NH
3
=
R—
NH
2
+
H
R
R
O
O
While both RCOOH and RNH3+ are weak acids,
A
B
RCOOH is a far stronger acid than RNH3+. At
physiologic pH (pH 7.4), carboxyl groups exist almost
Structure B cannot exist in aqueous solution because at
entirely as RCOO− and amino groups predomi-
any pH low enough to protonate the carboxyl group
nantly as RNH3+. Figure 3-1 illustrates the effect of
the amino group would also be protonated. Similarly,
pH on the charged state of aspartic acid.
at any pH sufficiently high for an uncharged amino
AMINO ACIDS & PEPTIDES
/
17
O
O
O
O
H+
H+
H+
OH
OH
O-
O-
pK1 = 2.09
pK2 = 3.86
pK3 = 9.82
NH3+
(α-COOH)
NH3+
(β-COOH)
NH3+
(— NH3+)
NH2
HO
-O
-O
-O
A
B
C
D
In strong acid
Around pH 3;
Around pH 6-8;
In strong alkali
(below pH 1);
net charge = 0
net charge = -1
(above pH 11);
net charge = +1
net charge = -2
Figure 3-1. Protonic equilibria of aspartic acid.
group to predominate, a carboxyl group will be present
At Its Isoelectric pH (pI), an Amino Acid
as RCOO−. The uncharged representation B (above)
Bears No Net Charge
is, however, often used for reactions that do not involve
The isoelectric species is the form of a molecule that
protonic equilibria.
has an equal number of positive and negative charges
and thus is electrically neutral. The isoelectric pH, also
pKa Values Express the Strengths
called the pI, is the pH midway between pKa values on
of Weak Acids
either side of the isoelectric species. For an amino acid
such as alanine that has only two dissociating groups,
The acid strengths of weak acids are expressed as their
there is no ambiguity. The first pKa (R COOH) is
pKa (Table 3-1). The imidazole group of histidine and
2.35 and the second pKa (RNH3+) is 9.69. The iso-
the guanidino group of arginine exist as resonance hy-
electric pH (pI) of alanine thus is
brids with positive charge distributed between both ni-
trogens (histidine) or all three nitrogens (arginine) (Fig-
ure
3-2). The net charge on an amino acid—the
1
pK +pK
2
2.35+9.69
pl=
=
=
6.02
algebraic sum of all the positively and negatively
2
2
charged groups present—depends upon the pKa values
of its functional groups and on the pH of the surround-
For polyfunctional acids, pI is also the pH midway be-
ing medium. Altering the charge on amino acids and
tween the pKa values on either side of the isoionic
their derivatives by varying the pH facilitates the physi-
species. For example, the pI for aspartic acid is
cal separation of amino acids, peptides, and proteins
(see Chapter 4).
1
pK +pK
2
2.09+
3.96
pl=
=
=
3.02
2
2
For lysine, pI is calculated from:
R
R
pK +pK
2
3
N H
N H
pl=
2
N
N
H
H
Similar considerations apply to all polyprotic acids (eg,
proteins), regardless of the number of dissociating
groups present. In the clinical laboratory, knowledge of
R
R
R
the pI guides selection of conditions for electrophoretic
NH
NH
NH
separations. For example, electrophoresis at pH 7.0 will
separate two molecules with pI values of 6.0 and 8.0
C NH2
C NH2
C NH2
because at pH 8.0 the molecule with a pI of 6.0 will
NH2
NH2
NH2
have a net positive charge, and that with pI of 8.0 a net
negative charge. Similar considerations apply to under-
Figure 3-2. Resonance hybrids of the protonated
standing chromatographic separations on ionic sup-
forms of the R groups of histidine and arginine.
ports such as DEAE cellulose (see Chapter 4).
18
/
CHAPTER 3
pKa Values Vary With the Environment
THE
-R GROUPS DETERMINE THE
The environment of a dissociable group affects its pKa.
PROPERTIES OF AMINO ACIDS
The pKa values of the R groups of free amino acids in
Since glycine, the smallest amino acid, can be accommo-
aqueous solution (Table 3-1) thus provide only an ap-
dated in places inaccessible to other amino acids, it often
proximate guide to the pKa values of the same amino
occurs where peptides bend sharply. The hydrophobic R
acids when present in proteins. A polar environment
groups of alanine, valine, leucine, and isoleucine and the
favors the charged form (R COO− or RNH3+),
aromatic R groups of phenylalanine, tyrosine, and tryp-
and a nonpolar environment favors the uncharged form
tophan typically occur primarily in the interior of cy-
(R COOH or RNH2). A nonpolar environment
tosolic proteins. The charged R groups of basic and
thus raises the pKa of a carboxyl group (making it a
acidic amino acids stabilize specific protein conforma-
weaker acid) but lowers that of an amino group (making
tions via ionic interactions, or salt bonds. These bonds
it a stronger acid). The presence of adjacent charged
also function in “charge relay” systems during enzymatic
groups can reinforce or counteract solvent effects. The
catalysis and electron transport in respiring mitochon-
pKa of a functional group thus will depend upon its lo-
dria. Histidine plays unique roles in enzymatic catalysis.
cation within a given protein. Variations in pKa can en-
The pKa of its imidazole proton permits it to function at
compass whole pH units (Table 3-2). pKa values that
neutral pH as either a base or an acid catalyst. The pri-
diverge from those listed by as much as three pH units
mary alcohol group of serine and the primary thioalco-
are common at the active sites of enzymes. An extreme
hol (SH) group of cysteine are excellent nucleophiles
example, a buried aspartic acid of thioredoxin, has a
and can function as such during enzymatic catalysis.
pKa above 9—a shift of over six pH units!
However, the secondary alcohol group of threonine,
while a good nucleophile, does not fulfill an analogous
The Solubility and Melting Points
role in catalysis. The OH groups of serine, tyrosine,
of Amino Acids Reflect
and threonine also participate in regulation of the activ-
Their Ionic Character
ity of enzymes whose catalytic activity depends on the
phosphorylation state of these residues.
The charged functional groups of amino acids ensure
that they are readily solvated by—and thus soluble in—
polar solvents such as water and ethanol but insoluble
FUNCTIONAL GROUPS DICTATE THE
in nonpolar solvents such as benzene, hexane, or ether.
CHEMICAL REACTIONS OF AMINO ACIDS
Similarly, the high amount of energy required to dis-
Each functional group of an amino acid exhibits all of
rupt the ionic forces that stabilize the crystal lattice
its characteristic chemical reactions. For carboxylic acid
account for the high melting points of amino acids
groups, these reactions include the formation of esters,
(> 200 °C).
amides, and acid anhydrides; for amino groups, acyla-
Amino acids do not absorb visible light and thus are
tion, amidation, and esterification; and for OH and
colorless. However, tyrosine, phenylalanine, and espe-
SH groups, oxidation and esterification. The most
cially tryptophan absorb high-wavelength
(250-290
important reaction of amino acids is the formation of a
nm) ultraviolet light. Tryptophan therefore makes the
peptide bond (shaded blue).
major contribution to the ability of most proteins to
absorb light in the region of 280 nm.
+H3N
O
H
N
O-
N
H
Table 3-2. Typical range of pKa values for
O
O
ionizable groups in proteins.
SH
Alanyl
Cysteinyl
Valine
Dissociating Group
pKa Range
α-Carboxyl
3.5-4.0
Amino Acid Sequence Determines
Non-α COOH of Asp or Glu
4.0-4.8
Primary Structure
Imidazole of His
6.5-7.4
SH of Cys
8.5-9.0
The number and order of all of the amino acid residues
OH of Tyr
9.5-10.5
in a polypeptide constitute its primary structure.
α-Amino
8.0-9.0
Amino acids present in peptides are called aminoacyl
ε-Amino of Lys
9.8-10.4
residues and are named by replacing the -ate or -ine suf-
Guanidinium of Arg
~12.0
fixes of free amino acids with -yl (eg, alanyl, aspartyl, ty-
AMINO ACIDS & PEPTIDES
/
19
rosyl). Peptides are then named as derivatives of the
SH
carboxyl terminal aminoacyl residue. For example, Lys-
O
CH2
H
Leu-Tyr-Gln is called lysyl-leucyl-tyrosyl-glutamine.
The -ine ending on glutamine indicates that its α-car-
C
CH
N
boxyl group is not involved in peptide bond formation.
CH2
N
C
CH2
CH2
H
O
COO-
Peptide Structures Are Easy to Draw
H
C
NH3+
Prefixes like tri- or octa- denote peptides with three or
eight residues, respectively, not those with three or
COO-
eight peptide bonds. By convention, peptides are writ-
ten with the residue that bears the free α-amino group
Figure 3-3. Glutathione (γ-glutamyl-cysteinyl-
at the left. To draw a peptide, use a zigzag to represent
glycine). Note the non-α peptide bond that links
the main chain or backbone. Add the main chain atoms,
Glu to Cys.
which occur in the repeating order: α-nitrogen, α-car-
bon, carbonyl carbon. Now add a hydrogen atom to
each α-carbon and to each peptide nitrogen, and an
releasing hormone (TRH) is cyclized to pyroglutamic
oxygen to the carbonyl carbon. Finally, add the appro-
acid, and the carboxyl group of the carboxyl terminal
priate R groups (shaded) to each α-carbon atom.
prolyl residue is amidated. Peptides elaborated by fungi,
bacteria, and lower animals can contain nonprotein
N
C
C
α
N
C
amino acids. The antibiotics tyrocidin and gramicidin S
Cα
N
C
Cα
are cyclic polypeptides that contain D-phenylalanine
and ornithine. The heptapeptide opioids dermorphin
O
H3C H
H
and deltophorin in the skin of South American tree
+H3N
C
C
N
COO-
frogs contain D-tyrosine and D-alanine.
C
N
C
C
H
H
H
CH2
CH2
Peptides Are Polyelectrolytes
O
OH
-OOC
The peptide bond is uncharged at any pH of physiologic
interest. Formation of peptides from amino acids is
Three-letter abbreviations linked by straight lines
therefore accompanied by a net loss of one positive and
represent an unambiguous primary structure. Lines are
one negative charge per peptide bond formed. Peptides
omitted for single-letter abbreviations.
nevertheless are charged at physiologic pH owing to their
Glu - Ala - Lys - Gly - Tyr - Ala
carboxyl and amino terminal groups and, where present,
their acidic or basic R groups. As for amino acids, the net
E A K G Y A
charge on a peptide depends on the pH of its environ-
Where there is uncertainty about the order of a portion
ment and on the pKa values of its dissociating groups.
of a polypeptide, the questionable residues are enclosed
in brackets and separated by commas.
The Peptide Bond Has Partial
Glu - Lys - (Ala, Gly, Tyr) - His - Ala
Double-Bond Character
Although peptides are written as if a single bond linked
the α-carboxyl and α-nitrogen atoms, this bond in fact
Some Peptides Contain Unusual
exhibits partial double-bond character:
Amino Acids
O
O-
In mammals, peptide hormones typically contain only
the α-amino acids of proteins linked by standard pep-
C
C
+
tide bonds. Other peptides may, however, contain non-
N
N
protein amino acids, derivatives of the protein amino
H
H
acids, or amino acids linked by an atypical peptide
bond. For example, the amino terminal glutamate of
There thus is no freedom of rotation about the bond
glutathione, which participates in protein folding and
that connects the carbonyl carbon and the nitrogen of a
in the metabolism of xenobiotics
(Chapter
53), is
peptide bond. Consequently, all four of the colored
linked to cysteine by a non-α peptide bond (Figure
atoms of Figure 3-4 are coplanar. The imposed semi-
3-3). The amino terminal glutamate of thyrotropin-
rigidity of the peptide bond has important conse-
20
/
CHAPTER 3
mixture of free amino acids is then treated with 6-amino-
O
R′
H
H
O
N-hydroxysuccinimidyl carbamate, which reacts with
their α-amino groups, forming fluorescent derivatives
that are then separated and identified using high-pressure
122°
121° C
C
N
C
liquid chromatography (see Chapter 5). Ninhydrin, also
120°
widely used for detecting amino acids, forms a purple
117°
110°
N
C
C
N
120°
120°
product with α-amino acids and a yellow adduct with
the imine groups of proline and hydroxyproline.
H
O
H
R′′
H
SUMMARY
0.36 nm
•
Both D-amino acids and non-α-amino acids occur
in nature, but only L-α-amino acids are present in
Figure 3-4. Dimensions of a fully extended poly-
proteins.
peptide chain. The four atoms of the peptide bond
•
All amino acids possess at least two weakly acidic
(colored blue) are coplanar. The unshaded atoms are
functional groups, RNH3+ and R COOH.
the α-carbon atom, the α-hydrogen atom, and the α-R
Many also possess additional weakly acidic functional
group of the particular amino acid. Free rotation can
groups such as OH, SH, guanidino, or imid-
occur about the bonds that connect the α-carbon with
azole groups.
the α-nitrogen and with the α-carbonyl carbon (blue
•
The pKa values of all functional groups of an amino
arrows). The extended polypeptide chain is thus a semi-
acid dictate its net charge at a given pH. pI is the pH
rigid structure with two-thirds of the atoms of the back-
at which an amino acid bears no net charge and thus
bone held in a fixed planar relationship one to another.
does not move in a direct current electrical field.
The distance between adjacent α-carbon atoms is 0.36
•
Of the biochemical reactions of amino acids, the
nm (3.6 Å). The interatomic distances and bond angles,
most important is the formation of peptide bonds.
which are not equivalent, are also shown. (Redrawn and
•
The R groups of amino acids determine their unique
reproduced, with permission, from Pauling L, Corey LP,
biochemical functions. Amino acids are classified as
Branson HR: The structure of proteins: Two hydrogen-
basic, acidic, aromatic, aliphatic, or sulfur-containing
bonded helical configurations of the polypeptide chain.
based on the properties of their R groups.
Proc Natl Acad Sci U S A 1951;37:205.)
•
Peptides are named for the number of amino acid
residues present, and as derivatives of the carboxyl
terminal residue. The primary structure of a peptide
quences for higher orders of protein structure. Encir-
is its amino acid sequence, starting from the amino-
cling arrows (Figure 3 - 4) indicate free rotation about
terminal residue.
the remaining bonds of the polypeptide backbone.
•
The partial double-bond character of the bond that
links the carbonyl carbon and the nitrogen of a pep-
Noncovalent Forces Constrain Peptide
tide renders four atoms of the peptide bond coplanar
Conformations
and restricts the number of possible peptide confor-
Folding of a peptide probably occurs coincident with
mations.
its biosynthesis (see Chapter 38). The physiologically
active conformation reflects the amino acid sequence,
REFERENCES
steric hindrance, and noncovalent interactions (eg, hy-
drogen bonding, hydrophobic interactions) between
Doolittle RF: Reconstructing history with amino acid sequences.
Protein Sci 1992;1:191.
residues. Common conformations include α-helices
and β-pleated sheets (see Chapter 5).
Kreil G: D-Amino acids in animal peptides. Annu Rev Biochem
1997;66:337.
Nokihara K, Gerhardt J: Development of an improved automated
ANALYSIS OF THE AMINO ACID
gas-chromatographic chiral analysis system: application to
CONTENT OF BIOLOGIC MATERIALS
non-natural amino acids and natural protein hydrolysates.
Chirality 2001;13:431.
In order to determine the identity and quantity of each
Sanger F: Sequences, sequences, and sequences. Annu Rev Biochem
amino acid in a sample of biologic material, it is first nec-
1988;57:1.
essary to hydrolyze the peptide bonds that link the amino
Wilson NA et al: Aspartic acid 26 in reduced Escherichia coli thiore-
acids together by treatment with hot HCl. The resulting
doxin has a pKa greater than 9. Biochemistry 1995;34:8931.
Proteins: Determination of
4
Primary Structure
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
Column Chromatography
Proteins perform multiple critically important roles. An
Column chromatography of proteins employs as the
internal protein network, the cytoskeleton
(Chapter
stationary phase a column containing small spherical
49), maintains cellular shape and physical integrity.
beads of modified cellulose, acrylamide, or silica whose
Actin and myosin filaments form the contractile ma-
surface typically has been coated with chemical func-
chinery of muscle (Chapter 49). Hemoglobin trans-
tional groups. These stationary phase matrices interact
ports oxygen (Chapter 6), while circulating antibodies
with proteins based on their charge, hydrophobicity,
search out foreign invaders (Chapter 50). Enzymes cat-
and ligand-binding properties. A protein mixture is ap-
alyze reactions that generate energy, synthesize and de-
plied to the column and the liquid mobile phase is per-
grade biomolecules, replicate and transcribe genes,
colated through it. Small portions of the mobile phase
process mRNAs, etc (Chapter 7). Receptors enable cells
or eluant are collected as they emerge (Figure 4-1).
to sense and respond to hormones and other environ-
mental cues (Chapters 42 and 43). An important goal
Partition Chromatography
of molecular medicine is the identification of proteins
whose presence, absence, or deficiency is associated
Column chromatographic separations depend on the
with specific physiologic states or diseases. The primary
relative affinity of different proteins for a given station-
sequence of a protein provides both a molecular finger-
ary phase and for the mobile phase. Association be-
print for its identification and information that can be
tween each protein and the matrix is weak and tran-
used to identify and clone the gene or genes that en-
sient. Proteins that interact more strongly with the
code it.
stationary phase are retained longer. The length of time
that a protein is associated with the stationary phase is a
function of the composition of both the stationary and
mobile phases. Optimal separation of the protein of in-
PROTEINS & PEPTIDES MUST BE
terest from other proteins thus can be achieved by care-
PURIFIED PRIOR TO ANALYSIS
ful manipulation of the composition of the two phases.
Highly purified protein is essential for determination of
its amino acid sequence. Cells contain thousands of dif-
Size Exclusion Chromatography
ferent proteins, each in widely varying amounts. The
isolation of a specific protein in quantities sufficient for
Size exclusion—or gel filtration—chromatography sep-
analysis thus presents a formidable challenge that may
arates proteins based on their Stokes radius, the diam-
require multiple successive purification techniques.
eter of the sphere they occupy as they tumble in solu-
Classic approaches exploit differences in relative solu-
tion. The Stokes radius is a function of molecular mass
bility of individual proteins as a function of pH (iso-
and shape. A tumbling elongated protein occupies a
electric precipitation), polarity
(precipitation with
larger volume than a spherical protein of the same mass.
ethanol or acetone), or salt concentration (salting out
Size exclusion chromatography employs porous beads
with ammonium sulfate). Chromatographic separations
(Figure 4-2). The pores are analogous to indentations
partition molecules between two phases, one mobile
in a river bank. As objects move downstream, those that
and the other stationary. For separation of amino acids
enter an indentation are retarded until they drift back
or sugars, the stationary phase, or matrix, may be a
into the main current. Similarly, proteins with Stokes
sheet of filter paper (paper chromatography) or a thin
radii too large to enter the pores (excluded proteins) re-
layer of cellulose, silica, or alumina (thin-layer chro-
main in the flowing mobile phase and emerge before
matography; TLC).
proteins that can enter the pores (included proteins).
21
22
/
CHAPTER 4
R
C
F
Figure 4-1. Components of a simple liquid chromatography apparatus. R: Reser-
voir of mobile phase liquid, delivered either by gravity or using a pump. C: Glass or
plastic column containing stationary phase. F: Fraction collector for collecting por-
tions, called fractions, of the eluant liquid in separate test tubes.
Proteins thus emerge from a gel filtration column in de-
Ion Exchange Chromatography
scending order of their Stokes radii.
In ion exchange chromatography, proteins interact with
the stationary phase by charge-charge interactions. Pro-
Absorption Chromatography
teins with a net positive charge at a given pH adhere to
For absorption chromatography, the protein mixture is
beads with negatively charged functional groups such as
applied to a column under conditions where the pro-
carboxylates or sulfates (cation exchangers). Similarly,
tein of interest associates with the stationary phase so
proteins with a net negative charge adhere to beads with
tightly that its partition coefficient is essentially unity.
positively charged functional groups, typically tertiary or
Nonadhering molecules are first eluted and discarded.
quaternary amines (anion exchangers). Proteins, which
Proteins are then sequentially released by disrupting the
are polyanions, compete against monovalent ions for
forces that stabilize the protein-stationary phase com-
binding to the support—thus the term “ion exchange.”
plex, most often by using a gradient of increasing salt
For example, proteins bind to diethylaminoethyl
concentration. The composition of the mobile phase is
(DEAE) cellulose by replacing the counter-ions (gener-
altered gradually so that molecules are selectively re-
ally Cl− or CH3COO−) that neutralize the protonated
leased in descending order of their affinity for the sta-
amine. Bound proteins are selectively displaced by grad-
tionary phase.
ually raising the concentration of monovalent ions in
PROTEINS: DETERMINATION OF PRIMARY STRUCTURE
/
23
A
B
C
Figure 4-2. Size-exclusion chromatography. A: A mixture of large molecules
(diamonds) and small molecules (circles) are applied to the top of a gel filtration
column. B: Upon entering the column, the small molecules enter pores in the sta-
tionary phase matrix from which the large molecules are excluded. C: As the mo-
bile phase flows down the column, the large, excluded molecules flow with it
while the small molecules, which are temporarily sheltered from the flow when in-
side the pores, lag farther and farther behind.
the mobile phase. Proteins elute in inverse order of the
fied by affinity chromatography using immobilized sub-
strength of their interactions with the stationary phase.
strates, products, coenzymes, or inhibitors. In theory,
Since the net charge on a protein is determined by
only proteins that interact with the immobilized ligand
the pH (see Chapter 3), sequential elution of proteins
adhere. Bound proteins are then eluted either by compe-
may be achieved by changing the pH of the mobile
tition with soluble ligand or, less selectively, by disrupt-
phase. Alternatively, a protein can be subjected to con-
ing protein-ligand interactions using urea, guanidine
secutive rounds of ion exchange chromatography, each
hydrochloride, mildly acidic pH, or high salt concentra-
at a different pH, such that proteins that co-elute at one
tions. Stationary phase matrices available commercially
pH elute at different salt concentrations at another pH.
contain ligands such as NAD+ or ATP analogs. Among
the most powerful and widely applicable affinity matri-
Hydrophobic Interaction Chromatography
ces are those used for the purification of suitably modi-
fied recombinant proteins. These include a Ni2+ matrix
Hydrophobic interaction chromatography separates
that binds proteins with an attached polyhistidine “tag”
proteins based on their tendency to associate with a sta-
and a glutathione matrix that binds a recombinant pro-
tionary phase matrix coated with hydrophobic groups
tein linked to glutathione S-transferase.
(eg, phenyl Sepharose, octyl Sepharose). Proteins with
exposed hydrophobic surfaces adhere to the matrix via
hydrophobic interactions that are enhanced by a mobile
Peptides Are Purified by Reversed-Phase
phase of high ionic strength. Nonadherent proteins are
High-Pressure Chromatography
first washed away. The polarity of the mobile phase is
The stationary phase matrices used in classic column
then decreased by gradually lowering the salt concentra-
chromatography are spongy materials whose compress-
tion. If the interaction between protein and stationary
ibility limits flow of the mobile phase. High-pressure liq-
phase is particularly strong, ethanol or glycerol may be
uid chromatography (HPLC) employs incompressible
added to the mobile phase to decrease its polarity and
silica or alumina microbeads as the stationary phase and
further weaken hydrophobic interactions.
pressures of up to a few thousand psi. Incompressible
matrices permit both high flow rates and enhanced reso-
Affinity Chromatography
lution. HPLC can resolve complex mixtures of lipids or
Affinity chromatography exploits the high selectivity of
peptides whose properties differ only slightly. Reversed-
most proteins for their ligands. Enzymes may be puri-
phase HPLC exploits a hydrophobic stationary phase of
24
/
CHAPTER 4
aliphatic polymers 3-18 carbon atoms in length. Peptide
through the acrylamide matrix determines the rate of
mixtures are eluted using a gradient of a water-miscible
migration. Since large complexes encounter greater re-
organic solvent such as acetonitrile or methanol.
sistance, polypeptides separate based on their relative
molecular mass (Mr). Individual polypeptides trapped
Protein Purity Is Assessed by
in the acrylamide gel are visualized by staining with
dyes such as Coomassie blue (Figure 4-4).
Polyacrylamide Gel Electrophoresis
(PAGE)
Isoelectric Focusing (IEF)
The most widely used method for determining the pu-
rity of a protein is SDS-PAGE—polyacrylamide gel
Ionic buffers called ampholytes and an applied electric
electrophoresis (PAGE) in the presence of the anionic
field are used to generate a pH gradient within a poly-
detergent sodium dodecyl sulfate (SDS). Electrophore-
acrylamide matrix. Applied proteins migrate until they
sis separates charged biomolecules based on the rates at
reach the region of the matrix where the pH matches
which they migrate in an applied electrical field. For
their isoelectric point (pI), the pH at which a peptide’s
SDS-PAGE, acrylamide is polymerized and cross-
net charge is zero. IEF is used in conjunction with SDS-
linked to form a porous matrix. SDS denatures and
PAGE for two-dimensional electrophoresis, which sepa-
binds to proteins at a ratio of one molecule of SDS per
rates polypeptides based on pI in one dimension and
two peptide bonds. When used in conjunction with 2-
based on Mr in the second (Figure 4-5). Two-dimen-
mercaptoethanol or dithiothreitol to reduce and break
sional electrophoresis is particularly well suited for sepa-
disulfide bonds (Figure 4 -3), SDS separates the com-
rating the components of complex mixtures of proteins.
ponent polypeptides of multimeric proteins. The large
number of anionic SDS molecules, each bearing a
SANGER WAS THE FIRST TO DETERMINE
charge of
−1, on each polypeptide overwhelms the
THE SEQUENCE OF A POLYPEPTIDE
charge contributions of the amino acid functional
groups. Since the charge-to-mass ratio of each SDS-
Mature insulin consists of the 21-residue A chain and
polypeptide complex is approximately equal, the physi-
the 30-residue B chain linked by disulfide bonds. Fred-
cal resistance each peptide encounters as it moves
erick Sanger reduced the disulfide bonds (Figure 4-3),
NH
O
HN
H
S
HN
S
H
NH
O
O
SH
HCOOH
C2H5
OH
NH
O
H
HN
SO −
2
HN
HS
H
NH
O
Figure 4-4. Use of SDS-PAGE to observe successive
purification of a recombinant protein. The gel was
Figure 4-3. Oxidative cleavage of adjacent polypep-
stained with Coomassie blue. Shown are protein stan-
tide chains linked by disulfide bonds (shaded) by per-
dards (lane S) of the indicated mass, crude cell extract
formic acid (left) or reductive cleavage by β-mercap-
(E), high-speed supernatant liquid (H), and the DEAE-
toethanol (right) forms two peptides that contain
Sepharose fraction (D). The recombinant protein has a
cysteic acid residues or cysteinyl residues, respectively.
mass of about 45 kDa.
PROTEINS: DETERMINATION OF PRIMARY STRUCTURE
/
25
pH = 3
pH = 10
IEF
SDS
PAGE
Figure 4-5. Two-dimensional IEF-SDS-PAGE. The
gel was stained with Coomassie blue. A crude bacter-
ial extract was first subjected to isoelectric focusing
(IEF) in a pH 3-10 gradient. The IEF gel was then
placed horizontally on the top of an SDS gel, and the
proteins then further resolved by SDS-PAGE. Notice
the greatly improved resolution of distinct polypep-
tides relative to ordinary SDS-
PAGE gel (Figure 4-4).
separated the A and B chains, and cleaved each chain
Large Polypeptides Are First Cleaved Into
into smaller peptides using trypsin, chymotrypsin, and
Smaller Segments
pepsin. The resulting peptides were then isolated and
While the first 20-30 residues of a peptide can readily
treated with acid to hydrolyze peptide bonds and gener-
be determined by the Edman method, most polypep-
ate peptides with as few as two or three amino acids.
tides contain several hundred amino acids. Conse-
Each peptide was reacted with 1-fluoro-2,4-dinitroben-
quently, most polypeptides must first be cleaved into
zene (Sanger’s reagent), which derivatizes the exposed
smaller peptides prior to Edman sequencing. Cleavage
α-amino group of amino terminal residues. The amino
also may be necessary to circumvent posttranslational
acid content of each peptide was then determined.
modifications that render a protein’s α-amino group
While the ε-amino group of lysine also reacts with
“blocked”, or unreactive with the Edman reagent.
Sanger’s reagent, amino-terminal lysines can be distin-
It usually is necessary to generate several peptides
guished from those at other positions because they react
using more than one method of cleavage. This reflects
with 2 mol of Sanger’s reagent. Working backwards to
both inconsistency in the spacing of chemically or enzy-
larger fragments enabled Sanger to determine the com-
matically susceptible cleavage sites and the need for sets
plete sequence of insulin, an accomplishment for which
of peptides whose sequences overlap so one can infer
he received a Nobel Prize in 1958.
the sequence of the polypeptide from which they derive
(Figure 4-7). Reagents for the chemical or enzymatic
THE EDMAN REACTION ENABLES
cleavage of proteins include cyanogen bromide (CNBr),
PEPTIDES & PROTEINS
trypsin, and Staphylococcus aureus V8 protease (Table
TO BE SEQUENCED
4-1). Following cleavage, the resulting peptides are pu-
rified by reversed-phase HPLC—or occasionally by
Pehr Edman introduced phenylisothiocyanate (Edman’s
SDS-PAGE—and sequenced.
reagent) to selectively label the amino-terminal residue
of a peptide. In contrast to Sanger’s reagent, the
phenylthiohydantoin (PTH) derivative can be removed
MOLECULAR BIOLOGY HAS
under mild conditions to generate a new amino terminal
REVOLUTIONIZED THE DETERMINATION
residue (Figure 4-6). Successive rounds of derivatization
OF PRIMARY STRUCTURE
with Edman’s reagent can therefore be used to sequence
many residues of a single sample of peptide. Edman se-
Knowledge of DNA sequences permits deduction of
quencing has been automated, using a thin film or solid
the primary structures of polypeptides. DNA sequenc-
matrix to immobilize the peptide and HPLC to identify
ing requires only minute amounts of DNA and can
PTH amino acids. Modern gas-phase sequencers can
readily yield the sequence of hundreds of nucleotides.
analyze as little as a few picomoles of peptide.
To clone and sequence the DNA that encodes a partic-
26
/
CHAPTER 4
S
Peptide X Peptide Y
C
Peptide Z
N
+
O
NH2
Carboxyl terminal
Amino terminal
H
N
portion of
portion of
peptide X
peptide Y
N
R
H
O
R′
Figure 4-7. The overlapping peptide Z is used to de-
Phenylisothiocyanate (Edman reagent)
duce that peptides X and Y are present in the original
and a peptide
protein in the order X → Y, not Y ← X.
sequence can be determined and the genetic code used
to infer the primary structure of the encoded poly-
S
peptide.
The hybrid approach enhances the speed and effi-
N
NH
ciency of primary structure analysis and the range of
H
proteins that can be sequenced. It also circumvents ob-
O
H
stacles such as the presence of an amino-terminal block-
N
ing group or the lack of a key overlap peptide. Only a
N
R
O
few segments of primary structure must be determined
H
R′
by Edman analysis.
A phenylthiohydantoic acid
DNA sequencing reveals the order in which amino
acids are added to the nascent polypeptide chain as it is
H+, nitro-
H2O
synthesized on the ribosomes. However, it provides no
methane
information about posttranslational modifications such
as proteolytic processing, methylation, glycosylation,
S
O
phosphorylation, hydroxylation of proline and lysine,
NH2
and disulfide bond formation that accompany matura-
N
NH
+ N
tion. While Edman sequencing can detect the presence
H
R
of most posttranslational events, technical limitations
O
R
often prevent identification of a specific modification.
A phenylthiohydantoin and a peptide
shorter by one residue
Table 4-1. Methods for cleaving polypeptides.
Figure 4-6. The Edman reaction. Phenylisothio-
cyanate derivatizes the amino-terminal residue of a
Method
Bond Cleaved
peptide as a phenylthiohydantoic acid. Treatment with
CNBr
Met-X
acid in a nonhydroxylic solvent releases a phenylthio-
hydantoin, which is subsequently identified by its chro-
Trypsin
Lys-X and Arg-X
matographic mobility, and a peptide one residue
Chymotrypsin
Hydrophobic amino acid-X
shorter. The process is then repeated.
Endoproteinase Lys-C
Lys-X
Endoproteinase Arg-C
Arg-X
ular protein, some means of identifying the correct
clone—eg, knowledge of a portion of its nucleotide se-
Endoproteinase Asp-N
X-Asp
quence—is essential. A hybrid approach thus has
V8 protease
Glu-X, particularly where X is hydro-
emerged. Edman sequencing is used to provide a partial
phobic
amino acid sequence. Oligonucleotide primers modeled
Hydroxylamine
Asn-Gly
on this partial sequence can then be used to identify
clones or to amplify the appropriate gene by the poly-
o-Iodosobenzene
Trp-X
merase chain reaction (PCR) (see Chapter 40). Once an
Mild acid
Asp-Pro
authentic DNA clone is obtained, its oligonucleotide
PROTEINS: DETERMINATION OF PRIMARY STRUCTURE
/
27
MASS SPECTROMETRY DETECTS
COVALENT MODIFICATIONS
Mass spectrometry, which discriminates molecules
based solely on their mass, is ideal for detecting the
S
A
phosphate, hydroxyl, and other groups on posttransla-
tionally modified amino acids. Each adds a specific and
readily identified increment of mass to the modified
amino acid (Table 4-2). For analysis by mass spec-
trometry, a sample in a vacuum is vaporized under
E
conditions where protonation can occur, imparting
D
positive charge. An electrical field then propels the
cations through a magnetic field which deflects them
Figure 4-8. Basic components of a simple mass
at a right angle to their original direction of flight and
spectrometer. A mixture of molecules is vaporized in an
focuses them onto a detector (Figure 4-8). The mag-
ionized state in the sample chamber S. These mole-
netic force required to deflect the path of each ionic
cules are then accelerated down the flight tube by an
species onto the detector, measured as the current ap-
electrical potential applied to accelerator grid A. An ad-
plied to the electromagnet, is recorded. For ions of
justable electromagnet, E, applies a magnetic field that
identical net charge, this force is proportionate to their
deflects the flight of the individual ions until they strike
mass. In a time-of-flight mass spectrometer, a briefly
the detector, D. The greater the mass of the ion, the
applied electric field accelerates the ions towards a de-
higher the magnetic field required to focus it onto the
tector that records the time at which each ion arrives.
detector.
For molecules of identical charge, the velocity to which
they are accelerated—and hence the time required to
reach the detector—will be inversely proportionate to
their mass.
Conventional mass spectrometers generally are used
phase HPLC column are introduced directly into the
to determine the masses of molecules of 1000 Da or
mass spectrometer for immediate determination of
less, whereas time-of-flight mass spectrometers are
their masses.
suited for determining the large masses of proteins.
Peptides inside the mass spectrometer are broken
The analysis of peptides and proteins by mass spec-
down into smaller units by collisions with neutral he-
tometry initially was hindered by difficulties in
lium atoms (collision-induced dissociation), and the
volatilizing large organic molecules. However, matrix-
masses of the individual fragments are determined.
assisted laser-desorption
(MALDI) and electrospray
Since peptide bonds are much more labile than carbon-
dispersion (eg, nanospray) permit the masses of even
carbon bonds, the most abundant fragments will differ
large polypeptides (> 100,000 Da) to be determined
from one another by units equivalent to one or two
with extraordinary accuracy (± 1 Da). Using electro-
amino acids. Since—with the exception of leucine and
spray dispersion, peptides eluting from a reversed-
isoleucine—the molecular mass of each amino acid is
unique, the sequence of the peptide can be recon-
structed from the masses of its fragments.
Table 4-2. Mass increases resulting from
common posttranslational modifications.
Tandem Mass Spectrometry
Complex peptide mixtures can now be analyzed with-
Modification
Mass Increase (Da)
out prior purification by tandem mass spectrometry,
Phosphorylation
80
which employs the equivalent of two mass spectrome-
ters linked in series. The first spectrometer separates in-
Hydroxylation
16
dividual peptides based upon their differences in mass.
Methylation
14
By adjusting the field strength of the first magnet, a sin-
gle peptide can be directed into the second mass spec-
Acetylation
42
trometer, where fragments are generated and their
Myristylation
210
masses determined. As the sensitivity and versatility of
Palmitoylation
238
mass spectrometry continue to increase, it is displacing
Edman sequencers for the direct analysis of protein pri-
Glycosylation
162
mary structure.
28
/
CHAPTER 4
GENOMICS ENABLES PROTEINS TO BE
in the hemoglobin tetramer undergo change pre- and
postpartum. Many proteins undergo posttranslational
IDENTIFIED FROM SMALL AMOUNTS
modifications during maturation into functionally
OF SEQUENCE DATA
competent forms or as a means of regulating their prop-
Primary structure analysis has been revolutionized by
erties. Knowledge of the human genome therefore rep-
genomics, the application of automated oligonucleotide
resents only the beginning of the task of describing liv-
sequencing and computerized data retrieval and analysis
ing organisms in molecular detail and understanding
to sequence an organism’s entire genetic complement.
the dynamics of processes such as growth, aging, and
The first genome sequenced was that of Haemophilus
disease. As the human body contains thousands of cell
influenzae, in
1995. By mid
2001, the complete
types, each containing thousands of proteins, the pro-
genome sequences for over 50 organisms had been de-
teome—the set of all the proteins expressed by an indi-
termined. These include the human genome and those
vidual cell at a particular time—represents a moving
of several bacterial pathogens; the results and signifi-
target of formidable dimensions.
cance of the Human Genome Project are discussed in
Chapter 54. Where genome sequence is known, the
Two-Dimensional Electrophoresis &
task of determining a protein’s DNA-derived primary
Gene Array Chips Are Used to Survey
sequence is materially simplified. In essence, the second
Protein Expression
half of the hybrid approach has already been com-
pleted. All that remains is to acquire sufficient informa-
One goal of proteomics is the identification of proteins
tion to permit the open reading frame (ORF) that
whose levels of expression correlate with medically sig-
encodes the protein to be retrieved from an Internet-
nificant events. The presumption is that proteins whose
accessible genome database and identified. In some
appearance or disappearance is associated with a specific
cases, a segment of amino acid sequence only four or
physiologic condition or disease will provide insights
five residues in length may be sufficient to identify the
into root causes and mechanisms. Determination of the
correct ORF.
proteomes characteristic of each cell type requires the
Computerized search algorithms assist the identifi-
utmost efficiency in the isolation and identification of
cation of the gene encoding a given protein and clarify
individual proteins. The contemporary approach uti-
uncertainties that arise from Edman sequencing and
lizes robotic automation to speed sample preparation
mass spectrometry. By exploiting computers to solve
and large two-dimensional gels to resolve cellular pro-
complex puzzles, the spectrum of information suitable
teins. Individual polypeptides are then extracted and
for identification of the ORF that encodes a particular
analyzed by Edman sequencing or mass spectroscopy.
polypeptide is greatly expanded. In peptide mass profil-
While only about 1000 proteins can be resolved on a
ing, for example, a peptide digest is introduced into the
single gel, two-dimensional electrophoresis has a major
mass spectrometer and the sizes of the peptides are de-
advantage in that it examines the proteins themselves.
termined. A computer is then used to find an ORF
An alternative and complementary approach employs
whose predicted protein product would, if broken
gene arrays, sometimes called DNA chips, to detect the
down into peptides by the cleavage method selected,
expression of the mRNAs which encode proteins.
produce a set of peptides whose masses match those ob-
While changes in the expression of the mRNA encod-
served by mass spectrometry.
ing a protein do not necessarily reflect comparable
changes in the level of the corresponding protein, gene
arrays are more sensitive probes than two-dimensional
PROTEOMICS & THE PROTEOME
gels and thus can examine more gene products.
The Goal of Proteomics Is to Identify the
Entire Complement of Proteins Elaborated
Bioinformatics Assists Identification
by a Cell Under Diverse Conditions
of Protein Functions
While the sequence of the human genome is known,
The functions of a large proportion of the proteins en-
the picture provided by genomics alone is both static
coded by the human genome are presently unknown.
and incomplete. Proteomics aims to identify the entire
Recent advances in bioinformatics permit researchers to
complement of proteins elaborated by a cell under di-
compare amino acid sequences to discover clues to po-
verse conditions. As genes are switched on and off, pro-
tential properties, physiologic roles, and mechanisms of
teins are synthesized in particular cell types at specific
action of proteins. Algorithms exploit the tendency of
times of growth or differentiation and in response to
nature to employ variations of a structural theme to
external stimuli. Muscle cells express proteins not ex-
perform similar functions in several proteins (eg, the
pressed by neural cells, and the type of subunits present
Rossmann nucleotide binding fold to bind NAD(P)H,
PROTEINS: DETERMINATION OF PRIMARY STRUCTURE
/
29
nuclear targeting sequences, and EF hands to bind
• Scientists are now trying to determine the primary
Ca2+). These domains generally are detected in the pri-
sequence and functional role of every protein ex-
mary structure by conservation of particular amino
pressed in a living cell, known as its proteome.
acids at key positions. Insights into the properties and
• A major goal is the identification of proteins whose
physiologic role of a newly discovered protein thus may
appearance or disappearance correlates with physio-
be inferred by comparing its primary structure with
logic phenomena, aging, or specific diseases.
that of known proteins.
REFERENCES
SUMMARY
Deutscher MP (editor): Guide to Protein Purification. Methods En-
• Long amino acid polymers or polypeptides constitute
zymol 1990;182. (Entire volume.)
the basic structural unit of proteins, and the structure
Geveart K, Vandekerckhove J: Protein identification methods in
of a protein provides insight into how it fulfills its
proteomics. Electrophoresis 2000;21:1145.
functions.
Helmuth L: Genome research: map of the human genome 3.0. Sci-
• The Edman reaction enabled amino acid sequence
ence 2001;293:583.
analysis to be automated. Mass spectrometry pro-
Khan J et al: DNA microarray technology: the anticipated impact
on the study of human disease. Biochim Biophys Acta
vides a sensitive and versatile tool for determining
1999;1423:M17.
primary structure and for the identification of post-
McLafferty FW et al: Biomolecule mass spectrometry. Science
translational modifications.
1999;284:1289.
• DNA cloning and molecular biology coupled with
Patnaik SK, Blumenfeld OO: Use of on-line tools and databases for
protein chemistry provide a hybrid approach that
routine sequence analyses. Anal Biochem 2001;289:1.
greatly increases the speed and efficiency for determi-
Schena M et al: Quantitative monitoring of gene expression pat-
nation of primary structures of proteins.
terns with a complementary DNA microarray. Science
1995;270:467.
• Genomics—the analysis of the entire oligonucleotide
sequence of an organism’s complete genetic mater-
Semsarian C, Seidman CE: Molecular medicine in the 21st cen-
tury. Intern Med J 2001;31:53.
ial—has provided further enhancements.
Temple LK et al: Essays on science and society: defining disease in
• Computer algorithms facilitate identification of the
the genomics era. Science 2001;293:807.
open reading frames that encode a given protein by
Wilkins MR et al: High-throughput mass spectrometric discovery
using partial sequences and peptide mass profiling to
of protein post-translational modifications. J Mol Biol
search sequence databases.
1999;289:645.
Proteins: Higher Orders of Structure
5
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
Globular proteins are compact, are roughly spherical
or ovoid in shape, and have axial ratios (the ratio of
Proteins catalyze metabolic reactions, power cellular
their shortest to longest dimensions) of not over 3.
motion, and form macromolecular rods and cables that
Most enzymes are globular proteins, whose large inter-
provide structural integrity to hair, bones, tendons, and
nal volume provides ample space in which to con-
teeth. In nature, form follows function. The structural
struct cavities of the specific shape, charge, and hy-
variety of human proteins therefore reflects the sophis-
drophobicity or hydrophilicity required to bind
tication and diversity of their biologic roles. Maturation
substrates and promote catalysis. By contrast, many
of a newly synthesized polypeptide into a biologically
structural proteins adopt highly extended conforma-
functional protein requires that it be folded into a spe-
tions. These fibrous proteins possess axial ratios of 10
cific three-dimensional arrangement, or conformation.
or more.
During maturation, posttranslational modifications
Lipoproteins and glycoproteins contain covalently
may add new chemical groups or remove transiently
bound lipid and carbohydrate, respectively. Myoglobin,
needed peptide segments. Genetic or nutritional defi-
hemoglobin, cytochromes, and many other proteins
ciencies that impede protein maturation are deleterious
contain tightly associated metal ions and are termed
to health. Examples of the former include Creutzfeldt-
metalloproteins. With the development and applica-
Jakob disease, scrapie, Alzheimer’s disease, and bovine
tion of techniques for determining the amino acid se-
spongiform encephalopathy (mad cow disease). Scurvy
quences of proteins (Chapter 4), more precise classifica-
represents a nutritional deficiency that impairs protein
tion schemes have emerged based upon similarity, or
maturation.
homology, in amino acid sequence and structure.
However, many early classification terms remain in
common use.
CONFORMATION VERSUS
CONFIGURATION
The terms configuration and conformation are often
confused. Configuration refers to the geometric rela-
PROTEINS ARE CONSTRUCTED USING
tionship between a given set of atoms, for example,
MODULAR PRINCIPLES
those that distinguish L- from D-amino acids. Intercon-
Proteins perform complex physical and catalytic func-
version of configurational alternatives requires breaking
tions by positioning specific chemical groups in a pre-
covalent bonds. Conformation refers to the spatial re-
cise three-dimensional arrangement. The polypeptide
lationship of every atom in a molecule. Interconversion
scaffold containing these groups must adopt a confor-
between conformers occurs without covalent bond rup-
mation that is both functionally efficient and phys-
ture, with retention of configuration, and typically via
ically strong. At first glance, the biosynthesis of
rotation about single bonds.
polypeptides comprised of tens of thousands of indi-
vidual atoms would appear to be extremely challeng-
ing. When one considers that a typical polypeptide
PROTEINS WERE INITIALLY CLASSIFIED
can adopt ≥ 1050 distinct conformations, folding into
BY THEIR GROSS CHARACTERISTICS
the conformation appropriate to their biologic func-
Scientists initially approached structure-function rela-
tion would appear to be even more difficult. As de-
tionships in proteins by separating them into classes
scribed in Chapters 3 and 4, synthesis of the polypep-
based upon properties such as solubility, shape, or the
tide backbones of proteins employs a small set of
presence of nonprotein groups. For example, the pro-
common building blocks or modules, the amino acids,
teins that can be extracted from cells using solutions at
joined by a common linkage, the peptide bond. A
physiologic pH and ionic strength are classified as sol-
stepwise modular pathway simplifies the folding and
uble. Extraction of integral membrane proteins re-
processing of newly synthesized polypeptides into ma-
quires dissolution of the membrane with detergents.
ture proteins.
30
PROTEINS: HIGHER ORDERS OF STRUCTURE
/
31
THE FOUR ORDERS OF
PROTEIN STRUCTURE
The modular nature of protein synthesis and folding
are embodied in the concept of orders of protein struc-
ture: primary structure, the sequence of the amino
90
acids in a polypeptide chain; secondary structure, the
folding of short (3- to 30-residue), contiguous segments
of polypeptide into geometrically ordered units; ter-
tiary structure, the three-dimensional assembly of sec-
ψ0
ondary structural units to form larger functional units
such as the mature polypeptide and its component do-
mains; and quaternary structure, the number and
types of polypeptide units of oligomeric proteins and
- 90
their spatial arrangement.
SECONDARY STRUCTURE
Peptide Bonds Restrict Possible
- 90
0
90
Secondary Conformations
φ
Free rotation is possible about only two of the three co-
valent bonds of the polypeptide backbone: the α-car-
Figure 5-1. Ramachandran plot of the main chain
bon (Cα) to the carbonyl carbon (Co) bond and the
phi (Φ) and psi (Ψ) angles for approximately 1000
Cα to nitrogen bond (Figure 3-4). The partial double-
nonglycine residues in eight proteins whose structures
bond character of the peptide bond that links Co to the
were solved at high resolution. The dots represent al-
α-nitrogen requires that the carbonyl carbon, carbonyl
lowable combinations and the spaces prohibited com-
oxygen, and α-nitrogen remain coplanar, thus prevent-
binations of phi and psi angles. (Reproduced, with per-
ing rotation. The angle about the CαN bond is
mission, from Richardson JS: The anatomy and taxonomy
termed the phi (Φ) angle, and that about the CoCα
of protein structures. Adv Protein Chem 1981;34:167.)
bond the psi (Ψ) angle. For amino acids other than
glycine, most combinations of phi and psi angles are
disallowed because of steric hindrance (Figure 5-1).
occur in nature. Schematic diagrams of proteins repre-
The conformations of proline are even more restricted
sent α helices as cylinders.
due to the absence of free rotation of the NCα bond.
The stability of an α helix arises primarily from hy-
Regions of ordered secondary structure arise when a
drogen bonds formed between the oxygen of the pep-
series of aminoacyl residues adopt similar phi and psi
tide bond carbonyl and the hydrogen atom of the pep-
angles. Extended segments of polypeptide (eg, loops)
tide bond nitrogen of the fourth residue down the
can possess a variety of such angles. The angles that de-
polypeptide chain (Figure 5-4). The ability to form the
fine the two most common types of secondary struc-
maximum number of hydrogen bonds, supplemented
ture, the helix and the sheet, fall within the lower
by van der Waals interactions in the core of this tightly
and upper left-hand quadrants of a Ramachandran
packed structure, provides the thermodynamic driving
plot, respectively (Figure 5-1).
force for the formation of an α helix. Since the peptide
bond nitrogen of proline lacks a hydrogen atom to con-
The Alpha Helix
tribute to a hydrogen bond, proline can only be stably
The polypeptide backbone of an α helix is twisted by
accommodated within the first turn of an α helix.
an equal amount about each α-carbon with a phi angle
When present elsewhere, proline disrupts the confor-
of approximately −57 degrees and a psi angle of approx-
mation of the helix, producing a bend. Because of its
imately − 47 degrees. A complete turn of the helix con-
small size, glycine also often induces bends in α helices.
tains an average of 3.6 aminoacyl residues, and the dis-
Many α helices have predominantly hydrophobic R
tance it rises per turn (its pitch) is 0.54 nm (Figure
groups on one side of the axis of the helix and predomi-
5-2). The R groups of each aminoacyl residue in an α
nantly hydrophilic ones on the other. These amphi-
helix face outward (Figure 5-3). Proteins contain only
pathic helices are well adapted to the formation of in-
L-amino acids, for which a right-handed α helix is by
terfaces between polar and nonpolar regions such as the
far the more stable, and only right-handed α helices
hydrophobic interior of a protein and its aqueous envi-
32
/
CHAPTER 5
R
R
N
C
R
C
R
N
C
C
N
C
R
C
N
C
R
R
C
N
C
C
R
N
R
C
C
Figure 5-3. View down the axis of an α helix. The
N
C
side chains (R) are on the outside of the helix. The van
C
der Waals radii of the atoms are larger than shown here;
N
C
hence, there is almost no free space inside the helix.
C
(Slightly modified and reproduced, with permission, from
0.54-nm pitch
Stryer L: Biochemistry, 3rd ed. Freeman, 1995. Copyright
(3.6 residues)
N
C
© 1995 by W.H. Freeman and Co.)
C
N
C
0.15 nm
C
N
polypeptide chain proceed in the same direction amino
C
to carboxyl, or an antiparallel sheet, in which they pro-
ceed in opposite directions (Figure 5-5). Either config-
uration permits the maximum number of hydrogen
bonds between segments, or strands, of the sheet. Most
Figure 5-2. Orientation of the main chain atoms of a
β sheets are not perfectly flat but tend to have a right-
peptide about the axis of an α helix.
handed twist. Clusters of twisted strands of β sheet
form the core of many globular proteins (Figure 5-6).
Schematic diagrams represent β sheets as arrows that
point in the amino to carboxyl terminal direction.
ronment. Clusters of amphipathic helices can create a
channel, or pore, that permits specific polar molecules
to pass through hydrophobic cell membranes.
Loops & Bends
Roughly half of the residues in a “typical” globular pro-
The Beta Sheet
tein reside in α helices and β sheets and half in loops,
The second (hence “beta”) recognizable regular sec-
turns, bends, and other extended conformational fea-
ondary structure in proteins is the β sheet. The amino
tures. Turns and bends refer to short segments of
acid residues of a β sheet, when viewed edge-on, form a
amino acids that join two units of secondary structure,
zigzag or pleated pattern in which the R groups of adja-
such as two adjacent strands of an antiparallel β sheet.
cent residues point in opposite directions. Unlike the
A β turn involves four aminoacyl residues, in which the
compact backbone of the α helix, the peptide backbone
first residue is hydrogen-bonded to the fourth, resulting
of the β sheet is highly extended. But like the α helix,
in a tight 180-degree turn (Figure 5-7). Proline and
β sheets derive much of their stability from hydrogen
glycine often are present in β turns.
bonds between the carbonyl oxygens and amide hydro-
Loops are regions that contain residues beyond the
gens of peptide bonds. However, in contrast to the α
minimum number necessary to connect adjacent re-
helix, these bonds are formed with adjacent segments of
gions of secondary structure. Irregular in conformation,
β sheet (Figure 5-5).
loops nevertheless serve key biologic roles. For many
Interacting β sheets can be arranged either to form a
enzymes, the loops that bridge domains responsible for
parallel β sheet, in which the adjacent segments of the
binding substrates often contain aminoacyl residues
PROTEINS: HIGHER ORDERS OF STRUCTURE
/
33
N
C
C
R
N
C
R
C
N
R
C
C
N
R
C
C
N
C
R
C
N
R
C
C
N O
C
R
C
N
R
C
C
N
C
R
C
N
C
R
C
N
R
C
Figure 5-5.
Spacing and bond angles of the hydro-
C
gen bonds of antiparallel and parallel pleated β sheets.
Arrows indicate the direction of each strand. The hydro-
gen-donating α-nitrogen atoms are shown as blue cir-
cles. Hydrogen bonds are indicated by dotted lines. For
Figure 5-4. Hydrogen bonds (dotted lines) formed
clarity in presentation, R groups and hydrogens are
between H and O atoms stabilize a polypeptide in an
omitted. Top: Antiparallel β sheet. Pairs of hydrogen
α-helical conformation. (Reprinted, with permission,
bonds alternate between being close together and
from Haggis GH et al: Introduction to Molecular Biology.
wide apart and are oriented approximately perpendicu-
Wiley, 1964.)
lar to the polypeptide backbone. Bottom: Parallel β
sheet. The hydrogen bonds are evenly spaced but slant
in alternate directions.
that participate in catalysis. Helix-loop-helix motifs
provide the oligonucleotide-binding portion of DNA-
binding proteins such as repressors and transcription
dered regions assume an ordered conformation upon
factors. Structural motifs such as the helix-loop-helix
motif that are intermediate between secondary and ter-
binding of a ligand. This structural flexibility enables
such regions to act as ligand-controlled switches that af-
tiary structures are often termed supersecondary struc-
tures. Since many loops and bends reside on the surface
fect protein structure and function.
of proteins and are thus exposed to solvent, they consti-
tute readily accessible sites, or epitopes, for recognition
Tertiary & Quaternary Structure
and binding of antibodies.
While loops lack apparent structural regularity, they
The term “tertiary structure” refers to the entire three-
exist in a specific conformation stabilized through hy-
dimensional conformation of a polypeptide. It indicates,
drogen bonding, salt bridges, and hydrophobic interac-
in three-dimensional space, how secondary structural
tions with other portions of the protein. However, not
features—helices, sheets, bends, turns, and loops—
all portions of proteins are necessarily ordered. Proteins
assemble to form domains and how these domains re-
may contain “disordered” regions, often at the extreme
late spatially to one another. A domain is a section of
amino or carboxyl terminal, characterized by high con-
protein structure sufficient to perform a particular
formational flexibility. In many instances, these disor-
chemical or physical task such as binding of a substrate
34
/
CHAPTER 5
COOH
H
H
CH
2
N
H
Cα
H
Cα
C
H
N
O
C
O
C
O H
N
CH3
CH2OH
Cα
Cα
H
H
Figure 5-7. A β-turn that links two segments of an-
tiparallel β sheet. The dotted line indicates the hydro-
gen bond between the first and fourth amino acids of
the four-residue segment Ala-Gly-Asp-Ser.
30
15
or other ligand. Other domains may anchor a protein to
55
a membrane or interact with a regulatory molecule that
N
modulates its function. A small polypeptide such as
345
80
70
triose phosphate isomerase (Figure 5-6) or myoglobin
50
280
(Chapter 6) may consist of a single domain. By contrast,
330
90
protein kinases contain two domains. Protein kinases
150
185
350
catalyze the transfer of a phosphoryl group from ATP to
a peptide or protein. The amino terminal portion of the
145
C
230
377
polypeptide, which is rich in β sheet, binds ATP, while
245
310
320
the carboxyl terminal domain, which is rich in α helix,
110
220
260
binds the peptide or protein substrate (Figure 5-8). The
groups that catalyze phosphoryl transfer reside in a loop
300
258
positioned at the interface of the two domains.
120
In some cases, proteins are assembled from more
205
than one polypeptide, or protomer. Quaternary struc-
170
ture defines the polypeptide composition of a protein
125
and, for an oligomeric protein, the spatial relationships
Figure 5-6. Examples of tertiary structure of pro-
between its subunits or protomers. Monomeric pro-
teins. Top: The enzyme triose phosphate isomerase.
teins consist of a single polypeptide chain. Dimeric
Note the elegant and symmetrical arrangement of al-
proteins contain two polypeptide chains. Homodimers
contain two copies of the same polypeptide chain,
ternating β sheets and α helices. (Courtesy of J Richard-
while in a heterodimer the polypeptides differ. Greek
son.) Bottom: Two-domain structure of the subunit of a
letters (α, β, γ etc) are used to distinguish different sub-
homodimeric enzyme, a bacterial class II HMG-CoA re-
units of a heterooligomeric protein, and subscripts indi-
ductase. As indicated by the numbered residues, the
cate the number of each subunit type. For example, α4
single polypeptide begins in the large domain, enters
designates a homotetrameric protein, and α2β2γ a pro-
the small domain, and ends in the large domain. (Cour-
tein with five subunits of three different types.
tesy of C Lawrence, V Rodwell, and C Stauffacher, Purdue
Since even small proteins contain many thousands
University.)
of atoms, depictions of protein structure that indicate
the position of every atom are generally too complex to
be readily interpreted. Simplified schematic diagrams
thus are used to depict key features of a protein’s ter-
PROTEINS: HIGHER ORDERS OF STRUCTURE
/
35
from water. Other significant contributors include hy-
drogen bonds and salt bridges between the carboxylates
of aspartic and glutamic acid and the oppositely
charged side chains of protonated lysyl, argininyl, and
histidyl residues. While individually weak relative to a
typical covalent bond of 80-120 kcal/mol, collectively
these numerous interactions confer a high degree of sta-
bility to the biologically functional conformation of a
protein, just as a Velcro fastener harnesses the cumula-
tive strength of multiple plastic loops and hooks.
Some proteins contain covalent disulfide
(S S)
bonds that link the sulfhydryl groups of cysteinyl
residues. Formation of disulfide bonds involves oxida-
tion of the cysteinyl sulfhydryl groups and requires oxy-
gen. Intrapolypeptide disulfide bonds further enhance
the stability of the folded conformation of a peptide,
while interpolypeptide disulfide bonds stabilize the
quaternary structure of certain oligomeric proteins.
THREE-DIMENSIONAL STRUCTURE
IS DETERMINED BY X-RAY
CRYSTALLOGRAPHY OR BY
NMR SPECTROSCOPY
X-Ray Crystallography
Since the determination of the three-dimensional struc-
ture of myoglobin over 40 years ago, the three-dimen-
sional structures of thousands of proteins have been de-
Figure 5-8.
Domain structure. Protein kinases con-
termined by x-ray crystallography. The key to x-ray
crystallography is the precipitation of a protein under
tain two domains. The upper, amino terminal domain
conditions in which it forms ordered crystals that dif-
binds the phosphoryl donor ATP (light blue). The lower,
fract x-rays. This is generally accomplished by exposing
carboxyl terminal domain is shown binding a synthetic
small drops of the protein solution to various combina-
peptide substrate (dark blue).
tions of pH and precipitating agents such as salts and
organic solutes such as polyethylene glycol. A detailed
three-dimensional structure of a protein can be con-
tiary and quaternary structure. Ribbon diagrams (Fig-
structed from its primary structure using the pattern by
ures
5-6 and 5-8) trace the conformation of the
which it diffracts a beam of monochromatic x-rays.
polypeptide backbone, with cylinders and arrows indi-
While the development of increasingly capable com-
cating regions of α helix and β sheet, respectively. In an
puter-based tools has rendered the analysis of complex
even simpler representation, line segments that link the
x-ray diffraction patterns increasingly facile, a major
α carbons indicate the path of the polypeptide back-
stumbling block remains the requirement of inducing
bone. These schematic diagrams often include the side
highly purified samples of the protein of interest to
chains of selected amino acids that emphasize specific
crystallize. Several lines of evidence, including the abil-
structure-function relationships.
ity of some crystallized enzymes to catalyze chemical re-
actions, indicate that the vast majority of the structures
MULTIPLE FACTORS STABILIZE
determined by crystallography faithfully represent the
structures of proteins in free solution.
TERTIARY & QUATERNARY STRUCTURE
Higher orders of protein structure are stabilized primar-
Nuclear Magnetic Resonance
ily—and often exclusively—by noncovalent interac-
Spectroscopy
tions. Principal among these are hydrophobic interac-
tions that drive most hydrophobic amino acid side
Nuclear magnetic resonance (NMR) spectroscopy, a
chains into the interior of the protein, shielding them
powerful complement to x-ray crystallography, mea-
36
/
CHAPTER 5
sures the absorbance of radio frequency electromagnetic
Folding Is Modular
energy by certain atomic nuclei. “NMR-active” isotopes
Protein folding generally occurs via a stepwise process.
of biologically relevant atoms include 1H, 13C, 15N, and
In the first stage, the newly synthesized polypeptide
31P. The frequency, or chemical shift, at which a partic-
emerges from ribosomes, and short segments fold into
ular nucleus absorbs energy is a function of both the
secondary structural units that provide local regions of
functional group within which it resides and the prox-
organized structure. Folding is now reduced to the se-
imity of other NMR-active nuclei. Two-dimensional
lection of an appropriate arrangement of this relatively
NMR spectroscopy permits a three-dimensional repre-
small number of secondary structural elements. In the
sentation of a protein to be constructed by determining
second stage, the forces that drive hydrophobic regions
the proximity of these nuclei to one another. NMR
into the interior of the protein away from solvent drive
spectroscopy analyzes proteins in aqueous solution, ob-
the partially folded polypeptide into a “molten globule”
viating the need to form crystals. It thus is possible to
in which the modules of secondary structure rearrange
observe changes in conformation that accompany lig-
to arrive at the mature conformation of the protein.
and binding or catalysis using NMR spectroscopy.
This process is orderly but not rigid. Considerable flexi-
However, only the spectra of relatively small proteins,
bility exists in the ways and in the order in which ele-
≤ 20 kDa in size, can be analyzed with current tech-
ments of secondary structure can be rearranged. In gen-
nology.
eral, each element of secondary or supersecondary
structure facilitates proper folding by directing the fold-
Molecular Modeling
ing process toward the native conformation and away
An increasingly useful adjunct to the empirical determi-
from unproductive alternatives. For oligomeric pro-
nation of the three-dimensional structure of proteins is
teins, individual protomers tend to fold before they as-
the use of computer technology for molecular model-
sociate with other subunits.
ing. The types of models created take two forms. In the
first, the known three-dimensional structure of a pro-
Auxiliary Proteins Assist Folding
tein is used as a template to build a model of the proba-
ble structure of a homologous protein. In the second,
Under appropriate conditions, many proteins will
computer software is used to manipulate the static
spontaneously refold after being previously denatured
model provided by crystallography to explore how a
(ie, unfolded) by treatment with acid or base,
protein’s conformation might change when ligands are
chaotropic agents, or detergents. However, unlike the
bound or when temperature, pH, or ionic strength is
folding process in vivo, refolding under laboratory con-
altered. Scientists also are examining the library of
ditions is a far slower process. Moreover, some proteins
available protein structures in an attempt to devise
fail to spontaneously refold in vitro, often forming in-
computer programs that can predict the three-dimen-
soluble aggregates, disordered complexes of unfolded
sional conformation of a protein directly from its pri-
or partially folded polypeptides held together by hy-
mary sequence.
drophobic interactions. Aggregates represent unproduc-
tive dead ends in the folding process. Cells employ aux-
PROTEIN FOLDING
iliary proteins to speed the process of folding and to
guide it toward a productive conclusion.
The Native Conformation of a Protein
Is Thermodynamically Favored
Chaperones
The number of distinct combinations of phi and psi
angles specifying potential conformations of even a rel-
Chaperone proteins participate in the folding of over
atively small—15-kDa—polypeptide is unbelievably
half of mammalian proteins. The hsp70 (70-kDa heat
vast. Proteins are guided through this vast labyrinth of
shock protein) family of chaperones binds short se-
possibilities by thermodynamics. Since the biologically
quences of hydrophobic amino acids in newly syn-
relevant—or native—conformation of a protein gener-
thesized polypeptides, shielding them from solvent.
ally is that which is most energetically favored, knowl-
Chaperones prevent aggregation, thus providing an op-
edge of the native conformation is specified in the pri-
portunity for the formation of appropriate secondary
mary sequence. However, if one were to wait for a
structural elements and their subsequent coalescence
polypeptide to find its native conformation by random
into a molten globule. The hsp60 family of chaperones,
exploration of all possible conformations, the process
sometimes called chaperonins, differ in sequence and
would require billions of years to complete. Clearly,
structure from hsp70 and its homologs. Hsp60 acts
protein folding in cells takes place in a more orderly
later in the folding process, often together with an
and guided fashion.
hsp70 chaperone. The central cavity of the donut-
PROTEINS: HIGHER ORDERS OF STRUCTURE
/
37
shaped hsp60 chaperone provides a sheltered environ-
sheep, and bovine spongiform encephalopathy (mad
ment in which a polypeptide can fold until all hy-
cow disease) in cattle. Prion diseases may manifest
drophobic regions are buried in its interior, eliminating
themselves as infectious, genetic, or sporadic disorders.
aggregation. Chaperone proteins can also “rescue” pro-
Because no viral or bacterial gene encoding the patho-
teins that have become thermodynamically trapped in a
logic prion protein could be identified, the source and
misfolded dead end by unfolding hydrophobic regions
mechanism of transmission of prion disease long re-
and providing a second chance to fold productively.
mained elusive. Today it is believed that prion diseases
are protein conformation diseases transmitted by alter-
Protein Disulfide Isomerase
ing the conformation, and hence the physical proper-
ties, of proteins endogenous to the host. Human prion-
Disulfide bonds between and within polypeptides stabi-
related protein, PrP, a glycoprotein encoded on the
lize tertiary and quaternary structure. However, disul-
short arm of chromosome 20, normally is monomeric
fide bond formation is nonspecific. Under oxidizing
and rich in α helix. Pathologic prion proteins serve as
conditions, a given cysteine can form a disulfide bond
the templates for the conformational transformation of
with the SH of any accessible cysteinyl residue. By
normal PrP, known as PrPc, into PrPsc. PrPsc is rich in
catalyzing disulfide exchange, the rupture of an SS
β sheet with many hydrophobic aminoacyl side chains
bond and its reformation with a different partner cys-
exposed to solvent. PrPsc molecules therefore associate
teine, protein disulfide isomerase facilitates the forma-
strongly with one other, forming insoluble protease-re-
tion of disulfide bonds that stabilize their native confor-
sistant aggregates. Since one pathologic prion or prion-
mation.
related protein can serve as template for the conforma-
tional transformation of many times its number of PrPc
Proline-cis,trans-Isomerase
molecules, prion diseases can be transmitted by the pro-
tein alone without involvement of DNA or RNA.
All X-Pro peptide bonds—where X represents any
residue—are synthesized in the trans configuration.
However, of the X-Pro bonds of mature proteins, ap-
Alzheimer’s Disease
proximately 6% are cis. The cis configuration is partic-
Refolding or misfolding of another protein endogenous
ularly common in β-turns. Isomerization from trans to
to human brain tissue, β-amyloid, is also a prominent
cis is catalyzed by the enzyme proline-cis,trans-iso-
feature of Alzheimer’s disease. While the root cause of
merase (Figure 5-9).
Alzheimer’s disease remains elusive, the characteristic
senile plaques and neurofibrillary bundles contain ag-
SEVERAL NEUROLOGIC DISEASES
gregates of the protein β-amyloid, a 4.3-kDa polypep-
RESULT FROM ALTERED PROTEIN
tide produced by proteolytic cleavage of a larger protein
known as amyloid precursor protein. In Alzheimer’s
CONFORMATION
disease patients, levels of β-amyloid become elevated,
Prions
and this protein undergoes a conformational transfor-
mation from a soluble α helix-rich state to a state rich
The transmissible spongiform encephalopathies, or
in β sheet and prone to self-aggregation. Apolipopro-
prion diseases, are fatal neurodegenerative diseases
tein E has been implicated as a potential mediator of
characterized by spongiform changes, astrocytic gli-
this conformational transformation.
omas, and neuronal loss resulting from the deposition
of insoluble protein aggregates in neural cells. They in-
clude Creutzfeldt-Jakob disease in humans, scrapie in
COLLAGEN ILLUSTRATES THE ROLE OF
POSTTRANSLATIONAL PROCESSING IN
PROTEIN MATURATION
O
O
O
Protein Maturation Often Involves Making
H
H
′
N
N
& Breaking Covalent Bonds
α1
N
α1
N
The maturation of proteins into their final structural
R1
α1
′
R1
state often involves the cleavage or formation (or both)
O
of covalent bonds, a process termed posttranslational
modification. Many polypeptides are initially synthe-
Figure 5-9. Isomerization of the N-α1 prolyl peptide
sized as larger precursors, called proproteins. The
bond from a cis to a trans configuration relative to the
“extra” polypeptide segments in these proproteins
backbone of the polypeptide.
often serve as leader sequences that target a polypeptide
38
/
CHAPTER 5
to a particular organelle or facilitate its passage through
rise per residue nearly twice that of an α helix. The
a membrane. Others ensure that the potentially harm-
R groups of each polypeptide strand of the triple helix
ful activity of a protein such as the proteases trypsin
pack so closely that in order to fit, one must be glycine.
and chymotrypsin remains inhibited until these pro-
Thus, every third amino acid residue in collagen is a
teins reach their final destination. However, once these
glycine residue. Staggering of the three strands provides
transient requirements are fulfilled, the now superflu-
appropriate positioning of the requisite glycines
ous peptide regions are removed by selective proteoly-
throughout the helix. Collagen is also rich in proline
sis. Other covalent modifications may take place that
and hydroxyproline, yielding a repetitive Gly-X-Y pat-
add new chemical functionalities to a protein. The mat-
tern (Figure 5-10) in which Y generally is proline or
uration of collagen illustrates both of these processes.
hydroxyproline.
Collagen triple helices are stabilized by hydrogen
Collagen Is a Fibrous Protein
bonds between residues in different polypeptide chains.
The hydroxyl groups of hydroxyprolyl residues also par-
Collagen is the most abundant of the fibrous proteins
ticipate in interchain hydrogen bonding. Additional
that constitute more than 25% of the protein mass in
stability is provided by covalent cross-links formed be-
the human body. Other prominent fibrous proteins in-
tween modified lysyl residues both within and between
clude keratin and myosin. These proteins represent a
polypeptide chains.
primary source of structural strength for cells (ie, the
cytoskeleton) and tissues. Skin derives its strength and
Collagen Is Synthesized as a
flexibility from a crisscrossed mesh of collagen and ker-
atin fibers, while bones and teeth are buttressed by an
Larger Precursor
underlying network of collagen fibers analogous to the
Collagen is initially synthesized as a larger precursor
steel strands in reinforced concrete. Collagen also is
polypeptide, procollagen. Numerous prolyl and lysyl
present in connective tissues such as ligaments and ten-
residues of procollagen are hydroxylated by prolyl hy-
dons. The high degree of tensile strength required to
droxylase and lysyl hydroxylase, enzymes that require
fulfill these structural roles requires elongated proteins
ascorbic acid (vitamin C). Hydroxyprolyl and hydroxy-
characterized by repetitive amino acid sequences and a
lysyl residues provide additional hydrogen bonding ca-
regular secondary structure.
pability that stabilizes the mature protein. In addition,
glucosyl and galactosyl transferases attach glucosyl or
Collagen Forms a Unique Triple Helix
galactosyl residues to the hydroxyl groups of specific
hydroxylysyl residues.
Tropocollagen consists of three fibers, each containing
The central portion of the precursor polypeptide
about 1000 amino acids, bundled together in a unique
then associates with other molecules to form the char-
conformation, the collagen triple helix (Figure 5-10). A
acteristic triple helix. This process is accompanied by
mature collagen fiber forms an elongated rod with an
the removal of the globular amino terminal and car-
axial ratio of about 200. Three intertwined polypeptide
boxyl terminal extensions of the precursor polypeptide
strands, which twist to the left, wrap around one an-
by selective proteolysis. Certain lysyl residues are modi-
other in a right-handed fashion to form the collagen
fied by lysyl oxidase, a copper-containing protein that
triple helix. The opposing handedness of this superhelix
converts ε-amino groups to aldehydes. The aldehydes
and its component polypeptides makes the collagen
can either undergo an aldol condensation to form a
triple helix highly resistant to unwinding—the same
CC double bond or to form a Schiff base (eneimine)
principle used in the steel cables of suspension bridges.
with the ε-amino group of an unmodified lysyl residue,
A collagen triple helix has 3.3 residues per turn and a
which is subsequently reduced to form a CN single
bond. These covalent bonds cross-link the individual
Amino acid
polypeptides and imbue the fiber with exceptional
- Gly - X - Y - Gly - X - Y - Gly - X - Y -
sequence
strength and rigidity.
2º structure
Nutritional & Genetic Disorders Can Impair
Collagen Maturation
The complex series of events in collagen maturation
Triple helix
provide a model that illustrates the biologic conse-
quences of incomplete polypeptide maturation. The
Figure 5-10. Primary, secondary, and tertiary struc-
best-known defect in collagen biosynthesis is scurvy, a
tures of collagen.
result of a dietary deficiency of vitamin C required by
PROTEINS: HIGHER ORDERS OF STRUCTURE
/
39
prolyl and lysyl hydroxylases. The resulting deficit in
sized polypeptide fold into secondary structural
the number of hydroxyproline and hydroxylysine
units. Forces that bury hydrophobic regions from
residues undermines the conformational stability of col-
solvent then drive the partially folded polypeptide
lagen fibers, leading to bleeding gums, swelling joints,
into a “molten globule” in which the modules of sec-
poor wound healing, and ultimately to death. Menkes’
ondary structure are rearranged to give the native
syndrome, characterized by kinky hair and growth re-
conformation of the protein.
tardation, reflects a dietary deficiency of the copper re-
•
Proteins that assist folding include protein disulfide
quired by lysyl oxidase, which catalyzes a key step in
isomerase, proline-cis,trans,-isomerase, and the chap-
formation of the covalent cross-links that strengthen
erones that participate in the folding of over half of
collagen fibers.
mammalian proteins. Chaperones shield newly syn-
Genetic disorders of collagen biosynthesis include
thesized polypeptides from solvent and provide an
several forms of osteogenesis imperfecta, characterized
environment for elements of secondary structure to
by fragile bones. In Ehlers-Dahlos syndrome, a group
emerge and coalesce into molten globules.
of connective tissue disorders that involve impaired in-
•
Techniques for study of higher orders of protein
tegrity of supporting structures, defects in the genes
structure include x-ray crystallography, NMR spec-
that encode α collagen-1, procollagen N-peptidase, or
troscopy, analytical ultracentrifugation, gel filtration,
lysyl hydroxylase result in mobile joints and skin abnor-
and gel electrophoresis.
malities.
•
Silk fibroin and collagen illustrate the close linkage of
protein structure and biologic function. Diseases of
SUMMARY
collagen maturation include Ehlers-Danlos syndrome
•
Proteins may be classified on the basis of the solubil-
and the vitamin C deficiency disease scurvy.
ity, shape, or function or of the presence of a pros-
•
Prions—protein particles that lack nucleic acid—
thetic group such as heme. Proteins perform complex
cause fatal transmissible spongiform encephalopa-
physical and catalytic functions by positioning spe-
thies such as Creutzfeldt-Jakob disease, scrapie, and
cific chemical groups in a precise three-dimensional
bovine spongiform encephalopathy. Prion diseases
arrangement that is both functionally efficient and
involve an altered secondary-tertiary structure of a
physically strong.
naturally occurring protein, PrPc. When PrPc inter-
acts with its pathologic isoform PrPSc, its conforma-
•
The gene-encoded primary structure of a polypeptide
is the sequence of its amino acids. Its secondary
tion is transformed from a predominantly α-helical
structure to the β-sheet structure characteristic of
structure results from folding of polypeptides into
hydrogen-bonded motifs such as the α helix, the
PrPSc.
β-pleated sheet, β bends, and loops. Combinations
of these motifs can form supersecondary motifs.
•
Tertiary structure concerns the relationships between
REFERENCES
secondary structural domains. Quaternary structure
of proteins with two or more polypeptides
Branden C, Tooze J: Introduction to Protein Structure. Garland,
1991.
(oligomeric proteins) is a feature based on the spatial
Burkhard P, Stetefeld J, Strelkov SV: Coiled coils: A highly versa-
relationships between various types of polypeptides.
tile protein folding motif. Trends Cell Biol 2001;11:82.
•
Primary structures are stabilized by covalent peptide
Collinge J: Prion diseases of humans and animals: Their causes and
bonds. Higher orders of structure are stabilized by
molecular basis. Annu Rev Neurosci 2001;24:519.
weak forces—multiple hydrogen bonds, salt (electro-
Frydman J: Folding of newly translated proteins in vivo: The role
static) bonds, and association of hydrophobic R
of molecular chaperones. Annu Rev Biochem 2001;70:603.
groups.
Radord S: Protein folding: Progress made and promises ahead.
•
The phi (Φ) angle of a polypeptide is the angle about
Trends Biochem Sci 2000;25:611.
the CαN bond; the psi (Ψ) angle is that about the
Schmid FX: Proly isomerase: Enzymatic catalysis of slow protein
folding reactions. Ann Rev Biophys Biomol Struct 1993;22:
Cα-Co bond. Most combinations of phi-psi angles
123.
are disallowed due to steric hindrance. The phi-psi
Segrest MP et al: The amphipathic alpha-helix: A multifunctional
angles that form the α helix and the β sheet fall
structural motif in plasma lipoproteins. Adv Protein Chem
within the lower and upper left-hand quadrants of a
1995;45:1.
Ramachandran plot, respectively.
Soto C: Alzheimer’s and prion disease as disorders of protein con-
•
Protein folding is a poorly understood process.
formation: Implications for the design of novel therapeutic
Broadly speaking, short segments of newly synthe-
approaches. J Mol Med 1999;77:412.
Proteins: Myoglobin & Hemoglobin
6
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
Myoglobin Is Rich in α Helix
The heme proteins myoglobin and hemoglobin main-
Oxygen stored in red muscle myoglobin is released dur-
tain a supply of oxygen essential for oxidative metabo-
ing O2 deprivation (eg, severe exercise) for use in mus-
lism. Myoglobin, a monomeric protein of red muscle,
cle mitochondria for aerobic synthesis of ATP
(see
stores oxygen as a reserve against oxygen deprivation.
Chapter
12). A
153-aminoacyl residue polypeptide
Hemoglobin, a tetrameric protein of erythrocytes,
(MW 17,000), myoglobin folds into a compact shape
transports O2 to the tissues and returns CO2 and pro-
that measures 4.5 × 3.5 × 2.5 nm (Figure 6-2). Unusu-
tons to the lungs. Cyanide and carbon monoxide kill
ally high proportions, about 75%, of the residues are
because they disrupt the physiologic function of the
present in eight right-handed, 7-20 residue α helices.
heme proteins cytochrome oxidase and hemoglobin, re-
Starting at the amino terminal, these are termed helices
spectively. The secondary-tertiary structure of the sub-
A-H. Typical of globular proteins, the surface of myo-
units of hemoglobin resembles myoglobin. However,
globin is polar, while—with only two exceptions—the
the tetrameric structure of hemoglobin permits cooper-
interior contains only nonpolar residues such as Leu,
ative interactions that are central to its function. For ex-
Val, Phe, and Met. The exceptions are His E7 and His
ample, 2,3-bisphosphoglycerate
(BPG) promotes the
F8, the seventh and eighth residues in helices E and F,
efficient release of O2
by stabilizing the quaternary
which lie close to the heme iron where they function in
structure of deoxyhemoglobin. Hemoglobin and myo-
O2 binding.
globin illustrate both protein structure-function rela-
tionships and the molecular basis of genetic diseases
such as sickle cell disease and the thalassemias.
Histidines F8 & E7 Perform Unique Roles in
Oxygen Binding
The heme of myoglobin lies in a crevice between helices
HEME & FERROUS IRON CONFER THE
E and F oriented with its polar propionate groups fac-
ABILITY TO STORE & TO TRANSPORT
ing the surface of the globin (Figure 6-2). The remain-
OXYGEN
der resides in the nonpolar interior. The fifth coordina-
tion position of the iron is linked to a ring nitrogen of
Myoglobin and hemoglobin contain heme, a cyclic
the proximal histidine, His F8. The distal histidine,
tetrapyrrole consisting of four molecules of pyrrole
His E7, lies on the side of the heme ring opposite to
linked by α-methylene bridges. This planar network of
His F8.
conjugated double bonds absorbs visible light and col-
ors heme deep red. The substituents at the β-positions
of heme are methyl (M), vinyl (V), and propionate (Pr)
The Iron Moves Toward the Plane of the
groups arranged in the order M, V, M, V, M, Pr, Pr, M
Heme When Oxygen Is Bound
(Figure 6-1). One atom of ferrous iron (Fe2+) resides at
the center of the planar tetrapyrrole. Other proteins
The iron of unoxygenated myoglobin lies 0.03 nm
with metal-containing tetrapyrrole prosthetic groups
(0.3 Å) outside the plane of the heme ring, toward His
include the cytochromes (Fe and Cu) and chlorophyll
F8. The heme therefore “puckers” slightly. When O2
(Mg) (see Chapter 12). Oxidation and reduction of the
occupies the sixth coordination position, the iron
Fe and Cu atoms of cytochromes is essential to their bi-
moves to within 0.01 nm (0.1 Å) of the plane of the
ologic function as carriers of electrons. By contrast, oxi-
heme ring. Oxygenation of myoglobin thus is accompa-
+
dation of the Fe2+
of myoglobin or hemoglobin to Fe3
nied by motion of the iron, of His F8, and of residues
destroys their biologic activity.
linked to His F8.
40
PROTEINS: MYOGLOBIN & HEMOGLOBIN
/
41
Apomyoglobin Provides a Hindered
Environment for Heme Iron
When O2 binds to myoglobin, the bond between the first
N
oxygen atom and the Fe2+ is perpendicular to the plane of
the heme ring. The bond linking the first and second
Fe2+
oxygen atoms lies at an angle of 121 degrees to the plane
of the heme, orienting the second oxygen away from the
N
distal histidine (Figure 6-3, left). Isolated heme binds
-O
carbon monoxide (CO) 25,000 times more strongly than
O
oxygen. Since CO is present in small quantities in the at-
mosphere and arises in cells from the catabolism of heme,
why is it that CO does not completely displace O2 from
heme iron? The accepted explanation is that the apopro-
teins of myoglobin and hemoglobin create a hindered
O-
environment. While CO can bind to isolated heme in its
Figure 6-1. Heme. The pyrrole rings and methylene
preferred orientation, ie, with all three atoms (Fe, C, and
bridge carbons are coplanar, and the iron atom (Fe2+)
O) perpendicular to the plane of the heme, in myoglobin
resides in almost the same plane. The fifth and sixth co-
and hemoglobin the distal histidine sterically precludes
ordination positions of Fe2+ are directed perpendicular
this orientation. Binding at a less favored angle reduces
the strength of the heme-CO bond to about 200 times
to—and directly above and below—the plane of the
that of the heme-O2 bond (Figure 6 -3, right) at which
heme ring. Observe the nature of the substituent
level the great excess of O2 over CO normally present
groups on the β carbons of the pyrrole rings, the cen-
dominates. Nevertheless, about 1% of myoglobin typi-
tral iron atom, and the location of the polar side of the
cally is present combined with carbon monoxide.
heme ring (at about 7 o’clock) that faces the surface of
the myoglobin molecule.
THE OXYGEN DISSOCIATION CURVES
FOR MYOGLOBIN & HEMOGLOBIN SUIT
THEIR PHYSIOLOGIC ROLES
O
O-
FG2
CD2
Why is myoglobin unsuitable as an O2 transport pro-
C
F9
H24
tein but well suited for O2 storage? The relationship
HC5
F6
between the concentration, or partial pressure, of O2
C3
C7
CD1
F8
C5
(PO2) and the quantity of O2 bound is expressed as an
G1
CD7
O2 saturation isotherm
(Figure
6-4). The oxygen-
E7
C1
E1
F1
G5
B14
D1
B16
D7
H16
E5
E7
E7
G15
N
N
EF3
EF1
B5
O
O
B1
A16
NA1
E20
O
C
G19
+H3N
H5
AB1
Fe
Fe
A1
N
N
H1
GH4
F8
F8
Figure 6-2. A model of myoglobin at low resolution.
Only the α-carbon atoms are shown. The α-helical re-
Figure 6-3. Angles for bonding of oxygen and car-
gions are named A through H. (Based on Dickerson RE in:
bon monoxide to the heme iron of myoglobin. The dis-
The Proteins, 2nd ed. Vol 2. Neurath H [editor]. Academic
tal E7 histidine hinders bonding of CO at the preferred
Press, 1964. Reproduced with permission.)
(180 degree) angle to the plane of the heme ring.
42
/
CHAPTER 6
100
Hemoglobin Is Tetrameric
Myoglobin
Hemoglobins are tetramers comprised of pairs of two
80
Oxygenated blood
different polypeptide subunits. Greek letters are used to
leaving the lungs
designate each subunit type. The subunit composition
60
of the principal hemoglobins are α2β2 (HbA; normal
adult hemoglobin), α2γ2 (HbF; fetal hemoglobin), α2S2
Reduced blood
40
(HbS; sickle cell hemoglobin), and α2δ2
(HbA2; a
returning from tissues
minor adult hemoglobin). The primary structures of
20
the β, γ, and δ chains of human hemoglobin are highly
Hemoglobin
conserved.
0
20
40
60
80
100
120
140
Myoglobin & the Subunits
Gaseous pressure of oxygen (mm Hg)
of Hemoglobin Share Almost Identical
Secondary and Tertiary Structures
Figure 6-4. Oxygen-binding curves of both hemo-
Despite differences in the kind and number of amino
globin and myoglobin. Arterial oxygen tension is about
acids present, myoglobin and the β polypeptide of he-
100 mm Hg; mixed venous oxygen tension is about 40
moglobin A have almost identical secondary and ter-
mm Hg; capillary (active muscle) oxygen tension is
tiary structures. Similarities include the location of the
about 20 mm Hg; and the minimum oxygen tension re-
heme and the eight helical regions and the presence of
quired for cytochrome oxidase is about 5 mm Hg. Asso-
amino acids with similar properties at comparable loca-
ciation of chains into a tetrameric structure (hemoglo-
tions. Although it possesses seven rather than eight heli-
bin) results in much greater oxygen delivery than
cal regions, the α polypeptide of hemoglobin also
would be possible with single chains. (Modified, with
closely resembles myoglobin.
permission, from Scriver CR et al [editors]: The Molecular
and Metabolic Bases of Inherited Disease, 7th ed.
Oxygenation of Hemoglobin
McGraw-Hill, 1995.)
Triggers Conformational Changes
in the Apoprotein
Hemoglobins bind four molecules of O2 per tetramer,
binding curve for myoglobin is hyperbolic. Myoglobin
one per heme. A molecule of O2 binds to a hemoglobin
therefore loads O2 readily at the PO2 of the lung capil-
tetramer more readily if other O2 molecules are already
lary bed (100 mm Hg). However, since myoglobin re-
bound (Figure 6-4). Termed cooperative binding,
leases only a small fraction of its bound O2 at the PO2
this phenomenon permits hemoglobin to maximize
values typically encountered in active muscle (20 mm
both the quantity of O2 loaded at the PO2 of the lungs
Hg) or other tissues (40 mm Hg), it represents an inef-
and the quantity of O2 released at the PO2 of the pe-
fective vehicle for delivery of O2. However, when
ripheral tissues. Cooperative interactions, an exclusive
strenuous exercise lowers the PO2 of muscle tissue to
property of multimeric proteins, are critically impor-
about 5 mm Hg, myoglobin releases O2 for mitochon-
tant to aerobic life.
drial synthesis of ATP, permitting continued muscular
activity.
P50 Expresses the Relative Affinities
of Different Hemoglobins for Oxygen
THE ALLOSTERIC PROPERTIES OF
The quantity P50, a measure of O2 concentration, is the
HEMOGLOBINS RESULT FROM THEIR
partial pressure of O2 that half-saturates a given hemo-
QUATERNARY STRUCTURES
globin. Depending on the organism, P50
can vary
The properties of individual hemoglobins are conse-
widely, but in all instances it will exceed the PO2 of the
quences of their quaternary as well as of their secondary
peripheral tissues. For example, values of P50 for HbA
and tertiary structures. The quaternary structure of he-
and fetal HbF are 26 and 20 mm Hg, respectively. In
moglobin confers striking additional properties, absent
the placenta, this difference enables HbF to extract oxy-
from monomeric myoglobin, which adapts it to its
gen from the HbA in the mother’s blood. However,
unique biologic roles. The allosteric (Gk allos “other,”
HbF is suboptimal postpartum since its high affinity
steros “space”) properties of hemoglobin provide, in ad-
for O2 dictates that it can deliver less O2 to the tissues.
dition, a model for understanding other allosteric pro-
The subunit composition of hemoglobin tetramers
teins (see Chapter 11).
undergoes complex changes during development. The
PROTEINS: MYOGLOBIN & HEMOGLOBIN
/
43
human fetus initially synthesizes a ζ2ε2 tetramer. By the
Histidine F8
end of the first trimester, ζ and γ subunits have been re-
F helix
N
placed by α and ε subunits, forming HbF (α2γ2), the
C
CH
hemoglobin of late fetal life. While synthesis of β sub-
HC
units begins in the third trimester, β subunits do not
N
completely replace γ subunits to yield adult HbA (α2β2)
Steric
repulsion
until some weeks postpartum (Figure 6-5).
Fe
Porphyrin
plane
Oxygenation of Hemoglobin Is
Accompanied by Large
Conformational Changes
+O2
F helix
The binding of the first O2 molecule to deoxyHb shifts
the heme iron towards the plane of the heme ring from
C
N
a position about 0.6 nm beyond it (Figure 6-6). This
HC
CH
motion is transmitted to the proximal (F8) histidine
N
and to the residues attached thereto, which in turn
causes the rupture of salt bridges between the carboxyl
terminal residues of all four subunits. As a consequence,
Fe
one pair of α/β subunits rotates 15 degrees with respect
to the other, compacting the tetramer (Figure 6-7).
Profound changes in secondary, tertiary, and quater-
nary structure accompany the high-affinity O2-induced
transition of hemoglobin from the low-affinity T (taut)
Figure 6-6. The iron atom moves into the plane of
state to the R (relaxed) state. These changes signifi-
the heme on oxygenation. Histidine F8 and its associ-
cantly increase the affinity of the remaining unoxy-
ated residues are pulled along with the iron atom.
genated hemes for O2, as subsequent binding events re-
(Slightly modified and reproduced, with permission,
quire the rupture of fewer salt bridges (Figure 6-8).
from Stryer L: Biochemistry, 4th ed. Freeman, 1995.)
The terms T and R also are used to refer to the low-
affinity and high-affinity conformations of allosteric en-
zymes, respectively.
50
α chain
γ chain
α1
β2
α1
β2
(fetal)
40
Axis
β chain (adult)
30
α2
20
and ζ chains
α2
β1
β1
(embryonic)
10
15°
δ chain
T form
R form
0
3
6
Birth
3
6
Figure 6-7. During transition of the T form to the R
form of hemoglobin, one pair of subunits (α2/β2) ro-
Gestation (months)
Age (months)
tates through 15 degrees relative to the other pair
Figure 6-5. Developmental pattern of the quater-
(α1/β1). The axis of rotation is eccentric, and the α2/β2
nary structure of fetal and newborn hemoglobins. (Re-
pair also shifts toward the axis somewhat. In the dia-
produced, with permission, from Ganong WF: Review of
gram, the unshaded α1/β1 pair is shown fixed while the
Medical Physiology, 20th ed. McGraw-Hill, 2001.)
colored α2/β2 pair both shifts and rotates.
44
/
CHAPTER 6
T structure
α1
α2
O2
O2
O2
O2
O2
β1
β2
O2
O2
O2
O2
O2
O2
O2
O2
O2
O2
O2
R structure
Figure 6-8. Transition from the T structure to the R structure. In this model, salt
bridges (thin lines) linking the subunits in the T structure break progressively as oxy-
gen is added, and even those salt bridges that have not yet ruptured are progressively
weakened (wavy lines). The transition from T to R does not take place after a fixed
number of oxygen molecules have been bound but becomes more probable as each
successive oxygen binds. The transition between the two structures is influenced by
protons, carbon dioxide, chloride, and BPG; the higher their concentration, the more
oxygen must be bound to trigger the transition. Fully oxygenated molecules in the T
structure and fully deoxygenated molecules in the R structure are not shown because
they are unstable. (Modified and redrawn, with permission, from Perutz MF: Hemoglobin
structure and respiratory transport. Sci Am [Dec] 1978;239:92.)
After Releasing O2 at the Tissues,
Hemoglobin Transports CO2 & Protons
to the Lungs
In addition to transporting O2 from the lungs to pe-
ripheral tissues, hemoglobin transports CO2, the by-
product of respiration, and protons from peripheral tis-
Deoxyhemoglobin binds one proton for every two
sues to the lungs. Hemoglobin carries CO2
as
O2 molecules released, contributing significantly to the
carbamates formed with the amino terminal nitrogens
buffering capacity of blood. The somewhat lower pH of
of the polypeptide chains.
peripheral tissues, aided by carbamation, stabilizes the
T state and thus enhances the delivery of O2. In the
lungs, the process reverses. As O2 binds to deoxyhemo-
O
globin, protons are released and combine with bicar-
||
+
H
bonate to form carbonic acid. Dehydration of H2CO3,
+
−
CO2+ HbNH
3
=
2H
+
HbN O
catalyzed by carbonic anhydrase, forms CO2, which is
exhaled. Binding of oxygen thus drives the exhalation
of CO2 (Figure 6-9).This reciprocal coupling of proton
Carbamates change the charge on amino terminals
and O2 binding is termed the Bohr effect. The Bohr
from positive to negative, favoring salt bond formation
effect is dependent upon cooperative interactions be-
between the α and β chains.
tween the hemes of the hemoglobin tetramer. Myo-
Hemoglobin carbamates account for about 15% of
globin, a monomer, exhibits no Bohr effect.
the CO2 in venous blood. Much of the remaining CO2
is carried as bicarbonate, which is formed in erythro-
Protons Arise From Rupture of Salt Bonds
cytes by the hydration of CO2
to carbonic acid
When O2 Binds
(H2CO3), a process catalyzed by carbonic anhydrase. At
the pH of venous blood, H2CO3 dissociates into bicar-
Protons responsible for the Bohr effect arise from rup-
bonate and a proton.
ture of salt bridges during the binding of O2 to T state
PROTEINS: MYOGLOBIN & HEMOGLOBIN
/
45
Exhaled
2CO2 + 2H2O
CARBONIC
ANHYDRASE
2H2CO3
PERIPHERAL
TISSUES
2HCO3- + 2H+
Hb • 4O2
The hemoglobin tetramer binds one molecule of
4O2
BPG in the central cavity formed by its four subunits.
–
However, the space between the H helices of the β
2H+
+ 2HCO
3
chains lining the cavity is sufficiently wide to accom-
4O2
Hb • 2H+
modate BPG only when hemoglobin is in the T state.
(buffer)
BPG forms salt bridges with the terminal amino groups
2H2CO
3
of both β chains via Val NA1 and with Lys EF6 and
LUNGS
CARBONIC
ANHYDRASE
His H21 (Figure 6-10). BPG therefore stabilizes de-
oxygenated (T state) hemoglobin by forming additional
2CO2 + 2H2O
salt bridges that must be broken prior to conversion to
the R state.
Generated by
Residue H21 of the γ subunit of fetal hemoglobin
the Krebs cycle
(HbF) is Ser rather than His. Since Ser cannot form a
Figure 6-9. The Bohr effect. Carbon dioxide gener-
salt bridge, BPG binds more weakly to HbF than to
ated in peripheral tissues combines with water to form
HbA. The lower stabilization afforded to the T state by
carbonic acid, which dissociates into protons and bicar-
BPG accounts for HbF having a higher affinity for O2
bonate ions. Deoxyhemoglobin acts as a buffer by
than HbA.
binding protons and delivering them to the lungs. In
the lungs, the uptake of oxygen by hemoglobin re-
leases protons that combine with bicarbonate ion,
forming carbonic acid, which when dehydrated by car-
bonic anhydrase becomes carbon dioxide, which then
is exhaled.
His H21
Lys EF6
hemoglobin. Conversion to the oxygenated R state
breaks salt bridges involving β-chain residue His 146.
BPG
Val NA1
The subsequent dissociation of protons from His 146
+
Val NA1
α-NH3
drives the conversion of bicarbonate to carbonic acid
(Figure 6-9). Upon the release of O2, the T structure
and its salt bridges re-form. This conformational
Lys EF6
change increases the pKa of the β-chain His
146
residues, which bind protons. By facilitating the re-for-
mation of salt bridges, an increase in proton concentra-
tion enhances the release of O2 from oxygenated (R
His H21
state) hemoglobin. Conversely, an increase in PO2 pro-
motes proton release.
Figure 6-10. Mode of binding of 2,3-bisphospho-
2,3-Bisphosphoglycerate (BPG) Stabilizes
glycerate to human deoxyhemoglobin. BPG interacts
the T Structure of Hemoglobin
with three positively charged groups on each β chain.
A low PO2 in peripheral tissues promotes the synthesis
(Based on Arnone A: X-ray diffraction study of binding of
in erythrocytes of 2,3-bisphosphoglycerate (BPG) from
2,3-diphosphoglycerate to human deoxyhemoglobin. Na-
the glycolytic intermediate 1,3-bisphosphoglycerate.
ture 1972;237:146. Reproduced with permission.)
46
/
CHAPTER 6
Adaptation to High Altitude
In hemoglobin M, histidine F8 (His F8) has been
replaced by tyrosine. The iron of HbM forms a tight
Physiologic changes that accompany prolonged expo-
ionic complex with the phenolate anion of tyrosine that
sure to high altitude include an increase in the number
stabilizes the Fe3+ form. In α-chain hemoglobin M vari-
of erythrocytes and in their concentrations of hemoglo-
ants, the R-T equilibrium favors the T state. Oxygen
bin and of BPG. Elevated BPG lowers the affinity of
affinity is reduced, and the Bohr effect is absent.
HbA for O2 (decreases P50), which enhances release of
β-Chain hemoglobin M variants exhibit R-T switching,
O2 at the tissues.
and the Bohr effect is therefore present.
Mutations (eg, hemoglobin Chesapeake) that favor
NUMEROUS MUTANT HUMAN
the R state increase O2 affinity. These hemoglobins
HEMOGLOBINS HAVE BEEN IDENTIFIED
therefore fail to deliver adequate O2 to peripheral tis-
sues. The resulting tissue hypoxia leads to poly-
Mutations in the genes that encode the α or β subunits
cythemia, an increased concentration of erythrocytes.
of hemoglobin potentially can affect its biologic func-
tion. However, almost all of the over 800 known mu-
tant human hemoglobins are both extremely rare and
Hemoglobin S
benign, presenting no clinical abnormalities. When a
In HbS, the nonpolar amino acid valine has replaced
mutation does compromise biologic function, the con-
the polar surface residue Glu6 of the β subunit, gener-
dition is termed a hemoglobinopathy. The URL
ating a hydrophobic “sticky patch” on the surface of
(Globin Gene Server) pro-
the β subunit of both oxyHbS and deoxyHbS. Both
vides information about—and links for—normal and
HbA and HbS contain a complementary sticky patch
mutant hemoglobins.
on their surfaces that is exposed only in the deoxy-
genated, R state. Thus, at low PO2, deoxyHbS can poly-
Methemoglobin & Hemoglobin M
merize to form long, insoluble fibers. Binding of deoxy-
In methemoglobinemia, the heme iron is ferric rather
HbA terminates fiber polymerization, since HbA lacks
than ferrous. Methemoglobin thus can neither bind nor
the second sticky patch necessary to bind another Hb
transport O2. Normally, the enzyme methemoglobin
molecule (Figure 6-11). These twisted helical fibers
reductase reduces the Fe3+ of methemoglobin to Fe2+.
distort the erythrocyte into a characteristic sickle shape,
+
Methemoglobin can arise by oxidation of Fe2+
to Fe3
rendering it vulnerable to lysis in the interstices of the
as a side effect of agents such as sulfonamides, from
splenic sinusoids. They also cause multiple secondary
hereditary hemoglobin M, or consequent to reduced
clinical effects. A low PO2 such as that at high altitudes
activity of the enzyme methemoglobin reductase.
exacerbates the tendency to polymerize.
Oxy A
Deoxy A
Oxy S
Deoxy S
β
α
α
β
Deoxy A Deoxy S
Figure 6-11. Representation of the sticky patch (
) on hemoglobin S and its “receptor” (
)
on deoxyhemoglobin A and deoxyhemoglobin S. The complementary surfaces allow deoxyhe-
moglobin S to polymerize into a fibrous structure, but the presence of deoxyhemoglobin A will
terminate the polymerization by failing to provide sticky patches. (Modified and reproduced, with
permission, from Stryer L: Biochemistry, 4th ed. Freeman, 1995.)
PROTEINS: MYOGLOBIN & HEMOGLOBIN
/
47
BIOMEDICAL IMPLICATIONS
different primary structures, myoglobin and the sub-
units of hemoglobin have nearly identical secondary
Myoglobinuria
and tertiary structures.
Following massive crush injury, myoglobin released
•
Heme, an essentially planar, slightly puckered, cyclic
from damaged muscle fibers colors the urine dark red.
tetrapyrrole, has a central Fe2+ linked to all four ni-
Myoglobin can be detected in plasma following a my-
trogen atoms of the heme, to histidine F8, and, in
ocardial infarction, but assay of serum enzymes (see
oxyMb and oxyHb, also to O2.
Chapter 7) provides a more sensitive index of myocar-
•
The O2-binding curve for myoglobin is hyperbolic,
dial injury.
but for hemoglobin it is sigmoidal, a consequence of
cooperative interactions in the tetramer. Cooperativ-
ity maximizes the ability of hemoglobin both to load
Anemias
O2 at the PO2 of the lungs and to deliver O2 at the
Anemias, reductions in the number of red blood cells or
PO2 of the tissues.
of hemoglobin in the blood, can reflect impaired syn-
•
Relative affinities of different hemoglobins for oxy-
thesis of hemoglobin (eg, in iron deficiency; Chapter
gen are expressed as P50, the PO2 that half-saturates
51) or impaired production of erythrocytes (eg, in folic
them with O2. Hemoglobins saturate at the partial
acid or vitamin B12 deficiency; Chapter 45). Diagnosis
pressures of their respective respiratory organ, eg, the
of anemias begins with spectroscopic measurement of
lung or placenta.
blood hemoglobin levels.
•
On oxygenation of hemoglobin, the iron, histidine
F8, and linked residues move toward the heme ring.
Thalassemias
Conformational changes that accompany oxygena-
tion include rupture of salt bonds and loosening of
The genetic defects known as thalassemias result from
quaternary structure, facilitating binding of addi-
the partial or total absence of one or more α or β chains
tional O2.
of hemoglobin. Over 750 different mutations have
•
2,3-Bisphosphoglycerate (BPG) in the central cavity
been identified, but only three are common. Either the
of deoxyHb forms salt bonds with the β subunits
α chain (alpha thalassemias) or β chain (beta thal-
that stabilize deoxyHb. On oxygenation, the central
assemias) can be affected. A superscript indicates
cavity contracts, BPG is extruded, and the quaternary
whether a subunit is completely absent (α0 or β0) or
structure loosens.
whether its synthesis is reduced (α+ or β+). Apart from
•
Hemoglobin also functions in CO2
and proton
marrow transplantation, treatment is symptomatic.
transport from tissues to lungs. Release of O2 from
Certain mutant hemoglobins are common in many
oxyHb at the tissues is accompanied by uptake of
populations, and a patient may inherit more than one
protons due to lowering of the pKa of histidine
type. Hemoglobin disorders thus present a complex
residues.
pattern of clinical phenotypes. The use of DNA probes
for their diagnosis is considered in Chapter 40.
•
In sickle cell hemoglobin (HbS), Val replaces the β6
Glu of HbA, creating a “sticky patch” that has a
complement on deoxyHb (but not on oxyHb). De-
Glycosylated Hemoglobin (HbA1c)
oxyHbS polymerizes at low O2
concentrations,
When blood glucose enters the erythrocytes it glycosy-
forming fibers that distort erythrocytes into sickle
lates the ε-amino group of lysine residues and the
shapes.
amino terminals of hemoglobin. The fraction of hemo-
•
Alpha and beta thalassemias are anemias that result
globin glycosylated, normally about 5%, is proportion-
from reduced production of α and β subunits of
ate to blood glucose concentration. Since the half-life of
HbA, respectively.
an erythrocyte is typically 60 days, the level of glycosy-
lated hemoglobin (HbA1c) reflects the mean blood glu-
REFERENCES
cose concentration over the preceding
6-8 weeks.
Measurement of HbA1c therefore provides valuable in-
Bettati S et al: Allosteric mechanism of haemoglobin: Rupture of
salt-bridges raises the oxygen affinity of the T-structure. J
formation for management of diabetes mellitus.
Mol Biol 1998;281:581.
Bunn HF: Pathogenesis and treatment of sickle cell disease. N Engl
SUMMARY
J Med 1997;337:762.
Faustino P et al: Dominantly transmitted beta-thalassemia arising
• Myoglobin is monomeric; hemoglobin is a tetramer
from the production of several aberrant mRNA species and
of two subunit types (α2β2 in HbA). Despite having
one abnormal peptide. Blood 1998;91:685.
48
/
CHAPTER 6
Manning JM et al: Normal and abnormal protein subunit interac-
Unzai S et al: Rate constants for O2 and CO binding to the alpha
tions in hemoglobins. J Biol Chem 1998;273:19359.
and beta subunits within the R and T states of human hemo-
Mario N, Baudin B, Giboudeau J: Qualitative and quantitative
globin. J Biol Chem 1998;273:23150.
analysis of hemoglobin variants by capillary isoelectric focus-
Weatherall DJ et al: The hemoglobinopathies. Chapter 181 in The
ing. J Chromatogr B Biomed Sci Appl 1998;706:123.
Metabolic and Molecular Bases of Inherited Disease, 8th ed.
Reed W, Vichinsky EP: New considerations in the treatment of
Scriver CR et al (editors). McGraw-Hill, 2000.
sickle cell disease. Annu Rev Med 1998;49:461.
Enzymes: Mechanism of Action
7
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
with the ability to simultaneously conduct and inde-
pendently control a broad spectrum of chemical
Enzymes are biologic polymers that catalyze the chemi-
processes.
cal reactions which make life as we know it possible.
The presence and maintenance of a complete and bal-
anced set of enzymes is essential for the breakdown of
ENZYMES ARE CLASSIFIED BY REACTION
nutrients to supply energy and chemical building
TYPE & MECHANISM
blocks; the assembly of those building blocks into pro-
A system of enzyme nomenclature that is comprehen-
teins, DNA, membranes, cells, and tissues; and the har-
sive, consistent, and at the same time easy to use has
nessing of energy to power cell motility and muscle
proved elusive. The common names for most enzymes
contraction. With the exception of a few catalytic RNA
derive from their most distinctive characteristic: their
molecules, or ribozymes, the vast majority of enzymes
ability to catalyze a specific chemical reaction. In gen-
are proteins. Deficiencies in the quantity or catalytic ac-
eral, an enzyme’s name consists of a term that identifies
tivity of key enzymes can result from genetic defects,
the type of reaction catalyzed followed by the suffix
nutritional deficits, or toxins. Defective enzymes can re-
-ase. For example, dehydrogenases remove hydrogen
sult from genetic mutations or infection by viral or bac-
atoms, proteases hydrolyze proteins, and isomerases cat-
terial pathogens (eg, Vibrio cholerae). Medical scientists
alyze rearrangements in configuration. One or more
address imbalances in enzyme activity by using pharma-
modifiers usually precede this name. Unfortunately,
cologic agents to inhibit specific enzymes and are inves-
while many modifiers name the specific substrate in-
tigating gene therapy as a means to remedy deficits in
volved (xanthine oxidase), others identify the source of
enzyme level or function.
the enzyme (pancreatic ribonuclease), specify its mode
of regulation (hormone-sensitive lipase), or name a dis-
ENZYMES ARE EFFECTIVE & HIGHLY
tinguishing characteristic of its mechanism (a cysteine
SPECIFIC CATALYSTS
protease). When it was discovered that multiple forms
of some enzymes existed, alphanumeric designators
The enzymes that catalyze the conversion of one or
were added to distinguish between them (eg, RNA
more compounds (substrates) into one or more differ-
polymerase III; protein kinase Cβ). To address the am-
ent compounds (products) enhance the rates of the
biguity and confusion arising from these inconsistencies
corresponding noncatalyzed reaction by factors of at
in nomenclature and the continuing discovery of new
least 106. Like all catalysts, enzymes are neither con-
enzymes, the International Union of Biochemists (IUB)
sumed nor permanently altered as a consequence of
developed a complex but unambiguous system of en-
their participation in a reaction.
zyme nomenclature. In the IUB system, each enzyme
In addition to being highly efficient, enzymes are
has a unique name and code number that reflect the
also extremely selective catalysts. Unlike most catalysts
type of reaction catalyzed and the substrates involved.
used in synthetic chemistry, enzymes are specific both
Enzymes are grouped into six classes, each with several
for the type of reaction catalyzed and for a single sub-
subclasses. For example, the enzyme commonly called
strate or a small set of closely related substrates. En-
“hexokinase” is designated “ATP:D-hexose-6-phospho-
zymes are also stereospecific catalysts and typically cat-
transferase E.C. 2.7.1.1.” This identifies hexokinase as a
alyze reactions only of specific stereoisomers of a given
member of class 2 (transferases), subclass 7 (transfer of a
compound—for example, D- but not L-sugars, L- but
phosphoryl group), sub-subclass 1 (alcohol is the phos-
not D-amino acids. Since they bind substrates through
phoryl acceptor). Finally, the term “hexose-6” indicates
at least “three points of attachment,” enzymes can even
that the alcohol phosphorylated is that of carbon six of
convert nonchiral substrates to chiral products. Figure
a hexose. Listed below are the six IUB classes of en-
7-1 illustrates why the enzyme-catalyzed reduction of
zymes and the reactions they catalyze.
the nonchiral substrate pyruvate produces L-lactate
rather a racemic mixture of D- and L-lactate. The ex-
1. Oxidoreductases catalyze oxidations and reduc-
quisite specificity of enzyme catalysts imbues living cells
tions.
49
50
/
CHAPTER 7
4
Prosthetic Groups Are Tightly Integrated
Into an Enzyme’s Structure
Prosthetic groups are distinguished by their tight, stable
1
3
1
incorporation into a protein’s structure by covalent or
noncovalent forces. Examples include pyridoxal phos-
3
phate, flavin mononucleotide
(FMN), flavin dinu-
cleotide
(FAD), thiamin pyrophosphate, biotin, and
2
2
the metal ions of Co, Cu, Mg, Mn, Se, and Zn. Metals
are the most common prosthetic groups. The roughly
Enzyme site
Substrate
one-third of all enzymes that contain tightly bound
metal ions are termed metalloenzymes. Metal ions that
Figure 7-1. Planar representation of the “three-
participate in redox reactions generally are complexed
point attachment” of a substrate to the active site of an
to prosthetic groups such as heme (Chapter 6) or iron-
enzyme. Although atoms 1 and 4 are identical, once
sulfur clusters (Chapter 12). Metals also may facilitate
atoms 2 and 3 are bound to their complementary sites
the binding and orientation of substrates, the formation
on the enzyme, only atom 1 can bind. Once bound to
of covalent bonds with reaction intermediates (Co2+ in
an enzyme, apparently identical atoms thus may be dis-
coenzyme B12 ), or interaction with substrates to render
tinguishable, permitting a stereospecific chemical
them more electrophilic (electron-poor) or nucleo-
change.
philic (electron-rich).
Cofactors Associate Reversibly With
2. Transferases catalyze transfer of groups such as
Enzymes or Substrates
methyl or glycosyl groups from a donor molecule
Cofactors serve functions similar to those of prosthetic
to an acceptor molecule.
groups but bind in a transient, dissociable manner ei-
3. Hydrolases catalyze the hydrolytic cleavage of
ther to the enzyme or to a substrate such as ATP. Un-
C C, C O, CN, P O, and certain other
like the stably associated prosthetic groups, cofactors
bonds, including acid anhydride bonds.
therefore must be present in the medium surrounding
4. Lyases catalyze cleavage of C C, C O, CN,
the enzyme for catalysis to occur. The most common
and other bonds by elimination, leaving double
cofactors also are metal ions. Enzymes that require a
bonds, and also add groups to double bonds.
metal ion cofactor are termed metal-activated enzymes
5. Isomerases catalyze geometric or structural
to distinguish them from the metalloenzymes for
changes within a single molecule.
which metal ions serve as prosthetic groups.
6. Ligases catalyze the joining together of two mole-
cules, coupled to the hydrolysis of a pyrophospho-
Coenzymes Serve as Substrate Shuttles
ryl group in ATP or a similar nucleoside triphos-
phate.
Coenzymes serve as recyclable shuttles—or group
transfer reagents—that transport many substrates from
Despite the many advantages of the IUB system,
their point of generation to their point of utilization.
texts tend to refer to most enzymes by their older and
Association with the coenzyme also stabilizes substrates
shorter, albeit sometimes ambiguous names.
such as hydrogen atoms or hydride ions that are unsta-
ble in the aqueous environment of the cell. Other
chemical moieties transported by coenzymes include
methyl groups (folates), acyl groups (coenzyme A), and
PROSTHETIC GROUPS, COFACTORS,
oligosaccharides (dolichol).
& COENZYMES PLAY IMPORTANT
ROLES IN CATALYSIS
Many Coenzymes, Cofactors, & Prosthetic
Many enzymes contain small nonprotein molecules and
Groups Are Derivatives of B Vitamins
metal ions that participate directly in substrate binding
or catalysis. Termed prosthetic groups, cofactors, and
The water-soluble B vitamins supply important compo-
coenzymes, these extend the repertoire of catalytic ca-
nents of numerous coenzymes. Many coenzymes con-
pabilities beyond those afforded by the limited number
tain, in addition, the adenine, ribose, and phosphoryl
of functional groups present on the aminoacyl side
moieties of AMP or ADP (Figure 7-2). Nicotinamide
chains of peptides.
and riboflavin are components of the redox coenzymes
ENZYMES: MECHANISM OF ACTION
/
51
O
Arg 145
NH
NH2
+
OH
C
N
NH2
NH2
O
CH2
O
H
H
C O
O
O
N
H
C
C
N
H
Tyr 248
H H
H
N
HO OH
His 196
C
O
P
O-
O
CH2
Zn2+
NH
2
NH2
O
O
His 69
C
N
N
N
Glu 72
N
O
N
N
H
O
P
O
CH2
Figure 7-3. Two-dimensional representation of a
O-
O
dipeptide substrate, glycyl-tyrosine, bound within the
active site of carboxypeptidase A.
H H
HO
OR
Figure 7-2.
Structure of NAD+ and NADP+. For
tribute to the extensive size and three-dimensional char-
NAD+, R = H. For NADP+, R = PO32−.
acter of the active site.
ENZYMES EMPLOY MULTIPLE
NAD and NADP and FMN and FAD, respectively.
MECHANISMS TO FACILITATE
Pantothenic acid is a component of the acyl group car-
CATALYSIS
rier coenzyme A. As its pyrophosphate, thiamin partici-
Four general mechanisms account for the ability of en-
pates in decarboxylation of α-keto acids and folic acid
zymes to achieve dramatic catalytic enhancement of the
and cobamide coenzymes function in one-carbon me-
rates of chemical reactions.
tabolism.
Catalysis by Proximity
CATALYSIS OCCURS AT THE ACTIVE SITE
For molecules to react, they must come within bond-
The extreme substrate specificity and high catalytic effi-
forming distance of one another. The higher their con-
ciency of enzymes reflect the existence of an environ-
centration, the more frequently they will encounter one
ment that is exquisitely tailored to a single reaction.
another and the greater will be the rate of their reaction.
Termed the active site, this environment generally
When an enzyme binds substrate molecules in its active
takes the form of a cleft or pocket. The active sites of
site, it creates a region of high local substrate concentra-
multimeric enzymes often are located at the interface
tion. This environment also orients the substrate mole-
between subunits and recruit residues from more than
cules spatially in a position ideal for them to interact, re-
one monomer. The three-dimensional active site both
sulting in rate enhancements of at least a thousandfold.
shields substrates from solvent and facilitates catalysis.
Substrates bind to the active site at a region comple-
Acid-Base Catalysis
mentary to a portion of the substrate that will not un-
dergo chemical change during the course of the reac-
The ionizable functional groups of aminoacyl side
tion. This simultaneously aligns portions of the
chains and (where present) of prosthetic groups con-
substrate that will undergo change with the chemical
tribute to catalysis by acting as acids or bases. Acid-base
functional groups of peptidyl aminoacyl residues. The
catalysis can be either specific or general. By “specific”
active site also binds and orients cofactors or prosthetic
we mean only protons (H3O+) or OH- ions. In specific
groups. Many amino acyl residues drawn from diverse
acid or specific base catalysis, the rate of reaction is
portions of the polypeptide chain (Figure 7-3) con-
sensitive to changes in the concentration of protons but
52
/
CHAPTER 7
independent of the concentrations of other acids (pro-
enzyme’s active site failed to account for the dynamic
ton donors) or bases (proton acceptors) present in solu-
changes that accompany catalysis. This drawback was
tion or at the active site. Reactions whose rates are re-
addressed by Daniel Koshland’s induced fit model,
sponsive to all the acids or bases present are said to be
which states that when substrates approach and bind to
subject to general acid or general base catalysis.
an enzyme they induce a conformational change, a
change analogous to placing a hand (substrate) into a
Catalysis by Strain
glove (enzyme) (Figure 7-5). A corollary is that the en-
zyme induces reciprocal changes in its substrates, har-
Enzymes that catalyze lytic reactions which involve
nessing the energy of binding to facilitate the transfor-
breaking a covalent bond typically bind their substrates
mation of substrates into products. The induced fit
in a conformation slightly unfavorable for the bond
model has been amply confirmed by biophysical studies
that will undergo cleavage. The resulting strain
of enzyme motion during substrate binding.
stretches or distorts the targeted bond, weakening it
and making it more vulnerable to cleavage.
HIV PROTEASE ILLUSTRATES
ACID-BASE CATALYSIS
Covalent Catalysis
Enzymes of the aspartic protease family, which in-
The process of covalent catalysis involves the formation
cludes the digestive enzyme pepsin, the lysosomal
of a covalent bond between the enzyme and one or more
cathepsins, and the protease produced by the human im-
substrates. The modified enzyme then becomes a reac-
munodeficiency virus (HIV), share a common catalytic
tant. Covalent catalysis introduces a new reaction path-
mechanism. Catalysis involves two conserved aspartyl
way that is energetically more favorable—and therefore
residues which act as acid-base catalysts. In the first stage
faster—than the reaction pathway in homogeneous so-
of the reaction, an aspartate functioning as a general base
lution. The chemical modification of the enzyme is,
(Asp X, Figure 7-6) extracts a proton from a water mole-
however, transient. On completion of the reaction, the
cule, making it more nucleophilic. This resulting nucle-
enzyme returns to its original unmodified state. Its role
ophile then attacks the electrophilic carbonyl carbon of
thus remains catalytic. Covalent catalysis is particularly
the peptide bond targeted for hydrolysis, forming a
common among enzymes that catalyze group transfer
tetrahedral transition state intermediate. A second as-
reactions. Residues on the enzyme that participate in co-
partate (Asp Y, Figure 7-6) then facilitates the decompo-
valent catalysis generally are cysteine or serine and occa-
sition of this tetrahedral intermediate by donating a pro-
sionally histidine. Covalent catalysis often follows a
ton to the amino group produced by rupture of the
“ping-pong” mechanism—one in which the first sub-
peptide bond. Two different active site aspartates thus
strate is bound and its product released prior to the
can act simultaneously as a general base or as a general
binding of the second substrate (Figure 7-4).
acid. This is possible because their immediate environ-
ment favors ionization of one but not the other.
SUBSTRATES INDUCE
CONFORMATIONAL CHANGES
CHYMOTRYPSIN & FRUCTOSE-2,6-
IN ENZYMES
BISPHOSPHATASE ILLUSTRATE
Early in the last century, Emil Fischer compared the
COVALENT CATALYSIS
highly specific fit between enzymes and their substrates
Chymotrypsin
to that of a lock and its key. While the “lock and key
model” accounted for the exquisite specificity of en-
While catalysis by aspartic proteases involves the direct
zyme-substrate interactions, the implied rigidity of the
hydrolytic attack of water on a peptide bond, catalysis
Pyr
Glu
Ala
CHO
CH2NH2
KG
CH2NH2
CHO
E CHO
E
E
E CH2NH2
E
E
E CHO
Ala
Pyr
KG
Glu
Figure 7-4. Ping-pong mechanism for transamination. ECHO and ECH2NH2 represent the enzyme-
pyridoxal phosphate and enzyme-pyridoxamine complexes, respectively. (Ala, alanine; Pyr, pyruvate; KG,
α-ketoglutarate; Glu, glutamate).
ENZYMES: MECHANISM OF ACTION
/
53
O
R′
N
C
R
A
B
H
H
O
1
H
O
O
O
O
H
C
C
CH2
CH2
A
Asp X
Asp Y
O
R′
N
C
R
2
H
OH
A
H
O
O
O
O
C
C
CH2
CH2
Asp Y
Asp X
Figure 7-5.
Two-dimensional representation of
Koshland’s induced fit model of the active site of a
O
R′
lyase. Binding of the substrate AB induces conforma-
N
H
+
C
R
tional changes in the enzyme that aligns catalytic
H
HO
residues which participate in catalysis and strains the
bond between A and B, facilitating its cleavage.
3
O
O
O
O
C
C
by the serine protease chymotrypsin involves prior for-
mation of a covalent acyl enzyme intermediate. A
CH2
CH2
highly reactive seryl residue, serine 195, participates in
Asp Y
Asp X
a charge-relay network with histidine 57 and aspartate
102. Far apart in primary structure, in the active site
Figure 7-6.
Mechanism for catalysis by an aspartic
these residues are within bond-forming distance of one
protease such as HIV protease. Curved arrows indicate
another. Aligned in the order Asp 102-His 57-Ser 195,
directions of electron movement. 1
Aspartate X acts
they constitute a “charge-relay network” that functions
as a base to activate a water molecule by abstracting a
as a “proton shuttle.”
proton. 2
The activated water molecule attacks the
Binding of substrate initiates proton shifts that in ef-
peptide bond, forming a transient tetrahedral interme-
fect transfer the hydroxyl proton of Ser 195 to Asp 102
diate.
3 Aspartate Y acts as an acid to facilitate break-
(Figure 7-7). The enhanced nucleophilicity of the seryl
down of the tetrahedral intermediate and release of the
oxygen facilitates its attack on the carbonyl carbon of
split products by donating a proton to the newly
the peptide bond of the substrate, forming a covalent
formed amino group. Subsequent shuttling of the pro-
acyl-enzyme intermediate. The hydrogen on Asp 102
ton on Asp X to Asp Y restores the protease to its initial
then shuttles through His 57 to the amino group liber-
state.
ated when the peptide bond is cleaved. The portion of
the original peptide with a free amino group then leaves
the active site and is replaced by a water molecule. The
charge-relay network now activates the water molecule
by withdrawing a proton through His 57 to Asp 102.
The resulting hydroxide ion attacks the acyl-enzyme in-
54
/
CHAPTER 7
H
O
termediate and a reverse proton shuttle returns a proton
to Ser 195, restoring its original state. While modified
R1
N
C
R2
during the process of catalysis, chymotrypsin emerges
unchanged on completion of the reaction. Trypsin and
1
O
O
H
N N H O
elastase employ a similar catalytic mechanism, but the
C
Ser 195
numbers of the residues in their Ser-His-Asp proton
Asp 102
His 57
shuttles differ.
H
O
Fructose-2,6-Bisphosphatase
R1
N
C
R2
Fructose-2,6-bisphosphatase, a regulatory enzyme of
gluconeogenesis (Chapter 19), catalyzes the hydrolytic
2
O
O
H
N N H O
release of the phosphate on carbon 2 of fructose 2,6-
Ser 195
bisphosphate. Figure 7-8 illustrates the roles of seven
Asp 102
His 57
active site residues. Catalysis involves a “catalytic triad”
O
of one Glu and two His residues and a covalent phos-
R1
NH2
C
R2
phohistidyl intermediate.
3
O
O
H
N N
O
CATALYTIC RESIDUES ARE
Ser 195
HIGHLY CONSERVED
Asp 102
His 57
Members of an enzyme family such as the aspartic or
O
H
serine proteases employ a similar mechanism to catalyze
C
R2
a common reaction type but act on different substrates.
O
O
Enzyme families appear to arise through gene duplica-
4
O
O
H
N N
H
Ser 195
tion events that create a second copy of the gene which
encodes a particular enzyme. The proteins encoded by
Asp 102
His 57
the two genes can then evolve independently to recog-
nize different substrates—resulting, for example, in
O
chymotrypsin, which cleaves peptide bonds on the car-
H
O
C
R2
boxyl terminal side of large hydrophobic amino acids;
and trypsin, which cleaves peptide bonds on the car-
5
O
O
H
N N H O
boxyl terminal side of basic amino acids. The common
Ser 195
ancestry of enzymes can be inferred from the presence
Asp 102
His 57
of specific amino acids in the same position in each
HOOC
R2
family member. These residues are said to be conserved
residues. Proteins that share a large number of con-
served residues are said to be homologous to one an-
6
O
O
H
N N H O
other. Table 7-1 illustrates the primary structural con-
Ser 195
servation of two components of the charge-relay
Asp 102
His 57
network for several serine proteases. Among the most
Figure 7-7. Catalysis by chymotrypsin.
1 The
highly conserved residues are those that participate di-
charge-relay system removes a proton from Ser 195,
rectly in catalysis.
making it a stronger nucleophile. 2 Activated Ser 195
attacks the peptide bond, forming a transient tetrahedral
ISOZYMES ARE DISTINCT ENZYME
intermediate. 3
Release of the amino terminal peptide
FORMS THAT CATALYZE THE
is facilitated by donation of a proton to the newly
SAME REACTION
formed amino group by His 57 of the charge-relay sys-
tem, yielding an acyl-Ser 195 intermediate. 4 His 57 and
Higher organisms often elaborate several physically dis-
Asp 102 collaborate to activate a water molecule, which
tinct versions of a given enzyme, each of which cat-
attacks the acyl-Ser 195, forming a second tetrahedral in-
alyzes the same reaction. Like the members of other
termediate. 5 The charge-relay system donates a pro-
protein families, these protein catalysts or isozymes
ton to Ser 195, facilitating breakdown of tetrahedral in-
arise through gene duplication. Isozymes may exhibit
termediate to release the carboxyl terminal peptide 6 .
subtle differences in properties such as sensitivity to
ENZYMES: MECHANISM OF ACTION
/
55
particular regulatory factors (Chapter 9) or substrate
Lys 356
Lys 356
affinity
(eg, hexokinase and glucokinase) that adapt
Arg
Arg
+
352
+
352
them to specific tissues or circumstances. Some iso-
P
+
P
+
zymes may also enhance survival by providing a “back-
6-
6-
up” copy of an essential enzyme.
2-
2-
Arg 307
Arg 307
-
O
-
O H
Glu
+
Glu
+
+ H
P
P
327
327
+
+
THE CATALYTIC ACTIVITY OF ENZYMES
His
His
392
392
FACILITATES THEIR DETECTION
Arg 257
His 258
1
Arg 257
His 258
2
The minute quantities of enzymes present in cells com-
E • Fru-2,6-P2
E-P • Fru-6-P
plicate determination of their presence and concentra-
tion. However, the ability to rapidly transform thou-
sands of molecules of a specific substrate into products
Lys 356
Lys 356
imbues each enzyme with the ability to reveal its pres-
Arg
Arg
+
352
+
352
ence. Assays of the catalytic activity of enzymes are fre-
+
+
quently used in research and clinical laboratories.
Under appropriate conditions (see Chapter 8), the rate
Arg 307
Arg 307
of the catalytic reaction being monitored is proportion-
–
-
Glu
+
Glu
+
ate to the amount of enzyme present, which allows its
+ H
P
+
Pi
327
327
+
+
concentration to be inferred.
His
His
392
392
Arg 257
His 258
3
Arg 257
His 258
4
Enzyme-Linked Immunoassays
E-P • H2O
E • Pi
The sensitivity of enzyme assays can also be exploited to
Figure 7-8. Catalysis by fructose-2,6-bisphos-
detect proteins that lack catalytic activity. Enzyme-
phatase. (1) Lys 356 and Arg 257, 307, and 352 stabilize
linked immunoassays (ELISAs) use antibodies cova-
the quadruple negative charge of the substrate by
lently linked to a “reporter enzyme” such as alkaline
phosphatase or horseradish peroxidase, enzymes whose
charge-charge interactions. Glu 327 stabilizes the posi-
products are readily detected. When serum or other
tive charge on His 392. (2) The nucleophile His 392 at-
samples to be tested are placed in a plastic microtiter
tacks the C-2 phosphoryl group and transfers it to His
plate, the proteins adhere to the plastic surface and are
258, forming a phosphoryl-enzyme intermediate. Fruc-
immobilized. Any remaining absorbing areas of the well
tose 6-phosphate leaves the enzyme. (3) Nucleophilic
are then “blocked” by adding a nonantigenic protein
attack by a water molecule, possibly assisted by Glu 327
such as bovine serum albumin. A solution of antibody
acting as a base, forms inorganic phosphate. (4) Inor-
covalently linked to a reporter enzyme is then added.
ganic orthophosphate is released from Arg 257 and Arg
The antibodies adhere to the immobilized antigen and
307. (Reproduced, with permission, from Pilkis SJ et al: 6-
these are themselves immobilized. Excess free antibody
Phosphofructo-2-kinase/fructose-2,6-bisphosphatase: A
molecules are then removed by washing. The presence
metabolic signaling enzyme. Annu Rev Biochem
and quantity of bound antibody are then determined
1995;64:799.)
by adding the substrate for the reporter enzyme.
Table 7-1. Amino acid sequences in the neighborhood of the catalytic sites of several
bovine proteases. Regions shown are those on either side of the catalytic site seryl (S) and
histidyl (H) residues.
Enzyme
Sequence Around Serine S
Sequence Around Histidine H
Trypsin
D S C Q D G S G G P V V C S G K
V V S A A H C Y K S G
Chymotrypsin A
S S C M G D S G G P L V C K K N
V V T A A H G G V T T
Chymotrypsin B
S S C M G D S G G P L V C Q K N
V V T A A H C G V T T
Thrombin
D A C E G D S G G P F V M K S P
V L T A A H C L L Y P
56
/
CHAPTER 7
tated by the use of radioactive substrates. An alternative
NAD(P)+-Dependent Dehydrogenases Are
strategy is to devise a synthetic substrate whose product
Assayed Spectrophotometrically
absorbs light. For example, p-nitrophenyl phosphate is
The physicochemical properties of the reactants in an
an artificial substrate for certain phosphatases and for
enzyme-catalyzed reaction dictate the options for the
chymotrypsin that does not absorb visible light. How-
assay of enzyme activity. Spectrophotometric assays ex-
ever, following hydrolysis, the resulting p-nitrophen-
ploit the ability of a substrate or product to absorb
ylate anion absorbs light at 419 nm.
light. The reduced coenzymes NADH and NADPH,
Another quite general approach is to employ a “cou-
written as NAD(P)H, absorb light at a wavelength of
pled” assay (Figure 7-10). Typically, a dehydrogenase
340 nm, whereas their oxidized forms NAD(P)+ do not
whose substrate is the product of the enzyme of interest
(Figure
7-9). When NAD(P)+ is reduced, the ab-
is added in catalytic excess. The rate of appearance or
sorbance at 340 nm therefore increases in proportion
disappearance of NAD(P)H then depends on the rate
to—and at a rate determined by—the quantity of
of the enzyme reaction to which the dehydrogenase has
NAD(P)H produced. Conversely, for a dehydrogenase
been coupled.
that catalyzes the oxidation of NAD(P)H, a decrease in
absorbance at 340 nm will be observed. In each case,
the rate of change in optical density at 340 nm will be
THE ANALYSIS OF CERTAIN ENZYMES
proportionate to the quantity of enzyme present.
AIDS DIAGNOSIS
Of the thousands of different enzymes present in the
Many Enzymes Are Assayed by Coupling
human body, those that fulfill functions indispensable
to a Dehydrogenase
to cell vitality are present throughout the body tissues.
The assay of enzymes whose reactions are not accompa-
Other enzymes or isozymes are expressed only in spe-
nied by a change in absorbance or fluorescence is gener-
cific cell types, during certain periods of development,
ally more difficult. In some instances, the product or re-
or in response to specific physiologic or pathophysio-
maining substrate can be transformed into a more
logic changes. Analysis of the presence and distribution
readily detected compound. In other instances, the re-
of enzymes and isozymes—whose expression is nor-
action product may have to be separated from unre-
mally tissue-, time-, or circumstance-specific—often
acted substrate prior to measurement—a process facili-
aids diagnosis.
1.0
Glucose
ATP, Mg2+
0.8
HEXOKINASE
ADP, Mg2+
0.6
Glucose 6-phosphate
NADP+
0.4
GLUCOSE-6-PHOSPHATE
NADH
DEHYDROGENASE
NADPH + H+
0.2
6-Phosphogluconolactone
Figure 7-10. Coupled enzyme assay for hexokinase
NAD+
0
activity. The production of glucose 6-phosphate by
200
250
300
350
400
hexokinase is coupled to the oxidation of this product
by glucose-6-phosphate dehydrogenase in the pres-
Wavelength (nm)
ence of added enzyme and NADP+. When an excess of
Figure 7-9. Absorption spectra of NAD+ and NADH.
glucose-6-phosphate dehydrogenase is present, the
Densities are for a 44 mg/L solution in a cell with a 1 cm
rate of formation of NADPH, which can be measured at
light path. NADP+ and NADPH have spectrums analo-
340 nm, is governed by the rate of formation of glucose
gous to NAD+ and NADH, respectively.
6-phosphate by hexokinase.
ENZYMES: MECHANISM OF ACTION
/
57
Nonfunctional Plasma Enzymes Aid
heart) and M (for muscle). The subunits can combine
Diagnosis & Prognosis
as shown below to yield catalytically active isozymes of
L-lactate dehydrogenase:
Certain enzymes, proenzymes, and their substrates are
present at all times in the circulation of normal individ-
uals and perform a physiologic function in the blood.
Lactate
Examples of these functional plasma enzymes include
Dehydrogenase
lipoprotein lipase, pseudocholinesterase, and the proen-
Isozyme
Subunits
zymes of blood coagulation and blood clot dissolution
I1
HHHH
(Chapters 9 and 51). The majority of these enzymes are
I2
HHHM
synthesized in and secreted by the liver.
I3
HHMM
Plasma also contains numerous other enzymes that
I4
HMMM
perform no known physiologic function in blood.
I5
MMMM
These apparently nonfunctional plasma enzymes arise
from the routine normal destruction of erythrocytes,
leukocytes, and other cells. Tissue damage or necrosis
Distinct genes whose expression is differentially regu-
resulting from injury or disease is generally accompa-
lated in various tissues encode the H and M subunits.
nied by increases in the levels of several nonfunctional
Since heart expresses the H subunit almost exclusively,
plasma enzymes. Table 7-2 lists several enzymes used
isozyme I1 predominates in this tissue. By contrast,
in diagnostic enzymology.
isozyme I5 predominates in liver. Small quantities of
lactate dehydrogenase are normally present in plasma.
Isozymes of Lactate Dehydrogenase Are
Following a myocardial infarction or in liver disease,
Used to Detect Myocardial Infarctions
the damaged tissues release characteristic lactate dehy-
L-Lactate dehydrogenase is a tetrameric enzyme whose
drogenase isoforms into the blood. The resulting eleva-
four subunits occur in two isoforms, designated H (for
tion in the levels of the I1 or I5 isozymes is detected by
separating the different oligomers of lactate dehydroge-
nase by electrophoresis and assaying their catalytic ac-
tivity (Figure 7-11).
Table 7-2. Principal serum enzymes used in
clinical diagnosis. Many of the enzymes are not
ENZYMES FACILITATE DIAGNOSIS
specific for the disease listed.
OF GENETIC DISEASES
Serum Enzyme
Major Diagnostic Use
While many diseases have long been known to result
from alterations in an individual’s DNA, tools for the
Aminotransferases
detection of genetic mutations have only recently be-
Aspartate aminotransfer-
Myocardial infarction
come widely available. These techniques rely upon the
ase (AST, or SGOT)
catalytic efficiency and specificity of enzyme catalysts.
Alanine aminotransferase
Viral hepatitis
For example, the polymerase chain reaction (PCR) re-
(ALT, or SGPT)
lies upon the ability of enzymes to serve as catalytic am-
Amylase
Acute pancreatitis
plifiers to analyze the DNA present in biologic and
forensic samples. In the PCR technique, a thermostable
Ceruloplasmin
Hepatolenticular degeneration
(Wilson’s disease)
DNA polymerase, directed by appropriate oligonu-
cleotide primers, produces thousands of copies of a
Creatine kinase
Muscle disorders and myocar-
sample of DNA that was present initially at levels too
dial infarction
low for direct detection.
γ-Glutamyl transpeptidase
Various liver diseases
The detection of restriction fragment length poly-
morphisms (RFLPs) facilitates prenatal detection of
Lactate dehydrogenase
Myocardial infarction
hereditary disorders such as sickle cell trait, beta-
(isozymes)
thalassemia, infant phenylketonuria, and Huntington’s
Lipase
Acute pancreatitis
disease. Detection of RFLPs involves cleavage of dou-
ble-stranded DNA by restriction endonucleases, which
Phosphatase, acid
Metastatic carcinoma of the
prostate
can detect subtle alterations in DNA that affect their
recognized sites. Chapter 40 provides further details
Phosphatase, alkaline
Various bone disorders, ob-
concerning the use of PCR and restriction enzymes for
(isozymes)
structive liver diseases
diagnosis.
58
/
CHAPTER 7
+
-
LACTATE
(Lactate)
SH2
S
(Pyruvate)
DEHYDROGENASE
Heart
A
NAD+
NADH + H+
Normal
B
Reduced PMS
Oxidized PMS
Liver
C
Oxidized NBT
Reduced NBT
(colorless)
(blue formazan)
5
4
3
2
1
Figure 7-11. Normal and pathologic patterns of lactate dehydrogenase (LDH) isozymes in human
serum. LDH isozymes of serum were separated by electrophoresis and visualized using the coupled reac-
tion scheme shown on the left. (NBT, nitroblue tetrazolium; PMS, phenazine methylsulfate). At right is
shown the stained electropherogram. Pattern A is serum from a patient with a myocardial infarct; B is nor-
mal serum; and C is serum from a patient with liver disease. Arabic numerals denote specific LDH isozymes.
RECOMBINANT DNA PROVIDES AN
resulting modified protein, termed a fusion protein,
contains a domain tailored to interact with a specific
IMPORTANT TOOL FOR STUDYING
affinity support. One popular approach is to attach an
ENZYMES
oligonucleotide that encodes six consecutive histidine
Recombinant DNA technology has emerged as an im-
residues. The expressed “His tag” protein binds to chro-
portant asset in the study of enzymes. Highly purified
matographic supports that contain an immobilized diva-
samples of enzymes are necessary for the study of their
lent metal ion such as Ni2+. Alternatively, the substrate-
structure and function. The isolation of an individual
binding domain of glutathione S-transferase (GST) can
enzyme, particularly one present in low concentration,
serve as a “GST tag.” Figure 7-12 illustrates the purifi-
from among the thousands of proteins present in a cell
cation of a GST-fusion protein using an affinity support
can be extremely difficult. If the gene for the enzyme of
containing bound glutathione. Fusion proteins also
interest has been cloned, it generally is possible to pro-
often encode a cleavage site for a highly specific protease
duce large quantities of its encoded protein in Esch-
such as thrombin in the region that links the two por-
erichia coli or yeast. However, not all animal proteins
tions of the protein. This permits removal of the added
can be expressed in active form in microbial cells, nor
fusion domain following affinity purification.
do microbes perform certain posttranslational process-
ing tasks. For these reasons, a gene may be expressed in
Site-Directed Mutagenesis Provides
cultured animal cell systems employing the baculovirus
Mechanistic Insights
expression vector to transform cultured insect cells. For
more details concerning recombinant DNA techniques,
Once the ability to express a protein from its cloned
see Chapter 40.
gene has been established, it is possible to employ site-
directed mutagenesis to change specific aminoacyl
residues by altering their codons. Used in combination
Recombinant Fusion Proteins Are Purified
with kinetic analyses and x-ray crystallography, this ap-
by Affinity Chromatography
proach facilitates identification of the specific roles of
Recombinant DNA technology can also be used to cre-
given aminoacyl residues in substrate binding and catal-
ate modified proteins that are readily purified by affinity
ysis. For example, the inference that a particular
chromatography. The gene of interest is linked to an
aminoacyl residue functions as a general acid can be
oligonucleotide sequence that encodes a carboxyl or
tested by replacing it with an aminoacyl residue inca-
amino terminal extension to the encoded protein. The
pable of donating a proton.
ENZYMES: MECHANISM OF ACTION
/
59
GST
T
Enzyme
•
Catalytic mechanisms employed by enzymes include
the introduction of strain, approximation of reac-
tants, acid-base catalysis, and covalent catalysis.
Plasmid encoding GST
Cloned DNA
with thrombin site (T)
encoding enzyme
•
Aminoacyl residues that participate in catalysis are
highly conserved among all classes of a given enzyme
activity.
•
Substrates and enzymes induce mutual conforma-
Ligate together
tional changes in one another that facilitate substrate
recognition and catalysis.
•
The catalytic activity of enzymes reveals their pres-
GST
T
Enzyme
ence, facilitates their detection, and provides the basis
for enzyme-linked immunoassays.
Transfect cells, add
•
Many enzymes can be assayed spectrophotometri-
inducing agent, then
break cells
cally by coupling them to an NAD(P)+-dependent
dehydrogenase.
Apply to glutathione (GSH)
•
Assay of plasma enzymes aids diagnosis and progno-
affinity column
sis. For example, a myocardial infarction elevates
serum levels of lactate dehydrogenase isozyme I1
Sepharose
GSH
GST
T
Enzyme
bead
•
Restriction endonucleases facilitate diagnosis of ge-
netic diseases by revealing restriction fragment length
Elute with GSH,
polymorphisms.
treat with thrombin
•
Site-directed mutagenesis, used to change residues
suspected of being important in catalysis or substrate
GSH
GST
T
Enzyme
binding, provides insights into the mechanisms of
enzyme action.
•
Recombinant fusion proteins such as His-tagged or
Figure 7-12. Use of glutathione S-transferase (GST)
GST fusion enzymes are readily purified by affinity
fusion proteins to purify recombinant proteins. (GSH,
chromatography.
glutathione.)
REFERENCES
SUMMARY
Conyers GB et al: Metal requirements of a diadenosine pyrophos-
phatase from Bartonella bacilliformis. Magnetic resonance and
• Enzymes are highly effective and extremely specific
kinetic studies of the role of Mn2+. Biochemistry
2000;
catalysts.
39:2347.
• Organic and inorganic prosthetic groups, cofactors,
Fersht A: Structure and Mechanism in Protein Science: A Guide to
and coenzymes play important roles in catalysis.
Enzyme Catalysis and Protein Folding. Freeman, 1999.
Coenzymes, many of which are derivatives of B vita-
Suckling CJ: Enzyme Chemistry. Chapman & Hall, 1990.
mins, serve as “shuttles.”
Walsh CT: Enzymatic Reaction Mechanisms. Freeman, 1979.
Enzymes: Kinetics
8
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
A+B → P+Q
(2)
Enzyme kinetics is the field of biochemistry concerned
Unidirectional arrows are also used to describe reac-
with the quantitative measurement of the rates of en-
tions in living cells where the products of reaction (2)
zyme-catalyzed reactions and the systematic study of fac-
are immediately consumed by a subsequent enzyme-
tors that affect these rates. Kinetic analyses permit scien-
catalyzed reaction. The rapid removal of product P or
tists to reconstruct the number and order of the
Q therefore precludes occurrence of the reverse reac-
individual steps by which enzymes transform substrates
tion, rendering equation (2) functionally irreversible
into products. The study of enzyme kinetics also repre-
under physiologic conditions.
sents the principal way to identify potential therapeutic
agents that selectively enhance or inhibit the rates of spe-
cific enzyme-catalyzed processes. Together with site-
CHANGES IN FREE ENERGY DETERMINE
directed mutagenesis and other techniques that probe
THE DIRECTION & EQUILIBRIUM STATE
protein structure, kinetic analysis can also reveal details
OF CHEMICAL REACTIONS
of the catalytic mechanism. A complete, balanced set of
enzyme activities is of fundamental importance for main-
The Gibbs free energy change ∆G (also called either the
taining homeostasis. An understanding of enzyme kinet-
free energy or Gibbs energy) describes both the direc-
ics thus is important for understanding how physiologic
tion in which a chemical reaction will tend to proceed
stresses such as anoxia, metabolic acidosis or alkalosis,
and the concentrations of reactants and products that
toxins, and pharmacologic agents affect that balance.
will be present at equilibrium. ∆G for a chemical reac-
tion equals the sum of the free energies of formation of
the reaction products ∆GP minus the sum of the free
CHEMICAL REACTIONS ARE DESCRIBED
energies of formation of the substrates ∆GS. ∆G0 de-
USING BALANCED EQUATIONS
notes the change in free energy that accompanies transi-
A balanced chemical equation lists the initial chemical
tion from the standard state, one-molar concentrations
species
(substrates) present and the new chemical
of substrates and products, to equilibrium. A more use-
species (products) formed for a particular chemical re-
ful biochemical term is ∆G0′, which defines ∆G0 at a
action, all in their correct proportions or stoichiome-
standard state of 10−7 M protons, pH 7.0 (Chapter 10).
try. For example, balanced equation (1) below describes
If the free energy of the products is lower than that of
the reaction of one molecule each of substrates A and B
the substrates, the signs of ∆G0 and ∆G0′ will be nega-
to form one molecule each of products P and Q.
tive, indicating that the reaction as written is favored in
the direction left to right. Such reactions are referred to
A+B → P+ Q
(1)
as spontaneous. The sign and the magnitude of the
free energy change determine how far the reaction will
The double arrows indicate reversibility, an intrinsic
proceed. Equation (3)—
property of all chemical reactions. Thus, for reaction
(1), if A and B can form P and Q, then P and Q can
∆G0=−RT
ln K
eq
(3)
also form A and B. Designation of a particular reactant
as a “substrate” or “product” is therefore somewhat ar-
—illustrates the relationship between the equilibrium
bitrary since the products for a reaction written in one
constant Keq and ∆G0, where R is the gas constant (1.98
direction are the substrates for the reverse reaction. The
cal/mol/°K or 8.31 J/mol/°K) and T is the absolute
term “products” is, however, often used to designate
temperature in degrees Kelvin. Keq is equal to the prod-
the reactants whose formation is thermodynamically fa-
uct of the concentrations of the reaction products, each
vored. Reactions for which thermodynamic factors
raised to the power of their stoichiometry, divided by
strongly favor formation of the products to which the
the product of the substrates, each raised to the power
arrow points often are represented with a single arrow
of their stoichiometry.
as if they were “irreversible”:
60
ENZYMES: KINETICS
/
61
For the reaction A + B → P + Q—
characteristic changes in free energy, ∆G
F, and ∆GD are
associated with each partial reaction.
[P][Q]
Keq
=
(4)
[A][B]
E+R− L← EL L
∆
GF
(8)
and for reaction (5)
ELRLL
→
E−R+L
∆
(9)
←
GD
(5)
A+A← P
E+R−L
→←
E−R+L
∆G=
∆G
+
∆G
(8-10)
F
D
[P]
Keq
=
(6)
[A]2
For the overall reaction (10), ∆G is the sum of ∆G
F and
∆GD. As for any equation of two terms, it is not possi-
—∆G0 may be calculated from equation (3) if the con-
ble to infer from ∆G either the sign or the magnitude
centrations of substrates and products present at equi-
of ∆G
F or ∆GD.
librium are known. If ∆G0 is a negative number, Keq
Many reactions involve multiple transition states,
will be greater than unity and the concentration of
each with an associated change in free energy. For these
products at equilibrium will exceed that of substrates. If
reactions, the overall ∆G represents the sum of all of
∆G0 is positive, Keq will be less than unity and the for-
the free energy changes associated with the formation
mation of substrates will be favored.
and decay of all of the transition states. Therefore, it is
Notice that, since ∆G0 is a function exclusively of
not possible to infer from the overall G the num-
the initial and final states of the reacting species, it can
ber or type of transition states through which the re-
provide information only about the direction and equi-
action proceeds. Stated another way: overall thermo-
librium state of the reaction. ∆G0 is independent of the
dynamics tells us nothing about kinetics.
mechanism of the reaction and therefore provides no
information concerning rates of reactions. Conse-
∆GF Defines the Activation Energy
quently—and as explained below—although a reaction
may have a large negative ∆G0 or ∆G0′, it may never-
Regardless of the sign or magnitude of ∆G, ∆G
F for the
theless take place at a negligible rate.
overwhelming majority of chemical reactions has a pos-
itive sign. The formation of transition state intermedi-
ates therefore requires surmounting of energy barriers.
THE RATES OF REACTIONS
For this reason, ∆G
F is often termed the activation en-
ARE DETERMINED BY THEIR
ergy, Eact, the energy required to surmount a given en-
ACTIVATION ENERGY
ergy barrier. The ease—and hence the frequency—with
which this barrier is overcome is inversely related to
Reactions Proceed via Transition States
Eact. The thermodynamic parameters that determine
how fast a reaction proceeds thus are the ∆G
F values for
The concept of the transition state is fundamental to
formation of the transition states through which the re-
understanding the chemical and thermodynamic basis
action proceeds. For a simple reaction, where means
of catalysis. Equation (7) depicts a displacement reac-
“proportionate to,”
tion in which an entering group E displaces a leaving
group L, attached initially to R.
−E
act
(11)
→
Rate ∝ e RT
E+R−L
E−R+L
←
(7)
Midway through the displacement, the bond between
The activation energy for the reaction proceeding in the
R and L has weakened but has not yet been completely
opposite direction to that drawn is equal to −∆G
D.
severed, and the new bond between E and R is as yet
incompletely formed. This transient intermediate—in
NUMEROUS FACTORS AFFECT
which neither free substrate nor product exists—is
termed the transition state, E R L. Dotted lines
THE REACTION RATE
represent the “partial” bonds that are undergoing for-
The kinetic theory—also called the collision theory—
mation and rupture.
of chemical kinetics states that for two molecules to
Reaction (7) can be thought of as consisting of two
react they must (1) approach within bond-forming dis-
“partial reactions,” the first corresponding to the forma-
tance of one another, or “collide”; and (2) must possess
tion (F) and the second to the subsequent decay (D) of
sufficient kinetic energy to overcome the energy barrier
the transition state intermediate. As for all reactions,
for reaching the transition state. It therefore follows
62
/
CHAPTER 8
that anything which increases the frequency or energy of
which can also be written as
collision between substrates will increase the rate of the
reaction in which they participate.
A+B+B→P
(15)
the corresponding rate expression is
Temperature
Rate ∝ [A][B][B]
(16)
Raising the temperature increases the kinetic energy of
molecules. As illustrated in Figure 8-1, the total num-
or
ber of molecules whose kinetic energy exceeds the en-
ergy barrier Eact (vertical bar) for formation of products
Rate ∝ [A][B]2
(17)
increases from low (A), through intermediate (B), to
high (C) temperatures. Increasing the kinetic energy of
For the general case when n molecules of A react with
molecules also increases their motion and therefore the
m molecules of B,
frequency with which they collide. This combination of
more frequent and more highly energetic and produc-
nA + mB → P
(18)
tive collisions increases the reaction rate.
the rate expression is
Reactant Concentration
]
(19)
The frequency with which molecules collide is directly
proportionate to their concentrations. For two different
Replacing the proportionality constant with an equal
molecules A and B, the frequency with which they col-
sign by introducing a proportionality or rate constant
lide will double if the concentration of either A or B is
k characteristic of the reaction under study gives equa-
doubled. If the concentrations of both A and B are dou-
tions (20) and (21), in which the subscripts 1 and −1
bled, the probability of collision will increase fourfold.
refer to the rate constants for the forward and reverse
For a chemical reaction proceeding at constant tem-
reactions, respectively.
perature that involves one molecule each of A and B,
n
m
Rate
=
k
[A]
[B]
(20)
1
1
A+B→P
(12)
the number of molecules that possess kinetic energy
Rate
−1
=
k
−1
[P]
(21)
sufficient to overcome the activation energy barrier will
be a constant. The number of collisions with sufficient
Keq Is a Ratio of Rate Constants
energy to produce product P therefore will be directly
proportionate to the number of collisions between A
While all chemical reactions are to some extent re-
and B and thus to their molar concentrations, denoted
versible, at equilibrium the overall concentrations of re-
by square brackets.
actants and products remain constant. At equilibrium,
the rate of conversion of substrates to products there-
Rate ∝ [A][B]
(13)
fore equals the rate at which products are converted to
substrates.
Similarly, for the reaction represented by
A +2B→P
(14)
1
Rate =Rate
−1
(22)
Therefore,
m
1
k [A]n[B]
=
−
k [P
1
]
(23)
Energy barrier
∞
and
A
B
C
k
1
[P]
=
(24)
n
m
k
−1
[A]
[B]
The ratio of k1 to k−1 is termed the equilibrium con-
0
stant, Keq. The following important properties of a sys-
∞
Kinetic energy
tem at equilibrium must be kept in mind:
(1) The equilibrium constant is a ratio of the reaction
Figure 8-1. The energy barrier for chemical
rate constants (not the reaction rates).
reactions.
ENZYMES: KINETICS
/
63
(2) At equilibrium, the reaction rates (not the rate
∆Go =−RT
ln K
(25)
constants) of the forward and back reactions are
eq
equal.
If we include the presence of the enzyme (E) in the cal-
(3) Equilibrium is a dynamic state. Although there is
culation of the equilibrium constant for a reaction,
no net change in the concentration of substrates
or products, individual substrate and product
A+B+Enz← P+Q+Enz
(26)
molecules are continually being interconverted.
the expression for the equilibrium constant,
(4) The numeric value of the equilibrium constant
Keq can be calculated either from the concentra-
P][Q][Enz]
Keq
=[
(27)
tions of substrates and products at equilibrium or
A][B][Enz
[
]
from the ratio k1/k−1.
reduces to one identical to that for the reaction in the
absence of the enzyme:
THE KINETICS OF
ENZYMATIC CATALYSIS
P][Q]
Keq
=[
(28)
[A][B]
Enzymes Lower the Activation Energy
Barrier for a Reaction
Enzymes therefore have no effect on Keq.
All enzymes accelerate reaction rates by providing tran-
sition states with a lowered ∆G
F for formation of the
MULTIPLE FACTORS AFFECT THE RATES
transition states. However, they may differ in the way
this is achieved. Where the mechanism or the sequence
OF ENZYME-CATALYZED REACTIONS
of chemical steps at the active site is essentially the same
Temperature
as those for the same reaction proceeding in the absence
of a catalyst, the environment of the active site lowers
Raising the temperature increases the rate of both uncat-
G
F by stabilizing the transition state intermediates. As
alyzed and enzyme-catalyzed reactions by increasing the
discussed in Chapter 7, stabilization can involve (1)
kinetic energy and the collision frequency of the react-
acid-base groups suitably positioned to transfer protons
ing molecules. However, heat energy can also increase
to or from the developing transition state intermediate,
the kinetic energy of the enzyme to a point that exceeds
(2) suitably positioned charged groups or metal ions
the energy barrier for disrupting the noncovalent inter-
that stabilize developing charges, or (3) the imposition
actions that maintain the enzyme’s three-dimensional
of steric strain on substrates so that their geometry ap-
structure. The polypeptide chain then begins to unfold,
proaches that of the transition state. HIV protease (Fig-
or denature, with an accompanying rapid loss of cat-
ure 7-6) illustrates catalysis by an enzyme that lowers
alytic activity. The temperature range over which an
the activation barrier by stabilizing a transition state in-
enzyme maintains a stable, catalytically competent con-
termediate.
formation depends upon—and typically moderately
Catalysis by enzymes that proceeds via a unique re-
exceeds—the normal temperature of the cells in which
action mechanism typically occurs when the transition
it resides. Enzymes from humans generally exhibit sta-
state intermediate forms a covalent bond with the en-
bility at temperatures up to 45-55 °C. By contrast,
zyme (covalent catalysis). The catalytic mechanism of
enzymes from the thermophilic microorganisms that re-
the serine protease chymotrypsin (Figure 7-7) illus-
side in volcanic hot springs or undersea hydrothermal
trates how an enzyme utilizes covalent catalysis to pro-
vents may be stable up to or above 100 °C.
vide a unique reaction pathway.
The Q10, or temperature coefficient, is the factor
by which the rate of a biologic process increases for a
10 °C increase in temperature. For the temperatures
ENZYMES DO NOT AFFECT Keq
over which enzymes are stable, the rates of most bio-
Enzymes accelerate reaction rates by lowering the acti-
logic processes typically double for a 10 °C rise in tem-
vation barrier ∆G
F. While they may undergo transient
perature (Q10 = 2). Changes in the rates of enzyme-
modification during the process of catalysis, enzymes
catalyzed reactions that accompany a rise or fall in body
emerge unchanged at the completion of the reaction.
temperature constitute a prominent survival feature for
The presence of an enzyme therefore has no effect on
“cold-blooded” life forms such as lizards or fish, whose
∆G0 for the overall reaction, which is a function solely
body temperatures are dictated by the external environ-
of the initial and final states of the reactants. Equation
ment. However, for mammals and other homeothermic
(25) shows the relationship between the equilibrium
organisms, changes in enzyme reaction rates with tem-
constant for a reaction and the standard free energy
perature assume physiologic importance only in cir-
change for that reaction:
cumstances such as fever or hypothermia.
64
/
CHAPTER 8
Hydrogen Ion Concentration
the rate of the forward reaction. Assays of enzyme activ-
ity almost always employ a large (103-107) molar excess
The rate of almost all enzyme-catalyzed reactions ex-
of substrate over enzyme. Under these conditions, vi is
hibits a significant dependence on hydrogen ion con-
proportionate to the concentration of enzyme. Measur-
centration. Most intracellular enzymes exhibit optimal
ing the initial velocity therefore permits one to estimate
activity at pH values between 5 and 9. The relationship
the quantity of enzyme present in a biologic sample.
of activity to hydrogen ion concentration (Figure 8-2)
reflects the balance between enzyme denaturation at
SUBSTRATE CONCENTRATION AFFECTS
high or low pH and effects on the charged state of the
enzyme, the substrates, or both. For enzymes whose
REACTION RATE
mechanism involves acid-base catalysis, the residues in-
In what follows, enzyme reactions are treated as if they
volved must be in the appropriate state of protonation
had only a single substrate and a single product. While
for the reaction to proceed. The binding and recogni-
most enzymes have more than one substrate, the princi-
tion of substrate molecules with dissociable groups also
ples discussed below apply with equal validity to en-
typically involves the formation of salt bridges with the
zymes with multiple substrates.
enzyme. The most common charged groups are the
For a typical enzyme, as substrate concentration is
negative carboxylate groups and the positively charged
increased, vi increases until it reaches a maximum value
groups of protonated amines. Gain or loss of critical
Vmax (Figure 8-3). When further increases in substrate
charged groups thus will adversely affect substrate bind-
concentration do not further increase vi, the enzyme is
ing and thus will retard or abolish catalysis.
said to be “saturated” with substrate. Note that the
shape of the curve that relates activity to substrate con-
ASSAYS OF ENZYME-CATALYZED
centration (Figure 8-3) is hyperbolic. At any given in-
REACTIONS TYPICALLY MEASURE
stant, only substrate molecules that are combined with
the enzyme as an ES complex can be transformed into
THE INITIAL VELOCITY
product. Second, the equilibrium constant for the for-
Most measurements of the rates of enzyme-catalyzed re-
mation of the enzyme-substrate complex is not infi-
actions employ relatively short time periods, conditions
nitely large. Therefore, even when the substrate is pre-
that approximate initial rate conditions. Under these
sent in excess (points A and B of Figure 8-4), only a
conditions, only traces of product accumulate, hence
fraction of the enzyme may be present as an ES com-
the rate of the reverse reaction is negligible. The initial
plex. At points A or B, increasing or decreasing [S]
velocity (vi ) of the reaction thus is essentially that of
therefore will increase or decrease the number of ES
complexes with a corresponding change in vi. At point
C (Figure 8-4), essentially all the enzyme is present as
X
the ES complex. Since no free enzyme remains available
100
for forming ES, further increases in [S] cannot increase
the rate of the reaction. Under these saturating condi-
tions, vi depends solely on—and thus is limited by—
SH+
E -
the rapidity with which free enzyme is released to com-
bine with more substrate.
%
Vmax
0
Low
High
C
Vmax/2
pH
vi
B
Figure 8-2. Effect of pH on enzyme activity. Con-
sider, for example, a negatively charged enzyme (EH−)
A
Vmax/2
that binds a positively charged substrate (SH+). Shown
is the proportion (%) of SH+ [\\\] and of EH− [///] as a
Km
[S]
function of pH. Only in the cross-hatched area do both
the enzyme and the substrate bear an appropriate
Figure 8-3. Effect of substrate concentration on the
charge.
initial velocity of an enzyme-catalyzed reaction.
ENZYMES: KINETICS
/
65
= S
= E
A
B
C
Figure 8-4. Representation of an enzyme at low (A), at high (C), and at a substrate concentration
equal to Km (B). Points A, B, and C correspond to those points in Figure 8-3.
THE MICHAELIS-MENTEN & HILL
equal to [S]. Replacing Km + [S] with [S] reduces equa-
tion (29) to
EQUATIONS MODEL THE EFFECTS
OF SUBSTRATE CONCENTRATION
max
V [S]
V
max
[S]
v
=
v
≈
≈V
(31)
The Michaelis-Menten Equation
i
i
max
K
m
+
[S]
[S]
The Michaelis-Menten equation
(29) illustrates in
mathematical terms the relationship between initial re-
Thus, when [S] greatly exceeds Km, the reaction velocity
action velocity vi and substrate concentration
[S],
is maximal (Vmax) and unaffected by further increases in
shown graphically in Figure 8-3.
substrate concentration.
(3) When [S] = Km (point B in Figures 8-3 and
]
8-4).
v
i
= max[
(29)
K
m
+
[S]
V [S]
V
[S]
V
max
max
max
(32)
v
i
=
=
=
The Michaelis constant Km is the substrate concen-
K
m
+
[S]
2[S]
2
tration at which vi is half the maximal velocity
(Vmax/2) attainable at a particular concentration of
Equation (32) states that when [S] equals Km, the initial
enzyme. Km thus has the dimensions of substrate con-
velocity is half-maximal. Equation (32) also reveals that
centration. The dependence of initial reaction velocity
Km is—and may be determined experimentally from—
on [S] and Km may be illustrated by evaluating the
the substrate concentration at which the initial velocity
Michaelis-Menten equation under three conditions.
is half-maximal.
(1) When [S] is much less than Km (point A in Fig-
ures 8-3 and 8-4), the term Km + [S] is essentially equal
A Linear Form of the Michaelis-Menten
to Km. Replacing Km + [S] with Km reduces equation
Equation Is Used to Determine Km & Vmax
(29) to
The direct measurement of the numeric value of Vmax
V
[S]
[S]
V
and therefore the calculation of Km often requires im-
max
max
max
(30)
v
=
v ≈ V
≈
[S]
1
1
practically high concentrations of substrate to achieve
K
m
+[S]
K
m
K
m
saturating conditions. A linear form of the Michaelis-
where ≈ means “approximately equal to.” Since Vmax
Menten equation circumvents this difficulty and per-
and Km are both constants, their ratio is a constant. In
mits Vmax and Km to be extrapolated from initial veloc-
other words, when [S] is considerably below Km, vi ∝
ity data obtained at less than saturating concentrations
k[S]. The initial reaction velocity therefore is directly
of substrate. Starting with equation (29),
proportionate to [S].
(2) When [S] is much greater than Km (point C in
S]
v
= Vmax[
(29)
i
Figures 8-3 and 8-4), the term Km + [S] is essentially
K
+
[S]
m
66
/
CHAPTER 8
invert
Stated another way, the smaller the tendency of the en-
zyme and its substrate to dissociate, the greater the affin-
1
K
+
[S]
m
ity of the enzyme for its substrate. While the Michaelis
(33)
=
v
1
V
max
[S]
constant Km often approximates the dissociation con-
stant Kd, this is by no means always the case. For a typi-
factor
cal enzyme-catalyzed reaction,
1
K
m
[S]
=
+
(34)
k
1
k
2
v
i
Vmax
[S]
V
max
[S]
(39)
E+ S
←
→ +
and simplify
k
−1
1
K
1
1
the value of [S] that gives vi = Vmax/2 is
=
m
+
(35)
v
[S]
V
i
Vmax
max
k
−1
+k
2
[S]=
=
K
(40)
Equation (35) is the equation for a straight line, y = ax
m
k
1
+ b, where y = 1/vi and x = 1/[S]. A plot of 1/vi as y as a
function of 1/[S] as x therefore gives a straight line
When k−1 » k2, then
whose y intercept is 1/Vmax and whose slope is Km/Vmax.
Such a plot is called a double reciprocal or
k
+k ≈k
(41)
Lineweaver-Burk plot (Figure 8-5). Setting the y term
−1
2
−1
of equation (36) equal to zero and solving for x reveals
and
that the x intercept is −1/Km.
−b
−1
k
1
(42)
0= ax+b; therefore, x =
=
(36)
[S]
≈
≈Kd
a
K
k
m
−1
Km is thus most easily calculated from the x intercept.
Hence, 1/Km only approximates 1/Kd under conditions
where the association and dissociation of the ES com-
Km May Approximate a Binding Constant
plex is rapid relative to the rate-limiting step in cataly-
sis. For the many enzyme-catalyzed reactions for which
The affinity of an enzyme for its substrate is the inverse
k−1 + k2 is not approximately equal to k −1, 1/Km will
of the dissociation constant Kd for dissociation of the
underestimate 1/Kd.
enzyme substrate complex ES.
k
1
The Hill Equation Describes the Behavior
(37)
E+ S
←
of Enzymes That Exhibit Cooperative
k
−1
Binding of Substrate
−1
While most enzymes display the simple saturation ki-
Kd = k
(38)
netics depicted in Figure 8-3 and are adequately de-
k
1
scribed by the Michaelis-Menten expression, some en-
zymes bind their substrates in a cooperative fashion
analogous to the binding of oxygen by hemoglobin
(Chapter 6). Cooperative behavior may be encountered
Km
for multimeric enzymes that bind substrate at multiple
1
Slope =
Vmax
sites. For enzymes that display positive cooperativity in
vi
binding substrate, the shape of the curve that relates
changes in vi to changes in [S] is sigmoidal (Figure
8-6). Neither the Michaelis-Menten expression nor its
1
–
Km
1
derived double-reciprocal plots can be used to evaluate
Vmax
cooperative saturation kinetics. Enzymologists therefore
0
employ a graphic representation of the Hill equation
1
[S]
originally derived to describe the cooperative binding of
O2
by hemoglobin. Equation (43) represents the Hill
Figure 8-5. Double reciprocal or Lineweaver-Burk
equation arranged in a form that predicts a straight line,
plot of 1/vi versus 1/[S] used to evaluate Km and Vmax.
where k′ is a complex constant.
ENZYMES: KINETICS
/
67
first substrate molecule then enhances the affinity of the
∞
enzyme for binding additional substrate. The greater
the value for n, the higher the degree of cooperativity
and the more sigmoidal will be the plot of vi versus [S].
A perpendicular dropped from the point where the y
term log vi/(Vmax − vi) is zero intersects the x axis at a
substrate concentration termed S50, the substrate con-
vi
centration that results in half-maximal velocity. S50 thus
is analogous to the P50 for oxygen binding to hemoglo-
bin (Chapter 6).
KINETIC ANALYSIS DISTINGUISHES
COMPETITIVE FROM
0
∞
NONCOMPETITIVE INHIBITION
[S]
Inhibitors of the catalytic activities of enzymes provide
Figure 8-6. Representation of sigmoid substrate
both pharmacologic agents and research tools for study
saturation kinetics.
of the mechanism of enzyme action. Inhibitors can be
classified based upon their site of action on the enzyme,
on whether or not they chemically modify the enzyme,
or on the kinetic parameters they influence. Kinetically,
log
v
1
(43)
=
n
log[S]−log
k′
we distinguish two classes of inhibitors based upon
V
max
−v1
whether raising the substrate concentration does or
Equation (43) states that when [S] is low relative to k′,
does not overcome the inhibition.
the initial reaction velocity increases as the nth power
of [S].
Competitive Inhibitors Typically
A graph of log vi/(Vmax − vi) versus log[S] gives a
Resemble Substrates
straight line (Figure 8-7), where the slope of the line n
The effects of competitive inhibitors can be overcome
is the Hill coefficient, an empirical parameter whose
by raising the concentration of the substrate. Most fre-
value is a function of the number, kind, and strength of
quently, in competitive inhibition the inhibitor, I,
the interactions of the multiple substrate-binding sites
binds to the substrate-binding portion of the active site
on the enzyme. When n = 1, all binding sites behave in-
and blocks access by the substrate. The structures of
dependently, and simple Michaelis-Menten kinetic be-
most classic competitive inhibitors therefore tend to re-
havior is observed. If n is greater than 1, the enzyme is
semble the structures of a substrate and thus are termed
said to exhibit positive cooperativity. Binding of the
substrate analogs. Inhibition of the enzyme succinate
dehydrogenase by malonate illustrates competitive inhi-
bition by a substrate analog. Succinate dehydrogenase
1
catalyzes the removal of one hydrogen atom from each
of the two methylene carbons of succinate (Figure 8-8).
Both succinate and its structural analog malonate
(−OOC CH2 COO−) can bind to the active site of
0
Slope = n
succinate dehydrogenase, forming an ES or an EI com-
plex, respectively. However, since malonate contains
- 1
H
- 4
S50
– 3
-
H
C COO
-2H
H
C COO-
Log [S]
–OOC
C
H
-OOC
C H
SUCCINATE
DEHYDROGENASE
Figure 8-7. A graphic representation of a linear
H
form of the Hill equation is used to evaluate S50, the
Succinate
Fumarate
substrate concentration that produces half-maximal
velocity, and the degree of cooperativity n.
Figure 8-8. The succinate dehydrogenase reaction.
68
/
CHAPTER 8
only one methylene carbon, it cannot undergo dehy-
drogenation. The formation and dissociation of the EI
complex is a dynamic process described by
1
k
1
vi
→
(44)
EnzI
←
Enz+I
k
−1
1
-
K′m
1
1
for which the equilibrium constant Ki is
-
Km
Vmax
0
1
1
[S]
K = [En ][] = k
(45)
1
[EnzI]
k
−1
Figure 8-9. Lineweaver-Burk plot of competitive in-
hibition. Note the complete relief of inhibition at high
In effect, a competitive inhibitor acts by decreasing
[S] (ie, low 1/[S]).
the number of free enzyme molecules available to
bind substrate, ie, to form ES, and thus eventually
to form product, as described below:
For simple competitive inhibition, the intercept on
E-I
the x axis is
E
E-S
−1
[I]
x
=
1+
(47)
E + P
K
K
(46)
m
i
A competitive inhibitor and substrate exert reciprocal
Once Km has been determined in the absence of in-
effects on the concentration of the EI and ES com-
hibitor, Ki can be calculated from equation (47). Ki val-
plexes. Since binding substrate removes free enzyme
ues are used to compare different inhibitors of the same
available to combine with inhibitor, increasing the [S]
enzyme. The lower the value for Ki, the more effective
decreases the concentration of the EI complex and
the inhibitor. For example, the statin drugs that act as
raises the reaction velocity. The extent to which [S]
competitive inhibitors of HMG-CoA reductase (Chap-
must be increased to completely overcome the inhibi-
ter 26) have Ki values several orders of magnitude lower
tion depends upon the concentration of inhibitor pre-
than the Km for the substrate HMG-CoA.
sent, its affinity for the enzyme Ki, and the Km of the
enzyme for its substrate.
Simple Noncompetitive Inhibitors Lower
Double Reciprocal Plots Facilitate the
Vmax but Do Not Affect Km
Evaluation of Inhibitors
In noncompetitive inhibition, binding of the inhibitor
Double reciprocal plots distinguish between competi-
does not affect binding of substrate. Formation of both
tive and noncompetitive inhibitors and simplify evalua-
EI and EIS complexes is therefore possible. However,
tion of inhibition constants Ki. vi is determined at sev-
while the enzyme-inhibitor complex can still bind sub-
eral substrate concentrations both in the presence and
strate, its efficiency at transforming substrate to prod-
in the absence of inhibitor. For classic competitive inhi-
uct, reflected by Vmax, is decreased. Noncompetitive
bition, the lines that connect the experimental data
inhibitors bind enzymes at sites distinct from the sub-
points meet at the y axis (Figure 8-9). Since the y inter-
strate-binding site and generally bear little or no struc-
cept is equal to 1/Vmax, this pattern indicates that when
tural resemblance to the substrate.
1/[S] approaches 0, vi is independent of the presence
For simple noncompetitive inhibition, E and EI
of inhibitor. Note, however, that the intercept on the
possess identical affinity for substrate, and the EIS com-
x axis does vary with inhibitor concentration—and that
plex generates product at a negligible rate (Figure 8-10).
since −1/Km′ is smaller than 1/Km, Km′ (the “apparent
More complex noncompetitive inhibition occurs when
Km”) becomes larger in the presence of increasing con-
binding of the inhibitor does affect the apparent affinity
centrations of inhibitor. Thus, a competitive inhibitor
of the enzyme for substrate, causing the lines to inter-
has no effect on Vmax but raises K ′m, the apparent
cept in either the third or fourth quadrants of a double
Km for the substrate.
reciprocal plot (not shown).
ENZYMES: KINETICS
/
69
A
B
P
Q
1
E
EA
EAB-EPQ
EQ
E
vi
A B
P
Q
1
-
V′max
1
-
EA
EQ
Km
1
Vmax
E
EAB-EPQ
E
0
1
[S]
EB
EP
Figure 8-10. Lineweaver-Burk plot for simple non-
B A
Q P
competitive inhibition.
A
P
B
Q
E
EA-FP
F
FB-EQ
E
Irreversible Inhibitors “Poison” Enzymes
Figure 8-11. Representations of three classes of Bi-
In the above examples, the inhibitors form a dissocia-
Bi reaction mechanisms. Horizontal lines represent the
ble, dynamic complex with the enzyme. Fully active en-
enzyme. Arrows indicate the addition of substrates and
zyme can therefore be recovered simply by removing
departure of products. Top: An ordered Bi-Bi reaction,
the inhibitor from the surrounding medium. However,
characteristic of many NAD(P)H-dependent oxidore-
a variety of other inhibitors act irreversibly by chemi-
ductases. Center: A random Bi-Bi reaction, characteris-
cally modifying the enzyme. These modifications gen-
erally involve making or breaking covalent bonds with
tic of many kinases and some dehydrogenases. Bot-
aminoacyl residues essential for substrate binding, catal-
tom: A ping-pong reaction, characteristic of
ysis, or maintenance of the enzyme’s functional confor-
aminotransferases and serine proteases.
mation. Since these covalent changes are relatively sta-
ble, an enzyme that has been
“poisoned” by an
irreversible inhibitor remains inhibited even after re-
moval of the remaining inhibitor from the surrounding
reactions because the group undergoing transfer is usu-
medium.
ally passed directly, in a single step, from one substrate
to the other. Sequential Bi-Bi reactions can be further
distinguished based on whether the two substrates add
MOST ENZYME-CATALYZED REACTIONS
in a random or in a compulsory order. For random-
INVOLVE TWO OR MORE SUBSTRATES
order reactions, either substrate A or substrate B may
combine first with the enzyme to form an EA or an EB
While many enzymes have a single substrate, many oth-
complex (Figure 8-11, center). For compulsory-order
ers have two—and sometimes more than two—sub-
reactions, A must first combine with E before B can
strates and products. The fundamental principles dis-
combine with the EA complex. One explanation for a
cussed above, while illustrated for single-substrate
compulsory-order mechanism is that the addition of A
enzymes, apply also to multisubstrate enzymes. The
induces a conformational change in the enzyme that
mathematical expressions used to evaluate multisub-
aligns residues which recognize and bind B.
strate reactions are, however, complex. While detailed
kinetic analysis of multisubstrate reactions exceeds the
scope of this chapter, two-substrate, two-product reac-
Ping-Pong Reactions
tions (termed “Bi-Bi” reactions) are considered below.
The term
“ping-pong” applies to mechanisms in
which one or more products are released from the en-
Sequential or Single
zyme before all the substrates have been added. Ping-
Displacement Reactions
pong reactions involve covalent catalysis and a tran-
In sequential reactions, both substrates must combine
sient, modified form of the enzyme
(Figure
7-4).
with the enzyme to form a ternary complex before
Ping-pong Bi-Bi reactions are double displacement re-
catalysis can proceed (Figure 8-11, top). Sequential re-
actions. The group undergoing transfer is first dis-
actions are sometimes referred to as single displacement
placed from substrate A by the enzyme to form product
70
/
CHAPTER 8
Increasing
[S2]
1
vi
Figure 8-12. Lineweaver-Burk plot for a two-sub-
strate ping-pong reaction. An increase in concentra-
tion of one substrate (S1) while that of the other sub-
1
strate (S2) is maintained constant changes both the x
S1
and y intercepts, but not the slope.
P and a modified form of the enzyme (F). The subse-
other combinations of product inhibitor and variable
quent group transfer from F to the second substrate B,
substrate will produce forms of complex noncompeti-
forming product Q and regenerating E, constitutes the
tive inhibition.
second displacement (Figure 8-11, bottom).
Most Bi-Bi Reactions Conform to
SUMMARY
Michaelis-Menten Kinetics
•
The study of enzyme kinetics—the factors that affect
Most Bi-Bi reactions conform to a somewhat more
the rates of enzyme-catalyzed reactions—reveals the
complex form of Michaelis-Menten kinetics in which
individual steps by which enzymes transform sub-
strates into products.
Vmax refers to the reaction rate attained when both sub-
strates are present at saturating levels. Each substrate
•
∆G, the overall change in free energy for a reaction,
has its own characteristic Km value which corresponds
is independent of reaction mechanism and provides
to the concentration that yields half-maximal velocity
no information concerning rates of reactions.
when the second substrate is present at saturating levels.
•
Enzymes do not affect Keq. Keq, a ratio of reaction
As for single-substrate reactions, double-reciprocal plots
rate constants, may be calculated from the concentra-
can be used to determine Vmax and Km. vi is measured as
tions of substrates and products at equilibrium or
a function of the concentration of one substrate (the
from the ratio k1/k−1.
variable substrate) while the concentration of the other
•
Reactions proceed via transition states in which ∆G
F
substrate (the fixed substrate) is maintained constant. If
is the activation energy. Temperature, hydrogen ion
the lines obtained for several fixed-substrate concentra-
concentration, enzyme concentration, substrate con-
tions are plotted on the same graph, it is possible to dis-
centration, and inhibitors all affect the rates of en-
tinguish between a ping-pong enzyme, which yields
zyme-catalyzed reactions.
parallel lines, and a sequential mechanism, which yields
•
A measurement of the rate of an enzyme-catalyzed
a pattern of intersecting lines (Figure 8-12).
reaction generally employs initial rate conditions, for
Product inhibition studies are used to complement
which the essential absence of product precludes the
kinetic analyses and to distinguish between ordered and
reverse reaction.
random Bi-Bi reactions. For example, in a random-
order Bi-Bi reaction, each product will be a competitive
•
A linear form of the Michaelis-Menten equation sim-
inhibitor regardless of which substrate is designated the
plifies determination of Km and Vmax.
variable substrate. However, for a sequential mecha-
•
A linear form of the Hill equation is used to evaluate
nism (Figure 8-11, bottom), only product Q will give
the cooperative substrate-binding kinetics exhibited
the pattern indicative of competitive inhibition when A
by some multimeric enzymes. The slope n, the Hill
is the variable substrate, while only product P will pro-
coefficient, reflects the number, nature, and strength
duce this pattern with B as the variable substrate. The
of the interactions of the substrate-binding sites. A
ENZYMES: KINETICS
/
71
value of n greater than 1 indicates positive coopera-
REFERENCES
tivity.
Fersht A: Structure and Mechanism in Protein Science: A Guide to
• The effects of competitive inhibitors, which typically
Enzyme Catalysis and Protein Folding. Freeman, 1999.
resemble substrates, are overcome by raising the con-
Schultz AR: Enzyme Kinetics: From Diastase to Multi-enzyme Sys-
centration of the substrate. Noncompetitive in-
tems. Cambridge Univ Press, 1994.
hibitors lower Vmax but do not affect Km.
Segel IH: Enzyme Kinetics. Wiley Interscience, 1975.
• Substrates may add in a random order (either sub-
strate may combine first with the enzyme) or in a
compulsory order (substrate A must bind before sub-
strate B).
• In ping-pong reactions, one or more products are re-
leased from the enzyme before all the substrates have
added.
Enzymes: Regulation of Activities
9
Victor W. Rodwell, PhD, & Peter J. Kennelly, PhD
BIOMEDICAL IMPORTANCE
concentration generate corresponding changes in me-
tabolite flux (Figure 9-1). Responses to changes in sub-
The 19th-century physiologist Claude Bernard enunci-
strate level represent an important but passive means for
ated the conceptual basis for metabolic regulation. He
coordinating metabolite flow and maintaining homeo-
observed that living organisms respond in ways that are
stasis in quiescent cells. However, they offer limited
both quantitatively and temporally appropriate to per-
scope for responding to changes in environmental vari-
mit them to survive the multiple challenges posed by
ables. The mechanisms that regulate enzyme activity in
changes in their external and internal environments.
an active manner in response to internal and external
Walter Cannon subsequently coined the term “homeo-
signals are discussed below.
stasis” to describe the ability of animals to maintain a
constant intracellular environment despite changes in
their external environment. We now know that organ-
Metabolite Flow Tends
isms respond to changes in their external and internal
to Be Unidirectional
environment by balanced, coordinated changes in the
Despite the existence of short-term oscillations in
rates of specific metabolic reactions. Many human dis-
metabolite concentrations and enzyme levels, living
eases, including cancer, diabetes, cystic fibrosis, and
cells exist in a dynamic steady state in which the mean
Alzheimer’s disease, are characterized by regulatory dys-
concentrations of metabolic intermediates remain rela-
functions triggered by pathogenic agents or genetic mu-
tively constant over time (Figure 9-2). While all chemi-
tations. For example, many oncogenic viruses elaborate
cal reactions are to some extent reversible, in living cells
protein-tyrosine kinases that modify the regulatory
the reaction products serve as substrates for—and are
events which control patterns of gene expression, con-
removed by—other enzyme-catalyzed reactions. Many
tributing to the initiation and progression of cancer. The
nominally reversible reactions thus occur unidirection-
toxin from Vibrio cholerae, the causative agent of cholera,
ally. This succession of coupled metabolic reactions is
disables sensor-response pathways in intestinal epithelial
accompanied by an overall change in free energy that
cells by ADP-ribosylating the GTP-binding proteins
favors unidirectional metabolite flow (Chapter 10). The
(G-proteins) that link cell surface receptors to adenylyl
unidirectional flow of metabolites through a pathway
cyclase. The consequent activation of the cyclase triggers
with a large overall negative change in free energy is
the flow of water into the intestines, resulting in massive
analogous to the flow of water through a pipe in which
diarrhea and dehydration. Yersinia pestis, the causative
one end is lower than the other. Bends or kinks in the
agent of plague, elaborates a protein-tyrosine phos-
pipe simulate individual enzyme-catalyzed steps with a
phatase that hydrolyzes phosphoryl groups on key cy-
small negative or positive change in free energy. Flow of
toskeletal proteins. Knowledge of factors that control the
water through the pipe nevertheless remains unidirec-
rates of enzyme-catalyzed reactions thus is essential to an
tional due to the overall change in height, which corre-
understanding of the molecular basis of disease. This
sponds to the overall change in free energy in a pathway
chapter outlines the patterns by which metabolic
(Figure 9-3).
processes are controlled and provides illustrative exam-
ples. Subsequent chapters provide additional examples.
COMPARTMENTATION ENSURES
REGULATION OF METABOLITE FLOW
METABOLIC EFFICIENCY
CAN BE ACTIVE OR PASSIVE
& SIMPLIFIES REGULATION
Enzymes that operate at their maximal rate cannot re-
In eukaryotes, anabolic and catabolic pathways that in-
spond to an increase in substrate concentration, and
terconvert common products may take place in specific
can respond only to a precipitous decrease in substrate
subcellular compartments. For example, many of the
concentration. For most enzymes, therefore, the aver-
enzymes that degrade proteins and polysaccharides re-
age intracellular concentration of their substrate tends
side inside organelles called lysosomes. Similarly, fatty
to be close to the Km value, so that changes in substrate
acid biosynthesis occurs in the cytosol, whereas fatty
72
ENZYMES: REGULATION OF ACTIVITIES
/
73
∆VB
∆VA
V
A
Km
∆S
∆S
B
[ S ]
Figure 9-1. Differential response of the rate of an
enzyme-catalyzed reaction, ∆V, to the same incremen-
Figure 9-3. Hydrostatic analogy for a pathway with
tal change in substrate concentration at a substrate
a rate-limiting step (A) and a step with a ∆G value near
concentration of Km (∆VA) or far above Km (∆VB).
zero (B).
acid oxidation takes place within mitochondria (Chap-
generation from those of NADPH that participate in
ters 21 and 22). Segregation of certain metabolic path-
the reductive steps in many biosynthetic pathways.
ways within specialized cell types can provide further
physical compartmentation. Alternatively, possession of
Controlling an Enzyme That Catalyzes
one or more unique intermediates can permit apparently
a Rate-Limiting Reaction Regulates
opposing pathways to coexist even in the absence of
an Entire Metabolic Pathway
physical barriers. For example, despite many shared in-
termediates and enzymes, both glycolysis and gluconeo-
While the flux of metabolites through metabolic path-
genesis are favored energetically. This cannot be true if
ways involves catalysis by numerous enzymes, active
all the reactions were the same. If one pathway was fa-
control of homeostasis is achieved by regulation of only
vored energetically, the other would be accompanied by
a small number of enzymes. The ideal enzyme for regu-
a change in free energy G equal in magnitude but op-
latory intervention is one whose quantity or catalytic ef-
posite in sign. Simultaneous spontaneity of both path-
ficiency dictates that the reaction it catalyzes is slow rel-
ways results from substitution of one or more reactions
ative to all others in the pathway. Decreasing the
by different reactions favored thermodynamically in the
catalytic efficiency or the quantity of the catalyst for the
opposite direction. The glycolytic enzyme phospho-
“bottleneck” or rate-limiting reaction immediately re-
fructokinase
(Chapter 17) is replaced by the gluco-
duces metabolite flux through the entire pathway. Con-
neogenic enzyme fructose-1,6-bisphosphatase (Chapter
versely, an increase in either its quantity or catalytic ef-
19). The ability of enzymes to discriminate between the
ficiency enhances flux through the pathway as a whole.
structurally similar coenzymes NAD+ and NADP+ also
For example, acetyl-CoA carboxylase catalyzes the syn-
results in a form of compartmentation, since it segre-
thesis of malonyl-CoA, the first committed reaction of
gates the electrons of NADH that are destined for ATP
fatty acid biosynthesis (Chapter 21). When synthesis of
malonyl-CoA is inhibited, subsequent reactions of fatty
acid synthesis cease due to lack of substrates. Enzymes
that catalyze rate-limiting steps serve as natural “gover-
Large
nors” of metabolic flux. Thus, they constitute efficient
molecules
targets for regulatory intervention by drugs. For exam-
ple, inhibition by “statin” drugs of HMG-CoA reduc-
tase, which catalyzes the rate-limiting reaction of cho-
Small
Small
Nutrients
~P
~P
Wastes
molecules
molecules
lesterogenesis, curtails synthesis of cholesterol.
REGULATION OF ENZYME QUANTITY
Small
molecules
The catalytic capacity of the rate-limiting reaction in a
metabolic pathway is the product of the concentration
Figure 9-2. An idealized cell in steady state. Note
of enzyme molecules and their intrinsic catalytic effi-
that metabolite flow is unidirectional.
ciency. It therefore follows that catalytic capacity can be
74
/
CHAPTER 9
influenced both by changing the quantity of enzyme
Enzyme levels in mammalian tissues respond to a
present and by altering its intrinsic catalytic efficiency.
wide range of physiologic, hormonal, or dietary factors.
For example, glucocorticoids increase the concentration
of tyrosine aminotransferase by stimulating ks, and
Control of Enzyme Synthesis
glucagon—despite its antagonistic physiologic effects—
increases ks fourfold to fivefold. Regulation of liver
Enzymes whose concentrations remain essentially con-
arginase can involve changes either in ks or in kdeg. After
stant over time are termed constitutive enzymes. By
a protein-rich meal, liver arginase levels rise and argi-
contrast, the concentrations of many other enzymes de-
nine synthesis decreases (Chapter 29). Arginase levels
pend upon the presence of inducers, typically sub-
also rise in starvation, but here arginase degradation de-
strates or structurally related compounds, that initiate
creases, whereas ks remains unchanged. Similarly, injec-
their synthesis. Escherichia coli grown on glucose will,
tion of glucocorticoids and ingestion of tryptophan
for example, only catabolize lactose after addition of a
both elevate levels of tryptophan oxygenase. While the
β-galactoside, an inducer that initiates synthesis of a
hormone raises ks for oxygenase synthesis, tryptophan
β-galactosidase and a galactoside permease (Figure 39-3).
specifically lowers kdeg by stabilizing the oxygenase
Inducible enzymes of humans include tryptophan pyr-
against proteolytic digestion.
rolase, threonine dehydrase, tyrosine-α-ketoglutarate
aminotransferase, enzymes of the urea cycle, HMG-CoA
reductase, and cytochrome P450. Conversely, an excess
MULTIPLE OPTIONS ARE AVAILABLE FOR
of a metabolite may curtail synthesis of its cognate
enzyme via repression. Both induction and repression
REGULATING CATALYTIC ACTIVITY
involve cis elements, specific DNA sequences located up-
In humans, the induction of protein synthesis is a com-
stream of regulated genes, and trans-acting regulatory
plex multistep process that typically requires hours to
proteins. The molecular mechanisms of induction and
produce significant changes in overall enzyme level. By
repression are discussed in Chapter 39.
contrast, changes in intrinsic catalytic efficiency ef-
fected by binding of dissociable ligands (allosteric reg-
ulation) or by covalent modification achieve regula-
Control of Enzyme Degradation
tion of enzymic activity within seconds. Changes in
protein level serve long-term adaptive requirements,
The absolute quantity of an enzyme reflects the net bal-
whereas changes in catalytic efficiency are best suited
ance between enzyme synthesis and enzyme degrada-
for rapid and transient alterations in metabolite flux.
tion, where ks and kdeg represent the rate constants for
the overall processes of synthesis and degradation, re-
spectively. Changes in both the ks and kdeg of specific
enzymes occur in human subjects.
ALLOSTERIC EFFECTORS REGULATE
CERTAIN ENZYMES
Enzyme
Feedback inhibition refers to inhibition of an enzyme
ks
kdeg
in a biosynthetic pathway by an end product of that
pathway. For example, for the biosynthesis of D from A
Amino acids
catalyzed by enzymes Enz1 through Enz3,
Protein turnover represents the net result of en-
Enz1
Enz
2
Enz
3
zyme synthesis and degradation. By measuring the rates
A
→
B
→
C
→
D
of incorporation of 15N-labeled amino acids into pro-
tein and the rates of loss of 15N from protein, Schoen-
heimer deduced that body proteins are in a state of “dy-
high concentrations of D inhibit conversion of A to B.
namic equilibrium” in which they are continuously
Inhibition results not from the “backing up” of inter-
synthesized and degraded. Mammalian proteins are de-
mediates but from the ability of D to bind to and in-
graded both by ATP and ubiquitin-dependent path-
hibit Enz1. Typically, D binds at an allosteric site spa-
ways and by ATP-independent pathways (Chapter 29).
tially distinct from the catalytic site of the target
Susceptibility to proteolytic degradation can be influ-
enzyme. Feedback inhibitors thus are allosteric effectors
enced by the presence of ligands such as substrates,
and typically bear little or no structural similarity to the
coenzymes, or metal ions that alter protein conforma-
substrates of the enzymes they inhibit. In this example,
tion. Intracellular ligands thus can influence the rates at
the feedback inhibitor D acts as a negative allosteric
which specific enzymes are degraded.
effector of Enz1.
ENZYMES: REGULATION OF ACTIVITIES
/
75
In a branched biosynthetic pathway, the initial reac-
tions participate in the synthesis of several products.
A
B
Figure 9-4 shows a hypothetical branched biosynthetic
pathway in which curved arrows lead from feedback in-
hibitors to the enzymes whose activity they inhibit. The
S1
S2
S3
S4
sequences S3 → A, S4 → B, S4 → C, and S3 → → D
C
each represent linear reaction sequences that are feed-
S5
D
back-inhibited by their end products. The pathways of
nucleotide biosynthesis (Chapter 34) provide specific
examples.
The kinetics of feedback inhibition may be competi-
Figure 9-5. Multiple feedback inhibition in a
tive, noncompetitive, partially competitive, or mixed.
branched biosynthetic pathway. Superimposed on sim-
Feedback inhibitors, which frequently are the small
ple feedback loops (dashed, curved arrows) are multi-
molecule building blocks of macromolecules (eg, amino
ple feedback loops (solid, curved arrows) that regulate
acids for proteins, nucleotides for nucleic acids), typi-
enzymes common to biosynthesis of several end prod-
cally inhibit the first committed step in a particular
ucts.
biosynthetic sequence. A much-studied example is inhi-
bition of bacterial aspartate transcarbamoylase by CTP
(see below and Chapter 34).
phosphate (CTP). Following treatment with mercuri-
Multiple feedback loops can provide additional fine
als, ATCase loses its sensitivity to inhibition by CTP
control. For example, as shown in Figure 9-5, the pres-
but retains its full activity for synthesis of carbamoyl as-
ence of excess product B decreases the requirement for
partate. This suggests that CTP is bound at a different
substrate S2. However, S2 is also required for synthesis
(allosteric) site from either substrate. ATCase consists
of A, C, and D. Excess B should therefore also curtail
of multiple catalytic and regulatory subunits. Each cat-
synthesis of all four end products. To circumvent this
alytic subunit contains four aspartate (substrate) sites
potential difficulty, each end product typically only
and each regulatory subunit at least two CTP (regula-
partially inhibits catalytic activity. The effect of an ex-
tory) sites (Chapter 34).
cess of two or more end products may be strictly addi-
tive or, alternatively, may be greater than their individ-
ual effect (cooperative feedback inhibition).
Allosteric & Catalytic Sites Are
Spatially Distinct
The lack of structural similarity between a feedback in-
Aspartate Transcarbamoylase Is a Model
hibitor and the substrate for the enzyme whose activity
Allosteric Enzyme
it regulates suggests that these effectors are not isosteric
Aspartate transcarbamoylase (ATCase), the catalyst for
with a substrate but allosteric
(“occupy another
the first reaction unique to pyrimidine biosynthesis
space”). Jacques Monod therefore proposed the exis-
(Figure
34-7), is feedback-inhibited by cytidine tri-
tence of allosteric sites that are physically distinct from
the catalytic site. Allosteric enzymes thus are those
whose activity at the active site may be modulated by
the presence of effectors at an allosteric site. This hy-
A
B
pothesis has been confirmed by many lines of evidence,
including x-ray crystallography and site-directed muta-
genesis, demonstrating the existence of spatially distinct
S1
S2
S3
S4
active and allosteric sites on a variety of enzymes.
C
S5
D
Allosteric Effects May Be on Km or on Vmax
To refer to the kinetics of allosteric inhibition as “com-
Figure 9-4. Sites of feedback inhibition in a
petitive” or
“noncompetitive” with substrate carries
branched biosynthetic pathway. S1-S5 are intermedi-
misleading mechanistic implications. We refer instead
ates in the biosynthesis of end products A-D. Straight
to two classes of regulated enzymes: K-series and V-se-
arrows represent enzymes catalyzing the indicated con-
ries enzymes. For K-series allosteric enzymes, the sub-
versions. Curved arrows represent feedback loops and
strate saturation kinetics are competitive in the sense
indicate sites of feedback inhibition by specific end
that Km is raised without an effect on Vmax. For V-series
products.
allosteric enzymes, the allosteric inhibitor lowers Vmax
76
/
CHAPTER 9
without affecting the Km. Alterations in Km or Vmax
REGULATORY COVALENT
probably result from conformational changes at the cat-
MODIFICATIONS CAN BE
alytic site induced by binding of the allosteric effector
REVERSIBLE OR IRREVERSIBLE
at the allosteric site. For a K-series allosteric enzyme,
this conformational change may weaken the bonds be-
In mammalian cells, the two most common forms of
tween substrate and substrate-binding residues. For a
covalent modification are partial proteolysis and
V-series allosteric enzyme, the primary effect may be to
phosphorylation. Because cells lack the ability to re-
alter the orientation or charge of catalytic residues, low-
unite the two portions of a protein produced by hydrol-
ering Vmax. Intermediate effects on Km and Vmax, how-
ysis of a peptide bond, proteolysis constitutes an irre-
ever, may be observed consequent to these conforma-
versible modification. By contrast, phosphorylation is a
tional changes.
reversible modification process. The phosphorylation of
proteins on seryl, threonyl, or tyrosyl residues, catalyzed
FEEDBACK REGULATION
by protein kinases, is thermodynamically spontaneous.
Equally spontaneous is the hydrolytic removal of these
IS NOT SYNONYMOUS WITH
phosphoryl groups by enzymes called protein phos-
FEEDBACK INHIBITION
phatases.
In both mammalian and bacterial cells, end products
“feed back” and control their own synthesis, in many
PROTEASES MAY BE SECRETED AS
instances by feedback inhibition of an early biosyn-
CATALYTICALLY INACTIVE PROENZYMES
thetic enzyme. We must, however, distinguish between
feedback regulation, a phenomenologic term devoid
Certain proteins are synthesized and secreted as inactive
of mechanistic implications, and feedback inhibition,
precursor proteins known as proproteins. The propro-
a mechanism for regulation of enzyme activity. For ex-
teins of enzymes are termed proenzymes or zymogens.
ample, while dietary cholesterol decreases hepatic syn-
Selective proteolysis converts a proprotein by one or
thesis of cholesterol, this feedback regulation does not
more successive proteolytic “clips” to a form that ex-
involve feedback inhibition. HMG-CoA reductase, the
hibits the characteristic activity of the mature protein,
rate-limiting enzyme of cholesterologenesis, is affected,
eg, its enzymatic activity. Proteins synthesized as pro-
but cholesterol does not feedback-inhibit its activity.
proteins include the hormone insulin (proprotein =
Regulation in response to dietary cholesterol involves
proinsulin), the digestive enzymes pepsin, trypsin, and
curtailment by cholesterol or a cholesterol metabolite of
chymotrypsin (proproteins = pepsinogen, trypsinogen,
the expression of the gene that encodes HMG-CoA re-
and chymotrypsinogen, respectively), several factors of
ductase (enzyme repression) (Chapter 26).
the blood clotting and blood clot dissolution cascades
(see Chapter 51), and the connective tissue protein col-
lagen (proprotein = procollagen).
MANY HORMONES ACT THROUGH
ALLOSTERIC SECOND MESSENGERS
Proenzymes Facilitate Rapid
Nerve impulses—and binding of hormones to cell sur-
Mobilization of an Activity in Response
face receptors—elicit changes in the rate of enzyme-
to Physiologic Demand
catalyzed reactions within target cells by inducing the re-
lease or synthesis of specialized allosteric effectors called
The synthesis and secretion of proteases as catalytically
second messengers. The primary or “first” messenger is
inactive proenzymes protects the tissue of origin (eg,
the hormone molecule or nerve impulse. Second mes-
the pancreas) from autodigestion, such as can occur in
sengers include 3′,5′-cAMP, synthesized from ATP by
pancreatitis. Certain physiologic processes such as di-
the enzyme adenylyl cyclase in response to the hormone
gestion are intermittent but fairly regular and pre-
epinephrine, and Ca2+, which is stored inside the endo-
dictable. Others such as blood clot formation, clot dis-
plasmic reticulum of most cells. Membrane depolariza-
solution, and tissue repair are brought “on line” only in
tion resulting from a nerve impulse opens a membrane
response to pressing physiologic or pathophysiologic
channel that releases calcium ion into the cytoplasm,
need. The processes of blood clot formation and dis-
where it binds to and activates enzymes involved in the
solution clearly must be temporally coordinated to
regulation of contraction and the mobilization of stored
achieve homeostasis. Enzymes needed intermittently
glucose from glycogen. Glucose then supplies the in-
but rapidly often are secreted in an initially inactive
creased energy demands of muscle contraction. Other
form since the secretion process or new synthesis of the
second messengers include 3′,5′-cGMP and polyphos-
required proteins might be insufficiently rapid for re-
phoinositols, produced by the hydrolysis of inositol
sponse to a pressing pathophysiologic demand such as
phospholipids by hormone-regulated phospholipases.
the loss of blood.
ENZYMES: REGULATION OF ACTIVITIES
/
77
1
13 14 15
16
146
149
245
Pro-CT
1
13 14 15
16
146
149
245
π-CT
14-15
147-148
1
13
16
146
149
245
α-CT
S
S
S
S
Figure 9-6. Selective proteolysis and associated conformational changes form the
active site of chymotrypsin, which includes the Asp102-His57-Ser195 catalytic triad.
Successive proteolysis forms prochymotrypsin (pro-CT), π-chymotrypsin (π-CT), and ul-
timately α-chymotrypsin (α-CT), an active protease whose three peptides remain asso-
ciated by covalent inter-chain disulfide bonds.
Activation of Prochymotrypsin
catalyzing transfer of the terminal phosphoryl group of
Requires Selective Proteolysis
ATP to the hydroxyl groups of seryl, threonyl, or tyro-
syl residues, forming O-phosphoseryl, O-phosphothre-
Selective proteolysis involves one or more highly spe-
onyl, or O-phosphotyrosyl residues, respectively (Figure
cific proteolytic clips that may or may not be accompa-
9-7). Some protein kinases target the side chains of his-
nied by separation of the resulting peptides. Most im-
tidyl, lysyl, arginyl, and aspartyl residues. The unmodi-
portantly, selective proteolysis often results in
fied form of the protein can be regenerated by hy-
conformational changes that “create” the catalytic site
drolytic removal of phosphoryl groups, catalyzed by
of an enzyme. Note that while His 57 and Asp 102 re-
protein phosphatases.
side on the B peptide of α-chymotrypsin, Ser 195 re-
A typical mammalian cell possesses over 1000 phos-
sides on the C peptide (Figure 9-6). The conforma-
phorylated proteins and several hundred protein kinases
tional changes that accompany selective proteolysis of
and protein phosphatases that catalyze their intercon-
prochymotrypsin (chymotrypsinogen) align the three
version. The ease of interconversion of enzymes be-
residues of the charge-relay network, creating the cat-
tween their phospho- and dephospho- forms in part
alytic site. Note also that contact and catalytic residues
can be located on different peptide chains but still be
within bond-forming distance of bound substrate.
ATP
ADP
REVERSIBLE COVALENT MODIFICATION
Mg2+
REGULATES KEY MAMMALIAN ENZYMES
KINASE
Mammalian proteins are the targets of a wide range of
Enz Ser
OH
Enz Ser
O PO3
2-
covalent modification processes. Modifications such as
PHOSPHATASE
glycosylation, hydroxylation, and fatty acid acylation
introduce new structural features into newly synthe-
Mg2+
sized proteins that tend to persist for the lifetime of the
Pi
H2O
protein. Among the covalent modifications that regu-
late protein function (eg, methylation, adenylylation),
Figure 9-7. Covalent modification of a regulated en-
the most common by far is phosphorylation-dephos-
zyme by phosphorylation-dephosphorylation of a seryl
phorylation. Protein kinases phosphorylate proteins by
residue.
78
/
CHAPTER 9
accounts for the frequency of phosphorylation-dephos-
Table 9-1. Examples of mammalian enzymes
phorylation as a mechanism for regulatory control.
whose catalytic activity is altered by covalent
Phosphorylation-dephosphorylation permits the func-
phosphorylation-dephosphorylation.
tional properties of the affected enzyme to be altered
only for as long as it serves a specific need. Once the
Activity State1
need has passed, the enzyme can be converted back to
its original form, poised to respond to the next stimula-
Enzyme
Low
High
tory event. A second factor underlying the widespread
Acetyl-CoA carboxylase
EP
E
use of protein phosphorylation-dephosphorylation lies
Glycogen synthase
EP
E
in the chemical properties of the phosphoryl group it-
Pyruvate dehydrogenase
EP
E
self. In order to alter an enzyme’s functional properties,
HMG-CoA reductase
EP
E
any modification of its chemical structure must influ-
Glycogen phosphorylase
E
EP
ence the protein’s three-dimensional configuration.
Citrate lyase
E
EP
The high charge density of protein-bound phosphoryl
Phosphorylase b kinase
E
EP
groups—generally −2 at physiologic pH—and their
HMG-CoA reductase kinase
E
EP
propensity to form salt bridges with arginyl residues
1E, dephosphoenzyme; EP, phosphoenzyme.
make them potent agents for modifying protein struc-
ture and function. Phosphorylation generally targets
amino acids distant from the catalytic site itself. Conse-
quent conformational changes then influence an en-
phosphorylation at different sites, or between phosphory-
zyme’s intrinsic catalytic efficiency or other properties.
lation sites and allosteric sites provides the basis for
In this sense, the sites of phosphorylation and other co-
regulatory networks that integrate multiple environ-
valent modifications can be considered another form of
mental input signals to evoke an appropriate coordi-
allosteric site. However, in this case the “allosteric li-
nated cellular response. In these sophisticated regula-
gand” binds covalently to the protein.
tory networks, individual enzymes respond to different
environmental signals. For example, if an enzyme can
be phosphorylated at a single site by more than one
PROTEIN PHOSPHORYLATION
protein kinase, it can be converted from a catalytically
IS EXTREMELY VERSATILE
efficient to an inefficient (inactive) form, or vice versa,
Protein phosphorylation-dephosphorylation is a highly
in response to any one of several signals. If the protein
versatile and selective process. Not all proteins are sub-
kinase is activated in response to a signal different from
ject to phosphorylation, and of the many hydroxyl
the signal that activates the protein phosphatase, the
groups on a protein’s surface, only one or a small subset
phosphoprotein becomes a decision node. The func-
are targeted. While the most common enzyme function
tional output, generally catalytic activity, reflects the
affected is the protein’s catalytic efficiency, phosphory-
phosphorylation state. This state or degree of phos-
lation can also alter the affinity for substrates, location
phorylation is determined by the relative activities of
within the cell, or responsiveness to regulation by al-
the protein kinase and protein phosphatase, a reflection
losteric ligands. Phosphorylation can increase an en-
of the presence and relative strength of the environ-
zyme’s catalytic efficiency, converting it to its active
mental signals that act through each. The ability of
form in one protein, while phosphorylation of another
many protein kinases and protein phosphatases to tar-
converts it into an intrinsically inefficient, or inactive,
get more than one protein provides a means for an en-
form (Table 9-1).
vironmental signal to coordinately regulate multiple
Many proteins can be phosphorylated at multiple
metabolic processes. For example, the enzymes 3-hy-
sites or are subject to regulation both by phosphoryla-
droxy-3-methylglutaryl-CoA reductase and acetyl-CoA
tion-dephosphorylation and by the binding of allosteric
carboxylase—the rate-controlling enzymes for choles-
ligands. Phosphorylation-dephosphorylation at any one
terol and fatty acid biosynthesis, respectively—are
site can be catalyzed by multiple protein kinases or pro-
phosphorylated and inactivated by the AMP-activated
tein phosphatases. Many protein kinases and most pro-
protein kinase. When this protein kinase is activated ei-
tein phosphatases act on more than one protein and are
ther through phosphorylation by yet another protein
themselves interconverted between active and inactive
kinase or in response to the binding of its allosteric acti-
forms by the binding of second messengers or by cova-
vator 5′-AMP, the two major pathways responsible for
lent modification by phosphorylation-dephosphoryla-
the synthesis of lipids from acetyl-CoA both are inhib-
tion.
ited. Interconvertible enzymes and the enzymes respon-
The interplay between protein kinases and protein
sible for their interconversion do not act as mere on
phosphatases, between the functional consequences of
and off switches working independently of one another.
ENZYMES: REGULATION OF ACTIVITIES
/
79
They form the building blocks of biomolecular com-
active site. Secretion as an inactive proenzyme facili-
puters that maintain homeostasis in cells that carry out
tates rapid mobilization of activity in response to in-
a complex array of metabolic processes that must be
jury or physiologic need and may protect the tissue
regulated in response to a broad spectrum of environ-
of origin (eg, autodigestion by proteases).
mental factors.
• Binding of metabolites and second messengers to
sites distinct from the catalytic site of enzymes trig-
Covalent Modification Regulates
gers conformational changes that alter Vmax or the
Metabolite Flow
Km.
• Phosphorylation by protein kinases of specific seryl,
Regulation of enzyme activity by phosphorylation-
threonyl, or tyrosyl residues—and subsequent de-
dephosphorylation has analogies to regulation by feed-
phosphorylation by protein phosphatases—regulates
back inhibition. Both provide for short-term, readily
the activity of many human enzymes. The protein ki-
reversible regulation of metabolite flow in response to
nases and phosphatases that participate in regulatory
specific physiologic signals. Both act without altering
cascades which respond to hormonal or second mes-
gene expression. Both act on early enzymes of a pro-
senger signals constitute a “bio-organic computer”
tracted, often biosynthetic metabolic sequence, and
that can process and integrate complex environmen-
both act at allosteric rather than catalytic sites. Feed-
tal information to produce an appropriate and com-
back inhibition, however, involves a single protein and
prehensive cellular response.
lacks hormonal and neural features. By contrast, regula-
tion of mammalian enzymes by phosphorylation-
dephosphorylation involves several proteins and ATP
REFERENCES
and is under direct neural and hormonal control.
Bray D: Protein molecules as computational elements in living
cells. Nature 1995;376:307.
SUMMARY
Graves DJ, Martin BL, Wang JH: Co- and Post-translational Modi-
fication of Proteins: Chemical Principles and Biological Effects.
• Homeostasis involves maintaining a relatively con-
Oxford Univ Press, 1994.
stant intracellular and intra-organ environment de-
Johnson LN, Barford D: The effect of phosphorylation on the
spite wide fluctuations in the external environment
structure and function of proteins. Annu Rev Biophys Bio-
via appropriate changes in the rates of biochemical
mol Struct 1993;22:199.
reactions in response to physiologic need.
Marks F (editor): Protein Phosphorylation. VCH Publishers, 1996.
• The substrates for most enzymes are usually present
Pilkis SJ et al: 6-Phosphofructo-2-kinase/fructose-2,6-bisphospha-
at a concentration close to Km. This facilitates passive
tase: A metabolic signaling enzyme. Annu Rev Biochem
control of the rates of product formation response to
1995;64:799.
changes in levels of metabolic intermediates.
Scriver CR et al (editors): The Metabolic and Molecular Bases of
Inherited Disease, 8th ed. McGraw-Hill, 2000.
• Active control of metabolite flux involves changes in
Sitaramayya A (editor): Introduction to Cellular Signal Transduction.
the concentration, catalytic activity, or both of an en-
Birkhauser, 1999.
zyme that catalyzes a committed, rate-limiting reac-
Stadtman ER, Chock PB (editors): Current Topics in Cellular Regu-
tion.
lation. Academic Press, 1969 to the present.
• Selective proteolysis of catalytically inactive proen-
Weber G (editor): Advances in Enzyme Regulation. Pergamon Press,
zymes initiates conformational changes that form the
1963 to the present.
SECTION II
Bioenergetics & the Metabolism
of Carbohydrates & Lipids
Bioenergetics:The Role of ATP
10
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
the system to another or may be transformed into an-
other form of energy. In living systems, chemical en-
Bioenergetics, or biochemical thermodynamics, is the
ergy may be transformed into heat or into electrical, ra-
study of the energy changes accompanying biochemical
diant, or mechanical energy.
reactions. Biologic systems are essentially isothermic
The second law of thermodynamics states that the
and use chemical energy to power living processes.
total entropy of a system must increase if a process
How an animal obtains suitable fuel from its food to
is to occur spontaneously. Entropy is the extent of
provide this energy is basic to the understanding of nor-
disorder or randomness of the system and becomes
mal nutrition and metabolism. Death from starvation
maximum as equilibrium is approached. Under condi-
occurs when available energy reserves are depleted, and
tions of constant temperature and pressure, the rela-
certain forms of malnutrition are associated with energy
tionship between the free energy change (∆G) of a re-
imbalance (marasmus). Thyroid hormones control the
acting system and the change in entropy
(∆S) is
rate of energy release (metabolic rate), and disease re-
expressed by the following equation, which combines
sults when they malfunction. Excess storage of surplus
the two laws of thermodynamics:
energy causes obesity, one of the most common dis-
eases of Western society.
∆G = ∆H− T∆S
FREE ENERGY IS THE USEFUL ENERGY
where ∆H is the change in enthalpy (heat) and T is the
absolute temperature.
IN A SYSTEM
In biochemical reactions, because ∆H is approxi-
Gibbs change in free energy (∆G) is that portion of the
mately equal to ∆E, the total change in internal energy
total energy change in a system that is available for
of the reaction, the above relationship may be expressed
doing work—ie, the useful energy, also known as the
in the following way:
chemical potential.
∆G = ∆E − T∆S
Biologic Systems Conform to the General
If
∆G is negative, the reaction proceeds sponta-
Laws of Thermodynamics
neously with loss of free energy; ie, it is exergonic. If,
The first law of thermodynamics states that the total
in addition, ∆G is of great magnitude, the reaction goes
energy of a system, including its surroundings, re-
virtually to completion and is essentially irreversible.
mains constant. It implies that within the total system,
On the other hand, if ∆G is positive, the reaction pro-
energy is neither lost nor gained during any change.
ceeds only if free energy can be gained; ie, it is ender-
However, energy may be transferred from one part of
gonic. If, in addition, the magnitude of ∆G is great, the
80
BIOENERGETICS: THE ROLE OF ATP
/
81
system is stable, with little or no tendency for a reaction
occurs with release of free energy. It is coupled to an-
to occur. If ∆G is zero, the system is at equilibrium and
other reaction, in which free energy is required to con-
no net change takes place.
vert metabolite C to metabolite D. The terms exer-
When the reactants are present in concentrations of
gonic and endergonic rather than the normal chemical
1.0 mol/L, ∆G0 is the standard free energy change. For
terms “exothermic” and “endothermic” are used to in-
biochemical reactions, a standard state is defined as
dicate that a process is accompanied by loss or gain, re-
having a pH of 7.0. The standard free energy change at
spectively, of free energy in any form, not necessarily as
this standard state is denoted by ∆G0′.
heat. In practice, an endergonic process cannot exist in-
The standard free energy change can be calculated
dependently but must be a component of a coupled ex-
from the equilibrium constant Keq.
ergonic-endergonic system where the overall net change
is exergonic. The exergonic reactions are termed catab-
olism (generally, the breakdown or oxidation of fuel
0′ = −RT ln K′
∆G
eq
molecules), whereas the synthetic reactions that build
up substances are termed anabolism. The combined
where R is the gas constant and T is the absolute tem-
catabolic and anabolic processes constitute metabo-
perature (Chapter 8). It is important to note that the
lism.
actual ∆G may be larger or smaller than ∆G0′ depend-
If the reaction shown in Figure 10-1 is to go from
ing on the concentrations of the various reactants, in-
left to right, then the overall process must be accompa-
cluding the solvent, various ions, and proteins.
nied by loss of free energy as heat. One possible mecha-
In a biochemical system, an enzyme only speeds up
nism of coupling could be envisaged if a common oblig-
the attainment of equilibrium; it never alters the final
atory intermediate (I) took part in both reactions, ie,
concentrations of the reactants at equilibrium.
A+C→I→B+D
ENDERGONIC PROCESSES PROCEED BY
Some exergonic and endergonic reactions in biologic
COUPLING TO EXERGONIC PROCESSES
systems are coupled in this way. This type of system has
The vital processes—eg, synthetic reactions, muscular
a built-in mechanism for biologic control of the rate of
contraction, nerve impulse conduction, and active
oxidative processes since the common obligatory inter-
transport—obtain energy by chemical linkage, or cou-
mediate allows the rate of utilization of the product of
pling, to oxidative reactions. In its simplest form, this
the synthetic path (D) to determine by mass action the
type of coupling may be represented as shown in Figure
rate at which A is oxidized. Indeed, these relationships
10-1. The conversion of metabolite A to metabolite B
supply a basis for the concept of respiratory control,
the process that prevents an organism from burning out
of control. An extension of the coupling concept is pro-
vided by dehydrogenation reactions, which are coupled
to hydrogenations by an intermediate carrier (Figure
10-2).
An alternative method of coupling an exergonic to
an endergonic process is to synthesize a compound of
high-energy potential in the exergonic reaction and to
incorporate this new compound into the endergonic re-
action, thus effecting a transference of free energy from
the exergonic to the endergonic pathway (Figure 10-3).
The biologic advantage of this mechanism is that the
compound of high potential energy,
∼ E, unlike I
∆G = ∆H− T∆S
Figure 10-1. Coupling of an exergonic to an ender-
Figure 10-2. Coupling of dehydrogenation and hy-
gonic reaction.
drogenation reactions by an intermediate carrier.
82
/
CHAPTER 10
Figure 10-4. Adenosine triphosphate (ATP) shown
as the magnesium complex. ADP forms a similar com-
plex with Mg2+.
Figure 10-3. Transfer of free energy from an exer-
gonic to an endergonic reaction via a high-energy in-
The Intermediate Value for the Free
termediate compound (∼ E ).
Energy of Hydrolysis of ATP Has Important
Bioenergetic Significance
The standard free energy of hydrolysis of a number of
in the previous system, need not be structurally related
biochemically important phosphates is shown in Table
to A, B, C, or D, allowing E to serve as a transducer of
10-1. An estimate of the comparative tendency of each
energy from a wide range of exergonic reactions to an
of the phosphate groups to transfer to a suitable accep-
equally wide range of endergonic reactions or processes,
tor may be obtained from the ∆G0′ of hydrolysis at
such as biosyntheses, muscular contraction, nervous ex-
37 °C. The value for the hydrolysis of the terminal
citation, and active transport. In the living cell, the
principal high-energy intermediate or carrier com-
pound (designated ∼ E in Figure 10-3) is adenosine
triphosphate (ATP).
Table 10-1. Standard free energy of hydrolysis
of some organophosphates of biochemical
HIGH-ENERGY PHOSPHATES PLAY A
importance.1,2
CENTRAL ROLE IN ENERGY CAPTURE
AND TRANSFER
G0
Compound
kJ/mol
kcal/mol
In order to maintain living processes, all organisms
must obtain supplies of free energy from their environ-
Phosphoenolpyruvate
−61.9
−14.8
ment. Autotrophic organisms utilize simple exergonic
Carbamoyl phosphate
−51.4
−12.3
processes; eg, the energy of sunlight (green plants), the
1,3-Bisphosphoglycerate
−49.3
−11.8
reaction Fe2+ → Fe3+ (some bacteria). On the other
(to 3-phosphoglycerate)
hand, heterotrophic organisms obtain free energy by
Creatine phosphate
−43.1
−10.3
coupling their metabolism to the breakdown of com-
ATP → ADP + Pi
−30.5
−7.3
plex organic molecules in their environment. In all
ADP → AMP + Pi
−27.6
−6.6
these organisms, ATP plays a central role in the trans-
Pyrophosphate
−27.6
−6.6
ference of free energy from the exergonic to the ender-
Glucose 1-phosphate
−20.9
−5.0
gonic processes
(Figure
10-3). ATP is a nucleoside
Fructose 6-phosphate
−15.9
−3.8
triphosphate containing adenine, ribose, and three
AMP
−14.2
−3.4
phosphate groups. In its reactions in the cell, it func-
Glucose 6-phosphate
−13.8
−3.3
tions as the Mg2+ complex (Figure 10-4).
Glycerol 3-phosphate
−9.2
−2.2
The importance of phosphates in intermediary me-
1Pi, inorganic orthophosphate.
tabolism became evident with the discovery of the role
2Values for ATP and most others taken from Krebs and Kornberg
of ATP, adenosine diphosphate (ADP), and inorganic
(1957). They differ between investigators depending on the pre-
phosphate (Pi) in glycolysis (Chapter 17).
cise conditions under which the measurements are made.
BIOENERGETICS: THE ROLE OF ATP
/
83
phosphate of ATP divides the list into two groups.
Low-energy phosphates, exemplified by the ester
phosphates found in the intermediates of glycolysis,
have ∆G0′ values smaller than that of ATP, while in
high-energy phosphates the value is higher than that
of ATP. The components of this latter group, including
ATP, are usually anhydrides (eg, the 1-phosphate of
1,3-bisphosphoglycerate), enolphosphates
(eg, phos-
phoenolpyruvate), and phosphoguanidines (eg, creatine
phosphate, arginine phosphate). The intermediate posi-
tion of ATP allows it to play an important role in en-
ergy transfer. The high free energy change on hydrolysis
of ATP is due to relief of charge repulsion of adjacent
negatively charged oxygen atoms and to stabilization of
the reaction products, especially phosphate, as reso-
nance hybrids. Other “high-energy compounds” are
thiol esters involving coenzyme A (eg, acetyl-CoA), acyl
carrier protein, amino acid esters involved in protein
synthesis, S-adenosylmethionine
(active methionine),
UDPGlc (uridine diphosphate glucose), and PRPP
(5-phosphoribosyl-1-pyrophosphate).
High-Energy Phosphates Are
Designated by ~ P
Figure 10-5. Structure of ATP, ADP, and AMP show-
ing the position and the number of high-energy phos-
The symbol ∼ P indicates that the group attached to
phates (∼ P ).
the bond, on transfer to an appropriate acceptor, results
in transfer of the larger quantity of free energy. For this
reason, the term group transfer potential is preferred
by some to “high-energy bond.” Thus, ATP contains
two high-energy phosphate groups and ADP contains
one, whereas the phosphate in AMP (adenosine mono-
phosphate) is of the low-energy type, since it is a nor-
mal ester link (Figure 10-5).
HIGH-ENERGY PHOSPHATES ACT AS THE
“ENERGY CURRENCY” OF THE CELL
ATP is able to act as a donor of high-energy phosphate
to form those compounds below it in Table 10-1. Like-
wise, with the necessary enzymes, ADP can accept
high-energy phosphate to form ATP from those com-
pounds above ATP in the table. In effect, an ATP/
ADP cycle connects those processes that generate ∼ P
to those processes that utilize ∼ P
(Figure 10-6), con-
tinuously consuming and regenerating ATP. This oc-
curs at a very rapid rate, since the total ATP/ADP pool
is extremely small and sufficient to maintain an active
tissue for only a few seconds.
There are three major sources of ∼ P taking part in
energy conservation or energy capture:
(1) Oxidative phosphorylation: The greatest quan-
Figure 10-6.
Role of ATP/ADP cycle in transfer of
titative source of ∼ P in aerobic organisms. Free energy
high-energy phosphate.
84
/
CHAPTER 10
(1) Glucose+Pi → Glucose 6- phosphate+ H2O
( ∆G0′ = +13.8 k/ mol)
To take place, the reaction must be coupled with an-
other—more exergonic—reaction such as the hydroly-
sis of the terminal phosphate of ATP.
(2) ATP → ADP+Pi (∆G0′ = −30.5 kJ / mol)
Figure 10-7. Transfer of high-energy phosphate be-
tween ATP and creatine.
When (1) and (2) are coupled in a reaction catalyzed by
hexokinase, phosphorylation of glucose readily pro-
ceeds in a highly exergonic reaction that under physio-
logic conditions is irreversible. Many “activation” reac-
comes from respiratory chain oxidation using molecular
tions follow this pattern.
O2 within mitochondria (Chapter 11).
(2) Glycolysis: A net formation of two ∼ P results
from the formation of lactate from one molecule of glu-
Adenylyl Kinase (Myokinase)
cose, generated in two reactions catalyzed by phospho-
Interconverts Adenine Nucleotides
glycerate kinase and pyruvate kinase, respectively (Fig-
This enzyme is present in most cells. It catalyzes the fol-
ure 17-2).
lowing reaction:
(3) The citric acid cycle: One ∼ P is generated di-
rectly in the cycle at the succinyl thiokinase step (Figure
16-3).
Phosphagens act as storage forms of high-energy
phosphate and include creatine phosphate, occurring in
vertebrate skeletal muscle, heart, spermatozoa, and
This allows:
brain; and arginine phosphate, occurring in inverte-
(1) High-energy phosphate in ADP to be used in
brate muscle. When ATP is rapidly being utilized as a
the synthesis of ATP.
source of energy for muscular contraction, phosphagens
(2) AMP, formed as a consequence of several acti-
permit its concentrations to be maintained, but when
vating reactions involving ATP, to be recovered by
the ATP/ADP ratio is high, their concentration can in-
rephosphorylation to ADP.
crease to act as a store of high-energy phosphate (Figure
(3) AMP to increase in concentration when ATP
10-7).
becomes depleted and act as a metabolic (allosteric) sig-
When ATP acts as a phosphate donor to form those
nal to increase the rate of catabolic reactions, which in
compounds of lower free energy of hydrolysis (Table
turn lead to the generation of more ATP (Chapter 19).
10-1), the phosphate group is invariably converted to
one of low energy, eg,
When ATP Forms AMP, Inorganic
Pyrophosphate (PPi) Is Produced
This occurs, for example, in the activation of long-
chain fatty acids (Chapter 22):
ATP Allows the Coupling of
Thermodynamically Unfavorable
Reactions to Favorable Ones
This reaction is accompanied by loss of free energy
as heat, which ensures that the activation reaction will
The phosphorylation of glucose to glucose
6-phos-
go to the right; and is further aided by the hydrolytic
phate, the first reaction of glycolysis (Figure 17-2), is
splitting of PPi, catalyzed by inorganic pyrophospha-
highly endergonic and cannot proceed under physio-
tase, a reaction that itself has a large ∆G0′ of −27.6 kJ/
logic conditions.
BIOENERGETICS: THE ROLE OF ATP
/
85
Thus, adenylyl kinase is a specialized monophosphate
kinase.
SUMMARY
• Biologic systems use chemical energy to power the
living processes.
• Exergonic reactions take place spontaneously with
loss of free energy (∆G is negative). Endergonic reac-
tions require the gain of free energy (∆G is positive)
and only occur when coupled to exergonic reactions.
• ATP acts as the “energy currency” of the cell, trans-
Phosphate cycles and interchange of
Figure 10-8.
ferring free energy derived from substances of higher
adenine nucleotides.
energy potential to those of lower energy potential.
REFERENCES
mol. Note that activations via the pyrophosphate path-
way result in the loss of two ∼ P rather than one ∼ P as
de Meis L: The concept of energy-rich phosphate compounds:
Water, transport ATPases, and entropy energy. Arch Bio-
occurs when ADP and Pi are formed.
chem Biophys 1993;306:287.
Ernster L (editor): Bioenergetics. Elsevier, 1984.
Harold FM: The Vital Force: A Study of Bioenergetics. Freeman,
1986.
Klotz IM: Introduction to Biomolecular Energetics. Academic Press,
1986.
A combination of the above reactions makes it pos-
Krebs HA, Kornberg HL: Energy Transformations in Living Matter.
Springer, 1957.
sible for phosphate to be recycled and the adenine nu-
cleotides to interchange (Figure 10-8).
Other Nucleoside Triphosphates
Participate in the Transfer of
High-Energy Phosphate
By means of the enzyme nucleoside diphosphate ki-
nase, UTP, GTP, and CTP can be synthesized from
their diphosphates, eg,
All of these triphosphates take part in phosphoryla-
tions in the cell. Similarly, specific nucleoside mono-
phosphate kinases catalyze the formation of nucleoside
diphosphates from the corresponding monophosphates.
Biologic Oxidation
11
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
groups: oxidases, dehydrogenases, hydroperoxidases,
and oxygenases.
Chemically, oxidation is defined as the removal of elec-
trons and reduction as the gain of electrons. Thus, oxi-
dation is always accompanied by reduction of an elec-
OXIDASES USE OXYGEN AS A
tron acceptor. This principle of oxidation-reduction
HYDROGEN ACCEPTOR
applies equally to biochemical systems and is an impor-
tant concept underlying understanding of the nature of
Oxidases catalyze the removal of hydrogen from a sub-
biologic oxidation. Note that many biologic oxidations
strate using oxygen as a hydrogen acceptor.* They form
can take place without the participation of molecular
water or hydrogen peroxide as a reaction product (Fig-
oxygen, eg, dehydrogenations. The life of higher ani-
ure 11-1).
mals is absolutely dependent upon a supply of oxygen
for respiration, the process by which cells derive energy
Some Oxidases Contain Copper
in the form of ATP from the controlled reaction of hy-
drogen with oxygen to form water. In addition, molec-
Cytochrome oxidase is a hemoprotein widely distrib-
ular oxygen is incorporated into a variety of substrates
uted in many tissues, having the typical heme pros-
by enzymes designated as oxygenases; many drugs, pol-
thetic group present in myoglobin, hemoglobin, and
lutants, and chemical carcinogens (xenobiotics) are me-
other cytochromes (Chapter 6). It is the terminal com-
tabolized by enzymes of this class, known as the cy-
ponent of the chain of respiratory carriers found in mi-
tochrome P450 system. Administration of oxygen can
tochondria and transfers electrons resulting from the
be lifesaving in the treatment of patients with respira-
oxidation of substrate molecules by dehydrogenases to
tory or circulatory failure.
their final acceptor, oxygen. The enzyme is poisoned by
carbon monoxide, cyanide, and hydrogen sulfide. It has
. It is now known that
also been termed cytochrome a3
FREE ENERGY CHANGES CAN
cytochromes a and a3 are combined in a single protein,
and the complex is known as cytochrome aa3. It con-
BE EXPRESSED IN TERMS
tains two molecules of heme, each having one Fe atom
OF REDOX POTENTIAL
that oscillates between Fe3+ and Fe2+ during oxidation
In reactions involving oxidation and reduction, the free
and reduction. Furthermore, two atoms of Cu are pre-
energy change is proportionate to the tendency of reac-
sent, each associated with a heme unit.
tants to donate or accept electrons. Thus, in addition to
expressing free energy change in terms of ∆G0′ (Chapter
Other Oxidases Are Flavoproteins
10), it is possible, in an analogous manner, to express it
numerically as an oxidation-reduction or redox po-
Flavoprotein enzymes contain flavin mononucleotide
tential (E′0). The redox potential of a system (E0) is
(FMN) or flavin adenine dinucleotide (FAD) as pros-
usually compared with the potential of the hydrogen
thetic groups. FMN and FAD are formed in the body
electrode (0.0 volts at pH 0.0). However, for biologic
from the vitamin riboflavin (Chapter 45). FMN and
systems, the redox potential (E′0)is normally expressed
FAD are usually tightly—but not covalently—bound to
at pH 7.0, at which pH the electrode potential of the
their respective apoenzyme proteins. Metalloflavopro-
hydrogen electrode is −0.42 volts. The redox potentials
teins contain one or more metals as essential cofactors.
of some redox systems of special interest in mammalian
Examples of flavoprotein enzymes include L-amino
biochemistry are shown in Table 11-1. The relative po-
acid oxidase, an FMN-linked enzyme found in kidney
sitions of redox systems in the table allows prediction of
with general specificity for the oxidative deamination of
the direction of flow of electrons from one redox couple
to another.
Enzymes involved in oxidation and reduction are
* The term “oxidase” is sometimes used collectively to denote all
called oxidoreductases and are classified into four
enzymes that catalyze reactions involving molecular oxygen.
86
BIOLOGIC OXIDATION
/
87
Table 11-1. Some redox potentials of special
versible, these properties enable reducing equivalents to
interest in mammalian oxidation systems.
be freely transferred within the cell. This type of reac-
tion, which enables one substrate to be oxidized at the
expense of another, is particularly useful in enabling ox-
System
E0 Volts
idative processes to occur in the absence of oxygen,
H+/H2
−0.42
such as during the anaerobic phase of glycolysis (Figure
NAD+/NADH
−0.32
17-2).
Lipoate; ox/red
−0.29
(2) As components in the respiratory chain of elec-
Acetoacetate/3-hydroxybutyrate
−0.27
tron transport from substrate to oxygen (Figure 12-3).
Pyruvate/lactate
−0.19
Oxaloacetate/malate
−0.17
Fumarate/succinate
+0.03
Many Dehydrogenases Depend
Cytochrome b; Fe3+/Fe2+
+0.08
on Nicotinamide Coenzymes
Ubiquinone; ox/red
+0.10
Cytochrome c1; Fe3+/Fe2+
+0.22
These dehydrogenases use nicotinamide adenine di-
Cytochrome a; Fe3+/Fe2+
+0.29
nucleotide (NAD+) or nicotinamide adenine dinu-
Oxygen/water
+0.82
cleotide phosphate
(NADP+)—or both—and are
formed in the body from the vitamin niacin (Chapter
45). The coenzymes are reduced by the specific sub-
strate of the dehydrogenase and reoxidized by a suitable
the naturally occurring L-amino acids; xanthine oxi-
electron acceptor (Figure 11-4).They may freely and
dase, which contains molybdenum and plays an impor-
reversibly dissociate from their respective apoenzymes.
tant role in the conversion of purine bases to uric acid
Generally, NAD-linked dehydrogenases catalyze ox-
(Chapter 34), and is of particular significance in uri-
idoreduction reactions in the oxidative pathways of me-
cotelic animals (Chapter 29); and aldehyde dehydro-
tabolism, particularly in glycolysis, in the citric acid
genase, an FAD-linked enzyme present in mammalian
cycle, and in the respiratory chain of mitochondria.
livers, which contains molybdenum and nonheme iron
NADP-linked dehydrogenases are found characteristi-
and acts upon aldehydes and N-heterocyclic substrates.
cally in reductive syntheses, as in the extramitochon-
The mechanisms of oxidation and reduction of these
drial pathway of fatty acid synthesis and steroid synthe-
enzymes are complex. Evidence suggests a two-step re-
sis—and also in the pentose phosphate pathway.
action as shown in Figure 11-2.
Other Dehydrogenases Depend
DEHYDROGENASES CANNOT USE
on Riboflavin
OXYGEN AS A HYDROGEN ACCEPTOR
The flavin groups associated with these dehydrogenases
There are a large number of enzymes in this class. They
are similar to FMN and FAD occurring in oxidases.
perform two main functions:
They are generally more tightly bound to their apoen-
zymes than are the nicotinamide coenzymes. Most of
(1) Transfer of hydrogen from one substrate to an-
the riboflavin-linked dehydrogenases are concerned
other in a coupled oxidation-reduction reaction (Figure
with electron transport in (or to) the respiratory chain
11-3). These dehydrogenases are specific for their sub-
(Chapter 12). NADH dehydrogenase acts as a carrier
strates but often utilize common coenzymes or hydro-
of electrons between NADH and the components of
gen carriers, eg, NAD+. Since the reactions are re-
higher redox potential (Figure 12-3). Other dehydro-
genases such as succinate dehydrogenase, acyl-CoA
dehydrogenase, and mitochondrial glycerol-3-phos-
phate dehydrogenase transfer reducing equivalents di-
AH2
1/2O2
AH2
O2
rectly from the substrate to the respiratory chain (Fig-
(Red)
ure
12-4). Another role of the flavin-dependent
OXIDASE
OXIDASE
dehydrogenases is in the dehydrogenation (by dihy-
drolipoyl dehydrogenase) of reduced lipoate, an inter-
A
H2O
A
H2O2
mediate in the oxidative decarboxylation of pyruvate
(Ox)
and α-ketoglutarate
(Figures
12-4 and 17-5). The
A
B
electron-transferring flavoprotein is an intermediary
Figure 11-1. Oxidation of a metabolite catalyzed by
carrier of electrons between acyl-CoA dehydrogenase
an oxidase (A) forming H2O, (B) forming H2O2.
and the respiratory chain (Figure 12-4).
88
/
CHAPTER 11
R
R
R
H
H
H3C
N
N
O H3C
N
N
O
H3C
N
N
O
NH
NH
NH
H3C
H3C
H3C
N
N
N
O
O
H
O
H
H
(H+ + e-)
(H+ + e-)
Figure 11-2. Oxidoreduction of isoalloxazine ring in flavin nucleotides via a semi-
quinone (free radical) intermediate (center).
Cytochromes May Also Be Regarded
Peroxidases Reduce Peroxides Using
as Dehydrogenases
Various Electron Acceptors
The cytochromes are iron-containing hemoproteins in
Peroxidases are found in milk and in leukocytes,
which the iron atom oscillates between Fe3+ and Fe2+
platelets, and other tissues involved in eicosanoid me-
during oxidation and reduction. Except for cytochrome
tabolism (Chapter 23). The prosthetic group is proto-
oxidase (previously described), they are classified as de-
heme. In the reaction catalyzed by peroxidase, hydro-
hydrogenases. In the respiratory chain, they are in-
gen peroxide is reduced at the expense of several
volved as carriers of electrons from flavoproteins on the
substances that will act as electron acceptors, such as
one hand to cytochrome oxidase on the other (Figure
ascorbate, quinones, and cytochrome c. The reaction
12-4). Several identifiable cytochromes occur in the
catalyzed by peroxidase is complex, but the overall reac-
respiratory chain, ie, cytochromes b, c1, c, a, and a3 (cy-
tion is as follows:
tochrome oxidase). Cytochromes are also found in
other locations, eg, the endoplasmic reticulum
(cy-
PEROXIDASE
tochromes P450 and b5), and in plant cells, bacteria,
H2O2 + AH2
2H2O + A
and yeasts.
In erythrocytes and other tissues, the enzyme glu-
tathione peroxidase, containing selenium as a pros-
HYDROPEROXIDASES USE HYDROGEN
thetic group, catalyzes the destruction of H2O2 and
PEROXIDE OR AN ORGANIC PEROXIDE
lipid hydroperoxides by reduced glutathione, protecting
AS SUBSTRATE
membrane lipids and hemoglobin against oxidation by
Two type of enzymes found both in animals and plants
peroxides (Chapter 20).
fall into this category: peroxidases and catalase.
Hydroperoxidases protect the body against harmful
Catalase Uses Hydrogen Peroxide as
peroxides. Accumulation of peroxides can lead to gen-
Electron Donor & Electron Acceptor
eration of free radicals, which in turn can disrupt mem-
branes and perhaps cause cancer and atherosclerosis.
Catalase is a hemoprotein containing four heme groups.
(See Chapters 14 and 45.)
In addition to possessing peroxidase activity, it is able
to use one molecule of H2O2 as a substrate electron
donor and another molecule of H2O2 as an oxidant or
electron acceptor.
AH2
Carrier
BH2
(Red)
(Ox)
(Red)
CATALASE
2H2O2
2H2O + O2
A
Carrier-H2
B
(Ox)
(Red)
(Ox)
Under most conditions in vivo, the peroxidase activity
DEHYDROGENASE
DEHYDROGENASE
SPECIFIC FOR A
SPECIFIC FOR B
of catalase seems to be favored. Catalase is found in
blood, bone marrow, mucous membranes, kidney, and
Figure 11-3. Oxidation of a metabolite catalyzed by
liver. Its function is assumed to be the destruction of
coupled dehydrogenases.
hydrogen peroxide formed by the action of oxidases.
BIOLOGIC OXIDATION
/
89
H
4
Figure 11-4. Mechanism of oxidation
DEHYDROGENASE
CONH2
SPECIFIC FOR A
and reduction of nicotinamide coen-
zymes. There is stereospecificity about
AH2
N
A Form
position 4 of nicotinamide when it is re-
H
R
duced by a substrate AH2. One of the hy-
drogen atoms is removed from the sub-
4
CONH2
A + H+
strate as a hydrogen nucleus with two
electrons (hydride ion, H−) and is trans-
+
N
ferred to the 4 position, where it may be
H
attached in either the A or the B position
R
AH2
4
CONH2
according to the specificity determined
by the particular dehydrogenase catalyz-
DEHYDROGENASE
SPECIFIC FOR B
ing the reaction. The remaining hydro-
N
B Form
gen of the hydrogen pair removed from
R
the substrate remains free as a hydro-
NAD+ + AH2
NADH + H+ + A
gen ion.
Peroxisomes are found in many tissues, including liver.
Monooxygenases (Mixed-Function
They are rich in oxidases and in catalase, Thus, the en-
Oxidases, Hydroxylases) Incorporate
zymes that produce H2O2 are grouped with the enzyme
Only One Atom of Molecular Oxygen
that destroys it. However, mitochondrial and microso-
Into the Substrate
mal electron transport systems as well as xanthine oxi-
dase must be considered as additional sources of H2O2.
The other oxygen atom is reduced to water, an addi-
tional electron donor or cosubstrate (Z) being necessary
for this purpose.
OXYGENASES CATALYZE THE DIRECT
TRANSFER & INCORPORATION
A—H+O +ZH →A—OH+H O+Z
2
2
2
OF OXYGEN INTO A SUBSTRATE
MOLECULE
Cytochromes P450 Are Monooxygenases
Oxygenases are concerned with the synthesis or degra-
Important for the Detoxification of Many
dation of many different types of metabolites. They cat-
Drugs & for the Hydroxylation of Steroids
alyze the incorporation of oxygen into a substrate mole-
cule in two steps: (1) oxygen is bound to the enzyme at
Cytochromes P450 are an important superfamily of
the active site, and (2) the bound oxygen is reduced or
heme-containing monooxgenases, and more than 1000
transferred to the substrate. Oxygenases may be divided
such enzymes are known. Both NADH and NADPH
into two subgroups, as follows.
donate reducing equivalents for the reduction of these
cytochromes (Figure 11-5), which in turn are oxidized
by substrates in a series of enzymatic reactions collectively
Dioxygenases Incorporate Both Atoms
known as the hydroxylase cycle (Figure 11-6). In liver
of Molecular Oxygen Into the Substrate
microsomes, cytochromes P450 are found together with
The basic reaction is shown below:
cytochrome b5 and have an important role in detoxifica-
tion. Benzpyrene, aminopyrine, aniline, morphine, and
A + O →AO
benzphetamine are hydroxylated, increasing their solubil-
2
2
ity and aiding their excretion. Many drugs such as phe-
Examples include the liver enzymes, homogentisate
nobarbital have the ability to induce the formation of mi-
dioxygenase
(oxidase) and
3-hydroxyanthranilate
crosomal enzymes and of cytochromes P450.
dioxygenase (oxidase), that contain iron; and L-trypto-
Mitochondrial cytochrome P450 systems are found
phan dioxygenase
(tryptophan pyrrolase)
(Chapter
in steroidogenic tissues such as adrenal cortex, testis,
30), that utilizes heme.
ovary, and placenta and are concerned with the biosyn-
90
/
CHAPTER 11
CN-
NADH
Flavoprotein2
Cyt b5
Stearyl-CoA desaturase
-
Amine oxidase, etc
Flavoprotein3
NADPH
Flavoprotein1
Cyt P450
Hydroxylation
Lipid peroxidation
Heme oxygenase
Figure 11-5. Electron transport chain in microsomes. Cyanide (CN−) inhibits the
indicated step.
thesis of steroid hormones from cholesterol (hydroxyla-
ing rise to free radical chain reactions (Chapter 14).
tion at C22 and C20 in side-chain cleavage and at the
The ease with which superoxide can be formed from
11β and 18 positions). In addition, renal systems cat-
oxygen in tissues and the occurrence of superoxide dis-
alyzing 1α- and 24-hydroxylations of 25-hydroxychole-
mutase, the enzyme responsible for its removal in all
calciferol in vitamin D metabolism—and cholesterol
aerobic organisms (although not in obligate anaerobes)
7α-hydroxylase and sterol 27-hydroxylase involved in
indicate that the potential toxicity of oxygen is due to
bile acid biosynthesis in the liver (Chapter 26)—are
its conversion to superoxide.
P450 enzymes.
Superoxide is formed when reduced flavins—pre-
sent, for example, in xanthine oxidase—are reoxidized
univalently by molecular oxygen.
SUPEROXIDE DISMUTASE PROTECTS
AEROBIC ORGANISMS AGAINST
⋅
+
Enz − Flavin − H +O
2
2
→Enz − Flavin − H+O
2
+H
OXYGEN TOXICITY
Transfer of a single electron to O2 generates the poten-
Superoxide can reduce oxidized cytochrome c
tially damaging superoxide anion free radical (O2−⋅ ),
the destructive effects of which are amplified by its giv-
⋅
3+
2+
2
O +Cyt c (Fe
)→
2
O +Cyt c (Fe
)
Substrate A-H
P450-A-H
Fe3+
e-
P450-A-H
P450
NADPH-CYT P450 REDUCTASE
2+
Fe
Fe3+
NADP+
FADH2
2Fe2S23+
O2
e-
-
+
2+
NADPH + H
FAD
2Fe2S
2
CO
2H+
P450-A-H
Fe2+ O2
H2O
P450-A-H
-
Fe2+
O2
A-OH
Figure 11-6. Cytochrome P450 hydroxylase cycle in microsomes. The system shown is typical
of steroid hydroxylases of the adrenal cortex. Liver microsomal cytochrome P450 hydroxylase does
not require the iron-sulfur protein Fe2S2. Carbon monoxide (CO) inhibits the indicated step.
BIOLOGIC OXIDATION
/
91
or be removed by superoxide dismutase.
REFERENCES
SUPEROXIDE
Babcock GT, Wikstrom M: Oxygen activation and the conserva-
DISMUTASE
tion of energy in cell respiration. Nature 1992;356:301.
O2− + O2− + 2H+
H2O2 + O2
Coon MJ et al: Cytochrome P450: Progress and predictions.
FASEB J 1992;6:669.
In this reaction, superoxide acts as both oxidant and
Ernster L (editor): Bioenergetics. Elsevier, 1984.
reductant. Thus, superoxide dismutase protects aerobic
Mammaerts GP, Van Veldhoven PP: Role of peroxisomes in mam-
organisms against the potential deleterious effects of su-
malian metabolism. Cell Biochem Funct 1992;10:141.
peroxide. The enzyme occurs in all major aerobic tis-
Nicholls DG: Cytochromes and Cell Respiration. Carolina Biological
sues in the mitochondria and the cytosol. Although ex-
Supply Company, 1984.
posure of animals to an atmosphere of 100% oxygen
Raha S, Robinson BH: Mitochondria, oxygen free radicals, disease
causes an adaptive increase in superoxide dismutase,
and aging. Trends Biochem Sci 2000;25:502.
particularly in the lungs, prolonged exposure leads to
Tyler DD: The Mitochondrion in Health and Disease. VCH Pub-
lung damage and death. Antioxidants, eg, α-tocopherol
lishers, 1992.
(vitamin E), act as scavengers of free radicals and reduce
Tyler DD, Sutton CM: Respiratory enzyme systems in mitochon-
the toxicity of oxygen (Chapter 45).
drial membranes. In: Membrane Structure and Function, vol
5. Bittar EE (editor). Wiley, 1984.
Yang CS, Brady JF, Hong JY: Dietary effects on cytochromes
SUMMARY
P450, xenobiotic metabolism, and toxicity. FASEB J 1992;
6:737.
• In biologic systems, as in chemical systems, oxidation
(loss of electrons) is always accompanied by reduc-
tion of an electron acceptor.
• Oxidoreductases have a variety of functions in me-
tabolism; oxidases and dehydrogenases play major
roles in respiration; hydroperoxidases protect the
body against damage by free radicals; and oxygenases
mediate the hydroxylation of drugs and steroids.
• Tissues are protected from oxygen toxicity caused by
the superoxide free radical by the specific enzyme su-
peroxide dismutase.
The Respiratory Chain &
12
Oxidative Phosphorylation
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
trapping the liberated free energy as high-energy phos-
phate, and the enzymes of β-oxidation and of the citric
Aerobic organisms are able to capture a far greater pro-
acid cycle (Chapters 22 and 16) that produce most of
portion of the available free energy of respiratory sub-
the reducing equivalents.
strates than anaerobic organisms. Most of this takes
place inside mitochondria, which have been termed the
“powerhouses” of the cell. Respiration is coupled to the
Components of the Respiratory Chain
generation of the high-energy intermediate, ATP, by
Are Arranged in Order of Increasing
oxidative phosphorylation, and the chemiosmotic
Redox Potential
theory offers insight into how this is accomplished. A
Hydrogen and electrons flow through the respiratory
number of drugs (eg, amobarbital) and poisons (eg,
chain (Figure 12-3) through a redox span of 1.1 V
cyanide, carbon monoxide) inhibit oxidative phos-
from NAD+/NADH to O2/2H2O (Table 11-1). The
phorylation, usually with fatal consequences. Several in-
respiratory chain consists of a number of redox carriers
herited defects of mitochondria involving components
that proceed from the NAD-linked dehydrogenase sys-
of the respiratory chain and oxidative phosphorylation
tems, through flavoproteins and cytochromes, to mole-
have been reported. Patients present with myopathy
cular oxygen. Not all substrates are linked to the respi-
and encephalopathy and often have lactic acidosis.
ratory chain through NAD-specific dehydrogenases;
some, because their redox potentials are more positive
(eg, fumarate/succinate; Table
11-1), are linked di-
SPECIFIC ENZYMES ACT AS MARKERS
rectly to flavoprotein dehydrogenases, which in turn are
OF COMPARTMENTS SEPARATED BY
linked to the cytochromes of the respiratory chain (Fig-
THE MITOCHONDRIAL MEMBRANES
ure 12-4).
Ubiquinone or Q (coenzyme Q) (Figure 12-5)
Mitochondria have an outer membrane that is perme-
links the flavoproteins to cytochrome b, the member of
able to most metabolites, an inner membrane that is
the cytochrome chain of lowest redox potential. Q ex-
selectively permeable, and a matrix within
(Figure
ists in the oxidized quinone or reduced quinol form
12-1). The outer membrane is characterized by the
under aerobic or anaerobic conditions, respectively.
presence of various enzymes, including acyl-CoA syn-
The structure of Q is very similar to that of vitamin K
thetase and glycerolphosphate acyltransferase. Adenylyl
and vitamin E (Chapter 45) and of plastoquinone,
kinase and creatine kinase are found in the intermem-
found in chloroplasts. Q acts as a mobile component of
brane space. The phospholipid cardiolipin is concen-
the respiratory chain that collects reducing equivalents
trated in the inner membrane together with the en-
from the more fixed flavoprotein complexes and passes
zymes of the respiratory chain.
them on to the cytochromes.
An additional component is the iron-sulfur protein
(FeS; nonheme iron) (Figure 12-6). It is associated
THE RESPIRATORY CHAIN COLLECTS
with the flavoproteins (metalloflavoproteins) and with
& OXIDIZES REDUCING EQUIVALENTS
cytochrome b. The sulfur and iron are thought to take
Most of the energy liberated during the oxidation of
part in the oxidoreduction mechanism between flavin
carbohydrate, fatty acids, and amino acids is made
and Q, which involves only a single e− change, the iron
available within mitochondria as reducing equivalents
atom undergoing oxidoreduction between Fe2+ and
(H or electrons) (Figure 12-2). Mitochondria con-
Fe3+.
tain the respiratory chain, which collects and trans-
Pyruvate and α-ketoglutarate dehydrogenase have
ports reducing equivalents directing them to their final
complex systems involving lipoate and FAD prior to
reaction with oxygen to form water, the machinery for
the passage of electrons to NAD, while electron trans-
92
THE RESPIRATORY CHAIN & OXIDATIVE PHOSPHORYLATION
/
93
Electrons flow from Q through the series of cyto-
chromes in order of increasing redox potential to mole-
cular oxygen (Figure 12-4). The terminal cytochrome
aa3 (cytochrome oxidase), responsible for the final com-
Phosphorylating
bination of reducing equivalents with molecular oxy-
complexes
gen, has a very high affinity for oxygen, allowing the
respiratory chain to function at maximum rate until the
tissue has become depleted of O2. Since this is an irre-
MATRIX
versible reaction (the only one in the chain), it gives di-
rection to the movement of reducing equivalents and to
the production of ATP, to which it is coupled.
Functionally and structurally, the components of
Cristae
the respiratory chain are present in the inner mitochon-
drial membrane as four protein-lipid respiratory chain
complexes that span the membrane. Cytochrome c is
the only soluble cytochrome and, together with Q,
INNER
seems to be a more mobile component of the respira-
MEMBRANE
tory chain connecting the fixed complexes
(Figures
12-7 and 12-8).
OUTER
MEMBRANE
THE RESPIRATORY CHAIN PROVIDES
MOST OF THE ENERGY CAPTURED
DURING CATABOLISM
ADP captures, in the form of high-energy phosphate, a
Figure 12-1. Structure of the mitochondrial mem-
significant proportion of the free energy released by
catabolic processes. The resulting ATP has been called
branes. Note that the inner membrane contains many
the energy “currency” of the cell because it passes on
folds, or cristae.
this free energy to drive those processes requiring en-
ergy (Figure 10-6).
fers from other dehydrogenases, eg, L(+)-3-hydroxyacyl-
There is a net direct capture of two high-energy
CoA dehydrogenase, couple directly with NAD.
phosphate groups in the glycolytic reactions
(Table
The reduced NADH of the respiratory chain is in
17-1), equivalent to approximately 103.2 kJ/mol of
turn oxidized by a metalloflavoprotein enzyme—NADH
glucose. (In vivo, ∆G for the synthesis of ATP from
dehydrogenase. This enzyme contains FeS and FMN,
ADP has been calculated as approximately 51.6 kJ/mol.
is tightly bound to the respiratory chain, and passes re-
(It is greater than ∆G0′ for the hydrolysis of ATP as
ducing equivalents on to Q.
given in Table 10-1, which is obtained under standard
FOOD
ATP
Fat
Fatty acids
+
β-Oxidation
Glycerol
O2
Citric
Carbohydrate
Glucose, etc
Acetyl - CoA
acid
2H
H2O
cycle
Respiratory chain
Protein
Amino acids
MITOCHONDRION
ADP
Extramitochondrial sources of
reducing equivalents
Figure 12-2. Role of the respiratory chain of mitochondria in the conversion of food energy to ATP. Oxidation
of the major foodstuffs leads to the generation of reducing equivalents (2H) that are collected by the respiratory
chain for oxidation and coupled generation of ATP.
94
/
CHAPTER 12
AH2
NAD+
FpH2
2Fe3+
H2O
Substrate
Flavoprotein
Cytochromes
1
Figure 12-3. Transport of reducing
A
NADH
Fp
2Fe2+
/2 O2
equivalents through the respiratory
H+
H+
2H+
2H+
chain.
concentrations of 1.0 mol/L.) Since 1 mol of glucose
dent that the respiratory chain is responsible for a large
yields approximately 2870 kJ on complete combustion,
proportion of total ATP formation.
the energy captured by phosphorylation in glycolysis is
small. Two more high-energy phosphates per mole of
Respiratory Control Ensures
glucose are captured in the citric acid cycle during the
a Constant Supply of ATP
conversion of succinyl CoA to succinate. All of these
phosphorylations occur at the substrate level. When
The rate of respiration of mitochondria can be con-
substrates are oxidized via an NAD-linked dehydrogen-
trolled by the availability of ADP. This is because oxi-
ase and the respiratory chain, approximately 3 mol of
dation and phosphorylation are tightly coupled; ie, oxi-
inorganic phosphate are incorporated into 3 mol of
dation cannot proceed via the respiratory chain without
ADP to form 3 mol of ATP per half mol of O2 con-
concomitant phosphorylation of ADP. Table
12-1
sumed; ie, the P:O ratio = 3 (Figure 12-7). On the
shows the five conditions controlling the rate of respira-
other hand, when a substrate is oxidized via a flavopro-
tion in mitochondria. Most cells in the resting state are
tein-linked dehydrogenase, only
2 mol of ATP are
in state 4, and respiration is controlled by the availabil-
formed; ie, P:O = 2. These reactions are known as ox-
ity of ADP. When work is performed, ATP is con-
idative phosphorylation at the respiratory chain
verted to ADP, allowing more respiration to occur,
level. Such dehydrogenations plus phosphorylations at
which in turn replenishes the store of ATP. Under cer-
the substrate level can now account for 68% of the free
tain conditions, the concentration of inorganic phos-
energy resulting from the combustion of glucose, cap-
phate can also affect the rate of functioning of the respi-
tured in the form of high-energy phosphate. It is evi-
ratory chain. As respiration increases (as in exercise),
Succinate
Choline
Proline
3-Hydroxyacyl-CoA
3-Hydroxybutyrate
Glutamate
Fp
Malate
(FAD)
Isocitrate
FeS
Pyruvate
Fp
Lipoate
Fp
NAD
(FMN)
Q
Cyt b
Cyt c1
Cyt c
Cyt aa3
O2
(FAD)
FeS
FeS
Cu
α-Ketoglutarate
Fp
FeS
(FAD)
ETF
FeS
(FAD)
Fp
FeS: Iron-sulfur protein
(FAD)
ETF: Electron-transferring flavoprotein
Fp: Flavoprotein
Q: Ubiquinone
Cyt: Cytochrome
Glycerol 3-phosphate
Acyl-CoA
Sarcosine
Dimethylglycine
Figure 12-4. Components of the respiratory chain in mitochondria, showing the collecting points for reduc-
ing equivalents from important substrates. FeS occurs in the sequences on the O2 side of Fp or Cyt b.
THE RESPIRATORY CHAIN & OXIDATIVE PHOSPHORYLATION
/
95
O
H
OH
H
OH
(H+ + e- )
(H+ + e- )
CH3O
CH3
CH3
CH3O
[CH2CH CCH2]nH
O
•O
OH
Fully oxidized or
Semiquinone form
Reduced or quinol form
quinone form
(free radical)
(hydroquinone)
Figure 12-5. Structure of ubiquinone (Q). n = Number of isoprenoid units, which is
10 in higher animals, ie, Q10.
the cell approaches state 3 or state 5 when either the ca-
MANY POISONS INHIBIT THE
pacity of the respiratory chain becomes saturated or the
RESPIRATORY CHAIN
PO2 decreases below the Km for cytochrome a3. There is
also the possibility that the ADP/ATP transporter (Fig-
Much information about the respiratory chain has been
ure 12-9), which facilitates entry of cytosolic ADP into
obtained by the use of inhibitors, and, conversely, this
and ATP out of the mitochondrion, becomes rate-
has provided knowledge about the mechanism of action
limiting.
of several poisons (Figure 12-7). They may be classified
Thus, the manner in which biologic oxidative
as inhibitors of the respiratory chain, inhibitors of ox-
processes allow the free energy resulting from the oxida-
idative phosphorylation, and uncouplers of oxidative
tion of foodstuffs to become available and to be cap-
phosphorylation.
tured is stepwise, efficient (approximately 68%), and
Barbiturates such as amobarbital inhibit NAD-
controlled—rather than explosive, inefficient, and un-
linked dehydrogenases by blocking the transfer from
controlled, as in many nonbiologic processes. The re-
FeS to Q. At sufficient dosage, they are fatal in vivo.
maining free energy that is not captured as high-energy
Antimycin A and dimercaprol inhibit the respiratory
phosphate is liberated as heat. This need not be consid-
chain between cytochrome b and cytochrome c. The
ered “wasted,” since it ensures that the respiratory sys-
classic poisons H2S, carbon monoxide, and cyanide
tem as a whole is sufficiently exergonic to be removed
inhibit cytochrome oxidase and can therefore totally ar-
from equilibrium, allowing continuous unidirectional
rest respiration. Malonate is a competitive inhibitor of
flow and constant provision of ATP. It also contributes
succinate dehydrogenase.
to maintenance of body temperature.
Atractyloside inhibits oxidative phosphorylation by
inhibiting the transporter of ADP into and ATP out of
the mitochondrion (Figure 12-10).
Pr
The action of uncouplers is to dissociate oxidation
in the respiratory chain from phosphorylation. These
Cys
compounds are toxic in vivo, causing respiration to be-
S
come uncontrolled, since the rate is no longer limited
S
Fe
by the concentration of ADP or Pi. The uncoupler that
has been used most frequently is 2,4-dinitrophenol,
Pr Cys
S
Fe
S
but other compounds act in a similar manner. The an-
tibiotic oligomycin completely blocks oxidation and
phosphorylation by acting on a step in phosphorylation
Fe
S
(Figures 12-7 and 12-8).
S
S
Fe
THE CHEMIOSMOTIC THEORY EXPLAINS
Cys
S
THE MECHANISM OF OXIDATIVE
Pr
Cys
PHOSPHORYLATION
Pr
Mitchell’s chemiosmotic theory postulates that the
energy from oxidation of components in the respiratory
Figure 12-6. Iron-sulfur-protein complex (Fe4S4). S ,
chain is coupled to the translocation of hydrogen ions
acid-labile sulfur; Pr, apoprotein; Cys, cysteine residue.
(protons, H+) from the inside to the outside of the
Some iron-sulfur proteins contain two iron atoms and
inner mitochondrial membrane. The electrochemical
two sulfur atoms (Fe2S2).
potential difference resulting from the asymmetric dis-
96
/
CHAPTER 12
Malonate
Complex II
FAD
Succinate
FeS
Carboxin
-
TTFA
H2S
CO
BAL
–
CN-
Antimycin A
-
Complex IV
Complex I
Complex III
Cyt a
Cyt a3
NADH
FMN, FeS
Q
Cyt b, FeS, Cyt c1
Cyt c
O2
Cu
Cu
-
–
Piericidin A
Uncouplers
–
Amobarbital
-
Uncouplers
-
Rotenone
Oligomycin
-
–
Oligomycin
-
ADP + Pi
ATP
ADP + Pi
ATP
ADP + Pi
ATP
Figure 12-7. Proposed sites of inhibition ( − ) of the respiratory chain by specific drugs, chemicals, and antibi-
otics. The sites that appear to support phosphorylation are indicated. BAL, dimercaprol. TTFA, an Fe-chelating
agent. Complex I, NADH:ubiquinone oxidoreductase; complex II, succinate:ubiquinone oxidoreductase; complex
III, ubiquinol:ferricytochrome c oxidoreductase; complex IV, ferrocytochrome c:oxygen oxidoreductase. Other ab-
breviations as in Figure 12-4.
tribution of the hydrogen ions is used to drive the
units are attached to a membrane protein complex
mechanism responsible for the formation of ATP (Fig-
known as F0, which also consists of several protein sub-
ure 12-8).
units. F0 spans the membrane and forms the proton
channel. The flow of protons through F0 causes it to ro-
complex
tate, driving the production of ATP in the F1
The Respiratory Chain Is a Proton Pump
(Figure 12-9). Estimates suggest that for each NADH
oxidized, complex I translocates four protons and com-
Each of the respiratory chain complexes I, III, and IV
plexes III and IV translocate 6 between them. As four
(Figures 12-7 and 12-8) acts as a proton pump. The
protons are taken into the mitochondrion for each ATP
inner membrane is impermeable to ions in general but
exported, the P:O ratio would not necessarily be a com-
particularly to protons, which accumulate outside the
plete integer, ie, 3, but possibly 2.5. However, for sim-
membrane, creating an electrochemical potential dif-
plicity, a value of 3 for the oxidation of NADH + H+
ference across the membrane (∆µH+).This consists of a
and 2 for the oxidation of FADH2 will continue to be
chemical potential (difference in pH) and an electrical
used throughout this text.
potential.
Experimental Findings Support
A Membrane-Located ATP Synthase
the Chemiosmotic Theory
Functions as a Rotary Motor to Form ATP
(1) Addition of protons
(acid) to the external
The electrochemical potential difference is used to drive
medium of intact mitochondria leads to the generation
a membrane-located ATP synthase which in the pres-
of ATP.
ence of Pi + ADP forms ATP (Figure 12-8). Scattered
(2) Oxidative phosphorylation does not occur in solu-
over the surface of the inner membrane are the phos-
ble systems where there is no possibility of a vectorial
phorylating complexes, ATP synthase, responsible for
ATP synthase. A closed membrane must be present in
the production of ATP (Figure 12-1). These consist of
order to achieve oxidative phosphorylation (Figure 12-8).
several protein subunits, collectively known as F1,
(3) The respiratory chain contains components or-
which project into the matrix and which contain the
ganized in a sided manner (transverse asymmetry) as re-
phosphorylation mechanism (Figure 12-8). These sub-
quired by the chemiosmotic theory.
THE RESPIRATORY CHAIN & OXIDATIVE PHOSPHORYLATION
/
97
H+
Oligomycin
-
F0
Phospholipid
bilayer
F1
H+
I
ATP
SYNTHASE
NADH
+ H+
ADP + Pi
ATP + H2O
Q
NAD+
Mitochondrial
Respiratory
III
inner (coupling)
(electron
H+
H+
membrane
transport)
chain
1/2
O2
C
H2O
IV
H+
INSIDE
pH gradient (∆pH)
-
Electrical
potential
OUTSIDE
+
(∆Ψ)
Figure 12-8. Principles of the chemiosmotic theory of oxidative phosphorylation. The main proton circuit
is created by the coupling of oxidation in the respiratory chain to proton translocation from the inside to the
outside of the membrane, driven by the respiratory chain complexes I, III, and IV, each of which acts as a pro-
ton pump. Q, ubiquinone; C, cytochrome c; F1, F0, protein subunits which utilize energy from the proton gra-
dient to promote phosphorylation. Uncoupling agents such as dinitrophenol allow leakage of H+ across the
membrane, thus collapsing the electrochemical proton gradient. Oligomycin specifically blocks conduction
of H+ through F0.
The Chemiosmotic Theory Can Account
for Respiratory Control and the Action
of Uncouplers
The electrochemical potential difference across the mem-
brane, once established as a result of proton transloca-
Table 12-1. States of respiratory control.
tion, inhibits further transport of reducing equivalents
through the respiratory chain unless discharged by back-
translocation of protons across the membrane through
Conditions Limiting the Rate of Respiration
the vectorial ATP synthase. This in turn depends on
State 1
Availability of ADP and substrate
availability of ADP and Pi.
State 2
Availability of substrate only
Uncouplers
(eg, dinitrophenol) are amphipathic
State 3
The capacity of the respiratory chain itself, when
(Chapter 14) and increase the permeability of the lipoid
all substrates and components are present in
inner mitochondrial membrane to protons
(Figure
saturating amounts
12-8), thus reducing the electrochemical potential and
State 4
Availability of ADP only
short-circuiting the ATP synthase. In this way, oxida-
State 5
Availability of oxygen only
tion can proceed without phosphorylation.
98
/
CHAPTER 12
ing electrical and osmotic equilibrium. The inner
β
bilipoid mitochondrial membrane is freely permeable
ATP
α
α
to uncharged small molecules, such as oxygen, water,
γ
CO2, and NH3, and to monocarboxylic acids, such as
ADP
β
β
ATP
3-hydroxybutyric, acetoacetic, and acetic. Long-chain
+
α
fatty acids are transported into mitochondria via the
Pi
carnitine system (Figure 22-1), and there is also a spe-
cial carrier for pyruvate involving a symport that utilizes
the H+ gradient from outside to inside the mitochon-
drion (Figure 12-10). However, dicarboxylate and tri-
γ
H+
Inside
Inner
OUTSIDE
mitochondrial
INSIDE
membrane
Mitochondrial
N-Ethylmaleimide
inner
membrane
OH-
C
C
1
Outside
C
C
H2PO4-
C
C
N-Ethylmaleimide
-
Hydroxycinnamate
Pyruvate-
2
+
H
H+
-
2-
Figure 12-9. Mechanism of ATP production by ATP
HPO4
synthase. The enzyme complex consists of an F0 sub-
3
complex which is a disk of “C” protein subunits. At-
Malate2-
tached is a γ-subunit in the form of a “bent axle.” Pro-
tons passing through the disk of “C” units cause it and
Malate2-
the attached γ-subunit to rotate. The γ-subunit fits in-
4
Citrate3-
side the F1 subcomplex of three α- and three β-sub-
+ H+
units, which are fixed to the membrane and do not ro-
Malate2-
tate. ADP and Pi are taken up sequentially by the
5
β-subunits to form ATP, which is expelled as the rotat-
α-Ketoglutarate2-
ing γ-subunit squeezes each β-subunit in turn. Thus,
-
three ATP molecules are generated per revolution. For
ADP3-
clarity, not all the subunits that have been identified are
6
shown—eg, the “axle” also contains an ε-subunit.
ATP4-
Atractyloside
Figure 12-10. Transporter systems in the inner mi-
tochondrial membrane. 1 , phosphate transporter;
THE RELATIVE IMPERMEABILITY
2 , pyruvate symport; 3 , dicarboxylate transporter;
OF THE INNER MITOCHONDRIAL
4 , tricarboxylate transporter; 5 , α-ketoglutarate trans-
MEMBRANE NECESSITATES
porter; 6 , adenine nucleotide transporter. N-Ethyl-
EXCHANGE TRANSPORTERS
maleimide, hydroxycinnamate, and atractyloside inhibit
Exchange diffusion systems are present in the mem-
( − ) the indicated systems. Also present (but not
brane for exchange of anions against OH− ions and
shown) are transporter systems for glutamate/aspar-
cations against H+ ions. Such systems are necessary for
tate (Figure 12-13), glutamine, ornithine, neutral amino
uptake and output of ionized metabolites while preserv-
acids, and carnitine (Figure 22-1).
THE RESPIRATORY CHAIN & OXIDATIVE PHOSPHORYLATION
/
99
carboxylate anions and amino acids require specific
Ionophores Permit Specific Cations
transporter or carrier systems to facilitate their passage
to Penetrate Membranes
across the membrane. Monocarboxylic acids penetrate
Ionophores are lipophilic molecules that complex spe-
more readily in their undissociated and more lipid-solu-
cific cations and facilitate their transport through bio-
ble form.
logic membranes, eg, valinomycin (K+). The classic
The transport of di- and tricarboxylate anions is
uncouplers such as dinitrophenol are, in fact, proton
closely linked to that of inorganic phosphate, which
ionophores.
penetrates readily as the H2PO4− ion in exchange for
OH−. The net uptake of malate by the dicarboxylate
transporter requires inorganic phosphate for exchange
A Proton-Translocating Transhydrogenase
in the opposite direction. The net uptake of citrate,
Is a Source of Intramitochondrial NADPH
isocitrate, or cis-aconitate by the tricarboxylate trans-
Energy-linked transhydrogenase, a protein in the inner
porter requires malate in exchange. α-Ketoglutarate
mitochondrial membrane, couples the passage of pro-
transport also requires an exchange with malate. The
tons down the electrochemical gradient from outside to
adenine nucleotide transporter allows the exchange of
inside the mitochondrion with the transfer of H from
ATP and ADP but not AMP. It is vital in allowing
intramitochondrial NADH to NADPH for intramito-
ATP exit from mitochondria to the sites of extramito-
chondrial enzymes such as glutamate dehydrogenase
chondrial utilization and in allowing the return of ADP
and hydroxylases involved in steroid synthesis.
for ATP production within the mitochondrion (Figure
12-11). Na+ can be exchanged for H+, driven by the
Oxidation of Extramitochondrial NADH
proton gradient. It is believed that active uptake of Ca2+
Is Mediated by Substrate Shuttles
by mitochondria occurs with a net charge transfer of 1
(Ca+ uniport), possibly through a Ca2+/H+ antiport.
NADH cannot penetrate the mitochondrial mem-
Calcium release from mitochondria is facilitated by ex-
brane, but it is produced continuously in the cytosol by
change with Na+.
3-phosphoglyceraldehyde dehydrogenase, an enzyme in
the glycolysis sequence (Figure 17-2). However, under
aerobic conditions, extramitochondrial NADH does not
accumulate and is presumed to be oxidized by the respi-
ratory chain in mitochondria. The transfer of reducing
equivalents through the mitochondrial membrane re-
Inner
OUTSIDE
mitochondrial
INSIDE
quires substrate pairs, linked by suitable dehydrogen-
membrane
ases on each side of the mitochondrial membrane. The
F1
mechanism of transfer using the glycerophosphate
ATP SYNTHASE
shuttle is shown in Figure 12-12). Since the mitochon-
drial enzyme is linked to the respiratory chain via a
3H+
flavoprotein rather than NAD, only 2 mol rather than
3 mol of ATP are formed per atom of oxygen con-
sumed. Although this shuttle is present in some tissues
(eg, brain, white muscle), in others (eg, heart muscle) it
ATP4-
is deficient. It is therefore believed that the malate
shuttle system (Figure
12-13) is of more universal
2
utility. The complexity of this system is due to the im-
ADP3-
permeability of the mitochondrial membrane to oxalo-
-
Pi
acetate, which must react with glutamate and transami-
1
nate to aspartate and α-ketoglutarate before transport
H+
through the mitochondrial membrane and reconstitu-
tion to oxaloacetate in the cytosol.
Figure 12-11. Combination of phosphate trans-
porter ( 1 ) with the adenine nucleotide transporter ( 2 )
Ion Transport in Mitochondria
in ATP synthesis. The H+/Pi symport shown is equiva-
Is Energy-Linked
lent to the Pi/OH− antiport shown in Figure 12-10. Four
protons are taken into the mitochondrion for each ATP
Mitochondria maintain or accumulate cations such as
exported. However, one less proton would be taken in
K+, Na+, Ca2+, and Mg2+, and Pi. It is assumed that a
when ATP is used inside the mitochondrion.
primary proton pump drives cation exchange.
100
/
CHAPTER 12
OUTER
INNER
MEMBRANE
MEMBRANE
CYTOSOL
MITOCHONDRION
NAD+
Glycerol 3-phosphate
Glycerol 3-phosphate
FAD
GLYCEROL-3-PHOSPHATE
GLYCEROL-3-PHOSPHATE
DEHYDROGENASE
DEHYDROGENASE
(CYTOSOLIC)
(MITOCHONDRIAL)
NADH + H+
Dihydroxyacetone
Dihydroxyacetone
FADH2
phosphate
phosphate
Respiratory chain
Figure 12-12. Glycerophosphate shuttle for transfer of reducing equivalents from the cytosol into the
mitochondrion.
The Creatine Phosphate Shuttle
ported into the cytosol via protein pores in the outer
Facilitates Transport of High-Energy
mitochondrial membrane, becoming available for gen-
eration of extramitochondrial ATP.
Phosphate From Mitochondria
This shuttle (Figure 12-14) augments the functions of
CLINICAL ASPECTS
creatine phosphate as an energy buffer by acting as a
dynamic system for transfer of high-energy phosphate
The condition known as fatal infantile mitochondrial
from mitochondria in active tissues such as heart and
myopathy and renal dysfunction involves severe dim-
skeletal muscle. An isoenzyme of creatine kinase (CKm)
inution or absence of most oxidoreductases of the respi-
is found in the mitochondrial intermembrane space,
ratory chain. MELAS (mitochondrial encephalopathy,
catalyzing the transfer of high-energy phosphate to cre-
lactic acidosis, and stroke) is an inherited condition due
atine from ATP emerging from the adenine nucleotide
to NADH:ubiquinone oxidoreductase (complex I) or
transporter. In turn, the creatine phosphate is trans-
cytochrome oxidase deficiency. It is caused by a muta-
INNER
CYTOSOL
MEMBRANE
MITOCHONDRION
+
NAD
Malate
Malate
NAD+
1
MALATE DEHYDROGENASE
MALATE DEHYDROGENASE
NADH
Oxaloacetate
α-KG
α-KG
Oxaloacetate
NADH
+ H+
+ H+
TRANSAMINASE
TRANSAMINASE
Glutamate
Asp
Asp
Glutamate
2
H+
H+
Figure 12-13. Malate shuttle for transfer of reducing equivalents from the cytosol into the mitochondrion.
1 Ketoglutarate transporter; 2 , glutamate/aspartate transporter (note the proton symport with glutamate).
THE RESPIRATORY CHAIN & OXIDATIVE PHOSPHORYLATION
/
101
Energy-requiring
processes
(eg, muscle contraction)
A
TP
ADP
CKa
ATP
ADP
Creatine
Creatine-P
CKc
CKg
ATP
ADP
Glycolysis
ito
Cytosol
mb
P
Figure 12-14. The creatine phosphate shuttle of
P
heart and skeletal muscle. The shuttle allows rapid
CKm
transport of high-energy phosphate from the mito-
chondrial matrix into the cytosol. CKa, creatine kinase
Inter-membrane
concerned with large requirements for ATP, eg, mus-
A
TP
ADP
space
cular contraction; CKc, creatine kinase for maintaining
Adenine
equilibrium between creatine and creatine phosphate
nucleotide
and ATP/ADP; CKg, creatine kinase coupling glycolysis
transporter
to creatine phosphate synthesis; CKm, mitochondrial
creatine kinase mediating creatine phosphate produc-
Oxidative
phosphorylation
tion from ATP formed in oxidative phosphorylation; P,
pore protein in outer mitochondrial membrane.
Matrix
tion in mitochondrial DNA and may be involved in
•
Because the inner mitochondrial membrane is imper-
Alzheimer’s disease and diabetes mellitus. A number of
meable to protons and other ions, special exchange
drugs and poisons act by inhibition of oxidative phos-
transporters span the membrane to allow passage of
phorylation (see above).
ions such as OH-, Pi−, ATP4−, ADP3−, and metabo-
lites, without discharging the electrochemical gradi-
ent across the membrane.
SUMMARY
•
Many well-known poisons such as cyanide arrest res-
piration by inhibition of the respiratory chain.
• Virtually all energy released from the oxidation of
carbohydrate, fat, and protein is made available in
REFERENCES
mitochondria as reducing equivalents (H or e−).
These are funneled into the respiratory chain, where
Balaban RS: Regulation of oxidative phosphorylation in the mam-
they are passed down a redox gradient of carriers to
malian cell. Am J Physiol 1990;258:C377.
their final reaction with oxygen to form water.
Hinkle PC et al: Mechanistic stoichiometry of mitochondrial ox-
idative phosphorylation. Biochemistry 1991;30:3576.
• The redox carriers are grouped into respiratory chain
Mitchell P: Keilin’s respiratory chain concept and its chemiosmotic
complexes in the inner mitochondrial membrane.
consequences. Science 1979;206:1148.
These use the energy released in the redox gradient to
Smeitink J et al: The genetics and pathology of oxidative phosphor-
pump protons to the outside of the membrane, creat-
ylation. Nat Rev Genet 2001;2:342.
ing an electrochemical potential across the membrane.
Tyler DD: The Mitochondrion in Health and Disease. VCH Pub-
• Spanning the membrane are ATP synthase com-
lishers, 1992.
plexes that use the potential energy of the proton gra-
Wallace DC: Mitochondrial DNA in aging and disease. Sci Am
dient to synthesize ATP from ADP and Pi. In this
1997;277(2):22.
way, oxidation is closely coupled to phosphorylation
Yoshida M et al: ATP synthase—a marvellous rotary engine of the
to meet the energy needs of the cell.
cell. Nat Rev Mol Cell Biol 2001;2:669.
Carbohydrates of
13
Physiologic Significance
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
(4) Polysaccharides are condensation products of
more than ten monosaccharide units; examples are the
Carbohydrates are widely distributed in plants and ani-
starches and dextrins, which may be linear or branched
mals; they have important structural and metabolic
polymers. Polysaccharides are sometimes classified as
roles. In plants, glucose is synthesized from carbon
hexosans or pentosans, depending upon the identity of
dioxide and water by photosynthesis and stored as
the constituent monosaccharides.
starch or used to synthesize cellulose of the plant frame-
work. Animals can synthesize carbohydrate from lipid
glycerol and amino acids, but most animal carbohy-
BIOMEDICALLY, GLUCOSE IS THE MOST
drate is derived ultimately from plants. Glucose is the
IMPORTANT MONOSACCHARIDE
most important carbohydrate; most dietary carbohy-
drate is absorbed into the bloodstream as glucose, and
The Structure of Glucose Can Be
other sugars are converted into glucose in the liver.
Represented in Three Ways
Glucose is the major metabolic fuel of mammals (ex-
The straight-chain structural formula
(aldohexose;
cept ruminants) and a universal fuel of the fetus. It is
the precursor for synthesis of all the other carbohy-
Figure 13-1A) can account for some of the properties
of glucose, but a cyclic structure is favored on thermo-
drates in the body, including glycogen for storage; ri-
bose and deoxyribose in nucleic acids; and galactose
dynamic grounds and accounts for the remainder of its
chemical properties. For most purposes, the structural
in lactose of milk, in glycolipids, and in combination
with protein in glycoproteins and proteoglycans. Dis-
formula is represented as a simple ring in perspective as
proposed by Haworth (Figure 13-1B). In this represen-
eases associated with carbohydrate metabolism include
diabetes mellitus, galactosemia, glycogen storage
tation, the molecule is viewed from the side and above
the plane of the ring. By convention, the bonds nearest
diseases, and lactose intolerance.
to the viewer are bold and thickened. The six-mem-
bered ring containing one oxygen atom is in the form
CARBOHYDRATES ARE ALDEHYDE
of a chair (Figure 13-1C).
OR KETONE DERIVATIVES OF
POLYHYDRIC ALCOHOLS
Sugars Exhibit Various Forms of Isomerism
(1) Monosaccharides are those carbohydrates that
cannot be hydrolyzed into simpler carbohydrates: They
Glucose, with four asymmetric carbon atoms, can form
may be classified as trioses, tetroses, pentoses, hex-
16 isomers. The more important types of isomerism
oses, or heptoses, depending upon the number of car-
found with glucose are as follows.
bon atoms; and as aldoses or ketoses depending upon
(1) D and L isomerism: The designation of a sugar
whether they have an aldehyde or ketone group. Exam-
isomer as the D form or of its mirror image as the L form
ples are listed in Table 13-1.
(2) Disaccharides are condensation products of two
monosaccharide units. Examples are maltose and su-
Table 13-1. Classification of important sugars.
crose.
(3) Oligosaccharides are condensation products of
two to ten monosaccharides; maltotriose* is an exam-
Aldoses
Ketoses
ple.
Trioses (C3H6O3)
Glycerose
Dihydroxyacetone
Tetroses (C4H8O4)
Erythrose
Erythrulose
Pentoses (C5H10O5)
Ribose
Ribulose
*Note that this is not a true triose but a trisaccharide containing
Hexoses (C6H12O6)
Glucose
Fructose
three α-glucose residues.
102
CARBOHYDRATES OF PHYSIOLOGIC SIGNIFICANCE
/
103
A
O
O
O
1C H
H
2
C OH
HO
3
C H
Pyran
Furan
H
4
C OH
H
5
C OH
6
CH
HOCH2
2OH
HOCH2
6
HCOH
O
B
HOCH2
O
H
H H
H
5
O
H
H
H
HO OH H OH
H OH
H OH
4
1
HO
OH
H OH
H
OH
H
OH
3
2
H
OH
α-D-Glucopyranose
α-D-Glucofuranose
C
H
Figure 13-3. Pyranose and furanose forms of glu-
6
HOCH2
O
cose.
HO
4
5
H
H
2
H
HO
OH
1
3
OH
H
Figure 13-1.
D-Glucose. A: straight chain form.
B: α-D-glucose; Haworth projection. C: α-D-glucose;
chair form.
1
O
O
H
HOCH2
H
6
O
6
O
1C H
C
H
H H
H H
OH
HO
2C H
H
C
OH
5
2
5
2
3CH2OH
CH2OH
HO H HO
OH
HO H HO
1
L-Glycerose
D-Glycerose
4
3
4
3
COH
(L-glyceraldehyde)
(D-glyceraldehyde)
OH H
OH H
H2
α-D-Fructopyranose
β-D-Fructopyranose
O
O
1C H
C
H
6
1
6
HOCH2
HOCH2
HOCH2
OH
HO
2C H
H
C
OH
O
O
H
3C
OH
HO C
H
5
2
5
2
HO
4C H
H
C
OH
H H HO
OH
H H HO
HO
5C H
H
C
OH
4
3
4
3
1
COH
OH H
OH H
H
6CH2OH
CH2OH
2
L-Glucose
D-Glucose
α-D-Fructofuranose
β-D-Fructofuranose
Figure 13-2. D- and L-isomerism of glycerose and
Figure 13-4. Pyranose and furanose forms of fruc-
glucose.
tose.
104
/
CHAPTER 13
HOCH2
HOCH2
HOCH2
O
O
O
HO H
H
H H
H
H
H
H
4
4
H OH H OH
OH
OH H
OH
OH
OH
HO OH
2
2
H
OH
H
OH
H
H
Figure 13-5. Epimerization of
α-D-Galactose
α-D-Glucose
α-D-Mannose
glucose.
is determined by its spatial relationship to the parent
(3) Alpha and beta anomers: The ring structure of
compound of the carbohydrates, the three-carbon
an aldose is a hemiacetal, since it is formed by combina-
sugar glycerose (glyceraldehyde). The L and D forms of
tion of an aldehyde and an alcohol group. Similarly, the
this sugar, and of glucose, are shown in Figure 13-2.
ring structure of a ketose is a hemiketal. Crystalline glu-
The orientation of the H and OH groups around
cose is α-D-glucopyranose. The cyclic structure is re-
the carbon atom adjacent to the terminal primary alco-
tained in solution, but isomerism occurs about position
hol carbon (carbon 5 in glucose) determines whether
1, the carbonyl or anomeric carbon atom, to give a
the sugar belongs to the D or L series. When the OH
mixture of α-glucopyranose (38%) and β-glucopyra-
group on this carbon is on the right (as seen in Figure
nose (62%). Less than 0.3% is represented by α and β
13-2), the sugar is the D-isomer; when it is on the
anomers of glucofuranose.
left, it is the L-isomer. Most of the monosaccharides
(4) Epimers: Isomers differing as a result of varia-
occurring in mammals are D sugars, and the enzymes
tions in configuration of the OH and H on car-
responsible for their metabolism are specific for this
bon atoms 2, 3, and 4 of glucose are known as epimers.
configuration. In solution, glucose is dextrorotatory—
Biologically, the most important epimers of glucose are
hence the alternative name dextrose, often used in
mannose and galactose, formed by epimerization at car-
clinical practice.
bons 2 and 4, respectively (Figure 13-5).
The presence of asymmetric carbon atoms also con-
(5) Aldose-ketose isomerism: Fructose has the
fers optical activity on the compound. When a beam
same molecular formula as glucose but differs in its
of plane-polarized light is passed through a solution of
structural formula, since there is a potential keto group
an optical isomer, it will be rotated either to the right,
in position 2, the anomeric carbon of fructose (Figures
dextrorotatory (+); or to the left, levorotatory (−). The
13-4 and 13-7), whereas there is a potential aldehyde
direction of rotation is independent of the stereochem-
group in position 1, the anomeric carbon of glucose
istry of the sugar, so it may be designated D(−), D(+),
(Figures 13-2 and 13-6).
L(−), or L(+). For example, the naturally occurring form
of fructose is the D(−) isomer.
Many Monosaccharides Are
(2) Pyranose and furanose ring structures: The
Physiologically Important
stable ring structures of monosaccharides are similar to
the ring structures of either pyran
(a six-membered
Derivatives of trioses, tetroses, and pentoses and of a
ring) or furan (a five-membered ring) (Figures 13-3
seven-carbon sugar (sedoheptulose) are formed as meta-
and 13-4). For glucose in solution, more than 99% is
bolic intermediates in glycolysis and the pentose phos-
in the pyranose form.
phate pathway. Pentoses are important in nucleotides,
CHO
CHO
CHO
CHO
CHO
CHO
CHO
H C OH
HO C
H
H C OH
CHO
HO C
H
H C OH
HO
C
H
H C OH
HO C
H
HO C
H
HO C
H
CHO
H C OH
HO C
H
HO C
H
H
C OH
H C OH
HO C
H
H C OH
H C OH
H C OH
H C OH
H C OH
H C OH
H
C OH
H C OH
H C OH
H C OH
H C OH
CH2OH
CH2OH
CH2OH
CH2OH
CH2OH
CH2OH
CH2OH
CH2OH
CH2OH
D-Glycerose
(D-glyceraldehyde) D-Erythrose
D-Lyxose
D-Xylose
D-Arabinose
D-Ribose
D-Galactose
D-Mannose
D-Glucose
Figure 13-6. Examples of aldoses of physiologic significance.
CARBOHYDRATES OF PHYSIOLOGIC SIGNIFICANCE
/
105
Table 13-2. Pentoses of physiologic importance.
Sugar
Where Found
Biochemical Importance
Clinical Significance
D-Ribose
Nucleic acids.
Structural elements of nucleic acids and
coenzymes, eg, ATP, NAD, NADP, flavo-
proteins. Ribose phosphates are inter-
mediates in pentose phosphate pathway.
D-Ribulose
Formed in metabolic processes.
Ribulose phosphate is an intermediate in
pentose phosphate pathway.
D-Arabinose
Gum arabic. Plum and cherry gums.
Constituent of glycoproteins.
D-Xylose
Wood gums, proteoglycans,
Constituent of glycoproteins.
glycosaminoglycans.
D-Lyxose
Heart muscle.
A constituent of a lyxoflavin isolated from
human heart muscle.
L-Xylulose
Intermediate in uronic acid pathway.
Found in urine in essential
pentosuria.
nucleic acids, and several coenzymes
(Table
13-2).
Sugars Form Glycosides With Other
Glucose, galactose, fructose, and mannose are physio-
Compounds & With Each Other
logically the most important hexoses (Table 13-3). The
biochemically important aldoses are shown in Figure
Glycosides are formed by condensation between the hy-
13-6, and important ketoses in Figure 13-7.
droxyl group of the anomeric carbon of a monosaccha-
In addition, carboxylic acid derivatives of glucose are
ride, or monosaccharide residue, and a second compound
important, including D-glucuronate
(for glucuronide
that may—or may not (in the case of an aglycone)—be
formation and in glycosaminoglycans) and its meta-
another monosaccharide. If the second group is a hy-
bolic derivative, L-iduronate
(in glycosaminoglycans)
droxyl, the O-glycosidic bond is an acetal link because it
(Figure 13-8) and L-gulonate (an intermediate in the
results from a reaction between a hemiacetal group
uronic acid pathway; see Figure 20-4).
(formed from an aldehyde and an OH group) and an-
Table 13-3. Hexoses of physiologic importance.
Sugar
Source
Importance
Clinical Significance
D-Glucose
Fruit juices. Hydrolysis of starch, cane
The “sugar” of the body. The sugar carried
Present in the urine (glycosuria)
sugar, maltose, and lactose.
by the blood, and the principal one used
in diabetes mellitus owing to
by the tissues.
raised blood glucose (hyper-
glycemia).
D-Fructose
Fruit juices. Honey. Hydrolysis of
Can be changed to glucose in the liver
Hereditary fructose intolerance
cane sugar and of inulin (from the
and so used in the body.
leads to fructose accumulation
Jerusalem artichoke).
and hypoglycemia.
D-Galactose
Hydrolysis of lactose.
Can be changed to glucose in the liver
Failure to metabolize leads
and metabolized. Synthesized in the
to galactosemia and cataract.
mammary gland to make the lactose of
milk. A constituent of glycolipids and
glycoproteins.
D-Mannose
Hydrolysis of plant mannans and
A constituent of many glycoproteins.
gums.
106
/
CHAPTER 13
CH2OH
CH2OH
C O
CH2OH
CH2OH
C
O
HO
C H
C
O
C O
HO
C
H
H
C
OH
CH2OH
HO
C
H
H
C
OH
H
C
OH
H
C
OH
C
O
H C OH
H C OH
H C OH
H
C
OH
CH2OH
CH2OH
CH2OH
CH2OH
CH2OH
Dihydroxyacetone
D-Xylulose
D-Ribulose
D-Fructose
D-Sedoheptulose
Figure 13-7. Examples of ketoses of physiologic significance.
other OH group. If the hemiacetal portion is glucose,
COO-
H
the resulting compound is a glucoside; if galactose, a
O
O
galactoside; and so on. If the second group is an amine,
H
H
H
H
COO-
H
an N-glycosidic bond is formed, eg, between adenine and
ribose in nucleotides such as ATP (Figure 10-4).
HO OH H
OH
HO OH H
OH
Glycosides are widely distributed in nature; the agly-
cone may be methanol, glycerol, a sterol, a phenol, or a
H
OH
H
OH
base such as adenine. The glycosides that are important
in medicine because of their action on the heart (car-
Figure 13-8.
α-D-Glucuronate (left) and
diac glycosides) all contain steroids as the aglycone.
β-L-iduronate (right).
These include derivatives of digitalis and strophanthus
such as ouabain, an inhibitor of the Na+-K+ ATPase of
cell membranes. Other glycosides include antibiotics
such as streptomycin.
5
HOCH2
Deoxy Sugars Lack an Oxygen Atom
O
OH
Deoxy sugars are those in which a hydroxyl group has
4
1
been replaced by hydrogen. An example is deoxyribose
H H
H
H
(Figure 13-9) in DNA. The deoxy sugar L-fucose (Figure
3
2
13-15) occurs in glycoproteins; 2-deoxyglucose is used
OH H
experimentally as an inhibitor of glucose metabolism.
Figure 13-9.
2-Deoxy-D-ribofuranose (β form).
Amino Sugars (Hexosamines) Are
Components of Glycoproteins,
Gangliosides, & Glycosaminoglycans
HOCH2
The amino sugars include D-glucosamine, a constituent
of hyaluronic acid
(Figure
13-10), D-galactosamine
O
H H
H
(chondrosamine), a constituent of chondroitin; and
D-mannosamine. Several antibiotics (eg, erythromycin)
HO
OH
H
OH
contain amino sugars believed to be important for their
antibiotic activity.
H
+
NH
3
MALTOSE, SUCROSE, & LACTOSE ARE
Figure 13-10. Glucosamine (2-amino-D-glucopyra-
IMPORTANT DISACCHARIDES
nose) (α form). Galactosamine is 2-amino-D-galactopy-
The physiologically important disaccharides are mal-
ranose. Both glucosamine and galactosamine occur as
tose, sucrose, and lactose (Table 13-4; Figure 13-11).
N-acetyl derivatives in more complex carbohydrates,
Hydrolysis of sucrose yields a mixture of glucose and
eg, glycoproteins.
CARBOHYDRATES OF PHYSIOLOGIC SIGNIFICANCE
/
107
Table 13-4. Disaccharides.
Sugar
Source
Clinical Significance
Maltose
Digestion by amylase or hydrolysis of starch.
Germinating cereals and malt.
Lactose
Milk. May occur in urine during pregnancy.
In lactase deficiency, malabsorption leads to diarrhea and flatulence.
Sucrose
Cane and beet sugar. Sorghum. Pineapple.
In sucrase deficiency, malabsorption leads to diarrhea and flatulence.
Carrot roots.
Trehalose1
Fungi and yeasts. The major sugar of insect
hemolymph.
1O-α-D-Glucopyranosyl-(1 → 1)-α-D-glucopyranoside.
fructose which is called
“invert sugar” because the
toes, legumes, and other vegetables. The two main con-
strongly levorotatory fructose changes (inverts) the pre-
stituents are amylose
(15-20%), which has a non-
vious dextrorotatory action of sucrose.
branching helical structure (Figure 13-12); and amy-
lopectin (80-85%), which consists of branched chains
composed of 24-30 glucose residues united by 1 → 4
POLYSACCHARIDES SERVE STORAGE
linkages in the chains and by 1 → 6 linkages at the
& STRUCTURAL FUNCTIONS
branch points.
Polysaccharides include the following physiologically
Glycogen (Figure 13-13) is the storage polysaccha-
important carbohydrates.
ride in animals. It is a more highly branched structure
Starch is a homopolymer of glucose forming an α-
than amylopectin, with chains of 12-14 α-D-glucopyra-
glucosidic chain, called a glucosan or glucan. It is the
nose residues (in α[1 → 4]-glucosidic linkage), with
most abundant dietary carbohydrate in cereals, pota-
branching by means of α(1 → 6)-glucosidic bonds.
Maltose
Lactose
6
6
6
6
HOCH2
HOCH2
HOCH2
HOCH2
5
O
5
O
5
O
5
O
H H
H
H H
H
HO H
H
H
OH
4
1
4
1
4
1
O
4
1
HO OH H
OH
H
OH
H OH H
H
OH
H
H
3
2
3
2
3
2
3
2
O
H
OH
H
OH
H
OH
H
OH
O-α-D-Glucopyranosyl-(1 → 4)-α-D-glucopyranose
O-β-D-Galactopyranosyl-(1 → 4)-β-D-glucopyranose
Sucrose
Figure 13-11. Structures of important disaccharides. The α and β
6
1
HOCH2
HOCH2
refer to the configuration at the anomeric carbon atom (asterisk). When
5
O
the anomeric carbon of the second residue takes part in the formation
O
H H
H
H
of the glycosidic bond, as in sucrose, the residue becomes a glycoside
4
1
2
5
6
known as a furanoside or pyranoside. As the disaccharide no longer has
HO OH H
H HO
COH
an anomeric carbon with a free potential aldehyde or ketone group, it
3
2
3
4
H2
O
no longer exhibits reducing properties. The configuration of the
H
OH
OH H
β-fructofuranose residue in sucrose results from turning the β-fructofu-
ranose molecule depicted in Figure 13-4 through 180 degrees and in-
O-α-D-Glucopyranosyl-(1 → 2)-β-D-fructofuranoside verting it.
108
/
CHAPTER 13
A
B
O
O
6
6
6
6
HOCH2
HOCH2
CH2
HOCH2
O
O
O
O
O
O
4
1
4
1
4
1
4
1
O
O
O
O
O
Figure 13-12. Structure of starch. A: Amylose, showing helical coil structure. B: Amylopectin, showing 1 → 6
branch point.
O
H
O
O
4
CH2
6
4
O
O
1
1
O
4
HOCH2
O
O
1
6
4
CH2
O
HOCH2
1
1
O
2
G
O
3
H
O
4
4
CH2
6
O
1
O
A
B
Figure 13-13. The glycogen molecule. A: General structure. B: Enlargement of structure at a branch point. The
molecule is a sphere approximately 21 nm in diameter that can be visualized in electron micrographs. It has a mo-
lecular mass of 107 Da and consists of polysaccharide chains each containing about 13 glucose residues. The
chains are either branched or unbranched and are arranged in 12 concentric layers (only four are shown in the
figure). The branched chains (each has two branches) are found in the inner layers and the unbranched chains
in the outer layer. (G, glycogenin, the primer molecule for glycogen synthesis.)
CARBOHYDRATES OF PHYSIOLOGIC SIGNIFICANCE
/
109
Chitin
Inulin is a polysaccharide of fructose (and hence a fruc-
tosan) found in tubers and roots of dahlias, artichokes,
HOCH2
HOCH2
and dandelions. It is readily soluble in water and is used
O
O
to determine the glomerular filtration rate. Dextrins are
H H
H H
intermediates in the hydrolysis of starch. Cellulose is
O
1
O
4
O
the chief constituent of the framework of plants. It is in-
OH H H
OH H H
soluble and consists of β-D-glucopyranose units linked
by β(1 → 4) bonds to form long, straight chains
H HN
CO CH3
H
HN CO CH3
strengthened by cross-linked hydrogen bonds. Cellulose
n
cannot be digested by mammals because of the absence
N-Acetylglucosamine
N-Acetylglucosamine
of an enzyme that hydrolyzes the β linkage. It is an im-
portant source of “bulk” in the diet. Microorganisms in
the gut of ruminants and other herbivores can hydrolyze
Hyaluronic acid
the β linkage and ferment the products to short-chain
HOCH2
fatty acids as a major energy source. There is limited
O
bacterial metabolism of cellulose in the human colon.
COO
-
H H
Chitin is a structural polysaccharide in the exoskeleton
O
1
O
of crustaceans and insects and also in mushrooms. It
H H
HO O
H H
consists of N-acetyl-D-glucosamine units joined by
3
O
4
β (1 → 4)-glycosidic linkages (Figure 13-14).
1
OH H H
Glycosaminoglycans
(mucopolysaccharides) are
H HN CO CH3
complex carbohydrates characterized by their content
of amino sugars and uronic acids. When these chains
H
OH
n
are attached to a protein molecule, the result is a pro-
β-Glucuronic acid
N-Acetylglucosamine
teoglycan. Proteoglycans provide the ground or pack-
ing substance of connective tissues. Their property of
holding large quantities of water and occupying space,
Chondroitin 4-sulfate
(Note: There is also a 6-sulfate)
thus cushioning or lubricating other structures, is due
to the large number of OH groups and negative
HOCH2
charges on the molecules, which, by repulsion, keep the
O
carbohydrate chains apart. Examples are hyaluronic
COO
-
- SO3O H
acid, chondroitin sulfate, and heparin
(Figure
O
1
O
13-14).
H H
H O
H H
Glycoproteins (mucoproteins) occur in many dif-
3
O
4
1
ferent situations in fluids and tissues, including the cell
OH H H
membranes (Chapters 41 and 47). They are proteins
H HN CO CH3
H
OH
n
β-Glucuronic acid
N-Acetylgalactosamine sulfate
Table 13-5. Carbohydrates found in
glycoproteins.
Heparin
Hexoses
Mannose (Man)
-
COSO
3
H
Galactose (Gal)
O
O
-
Acetyl hexosamines
N-Acetylglucosamine (GlcNAc)
H H
H
H
COO
N-Acetylgalactosamine (GalNAc)
O
1
4
O
OH H
OH
H H
Pentoses
Arabinose (Ara)
Xylose (Xyl)
O
-
–
H
NH SO3
H
OSO
3
Methyl pentose
L-Fucose (Fuc; see Figure 13-15)
n
Sialic acids
N-Acyl derivatives of neuraminic acid,
Sulfated glucosamine
Sulfated iduronic acid
eg, N-acetylneuraminic acid (NeuAc; see
Figure 13-16), the predominant sialic
Figure 13-14. Structure of some complex polysac-
acid.
charides and glycosaminoglycans.
110
/
CHAPTER 13
H
outside both the external and internal (cytoplasmic)
O
surfaces. Carbohydrate chains are only attached to the
H
CH3
H
amino terminal portion outside the external surface
(Chapter 41).
HO H
HO
OH
SUMMARY
OH
H
•
Carbohydrates are major constituents of animal food
Figure 13-15. β-L-Fucose (6-deoxy-β-L-galactose).
and animal tissues. They are characterized by the
type and number of monosaccharide residues in their
molecules.
•
Glucose is the most important carbohydrate in mam-
malian biochemistry because nearly all carbohydrate
containing branched or unbranched oligosaccharide
in food is converted to glucose for metabolism.
chains (see Table 13-5). The sialic acids are N- or
O-acyl derivatives of neuraminic acid (Figure 13-16).
•
Sugars have large numbers of stereoisomers because
Neuraminic acid is a nine-carbon sugar derived from
they contain several asymmetric carbon atoms.
mannosamine (an epimer of glucosamine) and pyru-
•
The monosaccharides include glucose, the “blood
vate. Sialic acids are constituents of both glycoproteins
sugar”; and ribose, an important constituent of nu-
and gangliosides (Chapters 14 and 47).
cleotides and nucleic acids.
•
The disaccharides include maltose (glucosyl glucose),
CARBOHYDRATES OCCUR IN CELL
an intermediate in the digestion of starch; sucrose
MEMBRANES & IN LIPOPROTEINS
(glucosyl fructose), important as a dietary constituent
containing fructose; and lactose (galactosyl glucose),
In addition to the lipid of cell membranes (see Chapters
in milk.
14 and 41), approximately 5% is carbohydrate in glyco-
•
Starch and glycogen are storage polymers of glucose
proteins and glycolipids. Carbohydrates are also present
in plants and animals, respectively. Starch is the
in apo B of lipoproteins. Their presence on the outer
major source of energy in the diet.
surface of the plasma membrane (the glycocalyx) has
•
Complex carbohydrates contain other sugar deriva-
been shown with the use of plant lectins, protein agglu-
tives such as amino sugars, uronic acids, and sialic
tinins that bind with specific glycosyl residues. For
acids. They include proteoglycans and glycosamino-
example, concanavalin A binds α-glucosyl and α-man-
glycans, associated with structural elements of the tis-
nosyl residues. Glycophorin is a major integral mem-
sues; and glycoproteins, proteins containing attached
brane glycoprotein of human erythrocytes and spans
oligosaccharide chains. They are found in many situ-
the lipid membrane, having free polypeptide portions
ations including the cell membrane.
REFERENCES
H
Binkley RW: Modern Carbohydrate Chemistry. Marcel Dekker,
O
1988.
Ac
NH
CHOH
COO—
Collins PM (editor): Carbohydrates. Chapman & Hall, 1988.
CHOH
El-Khadem HS: Carbohydrate Chemistry: Monosaccharides and
CH2OH
Their Oligomers. Academic Press, 1988.
H
H
Lehman J (editor) (translated by Haines A.): Carbohydrates: Struc-
H
OH
ture and Biology. Thieme, 1998.
Lindahl U, Höök M: Glycosaminoglycans and their binding to bio-
OH
H
logical macromolecules. Annu Rev Biochem 1978;47:385.
Melendes-Hevia E, Waddell TG, Shelton ED: Optimization of
Figure 13-16.
Structure of N-acetylneuraminic acid,
molecular design in the evolution of metabolism: the glyco-
a sialic acid (Ac = CH3 CO ).
gen molecule. Biochem J 1993;295:477.
Lipids of Physiologic Significance
14
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
c.
Other complex lipids: Lipids such as sul-
folipids and aminolipids. Lipoproteins may
The lipids are a heterogeneous group of compounds,
also be placed in this category.
including fats, oils, steroids, waxes, and related com-
3. Precursor and derived lipids: These include fatty
pounds, which are related more by their physical than
acids, glycerol, steroids, other alcohols, fatty alde-
by their chemical properties. They have the common
hydes, and ketone bodies (Chapter 22), hydrocar-
property of being (1) relatively insoluble in water and
bons, lipid-soluble vitamins, and hormones.
(2) soluble in nonpolar solvents such as ether and
chloroform. They are important dietary constituents
Because they are uncharged, acylglycerols
(glyc-
not only because of their high energy value but also be-
erides), cholesterol, and cholesteryl esters are termed
cause of the fat-soluble vitamins and the essential fatty
neutral lipids.
acids contained in the fat of natural foods. Fat is stored
in adipose tissue, where it also serves as a thermal insu-
lator in the subcutaneous tissues and around certain or-
FATTY ACIDS ARE ALIPHATIC
gans. Nonpolar lipids act as electrical insulators, al-
CARBOXYLIC ACIDS
lowing rapid propagation of depolarization waves along
Fatty acids occur mainly as esters in natural fats and oils
myelinated nerves. Combinations of lipid and protein
but do occur in the unesterified form as free fatty
(lipoproteins) are important cellular constituents, oc-
acids, a transport form found in the plasma. Fatty acids
curring both in the cell membrane and in the mito-
that occur in natural fats are usually straight-chain de-
chondria, and serving also as the means of transporting
rivatives containing an even number of carbon atoms.
lipids in the blood. Knowledge of lipid biochemistry is
The chain may be saturated (containing no double
necessary in understanding many important biomedical
bonds) or unsaturated (containing one or more double
areas, eg, obesity, diabetes mellitus, atherosclerosis,
bonds).
and the role of various polyunsaturated fatty acids in
nutrition and health.
Fatty Acids Are Named After
LIPIDS ARE CLASSIFIED AS SIMPLE
Corresponding Hydrocarbons
OR COMPLEX
The most frequently used systematic nomenclature
1. Simple lipids: Esters of fatty acids with various al-
names the fatty acid after the hydrocarbon with the
cohols.
same number and arrangement of carbon atoms, with
a.
Fats: Esters of fatty acids with glycerol. Oils
-oic being substituted for the final -e (Genevan sys-
are fats in the liquid state.
tem). Thus, saturated acids end in -anoic, eg, octanoic
b. Waxes: Esters of fatty acids with higher mole-
acid, and unsaturated acids with double bonds end in
cular weight monohydric alcohols.
-enoic, eg, octadecenoic acid (oleic acid).
2. Complex lipids: Esters of fatty acids containing
Carbon atoms are numbered from the carboxyl car-
groups in addition to an alcohol and a fatty acid.
bon (carbon No. 1). The carbon atoms adjacent to the
a.
Phospholipids: Lipids containing, in addition
carboxyl carbon (Nos. 2, 3, and 4) are also known as
to fatty acids and an alcohol, a phosphoric
the α, β, and γ carbons, respectively, and the terminal
acid residue. They frequently have nitrogen-
methyl carbon is known as the ω or n-carbon.
containing bases and other substituents, eg, in
Various conventions use ∆ for indicating the num-
glycerophospholipids the alcohol is glycerol
ber and position of the double bonds (Figure 14-1); eg,
and in sphingophospholipids the alcohol is
∆9 indicates a double bond between carbons 9 and 10
sphingosine.
of the fatty acid; ω9 indicates a double bond on the
b. Glycolipids
(glycosphingolipids): Lipids
ninth carbon counting from the ω- carbon. In animals,
containing a fatty acid, sphingosine, and car-
additional double bonds are introduced only between
bohydrate.
the existing double bond (eg, ω9, ω6, or ω3) and the
111
112
/
CHAPTER 14
18:1;9 or ∆9 18:1
Unsaturated Fatty Acids Contain One
18
10
9
1
or More Double Bonds
CH3(CH2)7CH
CH(CH2)7COOH
(Table 14-2)
or
Fatty acids may be further subdivided as follows:
ω9,C18:1 or n-9, 18:1
ω
2
3
4
5
6
7
8
9
10
18
(1) Monounsaturated (monoethenoid, monoenoic)
CH3CH2CH2CH2CH2CH2CH2CH2CH
CH(CH2)7COOH
n
17
10
9
1
acids, containing one double bond.
(2) Polyunsaturated
(polyethenoid, polyenoic)
Figure 14-1. Oleic acid. n − 9 (n minus 9) is equiva-
acids, containing two or more double bonds.
lent to ω9.
(3) Eicosanoids: These compounds, derived from
eicosa-
(20-carbon) polyenoic fatty acids, com-
prise the prostanoids, leukotrienes (LTs), and
carboxyl carbon, leading to three series of fatty acids
lipoxins
(LXs). Prostanoids include prosta-
known as the ω9, ω6, and ω3 families, respectively.
glandins
(PGs), prostacyclins
(PGIs), and
thromboxanes (TXs).
Saturated Fatty Acids Contain
Prostaglandins exist in virtually every mammalian
tissue, acting as local hormones; they have important
No Double Bonds
physiologic and pharmacologic activities. They are syn-
Saturated fatty acids may be envisaged as based on
thesized in vivo by cyclization of the center of the car-
acetic acid (CH3 COOH) as the first member of the
bon chain of 20-carbon (eicosanoic) polyunsaturated
series in which CH2 is progressively added be-
fatty acids (eg, arachidonic acid) to form a cyclopentane
tween the terminal CH3 and COOH groups. Ex-
ring (Figure 14-2). A related series of compounds, the
amples are shown in Table 14-1. Other higher mem-
thromboxanes, have the cyclopentane ring interrupted
bers of the series are known to occur, particularly in
with an oxygen atom (oxane ring) (Figure 14-3). Three
waxes. A few branched-chain fatty acids have also been
different eicosanoic fatty acids give rise to three groups
isolated from both plant and animal sources.
of eicosanoids characterized by the number of double
bonds in the side chains, eg, PG1, PG2, PG3. Different
substituent groups attached to the rings give rise to se-
ries of prostaglandins and thromboxanes, labeled A, B,
Table 14-1. Saturated fatty acids.
etc—eg, the “E” type of prostaglandin (as in PGE2) has
a keto group in position 9, whereas the “F” type has a
Common
Number of
hydroxyl group in this position. The leukotrienes and
Name
C Atoms
lipoxins are a third group of eicosanoid derivatives
Acetic
2
Major end product of carbohy-
formed via the lipoxygenase pathway (Figure 14-4).
drate fermentation by rumen
They are characterized by the presence of three or four
organisms1
conjugated double bonds, respectively. Leukotrienes
cause bronchoconstriction as well as being potent
Propionic
3
An end product of carbohydrate
fermentation by rumen
proinflammatory agents and play a part in asthma.
organisms1
Butyric
4
In certain fats in small amounts
Most Naturally Occurring Unsaturated
(especially butter). An end product
Valeric
5
Fatty Acids Have cis Double Bonds
of carbohydrate fermentation by
Caproic
6
rumen organisms1
The carbon chains of saturated fatty acids form a zigzag
pattern when extended, as at low temperatures. At
Lauric
12
Spermaceti, cinnamon, palm ker-
higher temperatures, some bonds rotate, causing chain
nel, coconut oils, laurels, butter
shortening, which explains why biomembranes become
Myristic
14
Nutmeg, palm kernel, coconut oils,
thinner with increases in temperature. A type of geo-
myrtles, butter
metric isomerism occurs in unsaturated fatty acids, de-
Palmitic
16
Common in all animal and plant
pending on the orientation of atoms or groups around
fats
the axes of double bonds, which do not allow rotation.
Stearic
18
If the acyl chains are on the same side of the bond, it
1Also formed in the cecum of herbivores and to a lesser extent in
is cis-, as in oleic acid; if on opposite sides, it is trans-, as
the colon of humans.
in elaidic acid, the trans isomer of oleic acid
(Fig-
LIPIDS OF PHYSIOLOGIC SIGNIFICANCE
/
113
Table 14-2. Unsaturated fatty acids of physiologic and nutritional significance.
Number of C
Atoms and Number
and Position of
Common
Double Bonds
Family
Name
Systematic Name
Occurrence
Monoenoic acids (one double bond)
16:1;9
ω7
Palmitoleic
cis-9-Hexadecenoic
In nearly all fats.
18:1;9
ω9
Oleic
cis-9-Octadecenoic
Possibly the most common fatty acid in
natural fats.
18:1;9
ω9
Elaidic
trans-9-Octadecenoic
Hydrogenated and ruminant fats.
Dienoic acids (two double bonds)
18:2;9,12
ω6
Linoleic
all-cis-9,12-Octadecadienoic
Corn, peanut, cottonseed, soybean,
and many plant oils.
Trienoic acids (three double bonds)
18:3;6,9,12
ω6
γ-Linolenic
all-cis-6,9,12-Octadecatrienoic
Some plants, eg, oil of evening prim-
rose, borage oil; minor fatty acid in
animals.
18:3;9,12,15
ω3
α-Linolenic
all-cis-9,12,15-Octadecatrienoic
Frequently found with linoleic acid but
particularly in linseed oil.
Tetraenoic acids (four double bonds)
20:4;5,8,11,14
ω6
Arachidonic
all-cis-5,8,11,14-Eicosatetraenoic
Found in animal fats and in peanut oil;
important component of phospho-
lipids in animals.
Pentaenoic acids (five double bonds)
20:5;5,8,11,14,17
ω3
Timnodonic
all-cis-5,8,11,14,17-Eicosapentaenoic
Important component of fish oils, eg,
cod liver, mackerel, menhaden, salmon
oils.
Hexaenoic acids (six double bonds)
22:6;4,7,10,13,16,19
ω3
Cervonic
all-cis-4,7,10,13,16,19-Docosahexaenoic
Fish oils, phospholipids in brain.
ure 14-5). Naturally occurring unsaturated long-chain
U shape. This has profound significance on molecular
fatty acids are nearly all of the cis configuration, the
packing in membranes and on the positions occupied
molecules being
“bent”
120 degrees at the double
by fatty acids in more complex molecules such as phos-
bond. Thus, oleic acid has an L shape, whereas elaidic
pholipids. Trans double bonds alter these spatial rela-
acid remains “straight.” Increase in the number of cis
tionships. Trans fatty acids are present in certain foods,
double bonds in a fatty acid leads to a variety of possi-
arising as a by-product of the saturation of fatty acids
ble spatial configurations of the molecule—eg, arachi-
during hydrogenation, or “hardening,” of natural oils
donic acid, with four cis double bonds, has “kinks” or a
in the manufacture of margarine. An additional small
O
5
9
COO—
COO—
10
O
11
O
OH
OH
OH
Figure 14-2. Prostaglandin E2 (PGE2).
Figure 14-3. Thromboxane A2 (TXA2).
114
/
CHAPTER 14
O
more unsaturated than storage lipids. Lipids in tissues
that are subject to cooling, eg, in hibernators or in the
COO-
extremities of animals, are more unsaturated.
TRIACYLGLYCEROLS (TRIGLYCERIDES)*
ARE THE MAIN STORAGE FORMS OF
Figure 14-4. Leukotriene A4 (LTA4).
FATTY ACIDS
The triacylglycerols (Figure 14-6) are esters of the tri-
hydric alcohol glycerol and fatty acids. Mono- and di-
contribution comes from the ingestion of ruminant fat
acylglycerols wherein one or two fatty acids are esteri-
that contains trans fatty acids arising from the action of
fied with glycerol are also found in the tissues. These
microorganisms in the rumen.
are of particular significance in the synthesis and hy-
drolysis of triacylglycerols.
Physical and Physiologic Properties
of Fatty Acids Reflect Chain Length
Carbons 1 & 3 of Glycerol Are
and Degree of Unsaturation
Not Identical
The melting points of even-numbered-carbon fatty
To number the carbon atoms of glycerol unambigu-
acids increase with chain length and decrease according
ously, the -sn-
(stereochemical numbering) system is
to unsaturation. A triacylglycerol containing three satu-
used. It is important to realize that carbons 1 and 3 of
rated fatty acids of 12 carbons or more is solid at body
glycerol are not identical when viewed in three dimen-
temperature, whereas if the fatty acid residues are 18:2,
sions (shown as a projection formula in Figure 14-7).
it is liquid to below 0 °C. In practice, natural acylglyc-
Enzymes readily distinguish between them and are
erols contain a mixture of fatty acids tailored to suit
nearly always specific for one or the other carbon; eg,
their functional roles. The membrane lipids, which
glycerol is always phosphorylated on sn-3 by glycerol
must be fluid at all environmental temperatures, are
kinase to give glycerol 3-phosphate and not glycerol
1-phosphate.
18
CH3
CH3
PHOSPHOLIPIDS ARE THE MAIN LIPID
CONSTITUENTS OF MEMBRANES
Phospholipids may be regarded as derivatives of phos-
Trans form
(elaidic acid)
phatidic acid (Figure 14-8), in which the phosphate is
esterified with the OH of a suitable alcohol. Phos-
phatidic acid is important as an intermediate in the syn-
thesis of triacylglycerols as well as phosphoglycerols but
120
10
is not found in any great quantity in tissues.
H
H
C
C
Cis form
(oleic acid)
C
C
Phosphatidylcholines (Lecithins)
9
H
Occur in Cell Membranes
110
Phosphoacylglycerols containing choline (Figure 14-8)
are the most abundant phospholipids of the cell mem-
* According to the standardized terminology of the International
Union of Pure and Applied Chemistry (IUPAC) and the Interna-
1
tional Union of Biochemistry (IUB), the monoglycerides, diglyc-
COO-
COO-
erides, and triglycerides should be designated monoacylglycerols,
diacylglycerols, and triacylglycerols, respectively. However, the
Figure 14-5.
Geometric isomerism of ∆9, 18:1 fatty
older terminology is still widely used, particularly in clinical medi-
acids (oleic and elaidic acids).
cine.
LIPIDS OF PHYSIOLOGIC SIGNIFICANCE
/
115
O
O
O
1CH2
O C
R1
O
1CH2
O C
R1
R2
C
O
2CH
O
R2
C
O
2CH
O
3CH2
O C
R2
3CH2
O
P
O-
O-
Figure 14-6. Triacylglycerol.
Phosphatidic acid
brane and represent a large proportion of the body’s
store of choline. Choline is important in nervous trans-
CH3
+
mission, as acetylcholine, and as a store of labile methyl
A
O CH2
CH2
N
CH3
groups. Dipalmitoyl lecithin is a very effective surface-
CH3
active agent and a major constituent of the surfactant
preventing adherence, due to surface tension, of the
Choline
inner surfaces of the lungs. Its absence from the lungs
+
of premature infants causes respiratory distress syn-
O CH2
CH2NH3
B
drome. Most phospholipids have a saturated acyl radi-
cal in the sn-1 position but an unsaturated radical in the
Ethanolamine
sn-2 position of glycerol.
Phosphatidylethanolamine (cephalin) and phos-
NH3+
phatidylserine
(found in most tissues) differ from
phosphatidylcholine only in that ethanolamine or ser-
C
O CH2
CH COO-
ine, respectively, replaces choline (Figure 14-8).
Serine
Phosphatidylinositol Is a Precursor
OH
OH
of Second Messengers
2
3
O H
H
H
The inositol is present in phosphatidylinositol as the
1
4
stereoisomer, myoinositol (Figure 14-8). Phosphati-
D
H
H
OH
OH
dylinositol
4,5-bisphosphate is an important con-
6
5
stituent of cell membrane phospholipids; upon stimula-
tion by a suitable hormone agonist, it is cleaved into
OH
H
diacylglycerol and inositol trisphosphate, both of
which act as internal signals or second messengers.
Myoinositol
O-
Cardiolipin Is a Major Lipid
of Mitochondrial Membranes
CH2
O
P
O
CH2
O
Phosphatidic acid is a precursor of phosphatidylglyc-
E
H
C
OH
O
H
C
O
C
R3
erol which, in turn, gives rise to cardiolipin (Figure
O
CH2
R4
C
O
CH2
14-8).
O
Phosphatidylglycerol
1
H2C
O C
R1
Figure 14-8.
Phosphatidic acid and its derivatives.
O
The O− shown shaded in phosphatidic acid is substi-
R2
C
O
tuted by the substituents shown to form in (A) 3-phos-
2C
phatidylcholine, (B) 3-phosphatidylethanolamine,
O
(C) 3-phosphatidylserine, (D) 3-phosphatidylinositol,
H2
3C
O C
R3
and (E) cardiolipin (diphosphatidylglycerol).
Figure 14-7. Triacyl-sn-glycerol.
116
/
CHAPTER 14
Lysophospholipids Are Intermediates in
O
1CH2
O CH CH
R1
the Metabolism of Phosphoglycerols
R2
C O
2CH
O
These are phosphoacylglycerols containing only one
3
+
CH2
O P O CH2
CH2
NH3
acyl radical, eg, lysophosphatidylcholine
(lysoleci-
thin), important in the metabolism and interconver-
O-
Ethanolamine
sion of phospholipids (Figure 14-9).It is also found in
oxidized lipoproteins and has been implicated in some
Figure 14-10. Plasmalogen.
of their effects in promoting atherosclerosis.
Plasmalogens Occur in Brain & Muscle
sphingolipid of brain and other nervous tissue, found in
These compounds constitute as much as 10% of the
relatively low amounts elsewhere. It contains a number
phospholipids of brain and muscle. Structurally, the
of characteristic C24
fatty acids, eg, cerebronic acid.
plasmalogens resemble phosphatidylethanolamine but
Galactosylceramide (Figure 14-12) can be converted to
possess an ether link on the sn-1 carbon instead of the
sulfogalactosylceramide
(sulfatide), present in high
ester link found in acylglycerols. Typically, the alkyl
amounts in myelin. Glucosylceramide is the predomi-
radical is an unsaturated alcohol (Figure 14-10). In
nant simple glycosphingolipid of extraneural tissues,
some instances, choline, serine, or inositol may be sub-
also occurring in the brain in small amounts. Ganglio-
stituted for ethanolamine.
sides are complex glycosphingolipids derived from glu-
cosylceramide that contain in addition one or more
Sphingomyelins Are Found
molecules of a sialic acid. Neuraminic acid (NeuAc;
in the Nervous System
see Chapter 13) is the principal sialic acid found in
human tissues. Gangliosides are also present in nervous
Sphingomyelins are found in large quantities in brain
tissues in high concentration. They appear to have re-
and nerve tissue. On hydrolysis, the sphingomyelins
ceptor and other functions. The simplest ganglioside
yield a fatty acid, phosphoric acid, choline, and a com-
found in tissues is GM3, which contains ceramide, one
plex amino alcohol, sphingosine (Figure 14-11). No
molecule of glucose, one molecule of galactose, and one
glycerol is present. The combination of sphingosine
molecule of NeuAc. In the shorthand nomenclature
plus fatty acid is known as ceramide, a structure also
used, G represents ganglioside; M is a monosialo-
found in the glycosphingolipids (see below).
containing species; and the subscript 3 is a number as-
signed on the basis of chromatographic migration. GM1
GLYCOLIPIDS (GLYCOSPHINGOLIPIDS)
(Figure 14-13), a more complex ganglioside derived
ARE IMPORTANT IN NERVE TISSUES
from GM3, is of considerable biologic interest, as it is
& IN THE CELL MEMBRANE
known to be the receptor in human intestine for
cholera toxin. Other gangliosides can contain anywhere
Glycolipids are widely distributed in every tissue of the
from one to five molecules of sialic acid, giving rise to
body, particularly in nervous tissue such as brain. They
di-, trisialogangliosides, etc.
occur particularly in the outer leaflet of the plasma
membrane, where they contribute to cell surface car-
bohydrates.
The major glycolipids found in animal tissues are
Ceramide
glycosphingolipids. They contain ceramide and one or
Sphingosine
more sugars. Galactosylceramide is a major glyco-
OH
O
H
CH
3
(CH2)12
CH CH CH
CH
N
C R
O
1
CH2
Fatty acid
CH2
O C
R
2
O
HO
CH
O
CH
3
Phosphoric acid
+
O P
O-
3CH2
O P O CH2
CH2
N
CH3
+
CH3
O CH2
CH2
N(CH3)3
O-
Choline
Choline
Figure 14-9. Lysophosphatidylcholine (lysolecithin).
Figure 14-11. A sphingomyelin.
LIPIDS OF PHYSIOLOGIC SIGNIFICANCE
/
117
Ceramide
Sphingosine
OH
O
H
CH3
(CH
2 )12
CH CH
CH
CH
N
C
CH(OH)
(CH2 )21
CH3
CH
Fatty acid
2OH
(eg, cerebronic acid)
O
HO
H
Galactose
O
CH2
H
OR H
H
Figure 14-12. Structure of galactosylcer-
amide (galactocerebroside, R = H), and sul-
3
fogalactosylceramide (a sulfatide, R = SO42−).
H
OH
STEROIDS PLAY MANY
groups and no carbonyl or carboxyl groups, it is a
sterol, and the name terminates in -ol.
PHYSIOLOGICALLY IMPORTANT ROLES
Cholesterol is probably the best known steroid because
Because of Asymmetry in the Steroid
of its association with atherosclerosis. However, bio-
Molecule, Many Stereoisomers
chemically it is also of significance because it is the pre-
Are Possible
cursor of a large number of equally important steroids
that include the bile acids, adrenocortical hormones,
Each of the six-carbon rings of the steroid nucleus is ca-
sex hormones, D vitamins, cardiac glycosides, sitos-
pable of existing in the three-dimensional conformation
terols of the plant kingdom, and some alkaloids.
either of a “chair” or a “boat” (Figure 14-15). In natu-
All of the steroids have a similar cyclic nucleus re-
rally occurring steroids, virtually all the rings are in the
sembling phenanthrene (rings A, B, and C) to which a
“chair” form, which is the more stable conformation.
cyclopentane ring (D) is attached. The carbon positions
With respect to each other, the rings can be either cis or
on the steroid nucleus are numbered as shown in Figure
trans (Figure 14-16). The junction between the A and
14-14. It is important to realize that in structural for-
B rings can be cis or trans in naturally occurring
mulas of steroids, a simple hexagonal ring denotes a
steroids. That between B and C is trans, as is usually the
completely saturated six-carbon ring with all valences
C/D junction. Bonds attaching substituent groups
satisfied by hydrogen bonds unless shown otherwise; ie,
above the plane of the rings (β bonds) are shown with
it is not a benzene ring. All double bonds are shown as
bold solid lines, whereas those bonds attaching groups
such. Methyl side chains are shown as single bonds un-
below (α bonds) are indicated with broken lines. The A
attached at the farther (methyl) end. These occur typi-
ring of a
5α steroid is always trans to the B ring,
cally at positions 10 and 13 (constituting C atoms 19
whereas it is cis in a 5β steroid. The methyl groups at-
and 18). A side chain at position 17 is usual (as in cho-
tached to C10 and C13 are invariably in the β configura-
lesterol). If the compound has one or more hydroxyl
tion.
Ceramide Glucose Galactose N-Acetylgalactosamine
(Acyl-
sphingo-
NeuAc
Galactose
18
sine)
12
17
or
11
16
19
13
C
D
1
9
Cer Glc Gal GalNAc Gal
15
14
2
10
8
A
B
NeuAc
3
7
5
4
6
Figure 14-13. GM1 ganglioside, a monosialoganglio-
side, the receptor in human intestine for cholera toxin.
Figure 14-14. The steroid nucleus.
118
/
CHAPTER 14
chain alcohol dolichol (Figure 14-20), which takes
part in glycoprotein synthesis by transferring carbohy-
drate residues to asparagine residues of the polypeptide
(Chapter 47). Plant-derived isoprenoid compounds in-
“Chair” form
“Boat” form
clude rubber, camphor, the fat-soluble vitamins A, D,
E, and K, and β-carotene (provitamin A).
Figure 14-15. Conformations of stereoisomers of
the steroid nucleus.
LIPID PEROXIDATION IS A SOURCE
OF FREE RADICALS
Cholesterol Is a Significant Constituent
Peroxidation
(auto-oxidation) of lipids exposed to
of Many Tissues
oxygen is responsible not only for deterioration of foods
Cholesterol (Figure 14-17) is widely distributed in all
(rancidity) but also for damage to tissues in vivo,
cells of the body but particularly in nervous tissue. It is
where it may be a cause of cancer, inflammatory dis-
a major constituent of the plasma membrane and of
eases, atherosclerosis, and aging. The deleterious effects
plasma lipoproteins. It is often found as cholesteryl
are considered to be caused by free radicals (ROO•,
ester, where the hydroxyl group on position 3 is esteri-
RO•, OH•) produced during peroxide formation from
fied with a long-chain fatty acid. It occurs in animals
fatty acids containing methylene-interrupted double
but not in plants.
bonds, ie, those found in the naturally occurring
polyunsaturated fatty acids (Figure 14-21). Lipid per-
Ergosterol Is a Precursor of Vitamin D
oxidation is a chain reaction providing a continuous
supply of free radicals that initiate further peroxidation.
Ergosterol occurs in plants and yeast and is important
The whole process can be depicted as follows:
as a precursor of vitamin D (Figure 14-18). When irra-
(1) Initiation:
diated with ultraviolet light, it acquires antirachitic
properties consequent to the opening of ring B.
(n)+
(n–1)+
+
ROOH+Metal
→ROO•+Metal
+
H
Polyprenoids Share the Same Parent
X•+RH → R•+ XH
Compound as Cholesterol
Although not steroids, these compounds are related be-
(2) Propagation:
cause they are synthesized, like cholesterol
(Figure
26-2), from five-carbon isoprene units (Figure 14-19).
They include ubiquinone (Chapter 12), a member of
2
R•+ O → ROO•
the respiratory chain in mitochondria, and the long-
ROO•+ RH → ROOH+ R•, etc
A
B
H
13
H
10
D
B
10
C
5
9
8
14
B
A
A
3
5
H
H
3
or
H
17
or
13
C
D
1
9
H
1
14
10 H
8
H
A
10
5
B
A
B
5
3
3
H
H
Figure 14-16. Generalized steroid nucleus, showing (A) an all-trans configuration be-
tween adjacent rings and (B) a cis configuration between rings A and B.
LIPIDS OF PHYSIOLOGIC SIGNIFICANCE
/
119
CH3
CH C CH CH
17
Figure 14-19. Isoprene unit.
3
HO
5
6
Peroxidation is also catalyzed in vivo by heme com-
Figure 14-17. Cholesterol, 3-hydroxy-5,6-
pounds and by lipoxygenases found in platelets and
cholestene.
leukocytes. Other products of auto-oxidation or en-
zymic oxidation of physiologic significance include
oxysterols (formed from cholesterol) and isoprostanes
(3) Termination:
(prostanoids).
ROO•+ROO•→ROOR+O
AMPHIPATHIC LIPIDS SELF-ORIENT
2
AT OIL:WATER INTERFACES
ROO•+R•→ROOR
They Form Membranes, Micelles,
R•+ R• → RR
Liposomes, & Emulsions
Since the molecular precursor for the initiation
In general, lipids are insoluble in water since they
process is generally the hydroperoxide product ROOH,
contain a predominance of nonpolar
(hydrocarbon)
lipid peroxidation is a chain reaction with potentially
groups. However, fatty acids, phospholipids, sphin-
devastating effects. To control and reduce lipid peroxi-
golipids, bile salts, and, to a lesser extent, cholesterol
dation, both humans in their activities and nature in-
contain polar groups. Therefore, part of the molecule is
voke the use of antioxidants. Propyl gallate, butylated
hydrophobic, or water-insoluble; and part is hydro-
hydroxyanisole (BHA), and butylated hydroxytoluene
philic, or water-soluble. Such molecules are described
(BHT) are antioxidants used as food additives. Natu-
as amphipathic (Figure 14-22). They become oriented
rally occurring antioxidants include vitamin E (tocoph-
at oil:water interfaces with the polar group in the water
erol), which is lipid-soluble, and urate and vitamin C,
phase and the nonpolar group in the oil phase. A bi-
which are water-soluble. Beta-carotene is an antioxidant
layer of such amphipathic lipids has been regarded as a
at low PO2. Antioxidants fall into two classes: (1) pre-
basic structure in biologic membranes (Chapter 41).
ventive antioxidants, which reduce the rate of chain ini-
When a critical concentration of these lipids is present
tiation; and (2) chain-breaking antioxidants, which in-
in an aqueous medium, they form micelles. Aggrega-
terfere with chain propagation. Preventive antioxidants
tions of bile salts into micelles and liposomes and the
include catalase and other peroxidases that react with
formation of mixed micelles with the products of fat di-
ROOH and chelators of metal ions such as EDTA
gestion are important in facilitating absorption of lipids
(ethylenediaminetetraacetate) and DTPA (diethylene-
from the intestine. Liposomes may be formed by soni-
triaminepentaacetate). In vivo, the principal chain-
cating an amphipathic lipid in an aqueous medium.
breaking antioxidants are superoxide dismutase, which
They consist of spheres of lipid bilayers that enclose
acts in the aqueous phase to trap superoxide free radi-
part of the aqueous medium. They are of potential clin-
cals (O2•); perhaps urate; and vitamin E, which acts in
ical use—particularly when combined with tissue-
the lipid phase to trap ROO• radicals (Figure 45-6).
specific antibodies—as carriers of drugs in the circula-
tion, targeted to specific organs, eg, in cancer therapy.
In addition, they are being used for gene transfer into
vascular cells and as carriers for topical and transdermal
CH2OH
B
HO
16
Figure 14-18. Ergosterol.
Figure 14-20. Dolichol—a C95 alcohol.
120
/
CHAPTER 14
RH
R•
R•
ROO •
X•
XH
H
O2
H O O•
•
•
H H
H
RH
O
O
O O
H OOH
•
+R•
H
H
Malondialdehyde
Endoperoxide
Hydroperoxide
ROOH
Figure 14-21. Lipid peroxidation. The reaction is initiated by an existing free radical (X•), by light, or by
metal ions. Malondialdehyde is only formed by fatty acids with three or more double bonds and is used as a
measure of lipid peroxidation together with ethane from the terminal two carbons of ω3 fatty acids and pen-
tane from the terminal five carbons of ω6 fatty acids.
AMPHIPATHIC LIPID
A
Polar or
hydrophiIic groups
Nonpolar or
hydrophobic groups
Aqueous phase
Aqueous phase
Aqueous phase
“Oil” or
nonpolar phase
Nonpolar
phase
“Oil” or nonpolar phase
Aqueous phase
LIPID BILAYER
MICELLE
OIL IN WATER EMULSION
B
C
D
Nonpolar
phase
Aqueous
Aqueous
phase
phase
Lipid
Aqueous
Lipid
bilayer
compartments
bilayers
LIPOSOME
LIPOSOME
(UNILAMELLAR)
(MULTILAMELLAR)
E
F
Figure 14-22. Formation of lipid membranes, micelles, emulsions, and liposomes from am-
phipathic lipids, eg, phospholipids.
LIPIDS OF PHYSIOLOGIC SIGNIFICANCE
/
121
delivery of drugs and cosmetics. Emulsions are much
are amphipathic lipids and have important roles—as
larger particles, formed usually by nonpolar lipids in an
major constituents of membranes and the outer layer
aqueous medium. These are stabilized by emulsifying
of lipoproteins, as surfactant in the lung, as precur-
agents such as amphipathic lipids (eg, lecithin), which
sors of second messengers, and as constituents of ner-
form a surface layer separating the main bulk of the
vous tissue.
nonpolar material from the aqueous phase
(Figure
• Glycolipids are also important constituents of ner-
14-22).
vous tissue such as brain and the outer leaflet of the
cell membrane, where they contribute to the carbo-
SUMMARY
hydrates on the cell surface.
• Cholesterol, an amphipathic lipid, is an important
• Lipids have the common property of being relatively
component of membranes. It is the parent molecule
insoluble in water (hydrophobic) but soluble in non-
from which all other steroids in the body, including
polar solvents. Amphipathic lipids also contain one
major hormones such as the adrenocortical and sex
or more polar groups, making them suitable as con-
hormones, D vitamins, and bile acids, are synthe-
stituents of membranes at lipid:water interfaces.
sized.
• The lipids of major physiologic significance are fatty
• Peroxidation of lipids containing polyunsaturated
acids and their esters, together with cholesterol and
fatty acids leads to generation of free radicals that
other steroids.
may damage tissues and cause disease.
• Long-chain fatty acids may be saturated, monounsat-
urated, or polyunsaturated, according to the number
of double bonds present. Their fluidity decreases
REFERENCES
with chain length and increases according to degree
of unsaturation.
Benzie IFF: Lipid peroxidation: a review of causes, consequences,
• Eicosanoids are formed from 20-carbon polyunsatu-
measurement and dietary influences. Int J Food Sci Nutr
1996;47:233.
rated fatty acids and make up an important group of
Christie WW: Lipid Analysis, 2nd ed. Pergamon Press, 1982.
physiologically and pharmacologically active com-
pounds known as prostaglandins, thromboxanes,
Cullis PR, Fenske DB, Hope MJ: Physical properties and func-
tional roles of lipids in membranes. In: Biochemistry of Lipids,
leukotrienes, and lipoxins.
Lipoproteins and Membranes. Vance DE, Vance JE (editors).
• The esters of glycerol are quantitatively the most sig-
Elsevier, 1996.
nificant lipids, represented by triacylglycerol (“fat”),
Gunstone FD, Harwood JL, Padley FB: The Lipid Handbook.
a major constituent of lipoproteins and the storage
Chapman & Hall, 1986.
form of lipid in adipose tissue. Phosphoacylglycerols
Gurr MI, Harwood JL: Lipid Biochemistry: An Introduction, 4th ed.
Chapman & Hall, 1991.
Overview of Metabolism
15
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
Carbohydrate Metabolism Is Centered
on the Provision & Fate of Glucose
The fate of dietary components after digestion and ab-
(Figure 15-2)
sorption constitutes metabolism—the metabolic path-
ways taken by individual molecules, their interrelation-
Glucose is metabolized to pyruvate by the pathway of
ships, and the mechanisms that regulate the flow of
glycolysis, which can occur anaerobically (in the ab-
metabolites through the pathways. Metabolic pathways
sence of oxygen), when the end product is lactate. Aero-
fall into three categories: (1) Anabolic pathways are
bic tissues metabolize pyruvate to acetyl-CoA, which
those involved in the synthesis of compounds. Protein
can enter the citric acid cycle for complete oxidation
synthesis is such a pathway, as is the synthesis of fuel
to CO2 and H2O, linked to the formation of ATP
reserves of triacylglycerol and glycogen. Anabolic path-
in the process of oxidative phosphorylation (Figure
ways are endergonic. (2) Catabolic pathways are in-
16-2). Glucose is the major fuel of most tissues.
volved in the breakdown of larger molecules, com-
monly involving oxidative reactions; they are exergonic,
producing reducing equivalents and, mainly via the res-
piratory chain, ATP. (3) Amphibolic pathways occur
Carbohydrate
Protein
Fat
at the “crossroads” of metabolism, acting as links be-
tween the anabolic and catabolic pathways, eg, the cit-
ric acid cycle.
Digestion and absorption
A knowledge of normal metabolism is essential for
an understanding of abnormalities underlying disease.
Normal metabolism includes adaptation to periods of
Simple sugars
Fatty acids
Amino acids
starvation, exercise, pregnancy, and lactation. Abnor-
(mainly glucose)
+ glycerol
mal metabolism may result from nutritional deficiency,
enzyme deficiency, abnormal secretion of hormones, or
the actions of drugs and toxins. An important example
Catabolism
of a metabolic disease is diabetes mellitus.
Acetyl-CoA
PATHWAYS THAT PROCESS THE MAJOR
PRODUCTS OF DIGESTION
The nature of the diet sets the basic pattern of metabo-
Citric
lism. There is a need to process the products of diges-
acid
2H
ATP
cycle
tion of dietary carbohydrate, lipid, and protein. These
are mainly glucose, fatty acids and glycerol, and amino
acids, respectively. In ruminants (and to a lesser extent
in other herbivores), dietary cellulose is fermented by
2CO2
symbiotic microorganisms to short-chain fatty acids
(acetic, propionic, butyric), and metabolism in these
Figure 15-1.
Outline of the pathways for the catab-
animals is adapted to use these fatty acids as major sub-
olism of dietary carbohydrate, protein, and fat. All the
strates. All the products of digestion are metabolized to
pathways lead to the production of acetyl-CoA, which is
a common product, acetyl-CoA, which is then oxi-
oxidized in the citric acid cycle, ultimately yielding ATP
dized by the citric acid cycle (Figure 15-1).
in the process of oxidative phosphorylation.
122
OVERVIEW OF METABOLISM
/
123
Diet
cursor of fatty acids and cholesterol (and hence of all
steroids synthesized in the body). Gluconeogenesis is
the process of forming glucose from noncarbohydrate
Glucose
Glycogen
precursors, eg, lactate, amino acids, and glycerol.
Glucose
Lipid Metabolism Is Concerned Mainly
3CO2
phosphates
With Fatty Acids & Cholesterol
(Figure 15-3)
Pentose phosphate
pathway
The source of long-chain fatty acids is either dietary
lipid or de novo synthesis from acetyl-CoA derived from
Triose
carbohydrate. Fatty acids may be oxidized to acetyl-
Ribose
RNA
phosphates
CoA (β-oxidation) or esterified with glycerol, forming
phosphate
DNA
triacylglycerol (fat) as the body’s main fuel reserve.
Acetyl-CoA formed by β-oxidation may undergo
several fates:
(1) As with acetyl-CoA arising from glycolysis, it is
Pyruvate
Lactate
oxidized to CO2 + H2O via the citric acid cycle.
Acylglycerols
(fat)
CO2
Triacylglycerol
Steroids
Acetyl-CoA
Fatty
(fat)
acids
Cholesterol
Fatty acids
Diet
Citric
acid
cycle
Cholesterol
Carbohydrate
2CO2
Acetyl-CoA
Amino acids
Cholesterologenesis
Figure 15-2. Overview of carbohydrate metabolism
Ketogenesis
showing the major pathways and end products. Gluco-
neogenesis is not shown.
Ketone
bodies
Citric
Glucose and its metabolites also take part in other
acid
cycle
processes. Examples:
(1) Conversion to the storage
polymer glycogen in skeletal muscle and liver. (2) The
pentose phosphate pathway, an alternative to part of
the pathway of glycolysis, is a source of reducing equiv-
alents (NADPH) for biosynthesis and the source of ri-
2CO2
bose for nucleotide and nucleic acid synthesis.
(3) Triose phosphate gives rise to the glycerol moiety
Figure 15-3. Overview of fatty acid metabolism
of triacylglycerols.
(4) Pyruvate and intermediates of
showing the major pathways and end products. Ketone
the citric acid cycle provide the carbon skeletons for
bodies comprise the substances acetoacetate, 3-hy-
the synthesis of amino acids; and acetyl-CoA, the pre-
droxybutyrate, and acetone.
124
/
CHAPTER 15
(2) It is the precursor for synthesis of cholesterol and
METABOLIC PATHWAYS MAY BE
other steroids.
STUDIED AT DIFFERENT LEVELS
(3) In the liver, it forms ketone bodies (acetone, ace-
OF ORGANIZATION
toacetate, and 3-hydroxybutyrate) that are impor-
tant fuels in prolonged starvation.
In addition to studies in the whole organism, the loca-
tion and integration of metabolic pathways is revealed
by studies at several levels of organization. At the tissue
and organ level, the nature of the substrates entering
Much of Amino Acid Metabolism
and metabolites leaving tissues and organs is defined. At
Involves Transamination
the subcellular level, each cell organelle (eg, the mito-
(Figure 15-4)
chondrion) or compartment (eg, the cytosol) has spe-
cific roles that form part of a subcellular pattern of
The amino acids are required for protein synthesis.
metabolic pathways.
Some must be supplied in the diet (the essential amino
acids) since they cannot be synthesized in the body.
The remainder are nonessential amino acids that are
At the Tissue and Organ Level, the Blood
supplied in the diet but can be formed from metabolic
Circulation Integrates Metabolism
intermediates by transamination, using the amino ni-
trogen from other amino acids. After deamination,
Amino acids resulting from the digestion of dietary
amino nitrogen is excreted as urea, and the carbon
protein and glucose resulting from the digestion of car-
skeletons that remain after transamination (1) are oxi-
bohydrate are absorbed and directed to the liver via the
dized to CO2 via the citric acid cycle, (2) form glucose
hepatic portal vein. The liver has the role of regulating
(gluconeogenesis), or (3) form ketone bodies.
the blood concentration of most water-soluble metabo-
Several amino acids are also the precursors of other
lites
(Figure
15-5). In the case of glucose, this is
compounds, eg, purines, pyrimidines, hormones such
achieved by taking up glucose in excess of immediate
as epinephrine and thyroxine, and neurotransmitters.
requirements and converting it to glycogen (glycogene-
Diet protein
Nonprotein
Tissue protein
Amino acids
nitrogen derivatives
T R A N S A M I N A T I O N
Carbohydrate
Ketone bodies
(glucose)
Amino nitrogen in
Acetyl-CoA
glutamate
DEAMINATION
Citric
NH3
acid
cycle
Urea
2CO2
Figure 15-4. Overview of amino acid metabolism showing the major pathways and end products.
OVERVIEW OF METABOLISM
/
125
Plasma proteins
LIVER
Urea
Protein
Amino acids
CO2
Glucose
Amino
acids
Glycogen
Protein
Lactate
Urea
Amino
acids
Alanine,
etc
ERYTHROCYTES
Glucose
CO2
phosphate
Glucose
KIDNEY
Urine
Glycogen
Diet
Carbohydrate
Glucose
BLOOD PLASMA
Protein
MUSCLE
Amino acids
SMALL INTESTINE
Figure 15-5. Transport and fate of major carbohydrate and amino acid substrates and metabolites. Note that
there is little free glucose in muscle, since it is rapidly phosphorylated upon entry.
sis) or to fat (lipogenesis). Between meals, the liver
mucosa. Here they are packaged with protein and se-
acts to maintain the blood glucose concentration from
creted into the lymphatic system and thence into the
glycogen (glycogenolysis) and, together with the kid-
blood stream as chylomicrons, the largest of the plasma
ney, by converting noncarbohydrate metabolites such
lipoproteins. Chylomicrons also contain other lipid-
as lactate, glycerol, and amino acids to glucose (gluco-
soluble nutrients, eg, vitamins. Unlike glucose and
neogenesis). Maintenance of an adequate concentra-
amino acids, chylomicron triacylglycerol is not taken up
tion of blood glucose is vital for those tissues in which it
directly by the liver. It is first metabolized by tissues that
is the major fuel (the brain) or the only fuel (the eryth-
have lipoprotein lipase, which hydrolyzes the triacyl-
rocytes). The liver also synthesizes the major plasma
glycerol, releasing fatty acids that are incorporated into
proteins (eg, albumin) and deaminates amino acids
tissue lipids or oxidized as fuel. The other major source
that are in excess of requirements, forming urea, which
of long-chain fatty acid is synthesis (lipogenesis) from
is transported to the kidney and excreted.
carbohydrate, mainly in adipose tissue and the liver.
Skeletal muscle utilizes glucose as a fuel, forming
Adipose tissue triacylglycerol is the main fuel reserve
both lactate and CO2. It stores glycogen as a fuel for its
of the body. On hydrolysis (lipolysis) free fatty acids are
use in muscular contraction and synthesizes muscle
released into the circulation. These are taken up by most
protein from plasma amino acids. Muscle accounts for
tissues (but not brain or erythrocytes) and esterified to
approximately 50% of body mass and consequently
acylglycerols or oxidized as a fuel. In the liver, triacyl-
represents a considerable store of protein that can be
glycerol arising from lipogenesis, free fatty acids, and
drawn upon to supply amino acids for gluconeogenesis
chylomicron remnants (see Figures 25-3 and 25-4) is se-
in starvation.
creted into the circulation as very low density lipopro-
Lipids in the diet (Figure 15-6) are mainly triacyl-
tein (VLDL). This triacylglycerol undergoes a fate simi-
glycerol and are hydrolyzed to monoacylglycerols and
lar to that of chylomicrons. Partial oxidation of fatty
fatty acids in the gut, then reesterified in the intestinal
acids in the liver leads to ketone body production (keto-
126
/
CHAPTER 15
FFA
CO2
Glucose
Fatty
acids
Ketone
bodies
TG
BLOOD
PLASMA
CO2
LIVER
LPL
Fatty
acids
Lipoprotein
Fatty
TG
Glucose
acids
LPL
TG
MUSCLE
TG
Diet
MG +
ADIPOSE
TG
TG
fatty acids
TISSUE
SMALL INTESTINE
Figure 15-6. Transport and fate of major lipid substrates and metabolites. (FFA, free fatty acids; LPL, lipopro-
tein lipase; MG, monoacylglycerol; TG, triacylglycerol; VLDL, very low density lipoprotein.)
genesis). Ketone bodies are transported to extrahepatic
Glycolysis, the pentose phosphate pathway, and fatty
tissues, where they act as a fuel source in starvation.
acid synthesis are all found in the cytosol. In gluconeo-
genesis, substrates such as lactate and pyruvate, which
are formed in the cytosol, enter the mitochondrion to
At the Subcellular Level, Glycolysis Occurs
yield oxaloacetate before formation of glucose.
in the Cytosol & the Citric Acid Cycle
The membranes of the endoplasmic reticulum con-
in the Mitochondria
tain the enzyme system for acylglycerol synthesis, and
Compartmentation of pathways in separate subcellular
the ribosomes are responsible for protein synthesis.
compartments or organelles permits integration and
regulation of metabolism. Not all pathways are of equal
THE FLUX OF METABOLITES IN
importance in all cells. Figure 15-7 depicts the subcel-
METABOLIC PATHWAYS MUST BE
lular compartmentation of metabolic pathways in a he-
REGULATED IN A CONCERTED MANNER
patic parenchymal cell.
The central role of the mitochondrion is immedi-
Regulation of the overall flux through a pathway is im-
ately apparent, since it acts as the focus of carbohydrate,
portant to ensure an appropriate supply, when re-
lipid, and amino acid metabolism. It contains the en-
quired, of the products of that pathway. Regulation is
zymes of the citric acid cycle, β-oxidation of fatty acids,
achieved by control of one or more key reactions in
and ketogenesis, as well as the respiratory chain and
the pathway, catalyzed by “regulatory enzymes.” The
ATP synthase.
physicochemical factors that control the rate of an
OVERVIEW OF METABOLISM
/
127
CYTOSOL
Glycogen
AA
Protein
Ribosome
ENDOPLASMIC
Pentose
RETICULUM
Glucose
phosphate
pathway
Triose phosphate
Glycerol phosphate
Triacylglycerol
Fatty acids
Glycerol
Glycolysis
Phosphoenolpyruvate
Lactate
Pyruvate
AA
AA
Pyruvate
CO2
Oxaloacetate
Acetyl-CoA
Ketone
bodies
AA
Fumarate
AA
Citrate
Citric acid
cycle
AA
CO
2
AA
Succinyl-CoA
α-Ketoglutarate
CO2
AA
MITOCHONDRION
AA
AA
Figure 15-7. Intracellular location and overview of major metabolic pathways in a liver parenchymal
cell. (AA →, metabolism of one or more essential amino acids; AA ↔, metabolism of one or more
nonessential amino acids.)
128
/
CHAPTER 15
enzyme-catalyzed reaction, eg, substrate concentration,
In vivo, under “steady-state” conditions, there is a net
are of primary importance in the control of the overall
flux from left to right because there is a continuous sup-
rate of a metabolic pathway (Chapter 9).
ply of A and removal of D. In practice, there are invari-
ably one or more nonequilibrium reactions in a meta-
“Nonequilibrium” Reactions Are
bolic pathway, where the reactants are present in
Potential Control Points
concentrations that are far from equilibrium. In at-
tempting to reach equilibrium, large losses of free en-
In a reaction at equilibrium, the forward and reverse re-
ergy occur as heat, making this type of reaction essen-
actions occur at equal rates, and there is therefore no
tially irreversible, eg,
net flux in either direction:
Heat
A↔B↔C↔D
A↔B→C↔D
Inactive
Enz1
2
2
+
+
Ca2+/calmodulin
cAMP
Cell
membrane
X
Y
Active
Enz1
A
A
B
C
D
Enz2
+
-
1
Negative allosteric
Positive allosteric
feed-back
feed-forward
inhibition
activation
+
or
–
+
or
–
Ribosomal synthesis
of new enzyme protein
3
Nuclear production
of mRNA
+
-
4
5
Induction
Repression
Figure 15-8. Mechanisms of control of an enzyme-catalyzed reaction. Circled
numbers indicate possible sites of action of hormones. 1 , Alteration of mem-
brane permeability; 2 , conversion of an inactive to an active enzyme, usually in-
volving phosphorylation/dephosphorylation reactions; 3 , alteration of the rate
of translation of mRNA at the ribosomal level; 4 , induction of new mRNA forma-
tion; and 5 , repression of mRNA formation. 1 and 2 are rapid, whereas 3 - 5
are slower ways of regulating enzyme activity.
OVERVIEW OF METABOLISM
/
129
Such a pathway has both flow and direction. The
activity of existing enzyme molecules, or slowly, by al-
enzymes catalyzing nonequilibrium reactions are usu-
tering the rate of enzyme synthesis.
ally present in low concentrations and are subject to a
variety of regulatory mechanisms. However, many of
SUMMARY
the reactions in metabolic pathways cannot be classified
as equilibrium or nonequilibrium but fall somewhere
•
The products of digestion provide the tissues with
between the two extremes.
the building blocks for the biosynthesis of complex
molecules and also with the fuel to power the living
The Flux-Generating Reaction
processes.
Is the First Reaction in a Pathway
•
Nearly all products of digestion of carbohydrate, fat,
That Is Saturated With Substrate
and protein are metabolized to a common metabo-
lite, acetyl-CoA, before final oxidation to CO2 in the
It may be identified as a nonequilibrium reaction in
citric acid cycle.
which the Km of the enzyme is considerably lower than
•
Acetyl-CoA is also used as the precursor for biosyn-
the normal substrate concentration. The first reaction
in glycolysis, catalyzed by hexokinase (Figure 17-2), is
thesis of long-chain fatty acids; steroids, including
cholesterol; and ketone bodies.
such a flux-generating step because its Km for glucose of
0.05 mmol/L is well below the normal blood glucose
•
Glucose provides carbon skeletons for the glycerol
concentration of 5 mmol/L.
moiety of fat and of several nonessential amino acids.
•
Water-soluble products of digestion are transported
ALLOSTERIC & HORMONAL
directly to the liver via the hepatic portal vein. The
liver regulates the blood concentrations of glucose
MECHANISMS ARE IMPORTANT
and amino acids.
IN THE METABOLIC CONTROL OF
•
Pathways are compartmentalized within the cell.
ENZYME-CATALYZED REACTIONS
Glycolysis, glycogenesis, glycogenolysis, the pentose
A hypothetical metabolic pathway is shown in Figure
phosphate pathway, and lipogenesis occur in the cy-
15-8, in which reactions A ↔ B and C ↔ D are equi-
tosol. The mitochondrion contains the enzymes of
librium reactions and B → C is a nonequilibrium reac-
the citric acid cycle, β-oxidation of fatty acids, and of
tion. The flux through such a pathway can be regulated
oxidative phosphorylation. The endoplasmic reticu-
by the availability of substrate A. This depends on its
lum also contains the enzymes for many other
supply from the blood, which in turn depends on either
processes, including protein synthesis, glycerolipid
food intake or key reactions that maintain and release
formation, and drug metabolism.
substrates from tissue reserves to the blood, eg, the
•
Metabolic pathways are regulated by rapid mecha-
glycogen phosphorylase in liver (Figure 18-1) and hor-
nisms affecting the activity of existing enzymes, eg,
mone-sensitive lipase in adipose tissue (Figure 25-7).
allosteric and covalent modification
(often in re-
The flux also depends on the transport of substrate A
sponse to hormone action); and slow mechanisms af-
across the cell membrane. Flux is also determined by
fecting the synthesis of enzymes.
the removal of the end product D and the availability
of cosubstrate or cofactors represented by X and Y. En-
zymes catalyzing nonequilibrium reactions are often al-
REFERENCES
losteric proteins subject to the rapid actions of “feed-
Cohen P: Control of Enzyme Activity, 2nd ed. Chapman & Hall,
back” or “feed-forward” control by allosteric modifiers
1983.
in immediate response to the needs of the cell (Chap-
Fell D: Understanding the Control of Metabolism. Portland Press,
ter 9). Frequently, the product of a biosynthetic path-
1997.
way will inhibit the enzyme catalyzing the first reaction
Frayn KN: Metabolic Regulation—A Human Perspective. Portland
in the pathway. Other control mechanisms depend on
Press, 1996.
the action of hormones responding to the needs of the
Newsholme EA, Crabtree B: Flux-generating and regulatory steps
body as a whole; they may act rapidly, by altering the
in metabolic control. Trends Biochem Sci 1981;6:53.
The Citric Acid Cycle:
16
The Catabolism of Acetyl-CoA
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
cated in the mitochondrial matrix, either free or at-
tached to the inner mitochondrial membrane, where
The citric acid cycle (Krebs cycle, tricarboxylic acid
the enzymes of the respiratory chain are also found.
cycle) is a series of reactions in mitochondria that oxi-
dize acetyl residues (as acetyl-CoA) and reduce coen-
REACTIONS OF THE CITRIC ACID
zymes that upon reoxidation are linked to the forma-
tion of ATP.
CYCLE LIBERATE REDUCING
The citric acid cycle is the final common pathway
EQUIVALENTS & CO2
for the aerobic oxidation of carbohydrate, lipid, and
(Figure 16-3)*
protein because glucose, fatty acids, and most amino
The initial reaction between acetyl-CoA and oxaloac-
acids are metabolized to acetyl-CoA or intermediates of
the cycle. It also has a central role in gluconeogenesis,
etate to form citrate is catalyzed by citrate synthase
which forms a carbon-carbon bond between the methyl
lipogenesis, and interconversion of amino acids. Many
of these processes occur in most tissues, but the liver is
carbon of acetyl-CoA and the carbonyl carbon of ox-
aloacetate. The thioester bond of the resultant citryl-
the only tissue in which all occur to a significant extent.
The repercussions are therefore profound when, for ex-
CoA is hydrolyzed, releasing citrate and CoASH—an
exergonic reaction.
ample, large numbers of hepatic cells are damaged as in
acute hepatitis or replaced by connective tissue (as in
Citrate is isomerized to isocitrate by the enzyme
aconitase (aconitate hydratase); the reaction occurs in
cirrhosis). Very few, if any, genetic abnormalities of
citric acid cycle enzymes have been reported; such ab-
two steps: dehydration to cis-aconitate, some of which
remains bound to the enzyme; and rehydration to isoci-
normalities would be incompatible with life or normal
development.
trate. Although citrate is a symmetric molecule, aconi-
tase reacts with citrate asymmetrically, so that the two
carbon atoms that are lost in subsequent reactions of
THE CITRIC ACID CYCLE PROVIDES
the cycle are not those that were added from acetyl-
SUBSTRATE FOR THE
CoA. This asymmetric behavior is due to channeling—
transfer of the product of citrate synthase directly onto
RESPIRATORY CHAIN
the active site of aconitase without entering free solu-
The cycle starts with reaction between the acetyl moiety
tion. This provides integration of citric acid cycle activ-
of acetyl-CoA and the four-carbon dicarboxylic acid ox-
ity and the provision of citrate in the cytosol as a source
aloacetate, forming a six-carbon tricarboxylic acid, cit-
of acetyl-CoA for fatty acid synthesis. The poison fluo-
rate. In the subsequent reactions, two molecules of CO2
roacetate is toxic because fluoroacetyl-CoA condenses
are released and oxaloacetate is regenerated
(Figure
with oxaloacetate to form fluorocitrate, which inhibits
16-1). Only a small quantity of oxaloacetate is needed
aconitase, causing citrate to accumulate.
for the oxidation of a large quantity of acetyl-CoA; ox-
Isocitrate undergoes dehydrogenation catalyzed by
aloacetate may be considered to play a catalytic role.
isocitrate dehydrogenase to form, initially, oxalosucci-
The citric acid cycle is an integral part of the process
nate, which remains enzyme-bound and undergoes de-
by which much of the free energy liberated during the
carboxylation to α-ketoglutarate. The decarboxylation
oxidation of fuels is made available. During oxidation
of acetyl-CoA, coenzymes are reduced and subsequently
*From Circular No. 200 of the Committee of Editors of Biochemi-
reoxidized in the respiratory chain, linked to the forma-
cal Journals Recommendations (1975): “According to standard
tion of ATP (oxidative phosphorylation; see Figure
biochemical convention, the ending ate in, eg, palmitate, denotes
16-2 and also Chapter 12). This process is aerobic, re-
any mixture of free acid and the ionized form(s) (according to pH)
quiring oxygen as the final oxidant of the reduced
in which the cations are not specified.” The same convention is
coenzymes. The enzymes of the citric acid cycle are lo-
adopted in this text for all carboxylic acids.
130
THE CITRIC ACID CYCLE: THE CATABOLISM OF ACETYL-CoA
/
131
Acetyl-CoA
Carbohydrate
Protein
Lipids
(C2)
CoA
Acetyl-CoA
(C2)
H2O
Citrate
Oxaloacetate
Citrate
Oxaloacetate
(C6)
(C4)
(C6)
(C4)
H2O
Citric acid
cycle
Cis-aconitate
(C
6)
H2O
Malate
(C4)
2H
Isocitrate
H2O
CO2
CO2
(C6)
2H
CO2
Fumarate
Figure 16-1. Citric acid cycle, illustrating the cat-
(C4)
α-Ketoglutarate
alytic role of oxaloacetate.
(C5)
2H
NAD
CO2
Succinate
Succinyl-CoA
(C4)
requires Mg2+ or Mn2+ ions. There are three isoenzymes
(C4)
2H
P
F
of isocitrate dehydrogenase. One, which uses NAD+, is
p
+
H2O
P
found only in mitochondria. The other two use NADP
and are found in mitochondria and the cytosol. Respi-
Q
ratory chain-linked oxidation of isocitrate proceeds al-
most completely through the NAD+-dependent en-
zyme.
Oxidative
Cyt b
P
α-Ketoglutarate undergoes oxidative decarboxyla-
phosphorylation
tion in a reaction catalyzed by a multi-enzyme complex
Cyt c
similar to that involved in the oxidative decarboxylation
of pyruvate (Figure 17-5). The
-ketoglutarate dehy-
drogenase complex requires the same cofactors as the
Cyt aa3
P
pyruvate dehydrogenase complex—thiamin diphos-
1/2
O2
phate, lipoate, NAD+, FAD, and CoA—and results in
the formation of succinyl-CoA. The equilibrium of this
–
Anaerobiosis
reaction is so much in favor of succinyl-CoA formation
(hypoxia, anoxia)
that it must be considered physiologically unidirec-
H2O
Respiratory chain
tional. As in the case of pyruvate oxidation (Chapter
F
Flavoprotein
17), arsenite inhibits the reaction, causing the substrate,
p
-ketoglutarate, to accumulate.
Cyt
Cytochrome
Succinyl-CoA is converted to succinate by the en-
zyme succinate thiokinase
(succinyl-CoA synthe-
P
High-energy phosphate
tase). This is the only example in the citric acid cycle of
Figure 16-2. The citric acid cycle: the major catabo-
substrate-level phosphorylation. Tissues in which glu-
lic pathway for acetyl-CoA in aerobic organisms. Acetyl-
coneogenesis occurs (the liver and kidney) contain two
CoA, the product of carbohydrate, protein, and lipid ca-
isoenzymes of succinate thiokinase, one specific for
tabolism, is taken into the cycle, together with H2O, and
GDP and the other for ADP. The GTP formed is
oxidized to CO2 with the release of reducing equivalents
used for the decarboxylation of oxaloacetate to phos-
(2H). Subsequent oxidation of 2H in the respiratory
phoenolpyruvate in gluconeogenesis and provides a
chain leads to coupled phosphorylation of ADP to ATP.
regulatory link between citric acid cycle activity and
the withdrawal of oxaloacetate for gluconeogenesis.
For one turn of the cycle, 11~ P are generated via ox-
Nongluconeogenic tissues have only the isoenzyme that
idative phosphorylation and one ~ P arises at substrate
uses ADP.
level from the conversion of succinyl-CoA to succinate.
3
CH CO
S
CoA
Acetyl-CoA
O
CITRATE SYNTHASE
MALATE
DEHYDROGENASE
C COO-
CoA SH
CH2
*OO-
CH2
COO-
NADH + H+
Oxaloacetate
H2O
HO
C
COO-
NAD+
CH2
COO-
CH2
Citrate
L-Malate
ACONITASE
FUMARASE
Fe2+
H2O
Fluoroacetate
CH2
H2O
C
COO-
CH COO-
-
OOC*
C
H
Cis -aconitate
Fumarate
FADH2
H
2O
SUCCINATE
ACONITASE
Fe2+
DEHYDROGENASE
FAD
Malonate
CH2
*OO-
CH2
CH
COO-
CH2
HO CH COO-
Succinate
Isocitrate
ATP
NAD+
Mg2+
CoA SH
NADH + H+
ADP + P
i
ISOCITRATE
SUCCINATE
DEHYDROGENASE
THIOKINASE
CH
2
COO-
-
CH2
COO
CH2
Arsenite
+
NADH + H
CH COO-
O
C
S CoA
CO2
+
Succinyl-CoA
NAD
–
O
C
COO-
CH2
COO
α-KETOGLUTARATE
Oxalosuccinate
DEHYDROGENASE COMPLEX
CH2
ISOCITRATE
CoA SH
Mn2+
DEHYDROGENASE
-
O
C
COO
CO2
α-Ketoglutarate
Figure 16-3. Reactions of the citric acid (Krebs) cycle. Oxidation of NADH and FADH2 in the respiratory chain
leads to the generation of ATP via oxidative phosphorylation. In order to follow the passage of acetyl-CoA through
the cycle, the two carbon atoms of the acetyl radical are shown labeled on the carboxyl carbon (designated by as-
terisk) and on the methyl carbon (using the designation •). Although two carbon atoms are lost as CO2 in one revo-
lution of the cycle, these atoms are not derived from the acetyl-CoA that has immediately entered the cycle but
from that portion of the citrate molecule that was derived from oxaloacetate. However, on completion of a single
turn of the cycle, the oxaloacetate that is regenerated is now labeled, which leads to labeled CO2 being evolved
during the second turn of the cycle. Because succinate is a symmetric compound and because succinate dehydro-
genase does not differentiate between its two carboxyl groups, “randomization” of label occurs at this step such
that all four carbon atoms of oxaloacetate appear to be labeled after one turn of the cycle. During gluconeogene-
sis, some of the label in oxaloacetate is incorporated into glucose and glycogen (Figure 19-1). For a discussion of
the stereochemical aspects of the citric acid cycle, see Greville (1968). The sites of inhibition ( − ) by fluoroacetate,
malonate, and arsenite are indicated.
132
THE CITRIC ACID CYCLE: THE CATABOLISM OF ACETYL-CoA
/
133
When ketone bodies are being metabolized in extra-
the coenzyme for three dehydrogenases in the cycle—
hepatic tissues there is an alternative reaction catalyzed
isocitrate dehydrogenase, α-ketoglutarate dehydrogen-
by succinyl-CoA-acetoacetate-CoA transferase (thio-
ase, and malate dehydrogenase; (3) thiamin (vitamin
phorase)—involving transfer of CoA from succinyl-
B1), as thiamin diphosphate, the coenzyme for decar-
CoA to acetoacetate, forming acetoacetyl-CoA (Chap-
boxylation in the α-ketoglutarate dehydrogenase reac-
ter 22).
tion; and (4) pantothenic acid, as part of coenzyme A,
The onward metabolism of succinate, leading to the
the cofactor attached to “active” carboxylic acid resi-
regeneration of oxaloacetate, is the same sequence of
dues such as acetyl-CoA and succinyl-CoA.
chemical reactions as occurs in the β-oxidation of fatty
acids: dehydrogenation to form a carbon-carbon double
bond, addition of water to form a hydroxyl group, and
THE CITRIC ACID CYCLE PLAYS A
a further dehydrogenation to yield the oxo- group of
PIVOTAL ROLE IN METABOLISM
oxaloacetate.
The citric acid cycle is not only a pathway for oxidation
The first dehydrogenation reaction, forming fu-
of two-carbon units—it is also a major pathway for in-
marate, is catalyzed by succinate dehydrogenase, which
terconversion of metabolites arising from transamina-
is bound to the inner surface of the inner mitochondrial
tion and deamination of amino acids. It also provides
membrane. The enzyme contains FAD and iron-sulfur
the substrates for amino acid synthesis by transamina-
(Fe:S) protein and directly reduces ubiquinone in the
tion, as well as for gluconeogenesis and fatty acid syn-
respiratory chain. Fumarase (fumarate hydratase) cat-
thesis. Because it functions in both oxidative and syn-
alyzes the addition of water across the double bond of
thetic processes, it is amphibolic (Figure 16-4).
fumarate, yielding malate. Malate is converted to ox-
aloacetate by malate dehydrogenase, a reaction requir-
ing NAD+. Although the equilibrium of this reaction
The Citric Acid Cycle Takes Part in
strongly favors malate, the net flux is toward the direc-
Gluconeogenesis, Transamination,
tion of oxaloacetate because of the continual removal of
& Deamination
oxaloacetate (either to form citrate, as a substrate for
gluconeogenesis, or to undergo transamination to as-
All the intermediates of the cycle are potentially gluco-
partate) and also because of the continual reoxidation
genic, since they can give rise to oxaloacetate and thus
of NADH.
net production of glucose (in the liver and kidney, the
organs that carry out gluconeogenesis; see Chapter 19).
The key enzyme that catalyzes net transfer out of the
cycle into gluconeogenesis is phosphoenolpyruvate
TWELVE ATP ARE FORMED PER TURN
carboxykinase, which decarboxylates oxaloacetate to
OF THE CITRIC ACID CYCLE
phosphoenolpyruvate, with GTP acting as the donor
As a result of oxidations catalyzed by the dehydrogen-
phosphate (Figure 16-4).
ases of the citric acid cycle, three molecules of NADH
Net transfer into the cycle occurs as a result of sev-
and one of FADH2 are produced for each molecule of
eral different reactions. Among the most important of
acetyl-CoA catabolized in one turn of the cycle. These
such anaplerotic reactions is the formation of oxaloac-
reducing equivalents are transferred to the respiratory
etate by the carboxylation of pyruvate, catalyzed by
chain (Figure 16-2), where reoxidation of each NADH
pyruvate carboxylase. This reaction is important in
results in formation of
3 ATP and reoxidation of
maintaining an adequate concentration of oxaloacetate
FADH2 in formation of 2 ATP. In addition, 1 ATP
for the condensation reaction with acetyl-CoA. If acetyl-
(or GTP) is formed by substrate-level phosphorylation
CoA accumulates, it acts both as an allosteric activator
catalyzed by succinate thiokinase.
of pyruvate carboxylase and as an inhibitor of pyruvate
dehydrogenase, thereby ensuring a supply of oxaloac-
etate. Lactate, an important substrate for gluconeogene-
sis, enters the cycle via oxidation to pyruvate and then
VITAMINS PLAY KEY ROLES
carboxylation to oxaloacetate.
IN THE CITRIC ACID CYCLE
Aminotransferase
(transaminase) reactions form
Four of the B vitamins are essential in the citric acid
pyruvate from alanine, oxaloacetate from aspartate, and
cycle and therefore in energy-yielding metabolism: (1)
α-ketoglutarate from glutamate. Because these reac-
riboflavin, in the form of flavin adenine dinucleotide
tions are reversible, the cycle also serves as a source of
(FAD), a cofactor in the α-ketoglutarate dehydrogenase
carbon skeletons for the synthesis of these amino acids.
complex and in succinate dehydrogenase; (2) niacin, in
Other amino acids contribute to gluconeogenesis be-
the form of nicotinamide adenine dinucleotide (NAD),
cause their carbon skeletons give rise to citric acid cycle
134
/
CHAPTER 16
Hydroxyproline
Lactate
Serine
Cysteine
Threonine
Glycine
TRANSAMINASE
Tryptophan
Alanine
Pyruvate
Acetyl-CoA
PYRUVATE
CARBOXYLASE
PHOSPHOENOLPYRUVATE
CARBOXYKINASE
Phosphoenol-
Glucose
Oxaloacetate
pyruvate
TRANSAMINASE
Tyrosine
Fumarate
Phenylalanine
Aspartate
Citrate
Isoleucine
Methionine
Succinyl-CoA
Valine
CO2
α-Ketoglutarate
Propionate
CO2
TRANSAMINASE
Histidine
Proline
Glutamate
Glutamine
Arginine
Figure 16-4. Involvement of the citric acid cycle in transamination and gluconeo-
genesis. The bold arrows indicate the main pathway of gluconeogenesis.
intermediates. Alanine, cysteine, glycine, hydroxypro-
Pyruvate dehydrogenase is a mitochondrial enzyme,
line, serine, threonine, and tryptophan yield pyruvate;
and fatty acid synthesis is a cytosolic pathway, but the
arginine, histidine, glutamine, and proline yield α-ke-
mitochondrial membrane is impermeable to acetyl-
toglutarate; isoleucine, methionine, and valine yield
CoA. Acetyl-CoA is made available in the cytosol from
succinyl-CoA; and tyrosine and phenylalanine yield fu-
citrate synthesized in the mitochondrion, transported
marate (Figure 16-4).
into the cytosol and cleaved in a reaction catalyzed by
In ruminants, whose main metabolic fuel is short-
ATP-citrate lyase.
chain fatty acids formed by bacterial fermentation, the
conversion of propionate, the major glucogenic product
of rumen fermentation, to succinyl-CoA via the
Regulation of the Citric Acid Cycle
methylmalonyl-CoA pathway (Figure 19-2) is espe-
Depends Primarily on a Supply
cially important.
of Oxidized Cofactors
In most tissues, where the primary role of the citric acid
The Citric Acid Cycle Takes Part
cycle is in energy-yielding metabolism, respiratory
in Fatty Acid Synthesis
control via the respiratory chain and oxidative phos-
(Figure 16-5)
phorylation regulates citric acid cycle activity (Chap-
Acetyl-CoA, formed from pyruvate by the action of
ter 14). Thus, activity is immediately dependent on the
pyruvate dehydrogenase, is the major building block for
supply of NAD+, which in turn, because of the tight
long-chain fatty acid synthesis in nonruminants. (In ru-
coupling between oxidation and phosphorylation, is de-
minants, acetyl-CoA is derived directly from acetate.)
pendent on the availability of ADP and hence, ulti-
THE CITRIC ACID CYCLE: THE CATABOLISM OF ACETYL-CoA
/
135
Fatty
regulated in the same way as is pyruvate dehydrogenase
Pyruvate
Glucose
acids
(Figure 17-6). Succinate dehydrogenase is inhibited by
oxaloacetate, and the availability of oxaloacetate, as
controlled by malate dehydrogenase, depends on the
[NADH]/[NAD+] ratio. Since the Km for oxaloacetate
PYRUVATE
of citrate synthase is of the same order of magnitude as
DEHYDROGENASE
Acetyl-CoA
the intramitochondrial concentration, it is likely that
Acetyl-CoA
the concentration of oxaloacetate controls the rate of
citrate formation. Which of these mechanisms are im-
portant in vivo has still to be resolved.
Citric
ATP-CITRATE
acid
LYASE
cycle
SUMMARY
Oxaloacetate
Citrate
Citrate
• The citric acid cycle is the final pathway for the oxi-
dation of carbohydrate, lipid, and protein whose
common end-metabolite, acetyl-CoA, reacts with ox-
aloacetate to form citrate. By a series of dehydrogena-
tions and decarboxylations, citrate is degraded,
releasing reduced coenzymes and 2CO2 and regener-
CO2
CO2
ating oxaloacetate.
MITOCHONDRIAL
• The reduced coenzymes are oxidized by the respira-
MEMBRANE
tory chain linked to formation of ATP. Thus, the
cycle is the major route for the generation of ATP
Figure 16-5. Participation of the citric acid cycle in
and is located in the matrix of mitochondria adjacent
fatty acid synthesis from glucose. See also Figure 21-5.
to the enzymes of the respiratory chain and oxidative
phosphorylation.
• The citric acid cycle is amphibolic, since in addition
mately, on the rate of utilization of ATP in chemical
to oxidation it is important in the provision of car-
and physical work. In addition, individual enzymes of
bon skeletons for gluconeogenesis, fatty acid synthe-
the cycle are regulated. The most likely sites for regula-
sis, and interconversion of amino acids.
tion are the nonequilibrium reactions catalyzed by
pyruvate dehydrogenase, citrate synthase, isocitrate de-
REFERENCES
hydrogenase, and α-ketoglutarate dehydrogenase. The
dehydrogenases are activated by Ca2+, which increases
Baldwin JE, Krebs HA: The evolution of metabolic cycles. Nature
1981;291:381.
in concentration during muscular contraction and se-
cretion, when there is increased energy demand. In a
Goodwin TW (editor): The Metabolic Roles of Citrate. Academic
Press, 1968.
tissue such as brain, which is largely dependent on car-
Greville GD: Vol 1, p 297, in: Carbohydrate Metabolism and Its
bohydrate to supply acetyl-CoA, control of the citric
Disorders. Dickens F, Randle PJ, Whelan WJ (editors). Acad-
acid cycle may occur at pyruvate dehydrogenase. Sev-
emic Press, 1968.
eral enzymes are responsive to the energy status, as
Kay J, Weitzman PDJ (editors): Krebs’ Citric Acid Cycle—Half a
shown by the [ATP]/[ADP] and [NADH]/[NAD+] ra-
Century and Still Turning. Biochemical Society, London,
tios. Thus, there is allosteric inhibition of citrate syn-
1987.
thase by ATP and long-chain fatty acyl-CoA. Allosteric
Srere PA: The enzymology of the formation and breakdown of cit-
activation of mitochondrial NAD-dependent isocitrate
rate. Adv Enzymol 1975;43:57.
dehydrogenase by ADP is counteracted by ATP and
Tyler DD: The Mitochondrion in Health and Disease. VCH Pub-
NADH. The α-ketoglutarate dehydrogenase complex is
lishers, 1992.
Glycolysis & the Oxidation
17
of Pyruvate
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
GLYCOLYSIS CAN FUNCTION UNDER
ANAEROBIC CONDITIONS
Most tissues have at least some requirement for glucose.
In brain, the requirement is substantial. Glycolysis, the
When a muscle contracts in an anaerobic medium, ie,
major pathway for glucose metabolism, occurs in the
one from which oxygen is excluded, glycogen disap-
cytosol of all cells. It is unique in that it can function ei-
pears and lactate appears as the principal end product.
ther aerobically or anaerobically. Erythrocytes, which
When oxygen is admitted, aerobic recovery takes place
lack mitochondria, are completely reliant on glucose as
and lactate disappears. However, if contraction occurs
their metabolic fuel and metabolize it by anaerobic gly-
under aerobic conditions, lactate does not accumulate
colysis. However, to oxidize glucose beyond pyruvate
and pyruvate is the major end product of glycolysis.
(the end product of glycolysis) requires both oxygen
Pyruvate is oxidized further to CO2 and water (Figure
and mitochondrial enzyme systems such as the pyruvate
17-1). When oxygen is in short supply, mitochondrial
dehydrogenase complex, the citric acid cycle, and the
reoxidation of NADH formed from NAD+ during gly-
respiratory chain.
colysis is impaired, and NADH is reoxidized by reduc-
Glycolysis is both the principal route for glucose
ing pyruvate to lactate, so permitting glycolysis to pro-
metabolism and the main pathway for the metabolism
ceed (Figure 17-1). While glycolysis can occur under
of fructose, galactose, and other carbohydrates derived
anaerobic conditions, this has a price, for it limits the
from the diet. The ability of glycolysis to provide ATP
amount of ATP formed per mole of glucose oxidized,
in the absence of oxygen is especially important because
so that much more glucose must be metabolized under
it allows skeletal muscle to perform at very high levels
anaerobic than under aerobic conditions.
when oxygen supply is insufficient and because it allows
tissues to survive anoxic episodes. However, heart mus-
THE REACTIONS OF GLYCOLYSIS
cle, which is adapted for aerobic performance, has rela-
CONSTITUTE THE MAIN PATHWAY
tively low glycolytic activity and poor survival under
conditions of ischemia. Diseases in which enzymes of
OF GLUCOSE UTILIZATION
glycolysis (eg, pyruvate kinase) are deficient are mainly
The overall equation for glycolysis from glucose to lac-
seen as hemolytic anemias or, if the defect affects
tate is as follows:
skeletal muscle (eg, phosphofructokinase), as fatigue.
In fast-growing cancer cells, glycolysis proceeds at a
Glucose+2ADP+2P
→2
L
(+)−Lactate+2ATP+2H O
i
2
higher rate than is required by the citric acid cycle,
forming large amounts of pyruvate, which is reduced to
All of the enzymes of glycolysis (Figure 17-2) are
lactate and exported. This produces a relatively acidic
found in the cytosol. Glucose enters glycolysis by phos-
local environment in the tumor which may have impli-
phorylation to glucose 6-phosphate, catalyzed by hexo-
cations for cancer therapy. The lactate is used for gluco-
kinase, using ATP as the phosphate donor. Under
neogenesis in the liver, an energy-expensive process re-
physiologic conditions, the phosphorylation of glucose
sponsible for much of the hypermetabolism seen in
to glucose 6-phosphate can be regarded as irreversible.
cancer cachexia. Lactic acidosis results from several
Hexokinase is inhibited allosterically by its product,
causes, including impaired activity of pyruvate dehy-
glucose 6-phosphate. In tissues other than the liver and
drogenase.
pancreatic B islet cells, the availability of glucose for
136
GLYCOLYSIS & THE OXIDATION OF PYRUVATE
/
137
Glucose
Glycogen
This reaction is followed by another phosphorylation
C6
(C6 )n
with ATP catalyzed by the enzyme phosphofructoki-
nase (phosphofructokinase-1), forming fructose 1,6-
bisphosphate. The phosphofructokinase reaction may
be considered to be functionally irreversible under
physiologic conditions; it is both inducible and subject
to allosteric regulation and has a major role in regulat-
Hexose phosphates
ing the rate of glycolysis. Fructose 1,6-bisphosphate is
C6
cleaved by aldolase (fructose 1,6-bisphosphate aldolase)
into two triose phosphates, glyceraldehyde 3-phosphate
and dihydroxyacetone phosphate. Glyceraldehyde
3-phosphate and dihydroxyacetone phosphate are inter-
converted by the enzyme phosphotriose isomerase.
Glycolysis continues with the oxidation of glycer-
Triose phosphate
Triose phosphate
aldehyde 3-phosphate to 1,3-bisphosphoglycerate. The
C3
C3
enzyme catalyzing this oxidation, glyceraldehyde
NAD+
H2O
3-phosphate dehydrogenase, is NAD-dependent.
Structurally, it consists of four identical polypeptides
(monomers) forming a tetramer.
SH groups are
O2
NADH
1/2O2
present on each polypeptide, derived from cysteine
+ H+
residues within the polypeptide chain. One of the
CO
2
Pyruvate
Lactate
SH groups at the active site of the enzyme (Figure
+
C3
C3
H2O
17-3) combines with the substrate forming a thiohemi-
acetal that is oxidized to a thiol ester; the hydrogens re-
moved in this oxidation are transferred to NAD+. The
Figure 17-1. Summary of glycolysis. − , blocked by
thiol ester then undergoes phosphorolysis; inorganic
anaerobic conditions or by absence of mitochondria
phosphate (Pi) is added, forming 1,3-bisphosphoglycer-
containing key respiratory enzymes, eg, as in erythro-
ate, and the SH group is reconstituted.
cytes.
In the next reaction, catalyzed by phosphoglycerate
kinase, phosphate is transferred from 1,3-bisphospho-
glycerate onto ADP, forming ATP
(substrate-level
glycolysis (or glycogen synthesis in muscle and lipogen-
phosphorylation) and 3-phosphoglycerate. Since two
esis in adipose tissue) is controlled by transport into the
molecules of triose phosphate are formed per molecule
cell, which in turn is regulated by insulin. Hexokinase
of glucose, two molecules of ATP are generated at this
has a high affinity (low Km) for its substrate, glucose,
stage per molecule of glucose undergoing glycolysis.
and in the liver and pancreatic B islet cells is saturated
The toxicity of arsenic is due to competition of arsenate
under all normal conditions and so acts at a constant
with inorganic phosphate (Pi) in the above reactions to
rate to provide glucose 6-phosphate to meet the cell’s
give
1-arseno-3-phosphoglycerate, which hydrolyzes
need. Liver and pancreatic B islet cells also contain an
spontaneously to give
3-phosphoglycerate plus heat,
isoenzyme of hexokinase, glucokinase, which has a Km
without generating ATP. 3-Phosphoglycerate is isomer-
very much higher than the normal intracellular concen-
ized to 2-phosphoglycerate by phosphoglycerate mu-
tration of glucose. The function of glucokinase in the
tase. It is likely that 2,3-bisphosphoglycerate (diphos-
liver is to remove glucose from the blood following a
phoglycerate; DPG) is an intermediate in this reaction.
meal, providing glucose 6-phosphate in excess of re-
The subsequent step is catalyzed by enolase and in-
quirements for glycolysis, which will be used for glyco-
volves a dehydration, forming phosphoenolpyruvate.
gen synthesis and lipogenesis. In the pancreas, the
Enolase is inhibited by fluoride. To prevent glycolysis
glucose 6-phosphate formed by glucokinase signals in-
in the estimation of glucose, blood is collected in
creased glucose availability and leads to the secretion of
tubes containing fluoride. The enzyme is also depen-
insulin.
dent on the presence of either Mg2+ or Mn2+. The
Glucose 6-phosphate is an important compound at
phosphate of phosphoenolpyruvate is transferred to
the junction of several metabolic pathways (glycolysis,
ADP by pyruvate kinase to generate, at this stage,
gluconeogenesis, the pentose phosphate pathway, gly-
two molecules of ATP per molecule of glucose oxi-
cogenesis, and glycogenolysis). In glycolysis, it is con-
dized. The product of the enzyme-catalyzed reaction,
verted to fructose
6-phosphate by phosphohexose-
enolpyruvate, undergoes spontaneous
(nonenzymic)
isomerase, which involves an aldose-ketose isomerization.
isomerization to pyruvate and so is not available to
Glycogen
Glucose 1-phosphate
HEXOKINASE
CH2OH
CH2
O P
GLUCOKINASE
CH2
O P
O
O
PHOSPHOHEXOSE
O
H
H
H
H
ISOMERASE
CH2OH
H
Mg2+
H
OH
H
OH
H
H
HO
HO
OH
HO
OH
H
OH
H OH
H OH
OH
H
ATP
ADP
α-D-Glucose
α-D-Glucose 6-phosphate
D-Fructose 6-phosphate
ATP
2+
Mg
PHOSPHOFRUCTO-
ADP
KINASE
CH2
O P
O
CH2
O P
D-Fructose 1,6-bisphosphate
H
HO
H
OH
ALDOLASE
Iodoacetate
HO
H
CH2
O P
GLYCERALDEHYDE-3-PHOSPHATE
PHOSPHOGLYCERATE
O
C O
DEHYDROGENASE
KINASE
COO
-
C
O
P
H
C O
CH2OH
Mg2+
P
i
Dihydroxyacetone phosphate
H
C
OH
H
C
OH
H
C
OH
CH2
O P
CH2
O P
CH2
O P
PHOSPHOTRIOSE
ATP
ADP
NADH NAD+
ISOMERASE
3-Phosphoglycerate
1,3-Bisphosphoglycerate
+ H+
Glyceraldehyde
3-phosphate
1/2O
2
PHOSPHOGLYCERATE MUTASE
Mitochondrial
respiratory chain
H
2O
COO-
3ADP
3ATP
H
C
O
P
2-Phosphoglycerate
+ P
i
CH2OH
Anaerobiosis
Fluoride
Mg2+
H2O
ENOLASE
COO-
Phosphoenolpyruvate
C
O P
Oxidation
in citric
CH2
acid cycle
ADP
+
PYRUVATE
Mg2
KINASE
NADH + H+
NAD+
ATP
-
-
COO
COO
COO-
Spontaneous
C OH
C O
HO
C
H
LACTATE
CH2
CH3
CH3
DEHYDROGENASE
(Enol)
(Keto)
L(+)-Lactate
Pyruvate
Pyruvate
Figure 17-2. The pathway of glycolysis. ( P , PO32−; Pi, HOPO32−;
− , inhibition.) At asterisk: Carbon
atoms 1-3 of fructose bisphosphate form dihydroxyacetone phosphate, whereas carbons 4-6 form
glyceraldehyde 3-phosphate. The term “bis-,” as in bisphosphate, indicates that the phosphate groups
are separated, whereas diphosphate, as in adenosine diphosphate, indicates that they are joined.
138
GLYCOLYSIS & THE OXIDATION OF PYRUVATE
/
139
S Enz
H C O
H
C
OH
H C OH
NAD+
H
C
OH
CH2
O
P
CH2
O
P
Glyceraldehyde 3-phosphate
Enzyme-substrate complex
HS Enz
NAD+
Bound
coenzyme
Substrate
oxidation
O
P
by bound
NAD+
C O
H
C
OH
P
i
CH2
O
P
1,3-Bisphosphoglycerate
S Enz
S
Enz
C O
C O
+
NADH + H
NAD+
H
C
OH
H
C
OH
NADH + H+
NAD+
CH2
O
P
CH2
O
P
Energy-rich intermediate
Figure 17-3. Mechanism of oxidation of glyceraldehyde 3-phosphate. (Enz, glycer-
aldehyde-3-phosphate dehydrogenase.) The enzyme is inhibited by the SH poison
iodoacetate, which is thus able to inhibit glycolysis. The NADH produced on the enzyme
is not as firmly bound to the enzyme as is NAD+. Consequently, NADH is easily displaced
by another molecule of NAD+.
undergo the reverse reaction. The pyruvate kinase re-
up into mitochondria for oxidation via one of the two
action is thus also irreversible under physiologic con-
shuttles described in Chapter 12.
ditions.
The redox state of the tissue now determines which
of two pathways is followed. Under anaerobic condi-
Tissues That Function Under Hypoxic
tions, the reoxidation of NADH through the respira-
Circumstances Tend to Produce Lactate
tory chain to oxygen is prevented. Pyruvate is reduced
(Figure 17-2)
by the NADH to lactate, the reaction being catalyzed
by lactate dehydrogenase. Several tissue-specific isoen-
This is true of skeletal muscle, particularly the white
zymes of this enzyme have been described and have
fibers, where the rate of work output—and therefore
clinical significance
(Chapter 7). The reoxidation of
the need for ATP formation—may exceed the rate at
NADH via lactate formation allows glycolysis to pro-
which oxygen can be taken up and utilized. Glycolysis
ceed in the absence of oxygen by regenerating sufficient
in erythrocytes, even under aerobic conditions, always
NAD+ for another cycle of the reaction catalyzed by
terminates in lactate, because the subsequent reactions
glyceraldehyde-3-phosphate dehydrogenase. Under aer-
of pyruvate are mitochondrial, and erythrocytes lack
obic conditions, pyruvate is taken up into mitochon-
mitochondria. Other tissues that normally derive much
dria and after conversion to acetyl-CoA is oxidized to
of their energy from glycolysis and produce lactate in-
CO2 by the citric acid cycle. The reducing equivalents
clude brain, gastrointestinal tract, renal medulla, retina,
from the NADH + H+ formed in glycolysis are taken
and skin. The liver, kidneys, and heart usually take up
140
/
CHAPTER 17
lactate and oxidize it but will produce it under hypoxic
H C O
Glucose
conditions.
H C OH
CH2
O
P
Glycolysis Is Regulated at Three Steps
Involving Nonequilibrium Reactions
Glyceraldehyde 3-phosphate
Pi
NAD+
Although most of the reactions of glycolysis are re-
versible, three are markedly exergonic and must there-
GLYCERALDEHYDE-3-PHOSPHATE
fore be considered physiologically irreversible. These re-
DEHYDROGENASE
actions, catalyzed by hexokinase
(and glucokinase),
NADH + H+
phosphofructokinase, and pyruvate kinase, are the
O
major sites of regulation of glycolysis. Cells that are ca-
pable of reversing the glycolytic pathway (gluconeoge-
C O
P
nesis) have different enzymes that catalyze reactions
BISPHOSPHOGLYCERATE
H
C
OH
MUTASE
which effectively reverse these irreversible reactions.
The importance of these steps in the regulation of gly-
CH2
O
P
colysis and gluconeogenesis is discussed in Chapter 19.
1,3-Bisphosphoglycerate
In Erythrocytes, the First Site in Glycolysis
ADP
COO-
for ATP Generation May Be Bypassed
PHOSPHOGLYCERATE
H
C
O
P
KINASE
In the erythrocytes of many mammals, the reaction cat-
CH2
O
P
alyzed by phosphoglycerate kinase may be bypassed
ATP
by a process that effectively dissipates as heat the free
2,3-Bisphosphoglycerate
energy associated with the high-energy phosphate of
COO-
1,3-bisphosphoglycerate
(Figure
17-4). Bisphospho-
glycerate mutase catalyzes the conversion of 1,3-bis-
H
C
OH
P
i
phosphoglycerate to 2,3-bisphosphoglycerate, which is
2,3-BISPHOSPHOGLYCERATE
CH2
O
P
PHOSPHATASE
converted to 3-phosphoglycerate by 2,3-bisphospho-
glycerate phosphatase (and possibly also phosphoglyc-
3-Phosphoglycerate
erate mutase). This alternative pathway involves no net
Pyruvate
yield of ATP from glycolysis. However, it does serve to
Figure 17-4.
2,3-Bisphosphoglycerate pathway in
provide 2,3-bisphosphoglycerate, which binds to hemo-
erythrocytes.
globin, decreasing its affinity for oxygen and so making
oxygen more readily available to tissues (see Chapter 6).
in thiamin deficiency glucose metabolism is impaired
THE OXIDATION OF PYRUVATE TO
and there is significant (and potentially life-threatening)
ACETYL-CoA IS THE IRREVERSIBLE
lactic and pyruvic acidosis. Acetyl lipoamide reacts with
ROUTE FROM GLYCOLYSIS TO THE
coenzyme A to form acetyl-CoA and reduced lipoamide.
CITRIC ACID CYCLE
The cycle of reaction is completed when the reduced
lipoamide is reoxidized by a flavoprotein, dihydrolipoyl
Pyruvate, formed in the cytosol, is transported into the
dehydrogenase, containing FAD. Finally, the reduced
mitochondrion by a proton symporter (Figure 12-10).
flavoprotein is oxidized by NAD+, which in turn trans-
Inside the mitochondrion, pyruvate is oxidatively decar-
fers reducing equivalents to the respiratory chain.
boxylated to acetyl-CoA by a multienzyme complex that
is associated with the inner mitochondrial membrane.
+
+
Pyruvate + NAD
+ CoA → Acetyl−CoA + NADH + H
+ CO
This pyruvate dehydrogenase complex is analogous to
2
the α-ketoglutarate dehydrogenase complex of the citric
acid cycle (Figure 16-3). Pyruvate is decarboxylated by
The pyruvate dehydrogenase complex consists of a
the pyruvate dehydrogenase component of the enzyme
number of polypeptide chains of each of the three com-
complex to a hydroxyethyl derivative of the thiazole ring
ponent enzymes, all organized in a regular spatial con-
of enzyme-bound thiamin diphosphate, which in turn
figuration. Movement of the individual enzymes ap-
reacts with oxidized lipoamide, the prosthetic group of
pears to be restricted, and the metabolic intermediates
dihydrolipoyl transacetylase, to form acetyl lipoamide
do not dissociate freely but remain bound to the en-
(Figure 17-5). Thiamin is vitamin B1 (Chapter 45), and
zymes. Such a complex of enzymes, in which the sub-
GLYCOLYSIS & THE OXIDATION OF PYRUVATE
/
141
O
CH3
C
COO- + H+
TDP
Pyruvate
Acetyl
lipoamide
CoA-SH
PYRUVATE
DEHYDROGENASE
CO2
TDP
H3C C OH
Hydroxyethyl
H
C
H
H2
H2C
C
N
DIHYDROLIPOYL
Oxidized lipoamide
C
TRANSACETYLASE
S
S
O
Lipoic acid
Lysine
side chain
NAD+
FADH2
DIHYDROLIPOYL
DEHYDROGENASE
CH3
CO S CoA
Acetyl-CoA
Dihydrolipoamide
FAD
NADH + H+
Figure 17-5. Oxidative decarboxylation of pyruvate by the pyruvate dehydrogenase complex. Lipoic acid is
joined by an amide link to a lysine residue of the transacetylase component of the enzyme complex. It forms a long
flexible arm, allowing the lipoic acid prosthetic group to rotate sequentially between the active sites of each of the
enzymes of the complex. (NAD+, nicotinamide adenine dinucleotide; FAD, flavin adenine dinucleotide; TDP, thiamin
diphosphate.)
strates are handed on from one enzyme to the next, in-
lated by phosphorylation by a kinase of three serine
creases the reaction rate and eliminates side reactions,
residues on the pyruvate dehydrogenase component of
increasing overall efficiency.
the multienzyme complex, resulting in decreased activ-
ity, and by dephosphorylation by a phosphatase that
Pyruvate Dehydrogenase Is Regulated
causes an increase in activity. The kinase is activated by
increases in the
[ATP]/[ADP],
[acetyl-CoA]/[CoA],
by End-Product Inhibition
and [NADH]/[NAD+] ratios. Thus, pyruvate dehydro-
& Covalent Modification
genase—and therefore glycolysis—is inhibited not only
Pyruvate dehydrogenase is inhibited by its products,
by a high-energy potential but also when fatty acids are
acetyl-CoA and NADH (Figure 17-6). It is also regu-
being oxidized. Thus, in starvation, when free fatty acid
142
/
CHAPTER 17
[ Acetyl-CoA ]
[ NADH ]
[ ATP ]
[ CoA ]
[ NAD+ ]
[ ADP ]
+
+
+
-
Dichloroacetate
Acetyl-CoA
-
-
Ca2+
PDH KINASE
Pyruvate
NADH + H+
CO2
Mg2+
ATP
ADP
–
PDH
-
PDH-a
PDH-b
(Active DEPHOSPHO-ENZYME)
(Inactive PHOSPHO-ENZYME)
P
NAD+
CoA
Pi
H2O
Pyruvate
PDH PHOSPHATASE
+
A
B
+
Mg2+, Ca2+
Insulin
(in adipose tissue)
Figure 17-6. Regulation of pyruvate dehydrogenase (PDH). Arrows with wavy shafts indicate allosteric ef-
fects. A: Regulation by end-product inhibition. B: Regulation by interconversion of active and inactive forms.
concentrations increase, there is a decrease in the pro-
ATP synthase reaction has been calculated as approxi-
portion of the enzyme in the active form, leading to a
mately 51.6 kJ. It follows that the total energy captured
sparing of carbohydrate. In adipose tissue, where glu-
in ATP per mole of glucose oxidized is 1961 kJ, or ap-
cose provides acetyl CoA for lipogenesis, the enzyme is
proximately 68% of the energy of combustion. Most of
activated in response to insulin.
the ATP is formed by oxidative phosphorylation result-
ing from the reoxidation of reduced coenzymes by the
respiratory chain. The remainder is formed by substrate-
Oxidation of Glucose Yields Up to 38 Mol
level phosphorylation (Table 17-1).
of ATP Under Aerobic Conditions But Only
2 Mol When O2 Is Absent
CLINICAL ASPECTS
When 1 mol of glucose is combusted in a calorimeter
Inhibition of Pyruvate Metabolism
to CO2 and water, approximately 2870 kJ are liberated
Leads to Lactic Acidosis
as heat. When oxidation occurs in the tissues, approxi-
mately 38 mol of ATP are generated per molecule of
Arsenite and mercuric ions react with the SH groups
glucose oxidized to CO2 and water. In vivo, ∆G for the
of lipoic acid and inhibit pyruvate dehydrogenase, as
GLYCOLYSIS & THE OXIDATION OF PYRUVATE
/
143
Table 17-1. Generation of high-energy phosphate in the catabolism of glucose.
Number of ~ P
Formed per
Pathway
Reaction Catalyzed by
Method of ~ P Production
Mole of Glucose
Glycolysis
Glyceraldehyde-3-phosphate dehydrogenase Respiratory chain oxidation of 2 NADH
6*
Phosphoglycerate kinase
Phosphorylation at substrate level
2
Pyruvate kinase
Phosphorylation at substrate level
2
10
Allow for consumption of ATP by reactions catalyzed by hexokinase and phosphofructokinase
−2
Net 8
Pyruvate dehydrogenase
Respiratory chain oxidation of 2 NADH
6
Isocitrate dehydrogenase
Respiratory chain oxidation of 2 NADH
6
α-Ketoglutarate dehydrogenase
Respiratory chain oxidation of 2 NADH
6
Citric acid cycle Succinate thiokinase
Phosphorylation at substrate level
2
Succinate dehydrogenase
Respiratory chain oxidation of 2 FADH2
4
Malate dehydrogenase
Respiratory chain oxidation of 2 NADH
6
Net 30
Total per mole of glucose under aerobic conditions
38
Total per mole of glucose under anaerobic conditions
2
*It is assumed that NADH formed in glycolysis is transported into mitochondria via the malate shuttle (see Figure 12-13). If the glyc-
erophosphate shuttle is used, only 2 ~ P would be formed per mole of NADH, the total net production being 26 instead of 38. The
calculation ignores the small loss of ATP due to a transport of H+ into the mitochondrion with pyruvate and a similar transport of H+
in the operation of the malate shuttle, totaling about 1 mol of ATP. Note that there is a substantial benefit under anaerobic condi-
tions if glycogen is the starting point, since the net production of high-energy phosphate in glycolysis is increased from 2 to 3, as ATP
is no longer required by the hexokinase reaction.
does a dietary deficiency of thiamin, allowing pyru-
• It can function anaerobically by regenerating oxidized
vate to accumulate. Nutritionally deprived alcoholics
NAD+ (required in the glyceraldehyde-3-phosphate de-
are thiamin-deficient and may develop potentially fatal
hydrogenase reaction) by reducing pyruvate to lactate.
pyruvic and lactic acidosis. Patients with inherited
• Lactate is the end product of glycolysis under anaero-
pyruvate dehydrogenase deficiency, which can be due
bic conditions (eg, in exercising muscle) or when the
to defects in one or more of the components of the en-
metabolic machinery is absent for the further oxida-
zyme complex, also present with lactic acidosis, particu-
tion of pyruvate (eg, in erythrocytes).
larly after a glucose load. Because of its dependence on
• Glycolysis is regulated by three enzymes catalyzing
glucose as a fuel, brain is a prominent tissue where these
nonequilibrium reactions: hexokinase, phosphofruc-
metabolic defects manifest themselves in neurologic
tokinase, and pyruvate kinase.
disturbances.
• In erythrocytes, the first site in glycolysis for genera-
Inherited aldolase A deficiency and pyruvate kinase
tion of ATP may be bypassed, leading to the forma-
deficiency in erythrocytes cause hemolytic anemia.
tion of 2,3-bisphosphoglycerate, which is important
The exercise capacity of patients with muscle phos-
in decreasing the affinity of hemoglobin for O2.
phofructokinase deficiency is low, particularly on
high-carbohydrate diets. By providing an alternative
• Pyruvate is oxidized to acetyl-CoA by a multienzyme
complex, pyruvate dehydrogenase, that is dependent
lipid fuel, eg, during starvation, when blood free fatty
acids and ketone bodies are increased, work capacity is
on the vitamin cofactor thiamin diphosphate.
improved.
• Conditions that involve an inability to metabolize
pyruvate frequently lead to lactic acidosis.
SUMMARY
REFERENCES
• Glycolysis is the cytosolic pathway of all mammalian
cells for the metabolism of glucose (or glycogen) to
Behal RH et al: Regulation of the pyruvate dehydrogenase multien-
pyruvate and lactate.
zyme complex. Annu Rev Nutr 1993;13:497.
144
/
CHAPTER 17
Boiteux A, Hess B: Design of glycolysis. Phil Trans R Soc London
Sols A: Multimodulation of enzyme activity. Curr Top Cell Reg
B 1981;293:5.
1981;19:77.
Fothergill-Gilmore LA: The evolution of the glycolytic pathway.
Srere PA: Complexes of sequential metabolic enzymes. Annu Rev
Trends Biochem Sci 1986;11:47.
Biochem 1987;56:89.
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
herited Disease, 8th ed. McGraw-Hill, 2001.
Metabolism of Glycogen
18
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
phatase catalyzes hydrolysis of pyrophosphate to 2 mol
of inorganic phosphate, shifting the equilibrium of the
Glycogen is the major storage carbohydrate in animals,
main reaction by removing one of its products.
corresponding to starch in plants; it is a branched poly-
Glycogen synthase catalyzes the formation of a gly-
mer of α-D-glucose. It occurs mainly in liver (up to 6%)
coside bond between C1 of the activated glucose of
and muscle, where it rarely exceeds 1%. However, be-
UDPGlc and C4 of a terminal glucose residue of glyco-
cause of its greater mass, muscle contains about three to
gen, liberating uridine diphosphate (UDP). A preexist-
four times as much glycogen as does liver (Table 18-1).
ing glycogen molecule, or “glycogen primer,” must be
Muscle glycogen is a readily available source of glu-
present to initiate this reaction. The glycogen primer
cose for glycolysis within the muscle itself. Liver glyco-
may in turn be formed on a primer known as glyco-
gen functions to store and export glucose to maintain
genin, which is a 37-kDa protein that is glycosylated
blood glucose between meals. After 12-18 hours of
on a specific tyrosine residue by UDPGlc. Further glu-
fasting, the liver glycogen is almost totally depleted.
cose residues are attached in the 1→4 position to make
Glycogen storage diseases are a group of inherited dis-
a short chain that is a substrate for glycogen synthase.
orders characterized by deficient mobilization of glyco-
In skeletal muscle, glycogenin remains attached in the
gen or deposition of abnormal forms of glycogen, lead-
center of the glycogen molecule
(Figure
13-15),
ing to muscular weakness or even death.
whereas in liver the number of glycogen molecules is
greater than the number of glycogenin molecules.
GLYCOGENESIS OCCURS MAINLY
IN MUSCLE & LIVER
Branching Involves Detachment
of Existing Glycogen Chains
The Pathway of Glycogen Biosynthesis
Involves a Special Nucleotide of Glucose
The addition of a glucose residue to a preexisting glyco-
(Figure 18-1)
gen chain, or
“primer,” occurs at the nonreducing,
outer end of the molecule so that the “branches” of the
As in glycolysis, glucose is phosphorylated to glucose
glycogen “tree” become elongated as successive 1→4
6-phosphate, catalyzed by hexokinase in muscle and
linkages are formed (Figure 18-3). When the chain has
glucokinase in liver. Glucose 6-phosphate is isomer-
been lengthened to at least 11 glucose residues, branch-
ized to glucose 1-phosphate by phosphoglucomutase.
ing enzyme transfers a part of the 1→4 chain (at least
The enzyme itself is phosphorylated, and the phospho-
six glucose residues) to a neighboring chain to form a
group takes part in a reversible reaction in which glu-
1→6 linkage, establishing a branch point. The
cose 1,6-bisphosphate is an intermediate. Next, glucose
branches grow by further additions of 1→4-glucosyl
1-phosphate reacts with uridine triphosphate (UTP) to
units and further branching.
form the active nucleotide uridine diphosphate glu-
cose (UDPGlc)* and pyrophosphate (Figure 18-2),
GLYCOGENOLYSIS IS NOT THE REVERSE
catalyzed by UDPGlc pyrophosphorylase. Pyrophos-
OF GLYCOGENESIS BUT IS A SEPARATE
* Other nucleoside diphosphate sugar compounds are known, eg,
PATHWAY (Figure 18-1)
UDPGal. In addition, the same sugar may be linked to different
Glycogen phosphorylase catalyzes the rate-limiting
nucleotides. For example, glucose may be linked to uridine (as
shown above) as well as to guanosine, thymidine, adenosine, or cy-
step in glycogenolysis by promoting the phosphorylytic
tidine nucleotides.
cleavage by inorganic phosphate (phosphorylysis; cf hy-
145
146
/
CHAPTER 18
Glycogen
(1→ 4 and 1→ 6 glucosyl units)x
BRANCHING ENZYME
Pi
(1→ 4 Glucosyl units)x
Insulin
UDP
GLYCOGEN
GLYCOGEN
cAMP
SYNTHASE
PHOSPHORYLASE
Glycogen
primer
Glucagon
Epinephrine
GLUCAN
TRANSFERASE
Glycogenin
DEBRANCHING
ENZYME
Uridine
disphosphate
glucose (UDPGlc)
To uronic acid
Free glucose from
pathway
debranching
UDPGlc PYROPHOSPHORYLASE
enzyme
INORGANIC
PYROPHOSPHATASE
PPi
2
Pi
Uridine
UDP
triphosphate (UTP)
Glucose 1-phosphate
Mg2+
PHOSPHOGLUCOMUTASE
Glucose 6-phosphate
To glycolysis and pentose
phosphate pathway
H
2O
ADP
NUCLEOSIDE
DIPHOSPHO-
ATP
ADP
GLUCOSE-6-
KINASE
Mg2+
GLUCOKINASE
PHOSPHATASE
Pi
ATP
Glucose
Figure 18-1. Pathway of glycogenesis and of glycogenolysis in the liver. Two high-energy phosphates are
used in the incorporation of 1 mol of glucose into glycogen. + , stimulation; − , inhibition. Insulin decreases the
level of cAMP only after it has been raised by glucagon or epinephrine—ie, it antagonizes their action. Glucagon
is active in heart muscle but not in skeletal muscle. At asterisk: Glucan transferase and debranching enzyme ap-
pear to be two separate activities of the same enzyme.
Table 18-1. Storage of carbohydrate in
drolysis) of the 1→4 linkages of glycogen to yield glu-
postabsorptive normal adult humans (70 kg).
cose 1-phosphate. The terminal glucosyl residues from
the outermost chains of the glycogen molecule are re-
Liver glycogen
4.0%
=
72 g1
moved sequentially until approximately four glucose
Muscle glycogen
0.7%
=
245 g2
residues remain on either side of a 1→6 branch (Figure
Extracellular glucose
0.1%
=
10 g3
18-4). Another enzyme (
-[1v4]v -[1v4] glucan
327 g
transferase) transfers a trisaccharide unit from one
branch to the other, exposing the 1→6 branch point.
1Liver weight 1800 g.
2Muscle mass 35 kg.
Hydrolysis of the
1→6 linkages requires the de-
3Total volume 10 L.
branching enzyme. Further phosphorylase action can
METABOLISM OF GLYCOGEN
/
147
O
dephosphorylation of enzyme protein in response to
6CH2
OH
hormone action (Chapter 9).
HN
Uracil
H
O
Cyclic AMP (cAMP) (Figure 18-5) is formed from
H
H
1
ATP by adenylyl cyclase at the inner surface of cell
O
N
OH H
O
O
membranes and acts as an intracellular second messen-
HO
O P O P O CH2
ger in response to hormones such as epinephrine, nor-
H OH
O-
O-
O
epinephrine, and glucagon. cAMP is hydrolyzed by
phosphodiesterase, so terminating hormone action. In
liver, insulin increases the activity of phosphodiesterase.
H H
Ribose
H
H
HO OH
Phosphorylase Differs Between
Liver & Muscle
Glucose Diphosphate
Uridine
In liver, one of the serine hydroxyl groups of active
Figure 18-2. Uridine diphosphate glucose (UDPGlc).
phosphorylase a is phosphorylated. It is inactivated by
hydrolytic removal of the phosphate by protein phos-
phatase-1 to form phosphorylase b. Reactivation re-
then proceed. The combined action of phosphorylase
quires rephosphorylation catalyzed by phosphorylase
and these other enzymes leads to the complete break-
kinase.
down of glycogen. The reaction catalyzed by phospho-
Muscle phosphorylase is distinct from that of liver. It
glucomutase is reversible, so that glucose 6-phosphate
is a dimer, each monomer containing 1 mol of pyridoxal
can be formed from glucose 1-phosphate. In liver (and
phosphate (vitamin B6). It is present in two forms: phos-
kidney), but not in muscle, there is a specific enzyme,
phorylase a, which is phosphorylated and active in either
glucose-6-phosphatase, that hydrolyzes glucose
the presence or absence of 5′-AMP (its allosteric modi-
6-phosphate, yielding glucose that is exported, leading
fier); and phosphorylase b, which is dephosphorylated
to an increase in the blood glucose concentration.
and active only in the presence of 5′-AMP. This occurs
during exercise when the level of 5′-AMP rises, providing,
CYCLIC AMP INTEGRATES THE
by this mechanism, fuel for the muscle. Phosphorylase a is
the normal physiologically active form of the enzyme.
REGULATION OF GLYCOGENOLYSIS
& GLYCOGENESIS
cAMP Activates Muscle Phosphorylase
The principal enzymes controlling glycogen metabo-
lism—glycogen phosphorylase and glycogen synthase—
Phosphorylase in muscle is activated in response to epi-
are regulated by allosteric mechanisms and covalent
nephrine (Figure 18-6) acting via cAMP. Increasing
modifications due to reversible phosphorylation and
the concentration of cAMP activates cAMP-dependent
1→ 4- Glucosidic bond
Unlabeled glucose residue
1→ 6- Glucosidic bond
14C-labeled glucose residue
14C-Glucose
New 1→ 6- bond
added
GLYCOGEN
BRANCHING
SYNTHASE
ENZYME
Figure 18-3. The biosynthesis of glycogen. The mechanism of branching as revealed
by adding 14C-labeled glucose to the diet in the living animal and examining the liver
glycogen at further intervals.
148
/
CHAPTER 18
types of subunits—α, β, γ, and δ—in a structure repre-
sented as (αβγδ)4. The α and β subunits contain serine
residues that are phosphorylated by cAMP-dependent
protein kinase. The δ subunit binds four Ca2+ and is
identical to the Ca2+-binding protein calmodulin
(Chapter 43). The binding of Ca2+ activates the cat-
alytic site of the γ subunit while the molecule remains
in the dephosphorylated b configuration. However, the
phosphorylated a form is only fully activated in the
presence of Ca2+. A second molecule of calmodulin, or
TpC (the structurally similar Ca2+-binding protein in
muscle), can interact with phosphorylase kinase, caus-
ing further activation. Thus, activation of muscle con-
traction and glycogenolysis are carried out by the same
PHOSPHORYLASE
GLUCAN
DEBRANCHING
TRANSFERASE
ENZYME
Ca2+-binding protein, ensuring their synchronization.
Glucose residues joined by
Glycogenolysis in Liver Can
1 → 4- glucosidic bonds
Glucose residues joined by
Be cAMP-Independent
1 → 6- glucosidic bonds
In addition to the action of glucagon in causing forma-
tion of cAMP and activation of phosphorylase in liver,
Figure 18-4. Steps in glycogenolysis.
1-adrenergic receptors mediate stimulation of glyco-
genolysis by epinephrine and norepinephrine. This in-
volves a cAMP-independent mobilization of Ca2+
protein kinase, which catalyzes the phosphorylation by
from mitochondria into the cytosol, followed by the
ATP of inactive phosphorylase kinase b to active
stimulation of a Ca2+/calmodulin-sensitive phosphory-
phosphorylase kinase a, which in turn, by means of a
lase kinase. cAMP-independent glycogenolysis is also
further phosphorylation, activates phosphorylase b to
caused by vasopressin, oxytocin, and angiotensin II act-
phosphorylase a.
ing through calcium or the phosphatidylinositol bis-
phosphate pathway (Figure 43-7).
Ca2+ Synchronizes the Activation of
Phosphorylase With Muscle Contraction
Protein Phosphatase-1
Glycogenolysis increases in muscle several hundred-fold
Inactivates Phosphorylase
immediately after the onset of contraction. This in-
Both phosphorylase a and phosphorylase kinase a are
volves the rapid activation of phosphorylase by activa-
dephosphorylated and inactivated by protein phos-
tion of phosphorylase kinase by Ca2+, the same signal as
phatase-1. Protein phosphatase-1 is inhibited by a
that which initiates contraction in response to nerve
protein, inhibitor-1, which is active only after it has
stimulation. Muscle phosphorylase kinase has four
been phosphorylated by cAMP-dependent protein ki-
nase. Thus, cAMP controls both the activation and in-
activation of phosphorylase (Figure 18-6). Insulin re-
NH2
inforces this effect by inhibiting the activation of
N
N
phosphorylase b. It does this indirectly by increasing
uptake of glucose, leading to increased formation of
N
N
glucose 6-phosphate, which is an inhibitor of phosphor-
5′
ylase kinase.
O
CH2
Glycogen Synthase & Phosphorylase
O
-O
P O
Activity Are Reciprocally Regulated
(Figure 18-7)
H H
H
H
Like phosphorylase, glycogen synthase exists in either a
O
3′
OH
phosphorylated or nonphosphorylated state. However,
unlike phosphorylase, the active form is dephosphory-
Figure 18-5.
3′,5′-Adenylic acid (cyclic AMP; cAMP).
lated (glycogen synthase a) and may be inactivated to
150
/
CHAPTER 18
Epinephrine
β Receptor
+
Inactive
Active
adenylyl
adenylyl
cyclase
cyclase
+
PHOSPHODIESTERASE
ATP
cAMP
5′-AMP
PHOSPHORYLASE
KINASE
Ca2+
+
+
Inactive
Active
cAMP-DEPENDENT
cAMP-DEPENDENT
PROTEIN KINASE
PROTEIN KINASE
ATP
Glycogen(n+1)
Inhibitor-1
GSK
ADP
(inactive)
CALMODULIN-DEPENDENT
PROTEIN KINASE
ATP
GLYCOGEN SYNTHASE
GLYCOGEN SYNTHASE
+
b
a
(inactive)
Ca2+
(active)
+
Insulin
G6P
+
ADP
PROTEIN
PHOSPHATASE
Glycogen(n)
H2O
Pi
+ UDPG
Inhibitor-1-phosphate
PROTEIN
PHOSPHATASE-1
(active)
-
Figure 18-7. Control of glycogen synthase in muscle (n = number of glucose residues). The sequence of reac-
tions arranged in a cascade causes amplification at each step, allowing only nanomole quantities of hormone to
cause major changes in glycogen concentration. (GSK, glycogen synthase kinase-3, -4, and -5; G6P, glucose
6-phosphate.)
glycogen synthase b by phosphorylation on serine
REGULATION OF GLYCOGEN
residues by no fewer than six different protein kinases.
METABOLISM IS EFFECTED BY
Two of the protein kinases are Ca2+/calmodulin-
A BALANCE IN ACTIVITIES
dependent (one of these is phosphorylase kinase). An-
BETWEEN GLYCOGEN
other kinase is cAMP-dependent protein kinase, which
allows cAMP-mediated hormonal action to inhibit
SYNTHASE & PHOSPHORYLASE
glycogen synthesis synchronously with the activation of
(Figure 18-8)
glycogenolysis. Insulin also promotes glycogenesis in
muscle at the same time as inhibiting glycogenolysis by
Not only is phosphorylase activated by a rise in concen-
raising glucose
6-phosphate concentrations, which
tration of cAMP (via phosphorylase kinase), but glyco-
stimulates the dephosphorylation and activation of
gen synthase is at the same time converted to the
glycogen synthase. Dephosphorylation of glycogen syn-
inactive form; both effects are mediated via cAMP-
thase b is carried out by protein phosphatase-1, which
dependent protein kinase. Thus, inhibition of gly-
is under the control of cAMP-dependent protein ki-
cogenolysis enhances net glycogenesis, and inhibition of
nase.
glycogenesis enhances net glycogenolysis. Furthermore,
METABOLISM OF GLYCOGEN
/
151
Epinephrine
PHOSPHODIESTERASE
(liver, muscle)
Inhibitor-1
cAMP
5′-AMP
Inhibitor-1
Glucagon
phosphate
(liver)
GLYCOGEN
PHOSPHORYLASE
SYNTHASE b
KINASE b
cAMP-
PROTEIN
PROTEIN
DEPENDENT
PHOSPHATASE-1
PHOSPHATASE-1
PROTEIN KINASE
GLYCOGEN
PHOSPHORYLASE
SYNTHASE a
KINASE a
Glycogen
Glycogen
PHOSPHORYLASE
PHOSPHORYLASE
UDPGIc
cycle
a
b
Glucose 1-phosphate
PROTEIN
PHOSPHATASE-1
Glucose (liver)
Glucose Lactate (muscle)
Figure 18-8. Coordinated control of glycogenolysis and glycogenesis by cAMP-dependent protein ki-
nase. The reactions that lead to glycogenolysis as a result of an increase in cAMP concentrations are shown
with bold arrows, and those that are inhibited by activation of protein phosphatase-1 are shown as broken
arrows. The reverse occurs when cAMP concentrations decrease as a result of phosphodiesterase activity,
leading to glycogenesis.
the dephosphorylation of phosphorylase a, phosphory-
more, they allow insulin, via glucose 6-phosphate eleva-
lase kinase a, and glycogen synthase b is catalyzed by
tion, to have effects that act reciprocally to those of
a single enzyme of wide specificity—protein phos-
cAMP (Figures 18-6 and 18-7).
phatase-1. In turn, protein phosphatase-1 is inhibited
by cAMP-dependent protein kinase via inhibitor-1.
Thus, glycogenolysis can be terminated and glycogenesis
CLINICAL ASPECTS
can be stimulated synchronously, or vice versa, because
Glycogen Storage Diseases Are Inherited
both processes are keyed to the activity of cAMP-depen-
dent protein kinase. Both phosphorylase kinase and
“Glycogen storage disease” is a generic term to describe
glycogen synthase may be reversibly phosphorylated in
a group of inherited disorders characterized by deposi-
more than one site by separate kinases and phosphatases.
tion of an abnormal type or quantity of glycogen in the
These secondary phosphorylations modify the sensitivity
tissues. The principal glycogenoses are summarized in
of the primary sites to phosphorylation and dephos-
Table
18-2. Deficiencies of adenylyl kinase and
phorylation
(multisite phosphorylation). What is
cAMP-dependent protein kinase have also been re-
152
/
CHAPTER 18
Table 18-2. Glycogen storage diseases.
Glycogenosis
Name
Cause of Disorder
Characteristics
Type I
Von Gierke’s disease
Deficiency of glucose-6-phosphatase
Liver cells and renal tubule cells loaded
with glycogen. Hypoglycemia, lactic-
acidemia, ketosis, hyperlipemia.
Type II
Pompe’s disease
Deficiency of lysosomal α-1→4- and
Fatal, accumulation of glycogen in lyso-
1→6-glucosidase (acid maltase)
somes, heart failure.
Type III
Limit dextrinosis, Forbes’ or
Absence of debranching enzyme
Accumulation of a characteristic
Cori’s disease
branched polysaccharide.
Type IV
Amylopectinosis, Andersen’s
Absence of branching enzyme
Accumulation of a polysaccharide hav-
disease
ing few branch points. Death due to
cardiac or liver failure in first year of life.
Type V
Myophosphorylase deficiency,
Absence of muscle phosphorylase
Diminished exercise tolerance; muscles
McArdle’s syndrome
have abnormally high glycogen con-
tent (2.5-4.1%). Little or no lactate in
blood after exercise.
Type VI
Hers’ disease
Deficiency of liver phosphorylase
High glycogen content in liver, ten-
dency toward hypoglycemia.
Type VII
Tarui’s disease
Deficiency of phosphofructokinase
As for type V but also possibility of he-
in muscle and erythrocytes
molytic anemia.
Type VIII
Deficiency of liver phosphorylase
As for type VI.
kinase
ported. Some of the conditions described have bene-
REFERENCES
fited from liver transplantation.
Bollen M, Keppens S, Stalmans W: Specific features of glycogen
metabolism in the liver. Biochem J 1998;336:19.
SUMMARY
Cohen P: The role of protein phosphorylation in the hormonal
control of enzyme activity. Eur J Biochem 1985;151:439.
• Glycogen represents the principal storage form of
Ercan N, Gannon MC, Nuttall FQ: Incorporation of glycogenin
carbohydrate in the mammalian body, mainly in the
into a hepatic proteoglycogen after oral glucose administra-
liver and muscle.
tion. J Biol Chem 1994;269:22328.
• In the liver, its major function is to provide glucose
Geddes R: Glycogen: a metabolic viewpoint. Bioscience Rep
for extrahepatic tissues. In muscle, it serves mainly as
1986;6:415.
a ready source of metabolic fuel for use in muscle.
McGarry JD et al: From dietary glucose to liver glycogen: the full
• Glycogen is synthesized from glucose by the pathway
circle round. Annu Rev Nutr 1987;7:51.
of glycogenesis. It is broken down by a separate path-
Meléndez-Hevia E, Waddell TG, Shelton ED: Optimization of
molecular design in the evolution of metabolism: the glyco-
way known as glycogenolysis. Glycogenolysis leads to
gen molecule. Biochem J 1993;295:477.
glucose formation in liver and lactate formation in
Raz I, Katz A, Spencer MK: Epinephrine inhibits insulin-mediated
muscle owing to the respective presence or absence of
glycogenesis but enhances glycolysis in human skeletal mus-
glucose-6-phosphatase.
cle. Am J Physiol 1991;260:E430.
• Cyclic AMP integrates the regulation of glycogenoly-
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
sis and glycogenesis by promoting the simultaneous
herited Disease, 8th ed. McGraw-Hill, 2001.
activation of phosphorylase and inhibition of glyco-
Shulman GI, Landau BR: Pathways of glycogen repletion. Physiol
gen synthase. Insulin acts reciprocally by inhibiting
Rev 1992;72:1019.
glycogenolysis and stimulating glycogenesis.
Villar-Palasi C: On the mechanism of inactivation of muscle glyco-
gen phosphorylase by insulin. Biochim Biophys Acta 1994;
• Inherited deficiencies in specific enzymes of glycogen
1224:384.
metabolism in both liver and muscle are the causes of
glycogen storage diseases.
Gluconeogenesis & Control
19
of the Blood Glucose
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
vate
(Figure
45-17). A second enzyme, phospho-
enolpyruvate carboxykinase, catalyzes the decarboxy-
Gluconeogenesis is the term used to include all path-
lation and phosphorylation of oxaloacetate to phospho-
ways responsible for converting noncarbohydrate pre-
enolpyruvate using GTP (or ITP) as the phosphate
cursors to glucose or glycogen. The major substrates are
donor. Thus, reversal of the reaction catalyzed by pyru-
the glucogenic amino acids and lactate, glycerol, and
vate kinase in glycolysis involves two endergonic reac-
propionate. Liver and kidney are the major gluco-
tions.
neogenic tissues. Gluconeogenesis meets the needs of
In pigeon, chicken, and rabbit liver, phospho-
the body for glucose when carbohydrate is not available
enolpyruvate carboxykinase is a mitochondrial enzyme,
in sufficient amounts from the diet or from glycogen
and phosphoenolpyruvate is transported into the cy-
reserves. A supply of glucose is necessary especially for
tosol for gluconeogenesis. In the rat and the mouse, the
the nervous system and erythrocytes. Failure of gluco-
enzyme is cytosolic. Oxaloacetate does not cross the mi-
neogenesis is usually fatal. Hypoglycemia causes brain
tochondrial inner membrane; it is converted to malate,
dysfunction, which can lead to coma and death. Glu-
which is transported into the cytosol, and converted
cose is also important in maintaining the level of inter-
back to oxaloacetate by cytosolic malate dehydrogenase.
mediates of the citric acid cycle even when fatty acids
In humans, the guinea pig, and the cow, the enzyme is
are the main source of acetyl-CoA in the tissues. In ad-
equally distributed between mitochondria and cytosol.
dition, gluconeogenesis clears lactate produced by mus-
The main source of GTP for phosphoenolpyruvate
cle and erythrocytes and glycerol produced by adipose
carboxykinase inside the mitochondrion is the reaction
tissue. Propionate, the principal glucogenic fatty acid
of succinyl-CoA synthetase (Chapter 16). This provides
produced in the digestion of carbohydrates by rumi-
a link and limit between citric acid cycle activity and
nants, is a major substrate for gluconeogenesis in these
the extent of withdrawal of oxaloacetate for gluconeo-
species.
genesis.
GLUCONEOGENESIS INVOLVES
B. FRUCTOSE 1,6-BISPHOSPHATE
GLYCOLYSIS, THE CITRIC ACID CYCLE,
& FRUCTOSE 6-PHOSPHATE
& SOME SPECIAL REACTIONS
The conversion of fructose 1,6-bisphosphate to fructose
(Figure 19-1)
6-phosphate, to achieve a reversal of glycolysis, is cat-
alyzed by fructose-1,6-bisphosphatase. Its presence
Thermodynamic Barriers Prevent
determines whether or not a tissue is capable of synthe-
a Simple Reversal of Glycolysis
sizing glycogen not only from pyruvate but also from
Three nonequilibrium reactions catalyzed by hexoki-
triosephosphates. It is present in liver, kidney, and
nase, phosphofructokinase, and pyruvate kinase prevent
skeletal muscle but is probably absent from heart and
simple reversal of glycolysis for glucose synthesis
smooth muscle.
(Chapter 17). They are circumvented as follows:
A. PYRUVATE & PHOSPHOENOLPYRUVATE
C. GLUCOSE 6-PHOSPHATE & GLUCOSE
Mitochondrial pyruvate carboxylase catalyzes the car-
The conversion of glucose 6-phosphate to glucose is
boxylation of pyruvate to oxaloacetate, an ATP-requir-
catalyzed by glucose-6-phosphatase. It is present in
ing reaction in which the vitamin biotin is the co-
liver and kidney but absent from muscle and adipose
enzyme. Biotin binds CO2
from bicarbonate as
tissue, which, therefore, cannot export glucose into the
carboxybiotin prior to the addition of the CO2 to pyru-
bloodstream.
153
P
i
Glucose
ATP
GLUCOKINASE
GLUCOSE-6-PHOSPHATASE
HEXOKINASE
Glucose 6-
H2O
ADP
phosphate
Glycogen
AMP
AMP
P
i
Fructose 6-
ATP
phosphate
FRUCTOSE-1,6-
PHOSPHOFRUCTOKINASE
BISPHOSPHATASE
Fructose 1,6-
H2O
ADP
bisphosphate
Fructose
cAMP
2,6-bisphosphate
(glucagon)
Fructose
2,6-bisphosphate
Glyceraldehyde 3-phosphate
Dihydroxyacetone phosphate
NAD+
Pi
NADH + H+
+
GLYCEROL 3-PHOSPHATE
NADH + H
DEHYDROGENASE
cAMP
(glucagon)
1,3-Bisphosphoglycerate
NAD+
ADP
Glycerol 3-phosphate
ADP
ATP
GLYCEROL KINASE
3-Phosphoglycerate
ATP
Glycerol
2-Phosphoglycerate
cAMP
(glucagon)
Phosphoenolpyruvate
ADP
PYRUVATE KINASE
Alanine
GDP + CO
2
Fatty
ATP
acids
PHOSPHOENOLPYRUVATE
Pyruvate
Lactate
CARBOXYKINASE
Citrate
GTP
NADH + H+ NAD+
PYRUVATE
Oxaloacetate
DEHYDROGENASE
Pyruvate
Acetyl-CoA
NADH + H+
CO2 + ATP
Mg
2+
PYRUVATE CARBOXYLASE
NAD+
ADP + P
i
NADH + H+
Oxaloacetate
NAD+
Malate
Malate
Citrate
Citric acid cycle
α-Ketoglutarate
Fumarate
Succinyl-CoA
Propionate
Figure 19-1. Major pathways and regulation of gluconeogenesis and glycolysis in the liver. Entry
points of glucogenic amino acids after transamination are indicated by arrows extended from circles.
(See also Figure 16-4.) The key gluconeogenic enzymes are enclosed in double-bordered boxes. The
ATP required for gluconeogenesis is supplied by the oxidation of long-chain fatty acids. Propionate is
of quantitative importance only in ruminants. Arrows with wavy shafts signify allosteric effects; dash-
shafted arrows, covalent modification by reversible phosphorylation. High concentrations of alanine
act as a “gluconeogenic signal” by inhibiting glycolysis at the pyruvate kinase step.
154
GLUCONEOGENESIS & CONTROL OF THE BLOOD GLUCOSE
/
155
D. GLUCOSE 1-PHOSPHATE & GLYCOGEN
phosphate by NAD+ catalyzed by glycerol-3-phos-
The breakdown of glycogen to glucose 1-phosphate is
phate dehydrogenase.
catalyzed by phosphorylase. Glycogen synthesis in-
volves a different pathway via uridine diphosphate glu-
SINCE GLYCOLYSIS & GLUCONEOGENESIS
cose and glycogen synthase (Figure 18-1).
The relationships between gluconeogenesis and the
SHARE THE SAME PATHWAY BUT IN
glycolytic pathway are shown in Figure 19-1. After
OPPOSITE DIRECTIONS, THEY MUST
transamination or deamination, glucogenic amino acids
BE REGULATED RECIPROCALLY
yield either pyruvate or intermediates of the citric acid
Changes in the availability of substrates are responsible
cycle. Therefore, the reactions described above can ac-
for most changes in metabolism either directly or indi-
count for the conversion of both glucogenic amino
rectly acting via changes in hormone secretion. Three
acids and lactate to glucose or glycogen. Propionate is a
mechanisms are responsible for regulating the activity
major source of glucose in ruminants and enters gluco-
of enzymes in carbohydrate metabolism: (1) changes in
neogenesis via the citric acid cycle. Propionate is esteri-
the rate of enzyme synthesis, (2) covalent modification
fied with CoA, then propionyl-CoA, is carboxylated to
by reversible phosphorylation, and (3) allosteric effects.
D-methylmalonyl-CoA, catalyzed by propionyl-CoA
carboxylase, a biotin-dependent enzyme (Figure 19-2).
Methylmalonyl-CoA racemase catalyzes the conver-
Induction & Repression of Key Enzyme
sion of D-methylmalonyl-CoA to L-methylmalonyl-
Synthesis Requires Several Hours
CoA, which then undergoes isomerization to succinyl-
CoA catalyzed by methylmalonyl-CoA isomerase.
The changes in enzyme activity in the liver that occur
This enzyme requires vitamin B12 as a coenzyme, and
under various metabolic conditions are listed in Table
deficiency of this vitamin results in the excretion of
19-1. The enzymes involved catalyze nonequilibrium
methylmalonate (methylmalonic aciduria).
(physiologically irreversible) reactions. The effects are
C15 and C17 fatty acids are found particularly in the
generally reinforced because the activity of the enzymes
lipids of ruminants. Dietary odd-carbon fatty acids
catalyzing the changes in the opposite direction varies
upon oxidation yield propionate (Chapter 22), which is
reciprocally (Figure 19-1). The enzymes involved in
a substrate for gluconeogenesis in human liver.
the utilization of glucose (ie, those of glycolysis and li-
Glycerol is released from adipose tissue as a result of
pogenesis) all become more active when there is a su-
lipolysis, and only tissues such as liver and kidney that
perfluity of glucose, and under these conditions the en-
possess glycerol kinase, which catalyzes the conversion
zymes responsible for gluconeogenesis all have low
of glycerol to glycerol 3-phosphate, can utilize it. Glyc-
activity. The secretion of insulin, in response to in-
erol 3-phosphate may be oxidized to dihydroxyacetone
creased blood glucose, enhances the synthesis of the key
CoA SH
CO2 + H2O
ACYL-CoA
PROPIONYL-CoA
CH3
SYNTHETASE
CH3
CARBOXYLASE
CH3
CH2
CH2
H
C
COO-
Mg2+
Biotin
COO-
CO
S CoA
CO
S CoA
ATP
AMP + PP
i
ATP
ADP + P
i
Propionate
Propionyl-CoA
D-Methyl-
malonyl-CoA
METHYLMALONYL-CoA
RACEMASE
COO-
METHYLMALONYL-
CoA ISOMERASE
CH
3
CH2
Intermediates
-OOC
C H
of citric acid cycle
CH2
B12
coenzyme
CO S CoA
CO
S CoA
L-Methyl-
Succinyl-CoA
malonyl-CoA
Figure 19-2. Metabolism of propionate.
156
/
CHAPTER 19
Table 19-1. Regulatory and adaptive enzymes of the rat (mainly liver).
Activity In
Carbo-
Starva-
hydrate
tion and
Feeding
Diabetes
Inducer
Repressor
Activator
Inhibitor
Enzymes of glycogenesis, glycolysis, and pyruvate oxidation
Glycogen synthase
↑
↓
Insulin
Insulin
Glucagon (cAMP) phos-
system
Glucose 6-
phorylase, glycogen
phosphate1
Hexokinase
Glucose 6-phosphate1
Glucokinase
↑
↓
Insulin
Glucagon
(cAMP)
Phosphofructokinase-1
↑
↓
Insulin
Glucagon
AMP, fructose 6-
Citrate (fatty acids, ketone
(cAMP)
phosphate, Pi, fruc-
bodies),1 ATP,1 glucagon
tose 2,6-bisphos-
(cAMP)
phate1
Pyruvate kinase
↑
↓
Insulin, fructose
Glucagon
Fructose 1,6-
ATP, alanine, glucagon
(cAMP)
bisphosphate1, in-
(cAMP), epinephrine
sulin
Pyruvate dehydro-
↑
↓
CoA, NAD+, insu-
Acetyl-CoA, NADH, ATP
genase
lin,2 ADP, pyruvate
(fatty acids, ketone bodies)
Enzymes of gluconeogenesis
Pyruvate carboxylase
↓
↑
Glucocorticoids,
Insulin
Acetyl-CoA1
ADP1
glucagon, epi-
nephrine (cAMP)
Phosphoenolpyruvate
↓
↑
Glucocorticoids,
Insulin
Glucagon?
carboxykinase
glucagon, epi-
nephrine (cAMP)
Fructose-1,6-
↓
↑
Glucocorticoids,
Insulin
Glucagon (cAMP)
Fructose 1,6- bisphosphate,
bisphosphatase
glucagon, epi-
AMP, fructose 2,6-bisphos-
nephrine (cAMP)
phate1
Glucose-6-phosphatase
↓
↑
Glucocorticoids,
Insulin
glucagon, epi-
nephrine (cAMP)
Enzymes of the pentose phosphate pathway and lipogenesis
Glucose-6-phosphate
↑
↓
Insulin
dehydrogenase
6-Phosphogluconate
↑
↓
Insulin
dehydrogenase
“Malic enzyme”
↑
↓
Insulin
ATP-citrate lyase
↑
↓
Insulin
Acetyl-CoA carboxylase
↑
↓
Insulin?
Citrate,1 insulin
Long-chain acyl-CoA, cAMP,
glucagon
Fatty acid synthase
↑
↓
Insulin?
1Allosteric.
2In adipose tissue but not in liver.
GLUCONEOGENESIS & CONTROL OF THE BLOOD GLUCOSE
/
157
enzymes in glycolysis. Likewise, it antagonizes the effect
presence of adenylyl kinase in liver and many other
of the glucocorticoids and glucagon-stimulated cAMP,
tissues allows rapid equilibration of the reaction:
which induce synthesis of the key enzymes responsible
for gluconeogenesis.
ATP + AMP ↔ 2ADP
Both dehydrogenases of the pentose phosphate
pathway can be classified as adaptive enzymes, since
they increase in activity in the well-fed animal and
Thus, when ATP is used in energy-requiring processes
when insulin is given to a diabetic animal. Activity is
resulting in formation of ADP, [AMP] increases. As
low in diabetes or starvation.
“Malic enzyme” and
[ATP] may be 50 times [AMP] at equilibrium, a small
ATP-citrate lyase behave similarly, indicating that these
fractional decrease in [ATP] will cause a severalfold in-
two enzymes are involved in lipogenesis rather than
crease in [AMP]. Thus, a large change in [AMP] acts as
gluconeogenesis (Chapter 21).
a metabolic amplifier of a small change in [ATP]. This
mechanism allows the activity of phosphofructokinase-1
to be highly sensitive to even small changes in energy
Covalent Modification by Reversible
status of the cell and to control the quantity of carbohy-
Phosphorylation Is Rapid
drate undergoing glycolysis prior to its entry into the
citric acid cycle. The increase in [AMP] can also explain
Glucagon, and to a lesser extent epinephrine, hor-
why glycolysis is increased during hypoxia when [ATP]
mones that are responsive to decreases in blood glucose,
decreases. Simultaneously, AMP activates phosphory-
inhibit glycolysis and stimulate gluconeogenesis in the
lase, increasing glycogenolysis. The inhibition of phos-
liver by increasing the concentration of cAMP. This in
phofructokinase-1 by citrate and ATP is another expla-
turn activates cAMP-dependent protein kinase, leading
nation of the sparing action of fatty acid oxidation on
to the phosphorylation and inactivation of pyruvate
glucose oxidation and also of the Pasteur effect,
kinase. They also affect the concentration of fructose
whereby aerobic oxidation (via the citric acid cycle) in-
2,6-bisphosphate and therefore glycolysis and gluco-
hibits the anaerobic degradation of glucose. A conse-
neogenesis, as explained below.
quence of the inhibition of phosphofructokinase-1 is an
accumulation of glucose 6-phosphate that, in turn, in-
hibits further uptake of glucose in extrahepatic tissues
Allosteric Modification Is Instantaneous
by allosteric inhibition of hexokinase.
In gluconeogenesis, pyruvate carboxylase, which cata-
lyzes the synthesis of oxaloacetate from pyruvate, re-
quires acetyl-CoA as an allosteric activator. The pres-
Fructose 2,6-Bisphosphate Plays a Unique
ence of acetyl-CoA results in a change in the tertiary
Role in the Regulation of Glycolysis &
structure of the protein, lowering the Km value for bi-
Gluconeogenesis in Liver
carbonate. This means that as acetyl-CoA is formed
from pyruvate, it automatically ensures the provision of
The most potent positive allosteric effector of phospho-
oxaloacetate and, therefore, its further oxidation in the
fructokinase-1 and inhibitor of fructose-1,6-bisphos-
citric acid cycle. The activation of pyruvate carboxylase
phatase in liver is fructose
2,6-bisphosphate. It re-
and the reciprocal inhibition of pyruvate dehydrogen-
lieves inhibition of phosphofructokinase-1 by ATP and
ase by acetyl-CoA derived from the oxidation of fatty
increases affinity for fructose 6-phosphate. It inhibits
acids explains the action of fatty acid oxidation in spar-
fructose-1,6-bisphosphatase by increasing the Km for
ing the oxidation of pyruvate and in stimulating gluco-
fructose 1,6-bisphosphate. Its concentration is under
neogenesis. The reciprocal relationship between these
both substrate (allosteric) and hormonal control (cova-
two enzymes in both liver and kidney alters the meta-
lent modification) (Figure 19-3).
bolic fate of pyruvate as the tissue changes from carbo-
Fructose 2,6-bisphosphate is formed by phosphory-
hydrate oxidation, via glycolysis, to gluconeogenesis
lation of fructose
6-phosphate by phosphofructoki-
during transition from a fed to a starved state (Figure
nase-2. The same enzyme protein is also responsible for
19-1). A major role of fatty acid oxidation in promot-
its breakdown, since it has fructose-2,6-bisphos-
ing gluconeogenesis is to supply the requirement for
phatase activity. This bifunctional enzyme is under
ATP. Phosphofructokinase (phosphofructokinase-1)
the allosteric control of fructose
6-phosphate, which
occupies a key position in regulating glycolysis and is
stimulates the kinase and inhibits the phosphatase.
also subject to feedback control. It is inhibited by cit-
Hence, when glucose is abundant, the concentration of
rate and by ATP and is activated by 5′-AMP. 5′-AMP
fructose 2,6-bisphosphate increases, stimulating glycol-
acts as an indicator of the energy status of the cell. The
ysis by activating phosphofructokinase-1 and inhibiting
158
/
CHAPTER 19
Glycogen
Substrate (Futile) Cycles Allow Fine Tuning
Glucose
It will be apparent that the control points in glycolysis
and glycogen metabolism involve a cycle of phosphory-
Fructose 6-phosphate
lation and dephosphorylation catalyzed by: glucokinase
Glucagon
and glucose-6-phosphatase; phosphofructokinase-1 and
fructose-1,6-bisphosphatase; pyruvate kinase, pyruvate
cAMP
carboxylase, and phosphoenolypyruvate carboxykinase;
Pi
and glycogen synthase and phosphorylase. If these were
cAMP-DEPENDENT
allowed to cycle unchecked, they would amount to fu-
PROTEIN KINASE
tile cycles whose net result was hydrolysis of ATP. This
does not occur extensively due to the various control
ADP
ATP
mechanisms, which ensure that one reaction is inhib-
ited as the other is stimulated. However, there is a phys-
iologic advantage in allowing some cycling. The rate of
Active
Inactive
net glycolysis may increase several thousand-fold in re-
F-2,6-Pase
F-2,6-Pase
P
sponse to stimulation, and this is more readily achieved
Inactive
Active
PFK-2
PFK-2
by both increasing the activity of phosphofructokinase
and decreasing that of fructose bisphosphatase if both
are active, than by switching one enzyme “on” and the
H2O
Pi
other “off” completely. This “fine tuning” of metabolic
control occurs at the expense of some loss of ATP.
PROTEIN
ADP
PHOSPHATASE-2
Citrate
THE CONCENTRATION OF BLOOD
Fructose 2,6-bisphosphate
Pi
ATP
GLUCOSE IS REGULATED WITHIN
NARROW LIMITS
F-1,6-Pase
PFK-1
In the postabsorptive state, the concentration of blood
H2O
ADP
glucose in most mammals is maintained between 4.5
and 5.5 mmol/L. After the ingestion of a carbohydrate
meal, it may rise to 6.5-7.2 mmol/L, and in starvation,
Fructose 1,6-bisphosphate
it may fall to 3.3-3.9 mmol/L. A sudden decrease in
blood glucose will cause convulsions, as in insulin over-
Pyruvate
dose, owing to the immediate dependence of the brain
on a supply of glucose. However, much lower concen-
Figure 19-3. Control of glycolysis and gluconeoge-
trations can be tolerated, provided progressive adapta-
nesis in the liver by fructose 2,6-bisphosphate and the
tion is allowed. The blood glucose level in birds is con-
bifunctional enzyme PFK-2/F-2,6-Pase (6-phospho-
siderably higher
(14.0 mmol/L) and in ruminants
fructo-2-kinase/fructose-2,6-bisphosphatase). (PFK-1,
considerably lower
(approximately
2.2 mmol/L in
phosphofructokinase-1 [6-phosphofructo-1-kinase];
sheep and 3.3 mmol/L in cattle). These lower normal
F-1,6-Pase, fructose-1,6-bisphosphatase. Arrows with
levels appear to be associated with the fact that rumi-
wavy shafts indicate allosteric effects.)
nants ferment virtually all dietary carbohydrate to lower
(volatile) fatty acids, and these largely replace glucose as
the main metabolic fuel of the tissues in the fed condi-
fructose-1,6-bisphosphatase. When glucose is short,
tion.
glucagon stimulates the production of cAMP, activat-
ing cAMP-dependent protein kinase, which in turn in-
activates phosphofructokinase-2 and activates fructose
BLOOD GLUCOSE IS DERIVED FROM
2,6-bisphosphatase by phosphorylation. Therefore, glu-
THE DIET, GLUCONEOGENESIS,
coneogenesis is stimulated by a decrease in the concen-
& GLYCOGENOLYSIS
tration of fructose 2,6-bisphosphate, which deactivates
phosphofructokinase-1 and deinhibits fructose-1,6-bis-
The digestible dietary carbohydrates yield glucose,
phosphatase. This mechanism also ensures that glu-
galactose, and fructose that are transported via the
cagon stimulation of glycogenolysis in liver results in
hepatic portal vein to the liver where galactose and
glucose release rather than glycolysis.
fructose are readily converted to glucose (Chapter 20).
GLUCONEOGENESIS & CONTROL OF THE BLOOD GLUCOSE
/
159
Glucose is formed from two groups of compounds
Metabolic & Hormonal Mechanisms
that undergo gluconeogenesis (Figures 16-4 and 19-1):
Regulate the Concentration
(1) those which involve a direct net conversion to glu-
of the Blood Glucose
cose without significant recycling, such as some amino
The maintenance of stable levels of glucose in the blood
acids and propionate; and (2) those which are the
products of the metabolism of glucose in tissues. Thus,
is one of the most finely regulated of all homeostatic
mechanisms, involving the liver, extrahepatic tissues,
lactate, formed by glycolysis in skeletal muscle and
erythrocytes, is transported to the liver and kidney
and several hormones. Liver cells are freely permeable
to glucose (via the GLUT 2 transporter), whereas cells
where it re-forms glucose, which again becomes avail-
able via the circulation for oxidation in the tissues. This
of extrahepatic tissues (apart from pancreatic B islets)
are relatively impermeable, and their glucose trans-
process is known as the Cori cycle, or lactic acid cycle
(Figure 19-4). Triacylglycerol glycerol in adipose tissue
porters are regulated by insulin. As a result, uptake
from the bloodstream is the rate-limiting step in the
is derived from blood glucose. This triacylglycerol is
continuously undergoing hydrolysis to form free glyc-
utilization of glucose in extrahepatic tissues. The role of
various glucose transporter proteins found in cell mem-
erol, which cannot be utilized by adipose tissue and is
converted back to glucose by gluconeogenic mecha-
branes, each having
12 transmembrane domains, is
shown in Table 19-2.
nisms in the liver and kidney (Figure 19-1).
Of the amino acids transported from muscle to the
liver during starvation, alanine predominates. The glu-
Glucokinase Is Important in Regulating
cose-alanine cycle
(Figure
19-4) transports glucose
Blood Glucose After a Meal
from liver to muscle with formation of pyruvate, fol-
lowed by transamination to alanine, then transports
Hexokinase has a low Km for glucose and in the liver is
alanine to the liver, followed by gluconeogenesis back
saturated and acting at a constant rate under all normal
to glucose. A net transfer of amino nitrogen from mus-
conditions. Glucokinase has a considerably higher Km
cle to liver and of free energy from liver to muscle is ef-
(lower affinity) for glucose, so that its activity increases
fected. The energy required for the hepatic synthesis of
over the physiologic range of glucose concentrations
glucose from pyruvate is derived from the oxidation of
(Figure
19-5). It promotes hepatic uptake of large
fatty acids.
amounts of glucose at the high concentrations found in
Glucose is also formed from liver glycogen by
the hepatic portal vein after a carbohydrate meal. It is
glycogenolysis (Chapter 18).
absent from the liver of ruminants, which have little
BLOOD
Glucose
LIVER
MUSCLE
Glucose 6-phosphate
Glycogen
Glycogen
Glucose 6-phosphate
Urea
Pyruvate
Lactate
Lactate
Pyruvate
-NH2
-NH2
Lactate
BLOOD
Pyruvate
Alanine
Alanine
Alanine
Figure 19-4. The lactic acid (Cori) cycle and glucose-alanine cycle.
160
/
CHAPTER 19
Table 19-2. Glucose transporters.
Tissue Location
Functions
Facilitative bidirectional transporters
GLUT 1
Brain, kidney, colon, placenta, erythrocyte
Uptake of glucose
GLUT 2
Liver, pancreatic B cell, small intestine, kidney
Rapid uptake and release of glucose
GLUT 3
Brain, kidney, placenta
Uptake of glucose
GLUT 4
Heart and skeletal muscle, adipose tissue
Insulin-stimulated uptake of glucose
GLUT 5
Small intestine
Absorption of glucose
Sodium-dependent unidirectional transporter
SGLT 1
Small intestine and kidney
Active uptake of glucose from lumen of intestine and
reabsorption of glucose in proximal tubule of kidney
against a concentration gradient
cose via the GLUT 2 transporter, and the glucose is
glucose entering the portal circulation from the intes-
phosphorylated by glucokinase. Therefore, increasing
tines.
blood glucose increases metabolic flux through glycoly-
At normal systemic-blood glucose concentrations
sis, the citric acid cycle, and the generation of ATP. In-
(4.5-5.5 mmol/L), the liver is a net producer of glu-
crease in
[ATP] inhibits ATP-sensitive K+ channels,
cose. However, as the glucose level rises, the output of
causing depolarization of the B cell membrane, which
glucose ceases, and there is a net uptake.
channels,
increases Ca2+ influx via voltage-sensitive Ca2+
Insulin Plays a Central Role in
stimulating exocytosis of insulin. Thus, the concentra-
tion of insulin in the blood parallels that of the blood
Regulating Blood Glucose
glucose. Other substances causing release of insulin from
In addition to the direct effects of hyperglycemia in en-
the pancreas include amino acids, free fatty acids, ketone
hancing the uptake of glucose into the liver, the hor-
bodies, glucagon, secretin, and the sulfonylurea drugs
mone insulin plays a central role in regulating blood glu-
tolbutamide and glyburide. These drugs are used to
cose. It is produced by the B cells of the islets of
stimulate insulin secretion in type 2 diabetes mellitus
Langerhans in the pancreas in response to hyper-
(NIDDM, non-insulin-dependent diabetes mellitus);
glycemia. The B islet cells are freely permeable to glu-
they act by inhibiting the ATP-sensitive K+ channels.
Epinephrine and norepinephrine block the release of in-
sulin. Insulin lowers blood glucose immediately by en-
V
max
100
hancing glucose transport into adipose tissue and muscle
Hexokinase
by recruitment of glucose transporters (GLUT 4) from
the interior of the cell to the plasma membrane. Al-
though it does not affect glucose uptake into the liver
directly, insulin does enhance long-term uptake as a re-
sult of its actions on the enzymes controlling glycolysis,
50
Glucokinase
glycogenesis, and gluconeogenesis (Chapter 18).
Glucagon Opposes the Actions of Insulin
Glucagon is the hormone produced by the A cells of
0
5
10
15
20
25
the pancreatic islets. Its secretion is stimulated by hypo-
Blood glucose (mmol/L)
glycemia. In the liver, it stimulates glycogenolysis by ac-
tivating phosphorylase. Unlike epinephrine, glucagon
Figure 19-5. Variation in glucose phosphorylating
does not have an effect on muscle phosphorylase.
activity of hexokinase and glucokinase with increase of
Glucagon also enhances gluconeogenesis from amino
blood glucose concentration. The Km
for glucose of
acids and lactate. In all these actions, glucagon acts via
hexokinase is 0.05 mmol/L and of glucokinase is 10
generation of cAMP
(Table
19-1). Both hepatic
mmol/L.
glycogenolysis and gluconeogenesis contribute to the
GLUCONEOGENESIS & CONTROL OF THE BLOOD GLUCOSE
/
161
hyperglycemic effect of glucagon, whose actions op-
meals or at night. Furthermore, premature and low-
pose those of insulin. Most of the endogenous glucagon
birth-weight babies are more susceptible to hypo-
(and insulin) is cleared from the circulation by the liver.
glycemia, since they have little adipose tissue to gener-
ate alternative fuels such as free fatty acids or ketone
bodies during the transition from fetal dependency to
Other Hormones Affect Blood Glucose
the free-living state. The enzymes of gluconeogenesis
The anterior pituitary gland secretes hormones that
may not be completely functional at this time, and the
tend to elevate the blood glucose and therefore antago-
process is dependent on a supply of free fatty acids for
nize the action of insulin. These are growth hormone,
energy. Glycerol, which would normally be released
ACTH (corticotropin), and possibly other “diabeto-
from adipose tissue, is less available for gluconeogenesis.
genic” hormones. Growth hormone secretion is stimu-
lated by hypoglycemia; it decreases glucose uptake in
The Body’s Ability to Utilize Glucose
muscle. Some of this effect may not be direct, since it
May Be Ascertained by Measuring Its
stimulates mobilization of free fatty acids from adipose
Glucose Tolerance
tissue, which themselves inhibit glucose utilization. The
Glucose tolerance is the ability to regulate the blood
glucocorticoids (11-oxysteroids) are secreted by the
adrenal cortex and increase gluconeogenesis. This is a
glucose concentration after the administration of a test
dose of glucose (normally 1 g/kg body weight) (Figure
result of enhanced hepatic uptake of amino acids and
increased activity of aminotransferases and key enzymes
19-6). Diabetes mellitus (type 1, or insulin-dependent
diabetes mellitus; IDDM) is characterized by decreased
of gluconeogenesis. In addition, glucocorticoids inhibit
the utilization of glucose in extrahepatic tissues. In all
glucose tolerance due to decreased secretion of insulin
in response to the glucose challenge. Glucose tolerance
these actions, glucocorticoids act in a manner antago-
nistic to insulin.
is also impaired in type 2 diabetes mellitus (NIDDM),
which is often associated with obesity and raised levels
Epinephrine is secreted by the adrenal medulla as a
result of stressful stimuli (fear, excitement, hemorrhage,
of plasma free fatty acids and in conditions where the
liver is damaged; in some infections; and in response to
hypoxia, hypoglycemia, etc) and leads to glycogenolysis
in liver and muscle owing to stimulation of phosphory-
some drugs. Poor glucose tolerance can also be expected
lase via generation of cAMP. In muscle, glycogenolysis
results in increased glycolysis, whereas in liver glucose is
15
the main product leading to increase in blood glucose.
FURTHER CLINICAL ASPECTS
Glucosuria Occurs When the Renal
Threshold for Glucose Is Exceeded
10
When the blood glucose rises to relatively high levels,
the kidney also exerts a regulatory effect. Glucose is
continuously filtered by the glomeruli but is normally
completely reabsorbed in the renal tubules by active
transport. The capacity of the tubular system to reab-
5
sorb glucose is limited to a rate of about 350 mg/min,
and in hyperglycemia (as occurs in poorly controlled di-
abetes mellitus) the glomerular filtrate may contain
more glucose than can be reabsorbed, resulting in glu-
cosuria. Glucosuria occurs when the venous blood glu-
cose concentration exceeds 9.5-10.0 mmol/L; this is
termed the renal threshold for glucose.
0
1
2
Time (h)
Hypoglycemia May Occur During
Figure 19-6. Glucose tolerance test. Blood glucose
Pregnancy & in the Neonate
curves of a normal and a diabetic individual after oral
During pregnancy, fetal glucose consumption increases
administration of 50 g of glucose. Note the initial raised
and there is a risk of maternal and possibly fetal hypo-
concentration in the diabetic. A criterion of normality is
glycemia, particularly if there are long intervals between
the return of the curve to the initial value within 2 hours.
162
/
CHAPTER 19
due to hyperactivity of the pituitary or adrenal cortex
• Insulin is secreted as a direct response to hyper-
because of the antagonism of the hormones secreted by
glycemia; it stimulates the liver to store glucose as
these glands to the action of insulin.
glycogen and facilitates uptake of glucose into extra-
Administration of insulin (as in the treatment of di-
hepatic tissues.
abetes mellitus type 1) lowers the blood glucose and in-
• Glucagon is secreted as a response to hypoglycemia
creases its utilization and storage in the liver and muscle
and activates both glycogenolysis and gluconeogene-
as glycogen. An excess of insulin may cause hypo-
sis in the liver, causing release of glucose into the
glycemia, resulting in convulsions and even in death
blood.
unless glucose is administered promptly. Increased tol-
erance to glucose is observed in pituitary or adrenocor-
tical insufficiency—attributable to a decrease in the an-
tagonism to insulin by the hormones normally secreted
REFERENCES
by these glands.
Burant CF et al: Mammalian glucose transporters: structure and
molecular regulation. Recent Prog Horm Res 1991;47:349.
SUMMARY
Krebs HA: Gluconeogenesis. Proc R Soc London (Biol) 1964;
159:545.
• Gluconeogenesis is the process of converting noncar-
Lenzen S: Hexose recognition mechanisms in pancreatic B-cells.
bohydrates to glucose or glycogen. It is of particular
Biochem Soc Trans 1990;18:105.
importance when carbohydrate is not available from
Newgard CB, McGarry JD: Metabolic coupling factors in pancre-
the diet. Significant substrates are amino acids, lac-
atic beta-cell signal transduction. Annu Rev Biochem 1995;
tate, glycerol, and propionate.
64:689.
• The pathway of gluconeogenesis in the liver and kid-
Newsholme EA, Start C: Regulation in Metabolism. Wiley, 1973.
ney utilizes those reactions in glycolysis which are re-
Nordlie RC, Foster JD, Lange AJ: Regulation of glucose produc-
versible plus four additional reactions that circum-
tion by the liver. Annu Rev Nutr 1999;19:379.
vent the irreversible nonequilibrium reactions.
Pilkis SJ, El-Maghrabi MR, Claus TH: Hormonal regulation of he-
patic gluconeogenesis and glycolysis. Annu Rev Biochem
• Since glycolysis and gluconeogenesis share the same
1988;57:755.
pathway but operate in opposite directions, their ac-
Pilkis SJ, Granner DK: Molecular physiology of the regulation of
tivities are regulated reciprocally.
hepatic gluconeogenesis and glycolysis. Annu Rev Physiol
• The liver regulates the blood glucose after a meal be-
1992;54:885.
cause it contains the high-Km glucokinase that pro-
Yki-Jarvinen H: Action of insulin on glucose metabolism in vivo.
motes increased hepatic utilization of glucose.
Baillieres Clin Endocrinol Metab 1993;7:903.
The Pentose Phosphate
Pathway & Other Pathways
20
of Hexose Metabolism
Peter A. Mayes, PhD, DSc, & David A. Bender, PhD
BIOMEDICAL IMPORTANCE
REACTIONS OF THE PENTOSE
PHOSPHATE PATHWAY OCCUR
The pentose phosphate pathway is an alternative route
for the metabolism of glucose. It does not generate
IN THE CYTOSOL
ATP but has two major functions: (1) The formation of
The enzymes of the pentose phosphate pathway, as of
NADPH for synthesis of fatty acids and steroids and
glycolysis, are cytosolic. As in glycolysis, oxidation
(2) the synthesis of ribose for nucleotide and nucleic
is achieved by dehydrogenation; but NADP+ and not
acid formation. Glucose, fructose, and galactose are the
NAD+ is the hydrogen acceptor. The sequence of reac-
main hexoses absorbed from the gastrointestinal tract,
tions of the pathway may be divided into two phases: an
derived principally from dietary starch, sucrose, and
oxidative nonreversible phase and a nonoxidative re-
lactose, respectively. Fructose and galactose are con-
versible phase. In the first phase, glucose 6-phosphate
verted to glucose, mainly in the liver.
undergoes dehydrogenation and decarboxylation to yield
Genetic deficiency of glucose 6-phosphate dehydro-
a pentose, ribulose 5-phosphate. In the second phase,
genase, the first enzyme of the pentose phosphate path-
ribulose 5-phosphate is converted back to glucose 6-phos-
way, is a major cause of hemolysis of red blood cells, re-
phate by a series of reactions involving mainly two en-
sulting in hemolytic anemia and affecting approximately
zymes: transketolase and transaldolase (Figure 20-1).
100 million people worldwide. Glucuronic acid is synthe-
sized from glucose via the uronic acid pathway, of major
The Oxidative Phase Generates NADPH
significance for the excretion of metabolites and foreign
chemicals (xenobiotics) as glucuronides. A deficiency in
(Figures 20-1 and 20-2)
the pathway leads to essential pentosuria. The lack of
Dehydrogenation of glucose 6-phosphate to 6-phos-
one enzyme of the pathway (gulonolactone oxidase) in
phogluconate occurs via the formation of 6-phospho-
primates and some other animals explains why ascorbic
gluconolactone, catalyzed by glucose-6-phosphate
acid (vitamin C) is a dietary requirement for humans but
dehydrogenase, an NADP-dependent enzyme. The
not most other mammals. Deficiencies in the enzymes of
hydrolysis of 6-phosphogluconolactone is accomplished
fructose and galactose metabolism lead to essential fruc-
by the enzyme gluconolactone hydrolase. A second
tosuria and the galactosemias.
oxidative step is catalyzed by 6-phosphogluconate de-
hydrogenase, which also requires NADP+ as hydrogen
THE PENTOSE PHOSPHATE PATHWAY
acceptor and involves decarboxylation followed by for-
GENERATES NADPH & RIBOSE
mation of the ketopentose, ribulose 5-phosphate.
PHOSPHATE (Figure 20-1)
The Nonoxidative Phase Generates
The pentose phosphate pathway (hexose monophos-
Ribose Precursors
phate shunt) is a more complex pathway than glycoly-
sis. Three molecules of glucose 6-phosphate give rise to
Ribulose 5-phosphate is the substrate for two enzymes.
three molecules of CO2 and three five-carbon sugars.
Ribulose 5-phosphate 3-epimerase alters the configu-
These are rearranged to regenerate two molecules of
ration about carbon 3, forming another ketopentose,
glucose 6-phosphate and one molecule of the glycolytic
xylulose 5-phosphate. Ribose 5-phosphate ketoisom-
intermediate, glyceraldehyde
3-phosphate. Since two
erase converts ribulose 5-phosphate to the correspond-
molecules of glyceraldehyde 3-phosphate can regenerate
ing aldopentose, ribose 5-phosphate, which is the pre-
glucose 6-phosphate, the pathway can account for the
cursor of the ribose required for nucleotide and nucleic
complete oxidation of glucose.
acid synthesis. Transketolase transfers the two-carbon
163
164
/
CHAPTER 20
Glucose 6-phosphate
Glucose 6-phosphate
Glucose 6-phosphate
C6
C6
C6
NADP+ + H2O
NADP+ + H2O
NADP+ + H2O
GLUCOSE-6-PHOSPHATE
DEHYDROGENASE
NADPH + H+
NADPH + H+
NADPH + H+
6-Phosphogluconate
6-Phosphogluconate
6-Phosphogluconate
C6
C6
C6
NADP+
NADP+
NADP+
6-PHOSPHO-
GLUCONATE
DEHYDROGENASE
NADPH + H+
NADPH + H+
NADPH + H+
CO2
CO2
CO2
Ribulose 5-phosphate
Ribulose 5-phosphate
Ribulose 5-phosphate
C5
C5
C5
3-EPIMERASE
KETO-ISOMERASE
3-EPIMERASE
Xylulose 5-phosphate
Ribose 5-phosphate
Xylulose 5-phosphate
C5
C5
C5
TRANSKETOLASE
Synthesis of
nucleotides,
RNA, DNA
Glyceraldehyde 3-phosphate
Sedoheptulose 7-phosphate
C3
C7
TRANSALDOLASE
Fructose 6-phosphate
Erythrose 4-phosphate
C6
C4
TRANSKETOLASE
Fructose 6-phosphate
Glyceraldehyde 3-phosphate
C6
C3
PHOSPHOTRIOSE
ALDOLASE
ISOMERASE
PHOSPHOHEXOSE
PHOSPHOHEXOSE
ISOMERASE
ISOMERASE
1/2 Fructose 1,6-bisphosphate
C6
FRUCTOSE-1,6-
BISPHOSPHATASE
1/2 Fructose 6-phosphate
C6
PHOSPHOHEXOSE
ISOMERASE
Glucose 6-phosphate
Glucose 6-phosphate
1/2 Glucose 6-phosphate
C6
C6
C6
Figure 20-1. Flow chart of pentose phosphate pathway and its connections with the pathway
of glycolysis. The full pathway, as indicated, consists of three interconnected cycles in which glu-
cose 6-phosphate is both substrate and end product. The reactions above the broken line are
nonreversible, whereas all reactions under that line are freely reversible apart from that catalyzed
by fructose-1,6-bisphosphatase.
THE PENTOSE PHOSPHATE PATHWAY & OTHER PATHWAYS OF HEXOSE METABOLISM
/
165
O
HO
C
H
NADP+ NADPH + H+
C
H2O
COO-
Mg
2+
Mg2+, Mn2+,
H
C
OH
H
C
OH
H
C
OH
or Ca2+
or Ca2+
HO
C
H
HO
C
H
HO
C
H
O
O
H
C
OH
GLUCOSE-6-PHOSPHATE
H
C
OH
GLUCONOLACTONE
H
C
OH
DEHYDROGENASE
HYDROLASE
H
C
H
C
H
C
OH
CH2
O P
CH2
O P
CH2
O P
β-D-Glucose 6-phosphate
6-Phosphogluconolactone
6-Phosphogluconate
NADP+
6-PHOSPHOGLUCONATE
Mg2+, Mn2+,
DEHYDROGENASE
or Ca2+
NADP+ + H+
COO-
CHOH
CH2OH
H
C
OH
RIBOSE 5-PHOSPHATE
C
OH
KETOISOMERASE
C O
C
O
H
C
OH
H
C
OH
H
C
OH
H
C
OH
H
C
OH
H
C
OH
CO2
CH2
O P
CH2
O P
CH2
O P
Enediol form
Ribulose 5-phosphate
3-Keto 6-phosphogluconate
RIBULOSE 5-PHOSPHATE
3-EPIMERASE
CH2OH
CH2OH
C O
C
O
H
C
OH
HO
C
H
HO
*C
H
H
C
OH
H
C
OH
H
*C
OH
H
C
OH
O
H
C
OH
*CH2
O P
H
C
H
C
OH
Xylulose 5-phosphate
CH2
O P
CH2
O P
Ribose 5-phosphate
Sedoheptulose 7-phosphate
ATP
TRANSKETOLASE
PRPP
Mg2+
Thiamin- P
H
*C
O
SYNTHETASE
2
AMP
Mg2+
H
*C
OH
CH2OH
H
C
O P
P
*CH2
O P
C O
H
C
OH
Glyceraldehyde 3-phosphate
HO
C
H
H
C
OH
O
TRANSALDOLASE
H
*C
OH
H
C
H
C
O
H
*C
OH
CH2
O P
H C
OH
*CH2
O
P
PRPP
H
C
OH
Fructose 6-phosphate
CH2
O P
Erythrose 4-phosphate
CH2OH
CH2OH
C O
C O
TRANSKETOLASE
HO
C
H
HO
C
H
Thiamin- P2
H C O
H
C
OH
Mg
2+
H
C
OH
H
C
OH
H
C
OH
CH2
O P
CH2
O P
CH2
O P
Xylulose 5-phosphate
Glyceraldehyde 3-phosphate
Fructose 6-phosphate
Figure 20-2. The pentose phosphate pathway. (
, PO32-; PRPP, 5-phosphoribosyl 1-pyrophosphate.)
P
166
/
CHAPTER 20
unit comprising carbons 1 and 2 of a ketose onto the
Ribose Can Be Synthesized in Virtually
aldehyde carbon of an aldose sugar. It therefore effects
All Tissues
the conversion of a ketose sugar into an aldose with two
Little or no ribose circulates in the bloodstream, so tis-
carbons less and simultaneously converts an aldose sugar
sues must synthesize the ribose required for nucleotide
into a ketose with two carbons more. The reaction re-
and nucleic acid synthesis (Chapter 34). The source of
quires Mg2+ and thiamin diphosphate (vitamin B1) as
ribose 5-phosphate is the pentose phosphate pathway
coenzyme. Thus, transketolase catalyzes the transfer of
(Figure 20-2). Muscle has only low activity of glucose-
the two-carbon unit from xylulose 5-phosphate to ribose
6-phosphate dehydrogenase and
6-phosphogluconate
5-phosphate, producing the seven-carbon ketose sedo-
dehydrogenase. Nevertheless, like most other tissues, it
heptulose 7-phosphate and the aldose glyceraldehyde
is capable of synthesizing ribose 5-phosphate by reversal
3-phosphate. Transaldolase allows the transfer of a
of the nonoxidative phase of the pentose phosphate
three-carbon dihydroxyacetone moiety
(carbons
1-3)
pathway utilizing fructose 6-phosphate. It is not neces-
from the ketose sedoheptulose 7-phosphate onto the al-
sary to have a completely functioning pentose phosphate
dose glyceraldehyde
3-phosphate to form the ketose
pathway for a tissue to synthesize ribose phosphates.
fructose 6-phosphate and the four-carbon aldose erythrose
4-phosphate. In a further reaction catalyzed by transke-
tolase, xylulose 5-phosphate donates a two-carbon unit
THE PENTOSE PHOSPHATE PATHWAY
to erythrose 4-phosphate to form fructose 6-phosphate
& GLUTATHIONE PEROXIDASE PROTECT
and glyceraldehyde 3-phosphate.
ERYTHROCYTES AGAINST HEMOLYSIS
In order to oxidize glucose completely to CO2 via
the pentose phosphate pathway, there must be enzymes
In erythrocytes, the pentose phosphate pathway pro-
present in the tissue to convert glyceraldehyde 3-phos-
vides NADPH for the reduction of oxidized glu-
phate to glucose 6-phosphate. This involves reversal of
tathione catalyzed by glutathione reductase, a flavo-
glycolysis and the gluconeogenic enzyme fructose 1,6-
protein containing FAD. Reduced glutathione removes
bisphosphatase. In tissues that lack this enzyme, glyc-
H2O2 in a reaction catalyzed by glutathione peroxi-
eraldehyde 3-phosphate follows the normal pathway of
dase, an enzyme that contains the selenium analogue
glycolysis to pyruvate.
of cysteine (selenocysteine) at the active site
(Figure
20-3). This reaction is important, since accumulation
of H2O2 may decrease the life span of the erythrocyte
The Two Major Pathways for the
by causing oxidative damage to the cell membrane,
Catabolism of Glucose Have
leading to hemolysis.
Little in Common
Although glucose
6-phosphate is common to both
GLUCURONATE, A PRECURSOR OF
pathways, the pentose phosphate pathway is markedly
PROTEOGLYCANS & CONJUGATED
different from glycolysis. Oxidation utilizes NADP
GLUCURONIDES, IS A PRODUCT OF
rather than NAD, and CO2, which is not produced in
THE URONIC ACID PATHWAY
glycolysis, is a characteristic product. No ATP is gener-
ated in the pentose phosphate pathway, whereas ATP is
In liver, the uronic acid pathway catalyzes the conver-
a major product of glycolysis.
sion of glucose to glucuronic acid, ascorbic acid, and
pentoses (Figure 20-4). It is also an alternative oxidative
pathway for glucose, but—like the pentose phosphate
Reducing Equivalents Are Generated
pathway—it does not lead to the generation of ATP.
in Those Tissues Specializing
Glucose 6-phosphate is isomerized to glucose 1-phos-
in Reductive Syntheses
phate, which then reacts with uridine triphosphate
The pentose phosphate pathway is active in liver, adipose
(UTP) to form uridine diphosphate glucose (UDPGlc)
tissue, adrenal cortex, thyroid, erythrocytes, testis, and
in a reaction catalyzed by UDPGlc pyrophosphorylase,
lactating mammary gland. Its activity is low in nonlactat-
as occurs in glycogen synthesis (Chapter 18). UDPGlc is
ing mammary gland and skeletal muscle. Those tissues in
oxidized at carbon 6 by NAD-dependent UDPGlc de-
which the pathway is active use NADPH in reductive
hydrogenase in a two-step reaction to yield UDP-glu-
syntheses, eg, of fatty acids, steroids, amino acids via glu-
curonate. UDP-glucuronate is the “active” form of glu-
tamate dehydrogenase, and reduced glutathione. The
curonate for reactions involving incorporation of
synthesis of glucose-6-phosphate dehydrogenase and
glucuronic acid into proteoglycans or for reactions in
6-phosphogluconate dehydrogenase may also be induced
which substrates such as steroid hormones, bilirubin, and
by insulin during conditions associated with the “fed
a number of drugs are conjugated with glucuronate for
state” (Table 19-1), when lipogenesis increases.
excretion in urine or bile (Figure 32-14).
THE PENTOSE PHOSPHATE PATHWAY & OTHER PATHWAYS OF HEXOSE METABOLISM
/
167
NADPH + H+
G
S S
G
2H2O
PENTOSE
GLUTATHIONE
GLUTATHIONE
PHOSPHATE
FAD
Se
REDUCTASE
PEROXIDASE
PATHWAY
2H
NADP+
2G SH
H2O2
Figure 20-3. Role of the pentose phosphate pathway in the glutathione peroxidase re-
action of erythrocytes. (G-S-S-G, oxidized glutathione; G-SH, reduced glutathione; Se, sele-
nium cofactor.)
Glucuronate is reduced to L-gulonate in an NADPH-
tose. However, glucose inhibits the phosphorylation of
dependent reaction; L-gulonate is the direct precursor of
fructose since it is a better substrate for hexokinase.
ascorbate in those animals capable of synthesizing this
Nevertheless, some fructose can be metabolized in adi-
vitamin. In humans and other primates as well as guinea
pose tissue and muscle. Fructose, a potential fuel, is
pigs, ascorbic acid cannot be synthesized because of the
found in seminal plasma and in the fetal circulation of
absence of L-gulonolactone oxidase. L-Gulonate is me-
ungulates and whales. Aldose reductase is found in the
tabolized ultimately to D-xylulose 5-phosphate, a con-
placenta of the ewe and is responsible for the secretion
stituent of the pentose phosphate pathway.
of sorbitol into the fetal blood. The presence of sor-
bitol dehydrogenase in the liver, including the fetal
liver, is responsible for the conversion of sorbitol into
INGESTION OF LARGE QUANTITIES
fructose. This pathway is also responsible for the occur-
OF FRUCTOSE HAS PROFOUND
rence of fructose in seminal fluid.
METABOLIC CONSEQUENCES
Diets high in sucrose or in high-fructose syrups used in
manufactured foods and beverages lead to large amounts
GALACTOSE IS NEEDED FOR THE
of fructose (and glucose) entering the hepatic portal vein.
SYNTHESIS OF LACTOSE, GLYCOLIPIDS,
Fructose undergoes more rapid glycolysis in the liver than
PROTEOGLYCANS, & GLYCOPROTEINS
does glucose because it bypasses the regulatory step cat-
alyzed by phosphofructokinase (Figure 20-5). This allows
Galactose is derived from intestinal hydrolysis of the
fructose to flood the pathways in the liver, leading to en-
disaccharide lactose, the sugar of milk. It is readily con-
hanced fatty acid synthesis, increased esterification of fatty
verted in the liver to glucose. Galactokinase catalyzes
acids, and increased VLDL secretion, which may raise
the phosphorylation of galactose, using ATP as phos-
serum triacylglycerols and ultimately raise LDL choles-
phate donor (Figure 20-6A). Galactose 1-phosphate re-
terol concentrations
(Figure
25-6). A specific kinase,
acts with uridine diphosphate glucose (UDPGlc) to
fructokinase, in liver (and kidney and intestine) catalyzes
form uridine diphosphate galactose (UDPGal) and glu-
the phosphorylation of fructose to fructose 1-phosphate.
cose 1-phosphate, in a reaction catalyzed by galactose
This enzyme does not act on glucose, and, unlike glucoki-
1-phosphate uridyl transferase. The conversion of
nase, its activity is not affected by fasting or by insulin,
UDPGal to UDPGlc is catalyzed by UDPGal 4-epim-
which may explain why fructose is cleared from the blood
erase. Epimerization involves an oxidation and reduc-
of diabetic patients at a normal rate. Fructose 1-phos-
tion at carbon 4 with NAD+ as coenzyme. Finally, glu-
phate is cleaved to D-glyceraldehyde and dihydroxyace-
cose is liberated from UDPGlc after conversion to
tone phosphate by aldolase B, an enzyme found in the
glucose 1-phosphate, probably via incorporation into
liver, which also functions in glycolysis by cleaving fruc-
glycogen followed by phosphorolysis (Chapter 18).
tose 1,6-bisphosphate. D-Glyceraldehyde enters glycolysis
Since the epimerase reaction is freely reversible, glu-
via phosphorylation to glyceraldehyde 3-phosphate, cat-
cose can be converted to galactose, so that galactose is
alyzed by triokinase. The two triose phosphates, dihy-
not a dietary essential. Galactose is required in the body
droxyacetone phosphate and glyceraldehyde 3-phosphate,
not only in the formation of lactose but also as a con-
may be degraded by glycolysis or may be substrates for al-
stituent of glycolipids
(cerebrosides), proteoglycans,
dolase and hence gluconeogenesis, which is the fate of
and glycoproteins. In the synthesis of lactose in the
much of the fructose metabolized in the liver.
mammary gland, UDPGal condenses with glucose to
In extrahepatic tissues, hexokinase catalyzes the
yield lactose, catalyzed by lactose synthase
(Figure
phosphorylation of most hexose sugars, including fruc-
20-6B).
168
/
CHAPTER 20
H
*C
OH
H
*C
O P
H
*C
O UDP
H
*C
O UDP
PHOSPHO-
UDPGlc PYRO-
UDPGlc
H
C
OH
GLUCOMUTASE
H
C
OH
PHOSPHORYLASE
H
C
OH
DEHYDROGENASE
H
C
OH
HO
C
H
HO
C
H
HO
C
H
HO
C
H
O
O
O
O
H
C
OH
H
C
OH
H
C
OH
H
C
OH
+
H
C
H
C
UTP
PPi
H
C
2NAD
2NADH
H
C
+ H2O
+ 2H+
CH2
O P
CH2OH
CH2OH
C
O-
O
α-D-Glucose
Glucose
Uridine diphosphate
Uridine diphosphate
6-phosphate
1-phosphate
glucose (UDPGlc)
glucuronate
Glucuronides
H2O
Proteoglycans
UDP
O
O
C
O-
C
O-
H
*C
OH
NADH
NADPH
CH2OH
HO
C H
HO C H
H
C
OH
CO
2
+ H+
NAD+
NADP+
+ H+
C O
C O
HO
C
H
HO
C
H
O
H
C
OH
H
C
OH
H
C
OH
H
C
OH
HO C H
HO C H
HO C H
H
C
CH2OH
CH2OH
*CH2OH
C
O-
L-Xylulose
3-Keto-L-gulonate
L-Gulonate
O
D-Glucuronate
H2O
NADPH + H+
Oxalate
Glycolate
L-Gulonolactone
CO2
O2
NADP+
BLOCK IN PRIMATES
AND GUINEA PIGS
BLOCK IN HUMANS
Glycolaldehyde
2-Keto-L-gulonolactone
BLOCK IN
PENTOSURIA
D-Xylulose 1-phosphate
CH2OH
CH2OH
O
O
NADH
H
C
OH
+
C O
C
C
NAD
+ H+
[2H]
HO
C
H
HO
C
H D-Xylulose
HO C
O
C
O
O
H
C
OH
H
C
OH
HO C
O
C
CH2OH
D-XYLULOSE
CH2OH
H
C
H
C
REDUCTASE
Oxalate
Xylitol
ATP
HO C H
HO C H
Diet
Mg2+
CH2OH
CH2OH
ADP
L-Ascorbate
L-Dehydroascorbate
D-Xylulose 5-phosphate
Pentose phosphate pathway
Figure 20-4. Uronic acid pathway. (Asterisk indicates the fate of carbon 1 of glucose;
, PO32-.)
P
THE PENTOSE PHOSPHATE PATHWAY & OTHER PATHWAYS OF HEXOSE METABOLISM
/
169
ATP
HEXOKINASE
Glycogen
GLUCOKINASE
ALDOSE
REDUCTASE
Glucose 6-phosphate
D-Glucose
D-Sorbitol
+
+
NAD
NADPH
NADP
GLUCOSE-6-PHOSPHATASE
PHOSPHOHEXOSE
+ H+
ISOMERASE
SORBITOL
DEHYDROGENASE
NADH
+ H+
HEXOKINASE
Fructose 6-phosphate
D-Fructose
Diet
ATP
FRUCTOSE-1,6-
FRUCTOKINASE
ATP
PHOSPHOFRUCTOKINASE
ATP
BISPHOSPHATASE
BLOCK IN ESSENTIAL
FRUCTOSURIA
Fructose 1,6-bisphosphate
Fructose 1-phosphate
BLOCK IN HEREDITARY
FRUCTOSE INTOLERANCE
ALDOLASE B
Dihydroxyacetone-phosphate
ALDOLASE A
PHOSPHO-
Fatty acid
ALDOLASE B
TRIOSE
esterification
ISOMERASE
ATP
Glyceraldehyde 3-phosphate
D-Glyceraldehyde
TRIOKINASE
2-Phosphoglycerate
Pyruvate
Fatty acid synthesis
Figure 20-5. Metabolism of fructose. Aldolase A is found in all tissues, whereas aldolase B is
the predominant form in liver. (*, not found in liver.)
Glucose Is the Precursor of All
CLINICAL ASPECTS
Amino Sugars (Hexosamines)
Impairment of the Pentose Phosphate
Amino sugars are important components of glycopro-
Pathway Leads to Erythrocyte Hemolysis
teins (Chapter 47), of certain glycosphingolipids (eg,
gangliosides) (Chapter 14), and of glycosaminoglycans
Genetic deficiency of glucose-6-phosphate dehydrogen-
(Chapter 48). The major amino sugars are glucosa-
ase, with consequent impairment of the generation of
mine, galactosamine, and mannosamine and the
NADPH, is common in populations of Mediterranean
nine-carbon compound sialic acid. The principal sialic
and Afro-Caribbean origin. The defect is manifested as
acid found in human tissues is N-acetylneuraminic acid
red cell hemolysis (hemolytic anemia) when suscepti-
(NeuAc). A summary of the metabolic interrelationships
ble individuals are subjected to oxidants, such as the an-
among the amino sugars is shown in Figure 20-7.
timalarial primaquine, aspirin, or sulfonamides or when
170
/
CHAPTER 20
A
Galactose
Glycogen
GLYCOGEN SYNTHASE
ATP
Pi
PHOSPHORYLASE
Mg2+
GALACTOKINASE
Glucose 1-phosphate
ADP
BLOCK IN
Galactose
GALACTOSEMIA
PHOSPHOGLUCOMUTASE
1-phosphate
UDPGlc
GALACTOSE
URIDINE
1-PHOSPHATE
NAD+
DIPHOSPHOGALACTOSE
GLUCOSE-
URIDYL TRANSFERASE
4-EPIMERASE
6-PHOSPHATASE
Glucose
1-phosphate
UDPGal
Glucose 6-phosphate
Glucose
B
NAD+
Glucose
UDPGlc
UDPGal
URIDINE
ATP
DIPHOSPHOGALACTOSE
4-EPIMERASE
UDPGlc
Mg2+
HEXOKINASE
Lactose
PYROPHOSPHORYLASE
LACTOSE
PP
i
SYNTHASE
ADP
PHOSPHOGLUCOMUTASE
Glucose 6-phosphate
Glucose 1-phosphate
Glucose
Figure 20-6. Pathway of conversion of (A) galactose to glucose in the liver and (B) glucose to lactose in
the lactating mammary gland.
they have eaten fava beans (Vicia fava—hence the term
uronic acid pathway. For example, administration of
favism). Glutathione peroxidase is dependent upon a
barbital or of chlorobutanol to rats results in a signifi-
supply of NADPH, which in erythrocytes can be
cant increase in the conversion of glucose to glu-
formed only via the pentose phosphate pathway. It re-
curonate, L-gulonate, and ascorbate.
duces organic peroxides and H2O2 as part of the body’s
defense against lipid peroxidation
(Figure
14-21).
Measurement of erythrocyte transketolase and its acti-
Loading of the Liver With Fructose
vation by thiamin diphosphate is used to assess thiamin
May Potentiate Hyperlipidemia
nutritional status (Chapter 45).
& Hyperuricemia
In the liver, fructose increases triacylglycerol synthesis
Disruption of the Uronic Acid Pathway Is
and VLDL secretion, leading to hypertriacylglyc-
Caused by Enzyme Defects & Some Drugs
erolemia—and increased LDL cholesterol—which can
In the rare hereditary disease essential pentosuria, con-
be regarded as potentially atherogenic (Chapter 26). In
siderable quantities of L-xylulose appear in the urine
addition, acute loading of the liver with fructose, as can
because of absence of the enzyme necessary to reduce
occur with intravenous infusion or following very high
L-xylulose to xylitol. Parenteral administration of xylitol
fructose intakes, causes sequestration of inorganic phos-
may lead to oxalosis, involving calcium oxalate deposi-
phate in fructose
1-phosphate and diminished ATP
tion in brain and kidneys (Figure 20-4). Various drugs
synthesis. As a result there is less inhibition of de novo
markedly increase the rate at which glucose enters the
purine synthesis by ATP and uric acid formation is in-
THE PENTOSE PHOSPHATE PATHWAY & OTHER PATHWAYS OF HEXOSE METABOLISM
/
171
Glycogen
Glucose 1-phosphate
ATP ADP
Glucose
Glucose 6-phosphate
Fructose 6-phosphate
Glutamine
AMIDOTRANSFERASE
ATP ADP
UTP
Glutamate
Glucosamine
Glucosamine
Glucosamine
UDP-
6-phosphate
PHOSPHOGLUCO-
1-phosphate
glucosamine*
MUTASE
Acetyl-CoA
PP
i
-
Acetyl-CoA
ATP ADP
N -Acetyl-
N -Acetyl-
N -Acetyl-
glucosamine
glucosamine
glucosamine
Glycosaminoglycans
6-phosphate
1-phosphate
(eg, heparin)
UTP
EPIMERASE
PP
i
N -Acetyl-
UDP-
Glycosaminoglycans
mannosamine
6-phosphate
N -acetylglucosamine*
(hyaluronic acid),
glycoproteins
Phosphoenolpyruvate
NAD+
EPIMERASE
N -Acetyl-
UDP-
neuraminic acid
N -acetylgalactosamine*
9-phosphate
Inhibiting
-
allosteric
effect
Sialic acid,
Glycosaminoglycans
gangliosides,
(chondroitins),
glycoproteins
glycoproteins
Figure 20-7. Summary of the interrelationships in metabolism of amino sugars. (At asterisk: Analo-
gous to UDPGlc.) Other purine or pyrimidine nucleotides may be similarly linked to sugars or amino sug-
ars. Examples are thymidine diphosphate (TDP)-glucosamine and TDP-N-acetylglucosamine.
creased, causing hyperuricemia, which is a cause of gout
fructose 1-phosphate, leads to hereditary fructose in-
(Chapter 34).
tolerance. Diets low in fructose, sorbitol, and sucrose
are beneficial for both conditions. One consequence of
hereditary fructose intolerance and of another condi-
Defects in Fructose Metabolism Cause
tion due to fructose-1,6-bisphosphatase deficiency is
Disease (Figure 20-5)
fructose-induced hypoglycemia despite the presence of
Lack of hepatic fructokinase causes essential fructo-
high glycogen reserves. The accumulation of fructose
suria, and absence of hepatic aldolase B, which cleaves
1-phosphate and fructose 1,6-bisphosphate allosterically
172
/
CHAPTER 20
inhibits the activity of liver phosphorylase. The seques-
SUMMARY
tration of inorganic phosphate also leads to depletion of
•
The pentose phosphate pathway, present in the cy-
ATP and hyperuricemia.
tosol, can account for the complete oxidation of glu-
cose, producing NADPH and CO2 but not ATP.
Fructose & Sorbitol in the Lens Are
•
The pathway has an oxidative phase, which is irre-
Associated With Diabetic Cataract
versible and generates NADPH; and a nonoxidative
Both fructose and sorbitol are found in the lens of the
phase, which is reversible and provides ribose precur-
eye in increased concentrations in diabetes mellitus and
sors for nucleotide synthesis. The complete pathway
may be involved in the pathogenesis of diabetic
is present only in those tissues having a requirement
cataract. The sorbitol (polyol) pathway (not found in
for NADPH for reductive syntheses, eg, lipogenesis
liver) is responsible for fructose formation from glucose
or steroidogenesis, whereas the nonoxidative phase is
(Figure 20-5) and increases in activity as the glucose
present in all cells requiring ribose.
concentration rises in diabetes in those tissues that are
•
In erythrocytes, the pathway has a major function in
not insulin-sensitive, ie, the lens, peripheral nerves, and
preventing hemolysis by providing NADPH to
renal glomeruli. Glucose is reduced to sorbitol by al-
maintain glutathione in the reduced state as the sub-
dose reductase, followed by oxidation of sorbitol to
strate for glutathione peroxidase.
fructose in the presence of NAD+ and sorbitol dehydro-
•
The uronic acid pathway is the source of glucuronic
genase (polyol dehydrogenase). Sorbitol does not dif-
acid for conjugation of many endogenous and exoge-
fuse through cell membranes easily and accumulates,
nous substances before excretion as glucuronides in
causing osmotic damage. Simultaneously, myoinositol
urine and bile.
levels fall. Sorbitol accumulation, myoinositol deple-
•
Fructose bypasses the main regulatory step in glycol-
tion, and diabetic cataract can be prevented by aldose
ysis, catalyzed by phosphofructokinase, and stimu-
reductase inhibitors in diabetic rats, and promising re-
lates fatty acid synthesis and hepatic triacylglycerol
sults have been obtained in clinical trials.
secretion.
When sorbitol is administered intravenously, it is
•
Galactose is synthesized from glucose in the lactating
converted to fructose rather than to glucose. It is poorly
absorbed in the small intestine, and much is fermented
mammary gland and in other tissues where it is re-
quired for the synthesis of glycolipids, proteoglycans,
by colonic bacteria to short-chain fatty acids, CO2, and
H2, leading to abdominal pain and diarrhea (sorbitol
and glycoproteins.
intolerance).
Enzyme Deficiencies in the Galactose
REFERENCES
Pathway Cause Galactosemia
Couet C, Jan P, Debry G: Lactose and cataract in humans: a re-
view. J Am Coll Nutr 1991;10:79.
Inability to metabolize galactose occurs in the galac-
Cox TM: Aldolase B and fructose intolerance. FASEB J 1994;8:62.
tosemias, which may be caused by inherited defects in
Cross NCP, Cox TM: Hereditary fructose intolerance. Int J
galactokinase, uridyl transferase, or 4-epimerase (Figure
Biochem 1990;22:685.
20-6A), though a deficiency in uridyl transferase is
Kador PF: The role of aldose reductase in the development of dia-
the best known cause. The galactose concentration in
betic complications. Med Res Rev 1988;8:325.
the blood and in the eye is reduced by aldose reductase
Kaufman FR, Devgan S: Classical galactosemia: a review. Endocri-
to galactitol, which accumulates, causing cataract. In
nologist 1995;5:189.
uridyl transferase deficiency, galactose 1-phosphate ac-
Macdonald I, Vrana A (editors): Metabolic Effects of Dietary Carbo-
cumulates and depletes the liver of inorganic phos-
hydrates. Karger, 1986.
phate. Ultimately, liver failure and mental deterioration
Mayes PA: Intermediary metabolism of fructose. Am J Clin Nutr
result. As the epimerase is present in adequate amounts,
1993(5 Suppl);58:754S.
the galactosemic individual can still form UDPGal
Van den Berghe G: Inborn errors of fructose metabolism. Annu
from glucose, and normal growth and development can
Rev Nutr 1994;14:41.
occur regardless of the galactose-free diets used to con-
Wood T: Physiological functions of the pentose phosphate path-
trol the symptoms of the disease.
way. Cell Biol Funct 1986;4:241.
Biosynthesis of Fatty Acids
21
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
The Fatty Acid Synthase Complex
Is a Polypeptide Containing
Fatty acids are synthesized by an extramitochondrial
Seven Enzyme Activities
system, which is responsible for the complete synthesis
of palmitate from acetyl-CoA in the cytosol. In the rat,
In bacteria and plants, the individual enzymes of the
the pathway is well represented in adipose tissue and
fatty acid synthase system are separate, and the acyl
liver, whereas in humans adipose tissue may not be an
radicals are found in combination with a protein called
important site, and liver has only low activity. In birds,
the acyl carrier protein (ACP). However, in yeast,
lipogenesis is confined to the liver, where it is particu-
mammals, and birds, the synthase system is a multien-
larly important in providing lipids for egg formation. In
zyme polypeptide complex that incorporates ACP,
most mammals, glucose is the primary substrate for
which takes over the role of CoA. It contains the vita-
lipogenesis, but in ruminants it is acetate, the main fuel
min pantothenic acid in the form of 4′-phosphopan-
molecule produced by the diet. Critical diseases of the
tetheine (Figure 45-18). The use of one multienzyme
pathway have not been reported in humans. However,
functional unit has the advantages of achieving the ef-
inhibition of lipogenesis occurs in type 1 (insulin-de-
fect of compartmentalization of the process within the
pendent) diabetes mellitus, and variations in its activ-
cell without the erection of permeability barriers, and
ity may affect the nature and extent of obesity.
synthesis of all enzymes in the complex is coordinated
since it is encoded by a single gene.
THE MAIN PATHWAY FOR DE NOVO
In mammals, the fatty acid synthase complex is a
dimer comprising two identical monomers, each con-
SYNTHESIS OF FATTY ACIDS
taining all seven enzyme activities of fatty acid synthase
(LIPOGENESIS) OCCURS
on one polypeptide chain (Figure 21-2). Initially, a
IN THE CYTOSOL
priming molecule of acetyl-CoA combines with a cys-
This system is present in many tissues, including liver,
teine
SH group catalyzed by acetyl transacylase
kidney, brain, lung, mammary gland, and adipose tis-
(Figure
21-3, reaction
1a). Malonyl-CoA combines
sue. Its cofactor requirements include NADPH, ATP,
with the adjacent SH on the 4′-phosphopantetheine
Mn2+, biotin, and HCO3− (as a source of CO2). Acetyl-
of ACP of the other monomer, catalyzed by malonyl
CoA is the immediate substrate, and free palmitate is
transacylase (reaction 1b), to form acetyl (acyl)-mal-
the end product.
onyl enzyme. The acetyl group attacks the methylene
group of the malonyl residue, catalyzed by 3-ketoacyl
synthase, and liberates CO2, forming 3-ketoacyl en-
Production of Malonyl-CoA Is
zyme (acetoacetyl enzyme) (reaction 2), freeing the cys-
the Initial & Controlling Step
teine SH group. Decarboxylation allows the reaction
in Fatty Acid Synthesis
to go to completion, pulling the whole sequence of re-
Bicarbonate as a source of CO2 is required in the initial
actions in the forward direction. The 3-ketoacyl group
reaction for the carboxylation of acetyl-CoA to mal-
is reduced, dehydrated, and reduced again (reactions 3,
onyl-CoA in the presence of ATP and acetyl-CoA car-
4,
5) to form the corresponding saturated acyl-S-
boxylase. Acetyl-CoA carboxylase has a requirement
enzyme. A new malonyl-CoA molecule combines with
for the vitamin biotin (Figure 21-1). The enzyme is a
the SH of 4′-phosphopantetheine, displacing the sat-
multienzyme protein containing a variable number of
urated acyl residue onto the free cysteine SH group.
identical subunits, each containing biotin, biotin car-
The sequence of reactions is repeated six more times
boxylase, biotin carboxyl carrier protein, and transcar-
until a saturated 16-carbon acyl radical (palmityl) has
boxylase, as well as a regulatory allosteric site. The reac-
been assembled. It is liberated from the enzyme com-
tion takes place in two steps: (1) carboxylation of biotin
plex by the activity of a seventh enzyme in the complex,
involving ATP and (2) transfer of the carboxyl to
thioesterase (deacylase). The free palmitate must be ac-
acetyl-CoA to form malonyl-CoA.
tivated to acyl-CoA before it can proceed via any other
173
174
/
CHAPTER 21
CH3
CO S CoA
-OOC
CH2
CO
S CoA
Acetyl-CoA
Malonyl-CoA
Enz biotin COO-
Enz biotin
ADP + P i
ATP + HCO3-
Figure 21-1. Biosynthesis of malonyl-CoA. (Enz, acetyl-CoA carboxylase.)
metabolic pathway. Its usual fate is esterification into
+
CH CO⋅S⋅CoA+7HOOC⋅CH CO⋅S⋅CoA+14NADPH+14H
2
2
acylglycerols, chain elongation or desaturation, or ester-
+
ification to cholesteryl ester. In mammary gland, there
CH (CH ) COOH+7CO +6H O+8CoA⋅SH+14NADP
3
2 14
2
2
→
is a separate thioesterase specific for acyl residues of C8,
C10, or C12, which are subsequently found in milk
The acetyl-CoA used as a primer forms carbon
lipids.
atoms 15 and 16 of palmitate. The addition of all the
The equation for the overall synthesis of palmitate
subsequent C2 units is via malonyl-CoA. Propionyl-
from acetyl-CoA and malonyl-CoA is:
CoA acts as primer for the synthesis of long-chain fatty
Hydratase
Malonyl
Enoyl
transacylase
reductase
Acetyl
Ketoacyl
transacylase
reductase
Ketoacyl
1.
ACP
Thioesterase
synthase
4′-Phospho-
Cys
pantetheine
SH
Subunit
SH
SH
division
SH
4′-Phospho-
Cys
2.
pantetheine
Ketoacyl
Thioesterase
ACP
synthase
Ketoacyl
Acetyl
reductase
transacylase
Enoyl
Malonyl
reductase
transacylase
Hydratase
Figure 21-2. Fatty acid synthase multienzyme complex. The complex is a dimer of two identical polypeptide
monomers, 1 and 2, each consisting of seven enzyme activities and the acyl carrier protein (ACP). (CysSH, cys-
teine thiol.) The SH of the 4′-phosphopantetheine of one monomer is in close proximity to the SH of the cys-
teine residue of the ketoacyl synthase of the other monomer, suggesting a “head-to-tail” arrangement of the two
monomers. Though each monomer contains all the partial activities of the reaction sequence, the actual func-
tional unit consists of one-half of one monomer interacting with the complementary half of the other. Thus, two
acyl chains are produced simultaneously. The sequence of the enzymes in each monomer is based on Wakil.
*CO2
Acetyl-CoA
*Malonyl-CoA
C2
ACETYL-CoA
C3
CARBOXYLASE
1a
HS Pan
1
Cys SH
1b
CoA
ACETYL
Cn transfer from
TRANSACYLASE
MALONYL
TRANSACYLASE
CoA
2
to
1
HS
Cys
2
Pan SH
C2
Fatty acid synthase
O
multienzyme complex
1
Cys S
C CH
3
O
2
Pan S
C CH2
*COO-
(C3 )
Acyl(acetyl)-malonyl enzyme
3-KETOACYL
2
SYNTHASE
*CO2
1
Cys SH
O
O
2
Pan S
C CH2
C
CH3
3-Ketoacyl enzyme
(acetoacetyl enzyme)
NADPH + H+
3-KETOACYL
3
REDUCTASE
NADP+
1
Cys SH
O
OH
2
Pan S
C CH2
CH CH3
NADPH
GENERATORS
D
(-)-3-Hydroxyacyl enzyme
Pentose phosphate
pathway
HYDRATASE
4
Isocitrate
H2O
dehydrogenase
Malic
enzyme
1
Cys SH
O
2
Pan S
C CH CH CH3
2,3-Unsaturated acyl enzyme
+
NADPH + H
ENOYL REDUCTASE
5
NADP+
H2O
1
Cys SH
THIOESTERASE
O
After cycling through
2
Pan S
C CH2
CH2
CH3
)
steps
2
-
5
seven times
(Cn
Acyl enzyme
Palmitate
KEY:
1
,
2
, individual monomers of fatty acid synthase
Figure 21-3. Biosynthesis of long-chain fatty acids. Details of how addition of a malonyl residue
causes the acyl chain to grow by two carbon atoms. (Cys, cysteine residue; Pan, 4′-phosphopante-
theine.) The blocks shown in dark blue contain initially a C2 unit derived from acetyl-CoA (as illustrated)
and subsequently the Cn unit formed in reaction 5.
175
176
/
CHAPTER 21
acids having an odd number of carbon atoms, found
phate pathway. Moreover, both metabolic pathways are
particularly in ruminant fat and milk.
found in the cytosol of the cell, so there are no mem-
branes or permeability barriers against the transfer of
The Main Source of NADPH
NADPH. Other sources of NADPH include the reac-
for Lipogenesis Is the Pentose
tion that converts malate to pyruvate catalyzed by the
“malic enzyme” (NADP malate dehydrogenase) (Fig-
Phosphate Pathway
ure 21-4) and the extramitochondrial isocitrate dehy-
NADPH is involved as donor of reducing equivalents
drogenase reaction (probably not a substantial source,
in both the reduction of the 3-ketoacyl and of the 2,3-
except in ruminants).
unsaturated acyl derivatives (Figure 21-3, reactions 3
and 5). The oxidative reactions of the pentose phos-
Acetyl-CoA Is the Principal Building
phate pathway (see Chapter 20) are the chief source of
Block of Fatty Acids
the hydrogen required for the reductive synthesis of
fatty acids. Significantly, tissues specializing in active
Acetyl-CoA is formed from glucose via the oxidation of
lipogenesis—ie, liver, adipose tissue, and the lactating
pyruvate within the mitochondria. However, it does
mammary gland—also possess an active pentose phos-
not diffuse readily into the extramitochondrial cytosol,
Glucose
Palmitate
Glucose 6-phosphate
NADP+
PPP
NADP+
Fructose 6-phosphate
NADPH
NADPH + H+
Malic
+ H+
enzyme
MALATE
Glyceraldehyde
DEHYDROGENASE
Malonyl-CoA
3-phosphate
NAD+
Malate
CO
2
GLYCERALDEHYDE-
ACETYL-
ATP
3-PHOSPHATE
CoA
DEHYDROGENASE
CARBOXY-
NADH + H+
Oxaloacetate
CO
2
LASE
Pyruvate
Acetyl-CoA
CoA
CYTOSOL
ATP-
CITRATE
CoA
ATP
Acetate
LYASE
ATP
Citrate
H+
Citrate
Isocitrate
ISOCITRATE
Outside
DEHYDROGENASE
T
P
INNER MITOCHONDRIAL MEMBRANE
T
Inside
PYRUVATE
DEHYDROGENASE
Malate
Pyruvate
Acetyl-CoA
MITOCHONDRION
α-Ketoglutarate
NADH + H+
Oxaloacetate
Citrate
Citric acid cycle
NAD+
Malate
α-Ketoglutarate
K
Figure 21-4. The provision of acetyl-CoA and NADPH for lipogenesis. (PPP, pentose phosphate path-
way; T, tricarboxylate transporter; K, α-ketoglutarate transporter; P, pyruvate transporter.)
BIOSYNTHESIS OF FATTY ACIDS
/
177
the principal site of fatty acid synthesis. Citrate, formed
O
O
after condensation of acetyl-CoA with oxaloacetate in
R
CH2
C S CoA
+ CH2
C S CoA
the citric acid cycle within mitochondria, is translo-
cated into the extramitochondrial compartment via the
COOH
tricarboxylate transporter, where in the presence of
Acyl-CoA
Malonyl-CoA
CoA and ATP it undergoes cleavage to acetyl-CoA and
oxaloacetate catalyzed by ATP-citrate lyase, which in-
creases in activity in the well-fed state. The acetyl-CoA
3-KETOACYL-CoA
is then available for malonyl-CoA formation and syn-
SYNTHASE
CoA SH + CO2
thesis to palmitate
(Figure 21-4). The resulting ox-
aloacetate can form malate via NADH-linked malate
dehydrogenase, followed by the generation of NADPH
O
O
via the malic enzyme. The NADPH becomes available
R
CH2
C
CH2
C S CoA
for lipogenesis, and the pyruvate can be used to regen-
erate acetyl-CoA after transport into the mitochon-
3-Ketoacyl-CoA
drion. This pathway is a means of transferring reducing
NADPH + H+
equivalents from extramitochondrial NADH to NADP.
Alternatively, malate itself can be transported into the
3-KETOACYL-CoA
REDUCTASE
mitochondrion, where it is able to re-form oxaloacetate.
Note that the citrate (tricarboxylate) transporter in the
NADP+
mitochondrial membrane requires malate to exchange
OH
O
with citrate (see Figure 12-10). There is little ATP-
citrate lyase or malic enzyme in ruminants, probably
R
CH2
CH
CH2
C S CoA
because in these species acetate
(derived from the
3-Hydroxyacyl-CoA
rumen and activated to acetyl CoA extramitochondri-
ally) is the main source of acetyl-CoA.
3-HYDROXYACYL-CoA
DEHYDRASE
Elongation of Fatty Acid Chains Occurs
H
2O
in the Endoplasmic Reticulum
This pathway (the “microsomal system”) elongates sat-
O
urated and unsaturated fatty acyl-CoAs (from C10 up-
R
CH
CH
CH
C S CoA
2
ward) by two carbons, using malonyl-CoA as acetyl
2-trans-Enoyl-CoA
donor and NADPH as reductant, and is catalyzed by
the microsomal fatty acid elongase system of enzymes
NADPH + H+
(Figure 21-5). Elongation of stearyl-CoA in brain in-
2-trans-ENOYL-CoA
creases rapidly during myelination in order to provide
REDUCTASE
C22 and C24 fatty acids for sphingolipids.
NADP+
O
THE NUTRITIONAL STATE
REGULATES LIPOGENESIS
R
CH2
CH2
CH2
C S CoA
Acyl-CoA
Excess carbohydrate is stored as fat in many animals in
anticipation of periods of caloric deficiency such as star-
Figure 21-5. Microsomal elongase system for fatty
vation, hibernation, etc, and to provide energy for use
acid chain elongation. NADH is also used by the reduc-
between meals in animals, including humans, that take
tases, but NADPH is preferred.
their food at spaced intervals. Lipogenesis converts sur-
plus glucose and intermediates such as pyruvate, lactate,
and acetyl-CoA to fat, assisting the anabolic phase of
a fat diet, or when there is a deficiency of insulin, as in
this feeding cycle. The nutritional state of the organism
diabetes mellitus. These latter conditions are associated
is the main factor regulating the rate of lipogenesis.
with increased concentrations of plasma free fatty acids,
Thus, the rate is high in the well-fed animal whose diet
and an inverse relationship has been demonstrated be-
contains a high proportion of carbohydrate. It is de-
tween hepatic lipogenesis and the concentration of
pressed under conditions of restricted caloric intake, on
serum-free fatty acids. Lipogenesis is increased when su-
178
/
CHAPTER 21
crose is fed instead of glucose because fructose bypasses
PROTEIN
the phosphofructokinase control point in glycolysis and
Pi
PHOSPHATASE
floods the lipogenic pathway (Figure 20-5).
H2O
SHORT- & LONG-TERM MECHANISMS
ACETYL-CoA
ACETYL-CoA
REGULATE LIPOGENESIS
CARBOXYLASE
P
CARBOXYLASE
(active)
(inactive)
Long-chain fatty acid synthesis is controlled in the
short term by allosteric and covalent modification of
Acetyl-
enzymes and in the long term by changes in gene ex-
CoA
pression governing rates of synthesis of enzymes.
ATP
AMPK
ADP
Acetyl-CoA Carboxylase Is the Most
H2O
(active)
Important Enzyme in the Regulation
Malonyl-
CoA
of Lipogenesis
P
AMPKK
Acetyl-CoA carboxylase is an allosteric enzyme and is
activated by citrate, which increases in concentration in
AMPK
+
+
the well-fed state and is an indicator of a plentiful sup-
(inactive)
Pi
ATP
ply of acetyl-CoA. Citrate converts the enzyme from an
Acyl-CoA
inactive dimer to an active polymeric form, having a
molecular mass of several million. Inactivation is pro-
cAMP-DEPENDENT
Glucagon
cAMP
moted by phosphorylation of the enzyme and by long-
PROTEIN KINASE
+
+
chain acyl-CoA molecules, an example of negative feed-
back inhibition by a product of a reaction. Thus, if
Figure 21-6. Regulation of acetyl-CoA carboxylase
acyl-CoA accumulates because it is not esterified
by phosphorylation/dephosphorylation. The enzyme is
quickly enough or because of increased lipolysis or an
inactivated by phosphorylation by AMP-activated pro-
influx of free fatty acids into the tissue, it will automati-
tein kinase (AMPK), which in turn is phosphorylated
cally reduce the synthesis of new fatty acid. Acyl-CoA
and activated by AMP-activated protein kinase kinase
may also inhibit the mitochondrial tricarboxylate
(AMPKK). Glucagon (and epinephrine), after increasing
transporter, thus preventing activation of the enzyme
by egress of citrate from the mitochondria into the cy-
cAMP, activate this latter enzyme via cAMP-dependent
tosol.
protein kinase. The kinase kinase enzyme is also be-
Acetyl-CoA carboxylase is also regulated by hor-
lieved to be activated by acyl-CoA. Insulin activates
mones such as glucagon, epinephrine, and insulin via
acetyl-CoA carboxylase, probably through an “activa-
changes in its phosphorylation state (details in Figure
tor” protein and an insulin-stimulated protein kinase.
21-6).
Pyruvate Dehydrogenase Is Also
Insulin Also Regulates Lipogenesis
Regulated by Acyl-CoA
by Other Mechanisms
Acyl-CoA causes an inhibition of pyruvate dehydrogen-
Insulin stimulates lipogenesis by several other mecha-
ase by inhibiting the ATP-ADP exchange transporter of
nisms as well as by increasing acetyl-CoA carboxylase
the inner mitochondrial membrane, which leads to in-
activity. It increases the transport of glucose into the
creased intramitochondrial
[ATP]/[ADP] ratios and
cell (eg, in adipose tissue), increasing the availability of
therefore to conversion of active to inactive pyruvate
both pyruvate for fatty acid synthesis and glycerol
dehydrogenase (see Figure 17-6), thus regulating the
3-phosphate for esterification of the newly formed fatty
availability of acetyl-CoA for lipogenesis. Furthermore,
acids, and also converts the inactive form of pyruvate
oxidation of acyl-CoA due to increased levels of free
dehydrogenase to the active form in adipose tissue but
fatty acids may increase the ratios of
[acetyl-CoA]/
not in liver. Insulin also—by its ability to depress the
[CoA] and [NADH]/[NAD+] in mitochondria, inhibit-
level of intracellular cAMP—inhibits lipolysis in adi-
ing pyruvate dehydrogenase.
pose tissue and thereby reduces the concentration of
BIOSYNTHESIS OF FATTY ACIDS
/
179
plasma free fatty acids and therefore long-chain acyl-
• Acetyl-CoA carboxylase is required to convert acetyl-
CoA, an inhibitor of lipogenesis.
CoA to malonyl-CoA. In turn, fatty acid synthase, a
multienzyme complex of one polypeptide chain with
The Fatty Acid Synthase Complex
seven separate enzymatic activities, catalyzes the as-
sembly of palmitate from one acetyl-CoA and seven
& Acetyl-CoA Carboxylase Are
malonyl-CoA molecules.
Adaptive Enzymes
• Lipogenesis is regulated at the acetyl-CoA carboxy-
These enzymes adapt to the body’s physiologic needs
lase step by allosteric modifiers, phosphorylation/de-
by increasing in total amount in the fed state and by
phosphorylation, and induction and repression of en-
decreasing in starvation, feeding of fat, and in diabetes.
zyme synthesis. Citrate activates the enzyme, and
Insulin is an important hormone causing gene expres-
long-chain acyl-CoA inhibits its activity. Insulin acti-
sion and induction of enzyme biosynthesis, and
vates acetyl-CoA carboxylase whereas glucagon and
glucagon (via cAMP) antagonizes this effect. Feeding
epinephrine have opposite actions.
fats containing polyunsaturated fatty acids coordinately
regulates the inhibition of expression of key enzymes of
glycolysis and lipogenesis. These mechanisms for
REFERENCES
longer-term regulation of lipogenesis take several days
to become fully manifested and augment the direct and
Hudgins LC et al: Human fatty acid synthesis is stimulated by a
eucaloric low fat, high carbohydrate diet. J Clin Invest
immediate effect of free fatty acids and hormones such
1996;97:2081.
as insulin and glucagon.
Jump DB et al: Coordinate regulation of glycolytic and lipogenic
gene expression by polyunsaturated fatty acids. J Lipid Res
SUMMARY
1994;35:1076.
Kim KH: Regulation of mammalian acetyl-coenzyme A carboxy-
• The synthesis of long-chain fatty acids (lipogenesis) is
lase. Annu Rev Nutr 1997;17:77.
carried out by two enzyme systems: acetyl-CoA car-
Salati LM, Goodridge AG: Fatty acid synthesis in eukaryotes. In:
boxylase and fatty acid synthase.
Biochemistry of Lipids, Lipoproteins and Membranes. Vance
• The pathway converts acetyl-CoA to palmitate and
DE, Vance JE (editors). Elsevier, 1996.
requires NADPH, ATP, Mn2+, biotin, pantothenic
Wakil SJ: Fatty acid synthase, a proficient multifunctional enzyme.
acid, and HCO3− as cofactors.
Biochemistry 1989;28:4523.
Oxidation of Fatty Acids:
22
Ketogenesis
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
more water-soluble and exist as the un-ionized acid or
as a fatty acid anion.
Although fatty acids are both oxidized to acetyl-CoA
and synthesized from acetyl-CoA, fatty acid oxidation is
Fatty Acids Are Activated Before
not the simple reverse of fatty acid biosynthesis but an
Being Catabolized
entirely different process taking place in a separate com-
partment of the cell. The separation of fatty acid oxida-
Fatty acids must first be converted to an active interme-
tion in mitochondria from biosynthesis in the cytosol
diate before they can be catabolized. This is the only
allows each process to be individually controlled and
step in the complete degradation of a fatty acid that re-
integrated with tissue requirements. Each step in fatty
quires energy from ATP. In the presence of ATP and
acid oxidation involves acyl-CoA derivatives catalyzed
coenzyme A, the enzyme acyl-CoA synthetase (thioki-
by separate enzymes, utilizes NAD+ and FAD as coen-
nase) catalyzes the conversion of a fatty acid (or free
zymes, and generates ATP. It is an aerobic process, re-
fatty acid) to an “active fatty acid” or acyl-CoA, which
quiring the presence of oxygen.
uses one high-energy phosphate with the formation of
Increased fatty acid oxidation is a characteristic of
AMP and PPi (Figure 22-1). The PPi is hydrolyzed by
starvation and of diabetes mellitus, leading to ketone
inorganic pyrophosphatase with the loss of a further
body production by the liver (ketosis). Ketone bodies
high-energy phosphate, ensuring that the overall reac-
are acidic and when produced in excess over long peri-
tion goes to completion. Acyl-CoA synthetases are
ods, as in diabetes, cause ketoacidosis, which is ulti-
found in the endoplasmic reticulum, peroxisomes, and
mately fatal. Because gluconeogenesis is dependent
inside and on the outer membrane of mitochondria.
upon fatty acid oxidation, any impairment in fatty acid
oxidation leads to hypoglycemia. This occurs in vari-
ous states of carnitine deficiency or deficiency of es-
Long-Chain Fatty Acids Penetrate the
sential enzymes in fatty acid oxidation, eg, carnitine
Inner Mitochondrial Membrane as
palmitoyltransferase, or inhibition of fatty acid oxida-
Carnitine Derivatives
tion by poisons, eg, hypoglycin.
Carnitine
(β-hydroxy-γ-trimethylammonium buty-
rate), (CH3)3N+CH2CH(OH)CH2COO−, is
widely distributed and is particularly abundant in mus-
OXIDATION OF FATTY ACIDS OCCURS
cle. Long-chain acyl-CoA (or FFA) will not penetrate
IN MITOCHONDRIA
the inner membrane of mitochondria. However, car-
nitine palmitoyltransferase-I, present in the outer
Fatty Acids Are Transported in the
mitochondrial membrane, converts long-chain acyl-
Blood as Free Fatty Acids (FFA)
CoA to acylcarnitine, which is able to penetrate the
Free fatty acids—also called unesterified (UFA) or non-
inner membrane and gain access to the β-oxidation
esterified (NEFA) fatty acids—are fatty acids that are in
system of enzymes (Figure 22-1). Carnitine-acylcar-
the unesterified state. In plasma, longer-chain FFA are
nitine translocase acts as an inner membrane ex-
combined with albumin, and in the cell they are at-
change transporter. Acylcarnitine is transported in,
tached to a fatty acid-binding protein, so that in fact
coupled with the transport out of one molecule of car-
they are never really “free.” Shorter-chain fatty acids are
nitine. The acylcarnitine then reacts with CoA, cat-
180
OXIDATION OF FATTY ACIDS: KETOGENESIS
/
181
CoA SH
ATP
AMP + PP
i
H3C
α
+
CoA
FFA
Acyl-CoA
β
CO S CoA
Palmitoyl-CoA
CARNITINE
Acyl-CoA
PALMITOYL-
OUTER
H3C
α
SYNTHETASE
TRANSFERASE
MITOCHONDRIAL
I
MEMBRANE
β
CO
S CoA
+
CH3
CO S CoA
Acyl-CoA
CoA
Acetyl-CoA
Successive removal of acetyl-CoA (C2) units
Carnitine
Acylcarnitine
8 CH3
CO S CoA
Acetyl-CoA
CARNITINE
INNER
CARNITINE
ACYLCAR-
MITOCHONDRIAL
PALMITOYL-
NITINE
Figure 22-2. Overview of β-oxidation of fatty acids.
MEMBRANE
TRANSFERASE
TRANSLOCASE
II
The Cyclic Reaction Sequence Generates
CoA
FADH2 & NADH
Acylcarnitine
Carnitine
Several enzymes, known collectively as “fatty acid oxi-
dase,” are found in the mitochondrial matrix or inner
Acylcarnitine
Acyl-CoA
β-Oxidation
membrane adjacent to the respiratory chain. These cat-
alyze the oxidation of acyl-CoA to acetyl-CoA, the sys-
tem being coupled with the phosphorylation of ADP to
Figure 22-1. Role of carnitine in the transport of
ATP (Figure 22-3).
long-chain fatty acids through the inner mitochondrial
The first step is the removal of two hydrogen atoms
membrane. Long-chain acyl-CoA cannot pass through
from the 2(α)- and 3(β)-carbon atoms, catalyzed by
the inner mitochondrial membrane, but its metabolic
acyl-CoA dehydrogenase and requiring FAD. This re-
product, acylcarnitine, can.
sults in the formation of
∆2-trans-enoyl-CoA and
FADH2. The reoxidation of FADH2 by the respiratory
chain requires the mediation of another flavoprotein,
alyzed by carnitine palmitoyltransferase-II, located
termed electron-transferring flavoprotein
(Chapter
on the inside of the inner membrane. Acyl-CoA is re-
11). Water is added to saturate the double bond and
formed in the mitochondrial matrix, and carnitine is
form 3-hydroxyacyl-CoA, catalyzed by
2-enoyl-CoA
liberated.
hydratase. The 3-hydroxy derivative undergoes further
dehydrogenation on the 3-carbon catalyzed by L(+)-3-
hydroxyacyl-CoA dehydrogenase to form the corre-
-OXIDATION OF FATTY ACIDS
sponding
3-ketoacyl-CoA compound. In this case,
NAD+ is the coenzyme involved. Finally, 3-ketoacyl-
INVOLVES SUCCESSIVE CLEAVAGE
CoA is split at the 2,3- position by thiolase (3-keto-
WITH RELEASE OF ACETYL-CoA
acyl-CoA-thiolase), forming acetyl-CoA and a new acyl-
In β-oxidation (Figure 22-2), two carbons at a time are
CoA two carbons shorter than the original acyl-CoA
cleaved from acyl-CoA molecules, starting at the car-
molecule. The acyl-CoA formed in the cleavage reac-
boxyl end. The chain is broken between the α(2)- and
tion reenters the oxidative pathway at reaction 2 (Fig-
β(3)-carbon atoms—hence the name β-oxidation. The
ure 22-3). In this way, a long-chain fatty acid may be
two-carbon units formed are acetyl-CoA; thus, palmi-
degraded completely to acetyl-CoA (C2 units). Since
toyl-CoA forms eight acetyl-CoA molecules.
acetyl-CoA can be oxidized to CO2 and water via the
182
/
CHAPTER 22
O
Figure 22-3. β-Oxidation of fatty acids. Long-chain
R
3CH
2CH2
C O-
2
acyl-CoA is cycled through reactions 2-5, acetyl-CoA
Fatty acid
being split off, each cycle, by thiolase (reaction 5).
CoA SH
ATP
When the acyl radical is only four carbon atoms in
1
ACYL-CoA
Mg2+
length, two acetyl-CoA molecules are formed in reac-
SYNTHETASE
tion 5.
AMP + PP
i
O
R
3CH2
2CH
2
C
S CoA
citric acid cycle (which is also found within the mito-
Acyl-CoA
chondria), the complete oxidation of fatty acids is
achieved.
(outside)
C side
Oxidation of a Fatty Acid With an Odd
INNER MITOCHONDRIAL MEMBRANE
C CARNITINE TRANSPORTER
Number of Carbon Atoms Yields Acetyl-
M side
(inside)
CoA Plus a Molecule of Propionyl-CoA
Fatty acids with an odd number of carbon atoms are oxi-
O
dized by the pathway of β-oxidation, producing acetyl-
R
3CH2
2CH2
C
S CoA
CoA, until a three-carbon (propionyl-CoA) residue re-
mains. This compound is converted to succinyl-CoA, a
Acyl-CoA
constituent of the citric acid cycle (Figure 19-2). Hence,
FAD
the propionyl residue from an odd-chain fatty acid is
2
ACYL-CoA
the only part of a fatty acid that is glucogenic.
DEHYDROGENASE
2
P
FADH2
H2O
O
Respiratory
chain
3
Oxidation of Fatty Acids Produces
R
CH
2CH C
S CoA
a Large Quantity of ATP
∆2-trans-Enoyl-CoA
H2O
Transport in the respiratory chain of electrons from
∆2-ENOYL-CoA
FADH2
and NADH will lead to the synthesis of five
3
HYDRATASE
high-energy phosphates (Chapter 12) for each of the
first seven acetyl-CoA molecules formed by β-oxidation
OH
O
3
of palmitate (7 × 5 = 35). A total of 8 mol of acetyl-
R CH
2CH
2
C
S CoA
CoA is formed, and each will give rise to 12 mol of
L(+)-3-Hydroxy-
acyl-CoA
ATP on oxidation in the citric acid cycle, making 8 ×
12 = 96 mol. Two must be subtracted for the initial ac-
NAD+
tivation of the fatty acid, yielding a net gain of 129 mol
L(+)-3-HYDROXYACYL-
4
CoA DEHYDROGENASE
of ATP per mole of palmitate, or 129 × 51.6* = 6656
3
P
kJ. This represents 68% of the free energy of combus-
NADH + H+
H2O
Respiratory
tion of palmitic acid.
O
O
chain
R
3C
2CH2
C
S CoA
3-Ketoacyl-CoA
Peroxisomes Oxidize Very Long
CoA SH
Chain Fatty Acids
5
THIOLASE
A modified form of β-oxidation is found in peroxi-
somes and leads to the formation of acetyl-CoA and
O
O
H2O2 (from the flavoprotein-linked dehydrogenase
R C
S
CoA +CH
3
C
S CoA
step), which is broken down by catalase. Thus, this de-
Acyl-CoA
Acetyl-CoA
hydrogenation in peroxisomes is not linked directly to
phosphorylation and the generation of ATP. The sys-
tem facilitates the oxidation of very long chain fatty
Citric
acid
acids
(eg, C20, C22). These enzymes are induced by
cycle
2CO2
* ∆G for the ATP reaction, as explained in Chapter 17.
OXIDATION OF FATTY ACIDS: KETOGENESIS
/
183
O
Figure 22-4. Sequence of reactions in the oxidation
cis
cis
of unsaturated fatty acids, eg, linoleic acid. ∆4-cis-fatty
12
9
C
S CoA
acids or fatty acids forming ∆4-cis-enoyl-CoA enter the
pathway at the position shown. NADPH for the dienoyl-
Linoleyl-CoA
CoA reductase step is supplied by intramitochondrial
3 Cycles of
sources such as glutamate dehydrogenase, isocitrate
β-oxidation
3 Acetyl-CoA
dehydrogenase, and NAD(P)H transhydrogenase.
O
cis
cis
6
3
C
S CoA
high-fat diets and in some species by hypolipidemic
drugs such as clofibrate.
The enzymes in peroxisomes do not attack shorter-
∆3-cis-∆6-cis-Dienoyl-CoA
chain fatty acids; the β-oxidation sequence ends at oc-
∆3-cis (or trans) → ∆2-trans-ENOYL-CoA
tanoyl-CoA. Octanoyl and acetyl groups are both further
ISOMERASE
oxidized in mitochondria. Another role of peroxisomal
β-oxidation is to shorten the side chain of cholesterol in
cis
6
2
bile acid formation (Chapter 26). Peroxisomes also take
part in the synthesis of ether glycerolipids (Chapter 24),
C S CoA
cholesterol, and dolichol (Figure 26-2).
O
∆2-trans-∆6-cis-Dienoyl-CoA
OXIDATION OF UNSATURATED FATTY
(∆2-trans-Enoyl-CoA stage of β-oxidation)
ACIDS OCCURS BY A MODIFIED
1 Cycle of
-OXIDATION PATHWAY
β-oxidation
Acetyl-CoA
The CoA esters of these acids are degraded by the en-
cis
zymes normally responsible for β-oxidation until either
4
2
a ∆3-cis-acyl-CoA compound or a ∆4-cis-acyl-CoA com-
C S CoA
pound is formed, depending upon the position of the
ACYL-CoA
DEHYDROGENASE
double bonds (Figure 22-4). The former compound is
O
isomerized (
3cis v
2-trans-enoyl-CoA isomerase)
∆2-trans-∆4-cis- Dienoyl-CoA
∆4-cis-Enoyl-CoA
to the corresponding ∆2-trans-CoA stage of β-oxidation
H+ + NADPH
for subsequent hydration and oxidation. Any ∆4-cis-acyl-
∆2-trans-∆4-cis-DIENOYL-CoA
REDUCTASE
CoA either remaining, as in the case of linoleic acid, or
NADP+
entering the pathway at this point after conversion by
acyl-CoA dehydrogenase to
∆2-trans-∆4-cis-dienoyl-
O
CoA, is then metabolized as indicated in Figure 22-4.
3
C S CoA
KETOGENESIS OCCURS WHEN THERE
IS A HIGH RATE OF FATTY ACID
∆3-trans-Enoyl-CoA
OXIDATION IN THE LIVER
∆3-cis (or trans) → ∆2-trans-ENOYL-CoA
Under metabolic conditions associated with a high rate
ISOMERASE
of fatty acid oxidation, the liver produces considerable
quantities of acetoacetate and D(
)-3-hydroxybutyrate
(β-hydroxybutyrate). Acetoacetate continually under-
O
goes spontaneous decarboxylation to yield acetone.
C S CoA
These three substances are collectively known as the ke-
2
tone bodies (also called acetone bodies or [incorrectly*]
“ketones”) (Figure 22-5). Acetoacetate and 3-hydroxybu-
∆2-trans-Enoyl-CoA
4 Cycles of
β-oxidation
* The term “ketones” should not be used because 3-hydroxybu-
tyrate is not a ketone and there are ketones in blood that are not
ketone bodies, eg, pyruvate, fructose.
5 Acetyl-CoA
184
/
CHAPTER 22
O
tyrate are interconverted by the mitochondrial enzyme
D(
)-3-hydroxybutyrate dehydrogenase; the equi-
CH3
C
CH3
librium is controlled by the mitochondrial [NAD+]/
Acetone
CO2
[NADH] ratio, ie, the redox state. The concentration
of total ketone bodies in the blood of well-fed mam-
mals does not normally exceed 0.2 mmol/L except in
ruminants, where 3-hydroxybutyrate is formed contin-
O
uously from butyric acid (a product of ruminal fermen-
tation) in the rumen wall. In vivo, the liver appears to
CH3
C
CH2
COO-
be the only organ in nonruminants to add significant
Acetoacetate
quantities of ketone bodies to the blood. Extrahepatic
D(-)-3-HYDROXYBUTYRATE
tissues utilize them as respiratory substrates. The net
DEHYDROGENASE
NADH + H+
flow of ketone bodies from the liver to the extrahepatic
tissues results from active hepatic synthesis coupled
with very low utilization. The reverse situation occurs
OH
NAD+
in extrahepatic tissues (Figure 22-6).
CH3
CH
CH2
COO-
D(-)-3-Hydroxybutyrate
3-Hydroxy-3-Methylglutaryl-CoA
(HMG-CoA) Is an Intermediate
Figure 22-5. Interrelationships of the ketone bod-
in the Pathway of Ketogenesis
ies. D(−)-3-hydroxybutyrate dehydrogenase is a mito-
chondrial enzyme.
Enzymes responsible for ketone body formation are as-
sociated mainly with the mitochondria. Two acetyl-
CoA molecules formed in β-oxidation condense with
one another to form acetoacetyl-CoA by a reversal of
the thiolase reaction. Acetoacetyl-CoA, which is the
EXTRAHEPATIC
LIVER
BLOOD
TISSUES
Acyl-CoA
FFA
Glucose
Glucose
Acyl-CoA
URINE
Acetyl-CoA
Acetyl-CoA
Ketone
Ketone
Ketone bodies
bodies
bodies
Acetone
Citric
Citric
acid
acid
cycle
cycle
LUNGS
2CO2
2CO2
Figure 22-6. Formation, utilization, and excretion of ketone bodies. (The main
pathway is indicated by the solid arrows.)
OXIDATION OF FATTY ACIDS: KETOGENESIS
/
185
starting material for ketogenesis, also arises directly
predominant ketone body present in the blood and
from the terminal four carbons of a fatty acid during
urine in ketosis.
β-oxidation
(Figure
22-7). Condensation of ace-
toacetyl-CoA with another molecule of acetyl-CoA
Ketone Bodies Serve as a Fuel
by 3-hydroxy-3-methylglutaryl-CoA synthase forms
for Extrahepatic Tissues
HMG-CoA. 3-Hydroxy-3-methylglutaryl-CoA lyase
then causes acetyl-CoA to split off from the HMG-
While an active enzymatic mechanism produces ace-
CoA, leaving free acetoacetate. The carbon atoms split
toacetate from acetoacetyl-CoA in the liver, acetoac-
off in the acetyl-CoA molecule are derived from the
etate once formed cannot be reactivated directly except
original acetoacetyl-CoA molecule. Both enzymes
in the cytosol, where it is used in a much less active
must be present in mitochondria for ketogenesis to
pathway as a precursor in cholesterol synthesis. This ac-
take place. This occurs solely in liver and rumen ep-
counts for the net production of ketone bodies by the
ithelium. D(−)-3-Hydroxybutyrate is quantitatively the
liver.
FFA
ATP
CoA
ACYL-CoA
SYNTHETASE
Esterification
Triacylglycerol
Acyl-CoA
Phospholipid
β-Oxidation
(Acetyl-CoA)
n
O
O
CH3
C
2
CH C S CoA
Acetoacetyl-CoA
HMG-CoA
SYNTHASE
CoA SH
THIOLASE
OH
O
H2O
CH3
C
CH
2
C
S CoA
O
*CH2
*COO-
*CH3
*C
S CoA
CoA SH
3-Hydroxy-3-methyl-
Acetyl-CoA
glutaryl-CoA (HMG-CoA)
HMG-CoA
CH
CO S CoA
3
LYASE
Acetyl-CoA
Citric
O
acid
cycle
CH3
C
*COO-
*CH2
Acetoacetate
2CO2
NADH + H+
D(-)-3-HYDROXYBUTYRATE
DEHYDROGENASE
NAD+
OH
CH3
CH
*CH2
*COO-
D(-)-3-Hydroxybutyrate
Figure 22-7.
Pathways of ketogenesis in the liver. (FFA, free fatty acids; HMG, 3-hy-
droxy-3-methylglutaryl.)
186
/
CHAPTER 22
In extrahepatic tissues, acetoacetate is activated to
ketonemia, not the ketonuria, is the preferred method
acetoacetyl-CoA by succinyl-CoA-acetoacetate CoA
of assessing the severity of ketosis.
transferase. CoA is transferred from succinyl-CoA to
form acetoacetyl-CoA (Figure 22-8). The acetoacetyl-
KETOGENESIS IS REGULATED
CoA is split to acetyl-CoA by thiolase and oxidized in
AT THREE CRUCIAL STEPS
the citric acid cycle. If the blood level is raised, oxida-
tion of ketone bodies increases until, at a concentration
(1) Ketosis does not occur in vivo unless there is an
of approximately 12 mmol/L, they saturate the oxida-
increase in the level of circulating free fatty acids that
tive machinery. When this occurs, a large proportion of
arise from lipolysis of triacylglycerol in adipose tissue.
the oxygen consumption may be accounted for by the
Free fatty acids are the precursors of ketone bodies
oxidation of ketone bodies.
in the liver. The liver, both in fed and in fasting condi-
In most cases, ketonemia is due to increased pro-
tions, extracts about 30% of the free fatty acids passing
duction of ketone bodies by the liver rather than to a
through it, so that at high concentrations the flux pass-
deficiency in their utilization by extrahepatic tissues.
ing into the liver is substantial. Therefore, the factors
While acetoacetate and D(−)-3-hydroxybutyrate are
regulating mobilization of free fatty acids from adi-
readily oxidized by extrahepatic tissues, acetone is diffi-
pose tissue are important in controlling ketogenesis
cult to oxidize in vivo and to a large extent is volatilized
(Figures 22-9 and 25-8).
in the lungs.
(2) After uptake by the liver, free fatty acids are ei-
In moderate ketonemia, the loss of ketone bodies via
ther
-oxidized to CO2 or ketone bodies or esterified
the urine is only a few percent of the total ketone body
to triacylglycerol and phospholipid. There is regulation
production and utilization. Since there are renal thresh-
of entry of fatty acids into the oxidative pathway by car-
old-like effects (there is not a true threshold) that vary
nitine palmitoyltransferase-I (CPT-I), and the remain-
between species and individuals, measurement of the
der of the fatty acid uptake is esterified. CPT-I activity is
EXTRAHEPATIC TISSUES
eg, MUSCLE
FFA
Acyl-CoA
Acetyl-CoA
LIVER
β-Oxidation
THIOLASE
Acetyl-CoA
Acetoacetyl-CoA
Succinate
OAA
CoA
Citric acid cycle
HMG-CoA
TRANSFERASE
Succinyl-
Citrate
CoA
2CO2
Acetoacetate
Acetoacetate
NADH + H+
NADH
+ H+
NAD+
NAD+
3-Hydroxybutyrate
3-Hydroxybutyrate
Figure 22-8. Transport of ketone bodies from the liver and pathways of utilization and oxidation in extrahe-
patic tissues.
OXIDATION OF FATTY ACIDS: KETOGENESIS
/
187
Triacylglycerol
(3) In turn, the acetyl-CoA formed in β-oxidation is
oxidized in the citric acid cycle, or it enters the pathway
of ketogenesis to form ketone bodies. As the level of
ADIPOSE TISSUE
1
serum free fatty acids is raised, proportionately more
Lipolysis
free fatty acid is converted to ketone bodies and less is
oxidized via the citric acid cycle to CO2. The partition
FFA
of acetyl-CoA between the ketogenic pathway and the
is so regulated that the
pathway of oxidation to CO2
BLOOD
total free energy captured in ATP which results from
the oxidation of free fatty acids remains constant. This
FFA
may be appreciated when it is realized that complete
oxidation of 1 mol of palmitate involves a net produc-
LIVER
tion of 129 mol of ATP via β-oxidation and CO2 pro-
duction in the citric acid cycle (see above), whereas only
33 mol of ATP are produced when acetoacetate is the
Acyl-CoA
CPT-I
end product and only 21 mol when 3-hydroxybutyrate
gateway
Esterification
is the end product. Thus, ketogenesis may be regarded
2
as a mechanism that allows the liver to oxidize increas-
β-Oxidation
ing quantities of fatty acids within the constraints of a
tightly coupled system of oxidative phosphorylation—
Acylglycerols
without increasing its total energy expenditure.
Acetyl-CoA
Theoretically, a fall in concentration of oxaloacetate,
Citric acid
3
cycle
particularly within the mitochondria, could impair the
Ketogenesis
ability of the citric acid cycle to metabolize acetyl-CoA
and divert fatty acid oxidation toward ketogenesis.
CO2
Such a fall may occur because of an increase in the
[NADH]/[NAD+] ratio caused by increased β-oxida-
Ketone bodies
tion affecting the equilibrium between oxaloacetate and
malate and decreasing the concentration of oxaloac-
Figure 22-9.
Regulation of ketogenesis. 1 - 3
show
etate. However, pyruvate carboxylase, which catalyzes
three crucial steps in the pathway of metabolism of free
the conversion of pyruvate to oxaloacetate, is activated
fatty acids (FFA) that determine the magnitude of keto-
by acetyl-CoA. Consequently, when there are signifi-
genesis. (CPT-I, carnitine palmitoyltransferase-I.)
cant amounts of acetyl-CoA, there should be sufficient
oxaloacetate to initiate the condensing reaction of the
citric acid cycle.
low in the fed state, leading to depression of fatty acid
oxidation, and high in starvation, allowing fatty acid ox-
CLINICAL ASPECTS
idation to increase. Malonyl-CoA, the initial intermedi-
ate in fatty acid biosynthesis (Figure 21-1), formed by
Impaired Oxidation of Fatty Acids
acetyl-CoA carboxylase in the fed state, is a potent in-
Gives Rise to Diseases Often
hibitor of CPT-I (Figure 22-10). Under these condi-
Associated With Hypoglycemia
tions, free fatty acids enter the liver cell in low concen-
trations and are nearly all esterified to acylglycerols and
Carnitine deficiency can occur particularly in the new-
transported out of the liver in very low density lipopro-
born—and especially in preterm infants—owing to in-
teins (VLDL). However, as the concentration of free
adequate biosynthesis or renal leakage. Losses can also
fatty acids increases with the onset of starvation, acetyl-
occur in hemodialysis. This suggests a vitamin-like di-
CoA carboxylase is inhibited directly by acyl-CoA, and
etary requirement for carnitine in some individuals.
[malonyl-CoA] decreases, releasing the inhibition of
Symptoms of deficiency include hypoglycemia, which
CPT-I and allowing more acyl-CoA to be β-oxidized.
is a consequence of impaired fatty acid oxidation and
These events are reinforced in starvation by decrease in
lipid accumulation with muscular weakness. Treatment
the [insulin]/[glucagon] ratio. Thus, β-oxidation from
is by oral supplementation with carnitine.
free fatty acids is controlled by the CPT-I gateway into
Inherited CPT-I deficiency affects only the liver,
the mitochondria, and the balance of the free fatty acid
resulting in reduced fatty acid oxidation and ketogene-
uptake not oxidized is esterified.
sis, with hypoglycemia. CPT-II deficiency affects pri-
188
/
CHAPTER 22
Glucose
FFA
VLDL
BLOOD
LIVER
Acetyl-CoA
Acylglycerols
Insulin
+
−
Acyl-CoA
Lipogenesis
ACETYL-CoA
CARBOXYLASE
Cytosol
−
Glucagon
Malonyl-CoA
CARNITINE
−
PALMITOYL-
TRANSFERASE I
Palmitate
Figure 22-10. Regulation of
Acyl-CoA
long-chain fatty acid oxidation in the
Mitochondrion
β-Oxidation
liver. (FFA, free fatty acids; VLDL,
very low density lipoprotein.) Posi-
Acetyl-CoA
tive ( + ) and negative ( − ) regulatory
effects are represented by broken
CO2
arrows and substrate flow by solid
Ketone bodies
arrows.
marily skeletal muscle and, when severe, the liver. The
syndrome occurs in individuals with a rare inherited
sulfonylurea drugs
(glyburide
[glibenclamide] and
absence of peroxisomes in all tissues. They accumulate
tolbutamide), used in the treatment of type 2 diabetes
C26-C38 polyenoic acids in brain tissue and also exhibit
mellitus, reduce fatty acid oxidation and, therefore, hy-
a generalized loss of peroxisomal functions, eg, im-
perglycemia by inhibiting CPT-I.
paired bile acid and ether lipid synthesis.
Inherited defects in the enzymes of β-oxidation and
ketogenesis also lead to nonketotic hypoglycemia, coma,
Ketoacidosis Results From
and fatty liver. Defects are known in long- and short-
Prolonged Ketosis
chain 3-hydroxyacyl-CoA dehydrogenase (deficiency of
the long-chain enzyme may be a cause of acute fatty
Higher than normal quantities of ketone bodies present
liver of pregnancy).
3-Ketoacyl-CoA thiolase and
in the blood or urine constitute ketonemia (hyperke-
HMG-CoA lyase deficiency also affect the degradation
tonemia) or ketonuria, respectively. The overall condi-
of leucine, a ketogenic amino acid (Chapter 30).
tion is called ketosis. Acetoacetic and 3-hydroxybutyric
Jamaican vomiting sickness is caused by eating the
acids are both moderately strong acids and are buffered
unripe fruit of the akee tree, which contains a toxin,
when present in blood or other tissues. However, their
hypoglycin, that inactivates medium- and short-chain
continual excretion in quantity progressively depletes
acyl-CoA dehydrogenase, inhibiting β-oxidation and
the alkali reserve, causing ketoacidosis. This may be
causing hypoglycemia. Dicarboxylic aciduria is char-
fatal in uncontrolled diabetes mellitus.
acterized by the excretion of C6-C10 ω-dicarboxylic
The basic form of ketosis occurs in starvation and
acids and by nonketotic hypoglycemia. It is caused by a
involves depletion of available carbohydrate coupled
lack of mitochondrial medium-chain acyl-CoA dehy-
with mobilization of free fatty acids. This general pat-
drogenase. Refsum’s disease is a rare neurologic disor-
tern of metabolism is exaggerated to produce the patho-
der due to a defect that causes the accumulation of phy-
logic states found in diabetes mellitus, twin lamb dis-
tanic acid, which is found in plant foodstuffs and
ease, and ketosis in lactating cattle. Nonpathologic
blocks β-oxidation. Zellweger’s (cerebrohepatorenal)
forms of ketosis are found under conditions of high-fat
OXIDATION OF FATTY ACIDS: KETOGENESIS
/
189
feeding and after severe exercise in the postabsorptive
• Diseases associated with impairment of fatty acid oxi-
state.
dation lead to hypoglycemia, fatty infiltration of or-
gans, and hypoketonemia.
SUMMARY
• Ketosis is mild in starvation but severe in diabetes
mellitus and ruminant ketosis.
• Fatty acid oxidation in mitochondria leads to the gen-
eration of large quantities of ATP by a process called
β-oxidation that cleaves acetyl-CoA units sequentially
from fatty acyl chains. The acetyl-CoA is oxidized in
REFERENCES
the citric acid cycle, generating further ATP.
Eaton S, Bartlett K, Pourfarzam M: Mammalian mitochondrial β-
• The ketone bodies (acetoacetate, 3-hydroxybutyrate,
oxidation. Biochem J 1996;320:345.
and acetone) are formed in hepatic mitochondria
Mayes PA, Laker ME: Regulation of ketogenesis in the liver.
when there is a high rate of fatty acid oxidation. The
Biochem Soc Trans 1981;9:339.
pathway of ketogenesis involves synthesis and break-
McGarry JD, Foster DW: Regulation of hepatic fatty acid oxida-
down of 3-hydroxy-3-methylglutaryl-CoA (HMG-
tion and ketone body production. Annu Rev Biochem
CoA) by two key enzymes, HMG-CoA synthase and
1980;49:395.
HMG-CoA lyase.
Osmundsen H, Hovik R: β-Oxidation of polyunsaturated fatty
acids. Biochem Soc Trans 1988;16:420.
• Ketone bodies are important fuels in extrahepatic tis-
Reddy JK, Mannaerts GP: Peroxisomal lipid metabolism. Annu
sues.
Rev Nutr 1994;14:343.
• Ketogenesis is regulated at three crucial steps:
(1)
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
control of free fatty acid mobilization from adipose
herited Disease, 8th ed. McGraw-Hill, 2001.
tissue;
(2) the activity of carnitine palmitoyltrans-
Treem WR et al: Acute fatty liver of pregnancy and long-chain 3-
ferase-I in liver, which determines the proportion of
hydroxyacyl-coenzyme A dehydrogenase deficiency. Hepatol-
the fatty acid flux that is oxidized rather than esteri-
ogy 1994;19:339.
fied; and (3) partition of acetyl-CoA between the
Wood PA: Defects in mitochondrial beta-oxidation of fatty acids.
pathway of ketogenesis and the citric acid cycle.
Curr Opin Lipidol 1999;10:107.
Metabolism of Unsaturated Fatty
23
Acids & Eicosanoids
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
duced at the ∆4, ∆5, ∆6, and ∆9 positions (see Chapter
14) in most animals, but never beyond the ∆9 position.
Unsaturated fatty acids in phospholipids of the cell
In contrast, plants are able to synthesize the nutrition-
membrane are important in maintaining membrane
ally essential fatty acids by introducing double bonds at
fluidity. A high ratio of polyunsaturated fatty acids to
the ∆12
and ∆15 positions.
saturated fatty acids (P:S ratio) in the diet is a major
factor in lowering plasma cholesterol concentrations
and is considered to be beneficial in preventing coro-
nary heart disease. Animal tissues have limited capacity
16
9
COOH
for desaturating fatty acids, and that process requires
Palmitoleic acid (ω7, 16:1, ∆9)
certain dietary polyunsaturated fatty acids derived
from plants. These essential fatty acids are used to
form eicosanoic (C20) fatty acids, which in turn give
18
9
COOH
rise to the prostaglandins and thromboxanes and to
Oleic acid (ω 9, 18:1, ∆9)
leukotrienes and lipoxins—known collectively as
eicosanoids. The prostaglandins and thromboxanes are
12
9
COOH
local hormones that are synthesized rapidly when re-
quired. Prostaglandins mediate inflammation, produce
18
*Linoleic acid (ω 6, 18:2, ∆9,12)
pain, and induce sleep as well as being involved in the
regulation of blood coagulation and reproduction.
Nonsteroidal anti-inflammatory drugs such as aspirin
18
15
12
9
COOH
act by inhibiting prostaglandin synthesis. Leukotrienes
have muscle contractant and chemotactic properties
*α-Linolenic acid (ω 3, 18:3, ∆9,12,15)
and are important in allergic reactions and inflamma-
14
11
8
5
COOH
tion.
20
*Arachidonic acid (ω 6, 20:4, ∆5,8,11,14)
SOME POLYUNSATURATED FATTY
ACIDS CANNOT BE SYNTHESIZED
BY MAMMALS & ARE
20
17
14
11
8
5
COOH
NUTRITIONALLY ESSENTIAL
Eicosapentaenoic acid (ω 3, 20:5, ∆5,8,11,14,17)
Certain long-chain unsaturated fatty acids of metabolic
Figure 23-1. Structure of some unsaturated fatty
significance in mammals are shown in Figure 23-1.
acids. Although the carbon atoms in the molecules are
Other C20, C22, and C24 polyenoic fatty acids may be
conventionally numbered—ie, numbered from the car-
derived from oleic, linoleic, and α-linolenic acids by
boxyl terminal—the ω numbers (eg, ω7 in palmitoleic
chain elongation. Palmitoleic and oleic acids are not es-
acid) are calculated from the reverse end (the methyl
sential in the diet because the tissues can introduce a
double bond at the ∆9 position of a saturated fatty acid.
terminal) of the molecules. The information in paren-
Linoleic and
-linolenic acids are the only fatty acids
theses shows, for instance, that α-linolenic acid con-
known to be essential for the complete nutrition of
tains double bonds starting at the third carbon from
many species of animals, including humans, and are
the methyl terminal, has 18 carbons and 3 double
known as the nutritionally essential fatty acids. In
bonds, and has these double bonds at the 9th, 12th,
most mammals, arachidonic acid can be formed from
and 15th carbons from the carboxyl terminal. (Asterisks:
linoleic acid (Figure 23-4). Double bonds can be intro-
Classified as “essential fatty acids.”)
190
METABOLISM OF UNSATURATED FATTY ACIDS & EICOSANOIDS
/
191
Stearoyl CoA
are able to synthesize the ω9 (oleic acid) family of unsat-
urated fatty acids completely by a combination of chain
O2 + NADH + H+
elongation and desaturation (Figure 23-3). However, as
indicated above, linoleic (ω6) or α-linolenic (ω3) acids
∆9
DESATURASE
Cyt b5
required for the synthesis of the other members of the
ω6 or ω3 families must be supplied in the diet.
NAD++ 2H2O
Linoleate may be converted to arachidonate via
-
Oleoyl CoA
linolenate by the pathway shown in Figure 23-4. The
nutritional requirement for arachidonate may thus be
Figure 23-2. Microsomal ∆9 desaturase.
dispensed with if there is adequate linoleate in the diet.
The desaturation and chain elongation system is greatly
diminished in the starving state, in response to glucagon
MONOUNSATURATED FATTY
and epinephrine administration, and in the absence of
ACIDS ARE SYNTHESIZED BY
insulin as in type 1 diabetes mellitus.
A
9 DESATURASE SYSTEM
Several tissues including the liver are considered to be re-
DEFICIENCY SYMPTOMS ARE PRODUCED
sponsible for the formation of nonessential monounsatu-
WHEN THE ESSENTIAL FATTY ACIDS
rated fatty acids from saturated fatty acids. The first dou-
(EFA) ARE ABSENT FROM THE DIET
ble bond introduced into a saturated fatty acid is nearly
always in the ∆9 position. An enzyme system—
9 desat-
Rats fed a purified nonlipid diet containing vitamins A
urase (Figure 23-2)—in the endoplasmic reticulum will
and D exhibit a reduced growth rate and reproductive
catalyze the conversion of palmitoyl-CoA or stearoyl-CoA
deficiency which may be cured by the addition of
to palmitoleoyl-CoA or oleoyl-CoA, respectively. Oxygen
linoleic,
-linolenic, and arachidonic acids to the diet.
and either NADH or NADPH are necessary for the reac-
These fatty acids are found in high concentrations in
tion. The enzymes appear to be similar to a monooxyge-
vegetable oils (Table 14-2) and in small amounts in ani-
nase system involving cytochrome b5
(Chapter 11).
mal carcasses. These essential fatty acids are required for
prostaglandin, thromboxane, leukotriene, and lipoxin
SYNTHESIS OF POLYUNSATURATED
formation (see below), and they also have various other
functions which are less well defined. Essential fatty acids
FATTY ACIDS INVOLVES DESATURASE
are found in the structural lipids of the cell, often in the
& ELONGASE ENZYME SYSTEMS
2 position of phospholipids, and are concerned with the
Additional double bonds introduced into existing mo-
structural integrity of the mitochondrial membrane.
nounsaturated fatty acids are always separated from each
Arachidonic acid is present in membranes and ac-
other by a methylene group (methylene interrupted) ex-
counts for 5-15% of the fatty acids in phospholipids.
cept in bacteria. Since animals have a ∆9 desaturase, they
Docosahexaenoic acid (DHA; ω3, 22:6), which is syn-
ω9
2
1
3
1
4
Oleic acid
18:2
20:2
20:3
22:3
22:4
Family
18:1
Accumulates in essential
1
fatty acid deficiency
1
1
—
24:1
22:1
20:1
2
1
3
1
4
ω6
Linoleic acid
18:3
20:3
20:4
22:4
22:5
Family
18:2
1
—
20:2
ω3
2
1
3
1
4
α-Linolenic
18:3
20:3
20:4
22:4
22:5
Family
acid
18:3
Figure 23-3. Biosynthesis of the ω9, ω6, and ω3 families of polyunsaturated fatty
acids. Each step is catalyzed by the microsomal chain elongation or desaturase sys-
tem: 1, elongase; 2, ∆6 desaturase; 3, ∆5 desaturase; 4, ∆4 desaturase. (
, Inhibition.)
—
192
/
CHAPTER 23
O
fatty acids in phospholipids, other complex lipids, and
12
9
membranes, particularly with ∆5,8,11-eicosatrienoic acid
C
S CoA
(ω9 20:3) (Figure 23-3). The triene:tetraene ratio in
18
plasma lipids can be used to diagnose the extent of es-
Linoleoyl-CoA (∆9,12-octadecadienoyl-CoA)
sential fatty acid deficiency.
+
O2 + NADH + H
Trans Fatty Acids Are Implicated
in Various Disorders
∆6
DESATURASE
Small amounts of trans-unsaturated fatty acids are found
+
2H2O + NAD
in ruminant fat (eg, butter fat has 2-7%), where they
arise from the action of microorganisms in the rumen,
12
9
6
but the main source in the human diet is from partially
hydrogenated vegetable oils (eg, margarine). Trans fatty
18
C
S CoA
acids compete with essential fatty acids and may exacer-
O
bate essential fatty acid deficiency. Moreover, they are
γ-Linolenoyl-CoA (∆6,9,12-octadecatrienoyl-CoA)
structurally similar to saturated fatty acids (Chapter 14)
and have comparable effects in the promotion of hyper-
C2
cholesterolemia and atherosclerosis (Chapter 26).
(Malonyl-CoA,
MICROSOMAL CHAIN
NADPH)
ELONGATION SYSTEM
(ELONGASE)
EICOSANOIDS ARE FORMED FROM C20
POLYUNSATURATED FATTY ACIDS
Arachidonate and some other C20 polyunsaturated fatty
14
11
8
acids give rise to eicosanoids, physiologically and phar-
20
C S CoA
macologically active compounds known as prosta-
glandins
(PG), thromboxanes
(TX), leukotrienes
O
(LT), and lipoxins (LX) (Chapter 14). Physiologically,
they are considered to act as local hormones function-
Dihomo-γ-linolenoyl-CoA (∆8,11,14-eicosatrienoyl-CoA)
ing through G-protein-linked receptors to elicit their
+
O2
+ NADH + H
biochemical effects.
There are three groups of eicosanoids that are syn-
∆5
DESATURASE
thesized from C20 eicosanoic acids derived from the es-
sential fatty acids linoleate and
-linolenate, or di-
2H2O + NAD+
rectly from dietary arachidonate and eicosapentaenoate
O
(Figure 23-5). Arachidonate, usually derived from the
14
11
8
5
2 position of phospholipids in the plasma membrane by
C
S CoA
the action of phospholipase A2 (Figure 24-6)—but also
20
from the diet—is the substrate for the synthesis of the
PG2, TX2 series (prostanoids) by the cyclooxygenase
Arachidonoyl-CoA (∆5,8,11,14-eicosatetraenoyl-CoA)
pathway, or the LT4 and LX4 series by the lipoxyge-
Figure 23-4. Conversion of linoleate to arachido-
nase pathway, with the two pathways competing for
nate. Cats cannot carry out this conversion owing to ab-
the arachidonate substrate (Figure 23-5).
sence of ∆6 desaturase and must obtain arachidonate in
their diet.
THE CYCLOOXYGENASE
PATHWAY IS RESPONSIBLE FOR
PROSTANOID SYNTHESIS
thesized from α-linolenic acid or obtained directly from
fish oils, is present in high concentrations in retina,
Prostanoid synthesis (Figure 23-6) involves the con-
cerebral cortex, testis, and sperm. DHA is particularly
sumption of two molecules of O2
catalyzed by
needed for development of the brain and retina and is
prostaglandin H synthase (PGHS), which consists of
supplied via the placenta and milk. Patients with retini-
two enzymes, cyclooxygenase and peroxidase. PGHS
tis pigmentosa are reported to have low blood levels of
is present as two isoenzymes, PGHS-1 and PGHS-2.
DHA. In essential fatty acid deficiency, nonessential
The product, an endoperoxide (PGH), is converted to
polyenoic acids of the ω9 family replace the essential
prostaglandins D, E, and F as well as to a thromboxane
METABOLISM OF UNSATURATED FATTY ACIDS & EICOSANOIDS
/
193
Diet
Membrane phospholipid
PHOSPHOLIPASE
Angiotensin II
Linoleate
+
A2
Bradykinin
Epinephrine
Thrombin
-2H
γ-Linolenate
GROUP 1
Diet
GROUP 2
Prostanoids
Prostanoids
+2C
PGE1
PGD2
PGF1
PGE2
1
1
TXA1
PGF2
COOH
COOH
-2H
PGI2
TXA2
Leukotrienes
Leukotrienes
Lipoxins
LTA3
8,11,14-Eicosatrienoate
5,8,11,14-
LTA4
LXA4
2
LTC3
(dihomo γ-linolenate)
Eicosatetraenoate
2
LTB4
LXB4
LTD3
LTC4
LXC4
Arachidonate
LTD4
LXD4
LTE4
LXE4
GROUP 3
Prostanoids
PGD3
PGE3
1
PGF3
COOH
-2H
PGI
3
Eicosatetraenoate
TXA3
Leukotrienes
+2C
5,8,11,14,17-
LTA5
Eicosapentaenoate
2
LTB5
LTC5
Octadecatetraenoate
-2H
Diet
α-Linolenate
Diet
Figure 23-5. The three groups of eicosanoids and their biosynthetic origins. (PG, prostaglandin; PGI, prosta-
cyclin; TX, thromboxane; LT, leukotriene; LX, lipoxin; 1 , cyclooxygenase pathway; 2 , lipoxygenase pathway.)
The subscript denotes the total number of double bonds in the molecule and the series to which the compound
belongs.
(TXA2) and prostacyclin (PGI2). Each cell type pro-
Essential Fatty Acids Do Not Exert
duces only one type of prostanoid. Aspirin, a nons-
All Their Physiologic Effects Via
teroidal anti-inflammatory drug (NSAID), inhibits cy-
Prostaglandin Synthesis
clooxygenase of both PGHS-1 and PGHS-2 by
acetylation. Most other NSAIDs, such as indomethacin
The role of essential fatty acids in membrane formation
and ibuprofen, inhibit cyclooxygenases by competing
is unrelated to prostaglandin formation. Prostaglandins
with arachidonate. Transcription of PGHS-2—but not
do not relieve symptoms of essential fatty acid defi-
of PGHS-1—is completely inhibited by anti-inflam-
ciency, and an essential fatty acid deficiency is not
matory corticosteroids.
caused by inhibition of prostaglandin synthesis.
194
/
CHAPTER 23
COOH
Arachidonate
2O2
CYCLOOXYGENASE
Aspirin
- Indomethacin
O
Ibuprofen
COOH
COOH
O
PGI
2
OOH
PROSTACYCLIN
PGG2
PEROXIDASE
O
O
SYNTHASE
O
C
H
COOH
COOH
+
C
H
O
OH
OH
OH
O
OH
PGH
Malondialdehyde + HHT
2
ISOMERASE
THROMBOXANE
Imidazole
COOH
-
O O
O
SYNTHASE
COOH
COOH
O
O
OH
OH
OH
OH
OH
TXA2
6-Keto PGF1α
PGE2
ISOMERASE
REDUCTASE
OH
OH
OH
COOH
COOH
COOH
HO
O
OH
OH
O
OH
OH
PGF2 α
PGD2
TXB2
Figure 23-6. Conversion of arachidonic acid to prostaglandins and thromboxanes of series 2. (PG,
prostaglandin; TX, thromboxane; PGI, prostacyclin; HHT, hydroxyheptadecatrienoate.) (Asterisk: Both of these
starred activities are attributed to one enzyme: prostaglandin H synthase. Similar conversions occur in
prostaglandins and thromboxanes of series 1 and 3.)
Cyclooxygenase Is a “Suicide Enzyme”
nase pathway in response to both immunologic and
nonimmunologic stimuli. Three different lipoxygenases
“Switching off” of prostaglandin activity is partly achieved
(dioxygenases) insert oxygen into the 5, 12, and 15 po-
by a remarkable property of cyclooxygenase—that of
sitions of arachidonic acid, giving rise to hydroperox-
self-catalyzed destruction; ie, it is a “suicide enzyme.”
ides
(HPETE). Only 5-lipoxygenase forms leuko-
Furthermore, the inactivation of prostaglandins by 15-
trienes (details in Figure 23-7). Lipoxins are a family of
hydroxyprostaglandin dehydrogenase is rapid. Block-
conjugated tetraenes also arising in leukocytes. They are
ing the action of this enzyme with sulfasalazine or in-
formed by the combined action of more than one
domethacin can prolong the half-life of prostaglandins
lipoxygenase (Figure 23-7).
in the body.
CLINICAL ASPECTS
LEUKOTRIENES & LIPOXINS
ARE FORMED BY THE
Symptoms of Essential Fatty Acid
LIPOXYGENASE PATHWAY
Deficiency in Humans Include Skin
Lesions & Impairment of Lipid Transport
The leukotrienes are a family of conjugated trienes
formed from eicosanoic acids in leukocytes, mastocy-
In adults subsisting on ordinary diets, no signs of es-
toma cells, platelets, and macrophages by the lipoxyge-
sential fatty acid deficiencies have been reported. How-
METABOLISM OF UNSATURATED FATTY ACIDS & EICOSANOIDS
/
195
COOH
15-LIPOXYGENASE
12-LIPOXYGENASE
Arachidonate
COOH
COOH
O2
HOO
OOH
1
12-HPETE
15-HPETE
5-LIPOXYGENASE
COOH
1
COOH
OH
HO
15-HETE
OOH
OH
12-HETE
COOH
COOH
5-LIPOXYGENASE
1
5-HPETE
5-HETE
OH
H2O
OH OH
COOH
COOH
H2O
O
COOH
15-LIPOXYGENASE
OH
2
OH
Leukotriene B4
Leukotriene A4
Lipoxins, eg, LXA4
Glutathione
3
Glutamic acid
O
NH2
Glycine
Glycine
OH
O
NH
O
NH2
NH2
O
NH
NH
HO
HO
HO
Cysteine
Glutamic acid
Cysteine
Glycine
Cysteine
O
S
O
S
O
S
4
5
COOH
COOH
COOH
OH
OH
OH
Leukotriene C4
Leukotriene D4
Leukotriene E4
Figure 23-7. Conversion of arachidonic acid to leukotrienes and lipoxins of series 4 via the lipoxygenase path-
way. Some similar conversions occur in series 3 and 5 leukotrienes. (HPETE, hydroperoxyeicosatetraenoate; HETE,
hydroxyeicosatetraenoate; 1 , peroxidase; 2 , leukotriene A4 epoxide hydrolase; 3 , glutathione S-transferase;
4 , γ-glutamyltranspeptidase; 5 , cysteinyl-glycine dipeptidase.)
ever, infants receiving formula diets low in fat and pa-
Abnormal Metabolism of Essential Fatty
tients maintained for long periods exclusively by intra-
Acids Occurs in Several Diseases
venous nutrition low in essential fatty acids show defi-
ciency symptoms that can be prevented by an essential
Abnormal metabolism of essential fatty acids, which
fatty acid intake of 1-2% of the total caloric require-
may be connected with dietary insufficiency, has been
ment.
noted in cystic fibrosis, acrodermatitis enteropathica,
196
/
CHAPTER 23
hepatorenal syndrome, Sjögren-Larsson syndrome,
immediate hypersensitivity reactions, such as asthma.
multisystem neuronal degeneration, Crohn’s disease,
Leukotrienes are vasoactive, and
5-lipoxygenase has
cirrhosis and alcoholism, and Reye’s syndrome. Ele-
been found in arterial walls. Evidence supports a role
vated levels of very long chain polyenoic acids have
for lipoxins in vasoactive and immunoregulatory func-
been found in the brains of patients with Zellweger’s
tion, eg, as counterregulatory compounds (chalones) of
syndrome (Chapter 22). Diets with a high P:S (polyun-
the immune response.
saturated:saturated fatty acid) ratio reduce serum cho-
lesterol levels and are considered to be beneficial in
terms of the risk of development of coronary heart dis-
SUMMARY
ease.
• Biosynthesis of unsaturated long-chain fatty acids is
achieved by desaturase and elongase enzymes, which
Prostanoids Are Potent Biologically
introduce double bonds and lengthen existing acyl
Active Substances
chains, respectively.
Thromboxanes are synthesized in platelets and upon
• Higher animals have ∆4, ∆5, ∆6, and ∆9 desaturases
release cause vasoconstriction and platelet aggregation.
but cannot insert new double bonds beyond the 9
Their synthesis is specifically inhibited by low-dose as-
position of fatty acids. Thus, the essential fatty acids
pirin. Prostacyclins (PGI2) are produced by blood ves-
linoleic (ω6) and α-linolenic (ω3) must be obtained
sel walls and are potent inhibitors of platelet aggrega-
from the diet.
tion. Thus, thromboxanes and prostacyclins are
• Eicosanoids are derived from C20 (eicosanoic) fatty
antagonistic. PG3 and TX3, formed from eicosapen-
acids synthesized from the essential fatty acids and
taenoic acid (EPA) in fish oils, inhibit the release of
comprise important groups of physiologically and
arachidonate from phospholipids and the formation
pharmacologically active compounds, including the
of PG2 and TX2. PGI3 is as potent an antiaggregator of
prostaglandins, thromboxanes, leukotrienes, and
platelets as PGI2, but TXA3 is a weaker aggregator than
lipoxins.
TXA2, changing the balance of activity and favoring
longer clotting times. As little as 1 ng/mL of plasma
prostaglandins causes contraction of smooth muscle in
REFERENCES
animals. Potential therapeutic uses include prevention
Connor WE: The beneficial effects of omega-3 fatty acids: cardio-
of conception, induction of labor at term, termination
vascular disease and neurodevelopment. Curr Opin Lipidol
of pregnancy, prevention or alleviation of gastric ulcers,
1997;8:1.
control of inflammation and of blood pressure, and re-
Fischer S: Dietary polyunsaturated fatty acids and eicosanoid for-
lief of asthma and nasal congestion. In addition, PGD2
mation in humans. Adv Lipid Res 1989;23:169.
is a potent sleep-promoting substance. Prostaglandins
Lagarde M, Gualde N, Rigaud M: Metabolic interactions between
increase cAMP in platelets, thyroid, corpus luteum,
eicosanoids in blood and vascular cells. Biochem J 1989;
fetal bone, adenohypophysis, and lung but reduce
257:313.
cAMP in renal tubule cells and adipose tissue (Chap-
Neuringer M, Anderson GJ, Connor WE: The essentiality of n-3
ter 25).
fatty acids for the development and function of the retina and
brain. Annu Rev Nutr 1988;8:517.
Serhan CN: Lipoxin biosynthesis and its impact in inflammatory
Leukotrienes & Lipoxins Are Potent
and vascular events. Biochim Biophys Acta 1994;1212:1.
Regulators of Many Disease Processes
Smith WL, Fitzpatrick FA: The eicosanoids: Cyclooxygenase,
lipoxygenase, and epoxygenase pathways. In: Biochemistry of
Slow-reacting substance of anaphylaxis (SRS-A) is a
Lipids, Lipoproteins and Membranes. Vance DE, Vance JE
mixture of leukotrienes C4, D4, and E4. This mixture of
(editors). Elsevier, 1996.
leukotrienes is a potent constrictor of the bronchial air-
Tocher DR, Leaver MJ, Hodgson PA: Recent advances in the bio-
way musculature. These leukotrienes together with
chemistry and molecular biology of fatty acyl desaturases.
leukotriene B4 also cause vascular permeability and at-
Prog Lipid Res 1998;37:73.
traction and activation of leukocytes and are important
Valenzuela A, Morgado N: Trans fatty acid isomers in human
regulators in many diseases involving inflammatory or
health and the food industry. Biol Res 1999;32:273.
Metabolism of Acylglycerols
24
& Sphingolipids
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
possess glycerol kinase, found in significant amounts
in liver, kidney, intestine, brown adipose tissue, and
Acylglycerols constitute the majority of lipids in the
lactating mammary gland.
body. Triacylglycerols are the major lipids in fat de-
posits and in food, and their roles in lipid transport and
storage and in various diseases such as obesity, diabetes,
TRIACYLGLYCEROLS &
and hyperlipoproteinemia will be described in subse-
PHOSPHOGLYCEROLS ARE FORMED BY
quent chapters. The amphipathic nature of phospho-
ACYLATION OF TRIOSE PHOSPHATES
lipids and sphingolipids makes them ideally suitable as
the main lipid component of cell membranes. Phos-
The major pathways of triacylglycerol and phosphoglyc-
pholipids also take part in the metabolism of many
erol biosynthesis are outlined in Figure 24-1. Impor-
other lipids. Some phospholipids have specialized func-
tant substances such as triacylglycerols, phosphatidyl-
tions; eg, dipalmitoyl lecithin is a major component of
choline, phosphatidylethanolamine, phosphatidylinositol,
lung surfactant, which is lacking in respiratory distress
and cardiolipin, a constituent of mitochondrial mem-
syndrome of the newborn. Inositol phospholipids in the
branes, are formed from glycerol-3-phosphate. Significant
cell membrane act as precursors of hormone second
branch points in the pathway occur at the phosphati-
messengers, and platelet-activating factor is an alkyl-
date and diacylglycerol steps. From dihydroxyacetone
phospholipid. Glycosphingolipids, containing sphingo-
phosphate are derived phosphoglycerols containing an
sine and sugar residues as well as fatty acid and found in
ether link (COC), the best-known of which
the outer leaflet of the plasma membrane with their
are plasmalogens and platelet-activating factor (PAF).
oligosaccharide chains facing outward, form part of the
Glycerol 3-phosphate and dihydroxyacetone phosphate
glycocalyx of the cell surface and are important (1) in
are intermediates in glycolysis, making a very important
cell adhesion and cell recognition; (2) as receptors for
connection between carbohydrate and lipid metabo-
bacterial toxins (eg, the toxin that causes cholera); and
lism.
(3) as ABO blood group substances. A dozen or so gly-
colipid storage diseases have been described
(eg,
Gaucher’s disease, Tay-Sachs disease), each due to a ge-
netic defect in the pathway for glycolipid degradation
in the lysosomes.
Glycerol 3-phosphate
Dihydroxyacetone phosphate
HYDROLYSIS INITIATES CATABOLISM
OF TRIACYLGLYCEROLS
Phosphatidate
Plasmalogens
PAF
Triacylglycerols must be hydrolyzed by a lipase to their
constituent fatty acids and glycerol before further catab-
olism can proceed. Much of this hydrolysis (lipolysis)
Diacylglycerol
Cardiolipin
Phosphatidylinositol
occurs in adipose tissue with release of free fatty acids
into the plasma, where they are found combined with
serum albumin. This is followed by free fatty acid up-
Phosphatidylcholine
Triacylglycerol
Phosphatidylinositol
take into tissues (including liver, heart, kidney, muscle,
Phosphatidylethanolamine
4,5-bisphosphate
lung, testis, and adipose tissue, but not readily by
brain), where they are oxidized or reesterified. The uti-
Figure 24-1. Overview of acylglycerol biosynthesis.
lization of glycerol depends upon whether such tissues
(PAF, platelet-activating factor.)
197
ATP
ADP
NAD+
NADH + H+
H2C OH
H2C OH
H2C OH
HO C H
HO C H
C O
Glycolysis
H2C
OH
GLYCEROL KINASE
H2C
O
P
GLYCEROL-
H
2C
O
P
3-PHOSPHATE
Glycerol
sn-Glycerol
DEHYDROGENASE
Dihydroxyacetone
3-phosphate
phosphate
Acyl-CoA (mainly saturated)
GLYCEROL-
2
3-PHOSPHATE
ACYLTRANSFERASE
CoA
O
H2C O
C R1
HO CH
H2C
OH
H2C
O
P
R2
C
O
C
H
1-Acylglycerol-
3-phosphate
O
H2C OH
(lysophosphatidate)
2-Monoacylglycerol
Acyl-CoA (usually unsaturated)
1-ACYLGLYCEROL-
3-PHOSPHATE
ACYLTRANSFERASE
Acyl-CoA
1
CoA
MONOACYLGLYCEROL
ACYLTRANSFERASE
O
(INTESTINE)
H2C O
C R1
CoA
R
2
C
O C
H
O
H2C
O
P
1,2-Diacylglycerol
phosphate
(phosphatidate)
Choline
H2O
CTP
ATP
PHOSPHATIDATE
CDP-DG
CHOLINE
PHOSPHOHYDROLASE
SYNTHASE
KINASE
P1
PP1
ADP
O
O
Phosphocholine
H2C O
C R1
H2C
O
C R1
CTP
R
2
C
O
C
H
R
2
C
O
C
H
CTP:
PHOSPHOCHOLINE
O
H2COH
O
H2C
O P P
CYTIDYL
1,2-Diacylglycerol
TRANSFERASE
Cytidine
CDP-diacylglycerol
Cardiolipin
PP
1
CDP-choline
Acyl-CoA
Inositol
CDP-CHOLINE:
DIACYLGLYCEROL
DIACYLGLYCEROL
PHOSPHATIDYL-
PHOSPHOCHOLINE
ACYLTRANSFERASE
INOSITOL SYNTHASE
TRANSFERASE
CoA
CMP
CMP
ATP
ADP
O
O
O
KINASE
O
H2C O
C R1
H2C O C R1
H2C O C R1
H2C O
C
R1
R
2
C
O
C
H
R
2
C
O
C
H
O
R
2
C
O
C
H
R
2
C
O
C
H
O H C O
2
P
2
O H C O C R
3
O
2
H C O
P
O H C O
2
P
Inositol
P
Triacyglycerol
Choline
Inositol
Phosphatidylinositol 4-phosphate
Phosphatidylcholine
Phosphatidylinositol
ATP
PHOSPHATIDYLETHANOLAMINE
N-METHYLTRANSFERASE
(-CH3)3
KINASE
Phosphatidylethanolamine
Serine
CO
2
ADP
O
H2C O C R1
Phosphatidylserine
Ethanolamine
R
2
C
O
C
H
Figure 24-2 . Biosynthesis of triacylglycerol and phospholipids.
O H C O
2
P Inositol
P
( 1 , Monoacylglycerol pathway; 2, glycerol phosphate pathway.)
P
Phosphatidylinositol 4,5-bisphosphate
Phosphatidylethanolamine may be formed from ethanolamine by a
pathway similar to that shown for the formation of phosphatidyl-
choline from choline.
METABOLISM OF ACYLGLYCEROLS & SPHINGOLIPIDS
/
199
Phosphatidate Is the Common Precursor
from phosphatidylglycerol, which in turn is synthesized
in the Biosynthesis of Triacylglycerols,
from CDP-diacylglycerol (Figure 24-2) and glycerol
Many Phosphoglycerols, & Cardiolipin
3-phosphate according to the scheme shown in Figure
24-3. Cardiolipin, found in the inner membrane of
Both glycerol and fatty acids must be activated by ATP
mitochondria, is specifically required for the function-
before they can be incorporated into acylglycerols.
ing of the phosphate transporter and for cytochrome
Glycerol kinase catalyzes the activation of glycerol to
oxidase activity.
sn-glycerol 3-phosphate. If the activity of this enzyme is
absent or low, as in muscle or adipose tissue, most of
B. BIOSYNTHESIS OF GLYCEROL ETHER PHOSPHOLIPIDS
the glycerol 3-phosphate is formed from dihydroxyace-
tone phosphate by glycerol-3-phosphate dehydrogen-
This pathway is located in peroxisomes. Dihydroxyace-
ase (Figure 24-2).
tone phosphate is the precursor of the glycerol moiety
of glycerol ether phospholipids
(Figure
24-4). This
A. BIOSYNTHESIS OF TRIACYLGLYCEROLS
compound combines with acyl-CoA to give 1-acyldihy-
Two molecules of acyl-CoA, formed by the activation
droxyacetone phosphate. The ether link is formed in
of fatty acids by acyl-CoA synthetase (Chapter 22),
the next reaction, producing 1-alkyldihydroxyacetone
combine with glycerol 3-phosphate to form phosphati-
phosphate, which is then converted to 1-alkylglycerol
date (1,2-diacylglycerol phosphate). This takes place in
3-phosphate. After further acylation in the 2 position,
two stages, catalyzed by glycerol-3-phosphate acyl-
the resulting 1-alkyl-2-acylglycerol 3-phosphate (analo-
transferase and 1-acylglycerol-3-phosphate acyltrans-
gous to phosphatidate in Figure 24-2) is hydrolyzed to
ferase. Phosphatidate is converted by phosphatidate
give the free glycerol derivative. Plasmalogens, which
phosphohydrolase and diacylglycerol acyltransferase
comprise much of the phospholipid in mitochondria,
to 1,2-diacylglycerol and then triacylglycerol. In intesti-
are formed by desaturation of the analogous 3-phos-
nal mucosa, monoacylglycerol acyltransferase con-
phoethanolamine derivative
(Figure
24-4). Platelet-
verts monoacylglycerol to
1,2-diacylglycerol in the
activating factor (PAF) (1-alkyl-2-acetyl-sn-glycerol-3-
monoacylglycerol pathway. Most of the activity of
phosphocholine) is synthesized from the corresponding
these enzymes resides in the endoplasmic reticulum of
3-phosphocholine derivative. It is formed by many
the cell, but some is found in mitochondria. Phosphati-
blood cells and other tissues and aggregates platelets at
date phosphohydrolase is found mainly in the cytosol,
concentrations as low as 10−11 mol/L. It also has hy-
but the active form of the enzyme is membrane-bound.
potensive and ulcerogenic properties and is involved in
In the biosynthesis of phosphatidylcholine and
a variety of biologic responses, including inflammation,
phosphatidylethanolamine
(Figure
24-2), choline or
chemotaxis, and protein phosphorylation.
ethanolamine must first be activated by phosphoryla-
tion by ATP followed by linkage to CTP. The resulting
CDP-choline or CDP-ethanolamine reacts with 1,2-di-
acylglycerol to form either phosphatidylcholine or
CDP-Diacyl-
sn-Glycerol
phosphatidylethanolamine, respectively. Phosphatidyl-
glycerol
3-phosphate
serine is formed from phosphatidylethanolamine di-
rectly by reaction with serine
(Figure
24-2). Phos-
CMP
phatidylserine may re-form phosphatidylethanolamine
Phosphatidylglycerol phosphate
by decarboxylation. An alternative pathway in liver en-
ables phosphatidylethanolamine to give rise directly to
H2O
phosphatidylcholine by progressive methylation of the
ethanolamine residue. In spite of these sources of
choline, it is considered to be an essential nutrient in
Pi
many mammalian species, but this has not been estab-
Phosphatidylglycerol
lished in humans.
The regulation of triacylglycerol, phosphatidyl-
choline, and phosphatidylethanolamine biosynthesis is
driven by the availability of free fatty acids. Those that
escape oxidation are preferentially converted to phos-
CMP
pholipids, and when this requirement is satisfied they
Cardiolipin
are used for triacylglycerol synthesis.
(diphosphatidylglycerol)
A phospholipid present in mitochondria is cardio-
lipin (diphosphatidylglycerol; Figure 14-8). It is formed
Figure 24-3. Biosynthesis of cardiolipin.
200
/
CHAPTER 24
NADPH
O
R2
(CH2)2
OH
+ H+ NADP+
Acyl-CoA
H2COH
H2C
O C R1
H2C
O
(CH2)2
R2
H2C
O
(CH2)2
R2
O
C
O
C
O
C
HO
C
H
H2C O P
ACYL-
H2C O P
SYNTHASE
H2C O P
REDUCTASE
H2C O
P
TRANSFERASE
HOOC R1
Dihydroxyacetone
1-Acyldihydroxyacetone
1-Alkyldihydroxyacetone
1-Alkylglycerol 3-phosphate
phosphate
phosphate
phosphate
Acyl-CoA
ACYL-
TRANSFERASE
CDP-
CMP Ethanolamine
Pi
H2O
O
H2C
O
(CH2)2
R2
O
H2C
O
(CH2)2
R2
O
H2C
O
(CH2)2
R2
R3
C
O
C
H
R3
C
O
C
H
R3
C
O
C
H
H2C O P CH2
CH2
NH2
CDP-ETHANOLAMINE:
H2C OH
PHOSPHOHYDROLASE
H2C O P
ALKYLACYLGLYCEROL
PHOSPHOETHANOLAMINE
1-Alkyl-2-acylglycerol
TRANSFERASE
3-phosphoethanolamine
1-Alkyl-2-acylglycerol 3-phosphate
1-Alkyl-2-acylglycerol
CDP-choline
NADPH, O2,
CDP-CHOLINE:
DESATURASE
ALKYLACYLGLYCEROL
Cyt b5
Alkyl, diacyl glycerols
PHOSPHOCHOLINE
TRANSFERASE
CMP
O
H2C
O
CH CH
R2
O
H2C
O
(CH2)2
R2
R3
C
O
C
H
H2O R3
COOH
R3
C
O
C
H
H2C
O
(CH2)2
R2
H2C O P
(CH2)2
NH2
H2C O
P
HO
C
H
1-Alkenyl-2-acylglycerol
PHOSPHOLIPASE A2
H2C O P
Choline
3-phosphoethanolamine
1-Alkyl-2-acylglycerol
Choline
plasmalogen
3-phosphocholine
1-Alkyl-2-lysoglycerol
Acetyl-CoA
3-phosphocholine
ACETYLTRANSFERASE
O
H2C
O
(CH2)2
R2
H3C
C
O
C
H
H2C O
P
Choline
1-Alkyl-2-acetylglycerol 3-phosphocholine
PAF
Figure 24-4. Biosynthesis of ether lipids, including plasmalogens, and platelet-activating factor (PAF). In the
de novo pathway for PAF synthesis, acetyl-CoA is incorporated at stage *, avoiding the last two steps in the path-
way shown here.
Phospholipases Allow Degradation
solecithin) is attacked by lysophospholipase, forming
& Remodeling of Phosphoglycerols
the corresponding glyceryl phosphoryl base, which in
turn may be split by a hydrolase liberating glycerol
Although phospholipids are actively degraded, each
3-phosphate plus base. Phospholipases A1, A2, B, C,
portion of the molecule turns over at a different rate—
and D attack the bonds indicated in Figure 24-6.
eg, the turnover time of the phosphate group is differ-
Phospholipase A2 is found in pancreatic fluid and
ent from that of the 1-acyl group. This is due to the
snake venom as well as in many types of cells; phos-
presence of enzymes that allow partial degradation fol-
pholipase C is one of the major toxins secreted by bac-
lowed by resynthesis (Figure 24-5). Phospholipase A2
teria; and phospholipase D is known to be involved in
catalyzes the hydrolysis of glycerophospholipids to form
mammalian signal transduction.
a free fatty acid and lysophospholipid, which in turn
Lysolecithin
(lysophosphatidylcholine) may be
may be reacylated by acyl-CoA in the presence of an
formed by an alternative route that involves lecithin:
acyltransferase. Alternatively, lysophospholipid (eg, ly-
cholesterol acyltransferase
(LCAT). This enzyme,
METABOLISM OF ACYLGLYCEROLS & SPHINGOLIPIDS
/
201
O
found in plasma, catalyzes the transfer of a fatty acid
residue from the 2 position of lecithin to cholesterol to
O
H2C O C R1
form cholesteryl ester and lysolecithin and is considered
R2
C
O
C
H
to be responsible for much of the cholesteryl ester in
H2C O P Choline
plasma lipoproteins. Long-chain saturated fatty acids
are found predominantly in the 1 position of phospho-
Phosphatidylcholine
lipids, whereas the polyunsaturated acids (eg, the pre-
H2O
cursors of prostaglandins) are incorporated more into
the 2 position. The incorporation of fatty acids into
ACYLTRANSFERASE
PHOSPHOLIPASE A2
lecithin occurs by complete synthesis of the phospho-
2
R COOH
lipid, by transacylation between cholesteryl ester and
O
lysolecithin, and by direct acylation of lysolecithin by
acyl-CoA. Thus, a continuous exchange of the fatty
H2C O C R
1
acids is possible, particularly with regard to introducing
HO
C
H
essential fatty acids into phospholipid molecules.
H2C O P Choline
Acyl-CoA
Lysophosphatidylcholine (lysolecithin)
ALL SPHINGOLIPIDS ARE FORMED
H2O
FROM CERAMIDE
LYSOPHOSPHOLIPASE
Ceramide is synthesized in the endoplasmic reticulum
from the amino acid serine according to Figure 24-7.
1
R COOH
Ceramide is an important signaling molecule (second
messenger) regulating pathways including apoptosis
H2C OH
(processes leading to cell death), cell senescence, and
HO
C
H
differentiation, and opposes some of the actions of di-
H2C O P Choline
acylglycerol.
Sphingomyelins (Figure 14-11) are phospholipids
Glycerylphosphocholine
and are formed when ceramide reacts with phos-
H2O
phatidylcholine to form sphingomyelin plus diacylglyc-
GLYCERYLPHOSPHO-
erol (Figure 24-8A). This occurs mainly in the Golgi
CHOLINE HYDROLASE
apparatus and to a lesser extent in the plasma mem-
brane.
H2C OH
Glycosphingolipids Are a Combination
HO
C
H
+ Choline
of Ceramide With One or More
C O P
H2
Sugar Residues
sn-Glycerol 3-phosphate
The simplest glycosphingolipids
(cerebrosides) are
Figure 24-5.
Metabolism of phosphatidylcholine
galactosylceramide
(GalCer) and glucosylceramide
(lecithin).
(GlcCer). GalCer is a major lipid of myelin, whereas
GlcCer is the major glycosphingolipid of extraneural
tissues and a precursor of most of the more complex
PHOSPHOLIPASE B
PHOSPHOLIPASE A1
glycosphingolipids. Galactosylceramide (Figure 24-8B)
O
is formed in a reaction between ceramide and UDPGal
(formed by epimerization from UDPGlc—Figure
H2C O
C R
1
O
20-6). Sulfogalactosylceramide and other sulfolipids
PHOSPHOLIPASE D
such as the sulfo(galacto)-glycerolipids and the
R
2
C
O
C
H
O
steroid sulfates are formed after further reactions in-
volving 3′-phosphoadenosine-5′-phosphosulfate (PAPS;
H2C O
P
O
N-BASE
“active sulfate”). Gangliosides are synthesized from
PHOSPHOLIPASE A2
O-
ceramide by the stepwise addition of activated sugars (eg,
PHOSPHOLIPASE C
UDPGlc and UDPGal) and a sialic acid, usually N-
acetylneuraminic acid (Figure 24-9). A large number
Figure 24-6. Sites of the hydrolytic activity of phos-
of gangliosides of increasing molecular weight may be
pholipases on a phospholipid substrate.
formed. Most of the enzymes transferring sugars from
202
/
CHAPTER 24
nucleotide sugars (glycosyl transferases) are found in
O
+NH
3
the Golgi apparatus.
CH3
(CH2)14
C
S CoA
−OOC
CH
CH2
OH
Glycosphingolipids are constituents of the outer
Palmitoyl-CoA
Serine
leaflet of plasma membranes and are important in cell
adhesion and cell recognition. Some are antigens, eg,
ABO blood group substances. Certain gangliosides
Pyridoxal phosphate, Mn2+
function as receptors for bacterial toxins (eg, for cholera
toxin, which subsequently activates adenylyl cyclase).
SERINE
PALMITOYLTRANSFERASE
CLINICAL ASPECTS
CoA SH
CO
2
O
Deficiency of Lung Surfactant Causes
Respiratory Distress Syndrome
CH3
(CH2)12
CH
2
CH
2
C CH
CH2
OH
+
Lung surfactant is composed mainly of lipid with
NH3
3-Ketosphinganine
some proteins and carbohydrate and prevents the alve-
oli from collapsing. Surfactant activity is largely attrib-
NADPH + H+
uted to dipalmitoylphosphatidylcholine, which is
3-KETOSPHINGANINE
REDUCTASE
synthesized shortly before parturition in full-term in-
fants. Deficiency of lung surfactant in the lungs of
NADP+
many preterm newborns gives rise to respiratory dis-
CH3(CH2)12
CH2
CH2
CH CH
CH2
OH
tress syndrome. Administration of either natural or ar-
+
tificial surfactant has been of therapeutic benefit.
OH NH
3
Dihydrosphingosine (sphinganine)
Phospholipids & Sphingolipids
R CO
S CoA
Are Involved in Multiple Sclerosis
Acyl-CoA
DIHYDROSPHINGOSINE
N -ACYLTRANSFERASE
and Lipidoses
CoA SH
Certain diseases are characterized by abnormal quanti-
CH3
(CH2)12
CH2
CH2
CH
CH
CH2
OH
ties of these lipids in the tissues, often in the nervous
system. They may be classified into two groups: (1) true
OH
NH
CO
R
demyelinating diseases and (2) sphingolipidoses.
Dihydroceramide
In multiple sclerosis, which is a demyelinating dis-
DIHYDROCERAMIDE
ease, there is loss of both phospholipids (particularly
2H
DESATURASE
ethanolamine plasmalogen) and of sphingolipids from
white matter. Thus, the lipid composition of white
matter resembles that of gray matter. The cerebrospinal
CH3
(CH2)12
CH CH CH
CH
CH2
OH
fluid shows raised phospholipid levels.
OH
NH
CO
R
The sphingolipidoses (lipid storage diseases) are a
Ceramide
group of inherited diseases that are often manifested in
childhood. These diseases are part of a larger group of
Figure 24-7. Biosynthesis of ceramide.
lysosomal disorders and exhibit several constant fea-
tures: (1) Complex lipids containing ceramide accumu-
late in cells, particularly neurons, causing neurodegen-
A Ceramide
Sphingomyelin
Phosphatidylcholine
Diacylglycerol
Figure 24-8. Biosynthesis of sphingomyelin (A),
UDPGal UDP
PAPS
galactosylceramide and its sulfo derivative (B). (PAPS,
Sulfogalactosyl-
Galactosylceramide
ceramide
“active sulfate,” adenosine 3′-phosphate-5′-phospho-
B
Ceramide
(cerebroside)
(sulfatide)
sulfate.)
METABOLISM OF ACYLGLYCEROLS & SPHINGOLIPIDS
/
203
UDPGlc
UDP
UDPGal
UDP
CMP-NeuAc
CMP
Glucosyl
Ceramide
ceramide
Cer-Glc-Gal
Cer-Glc-Gal
(Cer-Glc)
NeuAc
(GM3)
UDP-N-acetyl
galactosamine
UDP
UDPGal
UDP
Higher gangliosides
Cer-Glc-Gal-GalNAc-Gal
Cer-Glc-Gal-GalNAc
(disialo- and trisialo-
gangliosides)
NeuAc
NeuAc
(GM1)
(GM2)
Figure 24-9. Biosynthesis of gangliosides. (NeuAc, N-acetylneuraminic acid.)
eration and shortening the life span. (2) The rate of
dase) in the treatment of Gaucher’s disease. A recent
synthesis of the stored lipid is normal. (3) The enzy-
promising approach is substrate reduction therapy to
matic defect is in the lysosomal degradation pathway
inhibit the synthesis of sphingolipids, and gene therapy
of sphingolipids. (4) The extent to which the activity of
for lysosomal disorders is currently under investigation.
the affected enzyme is decreased is similar in all tissues.
Some examples of the more important lipid storage dis-
There is no effective treatment for many of the diseases,
eases are shown in Table 24-1.
though some success has been achieved with enzymes
Multiple sulfatase deficiency results in accumula-
that have been chemically modified to ensure binding
tion of sulfogalactosylceramide, steroid sulfates, and
to receptors of target cells, eg, to macrophages in the
proteoglycans owing to a combined deficiency of aryl-
liver in order to deliver β-glucosidase (glucocerebrosi-
sulfatases A, B, and C and steroid sulfatase.
Table 24-1. Examples of sphingolipidoses.
Disease
Enzyme Deficiency
Lipid Accumulating1
Clinical Symptoms
:
Tay-Sachs disease
Hexosaminidase A
Cer—Glc—Gal(NeuAc)—
Mental retardation, blindness, muscular weakness.
:GalNAc
GM2 Ganglioside
:
Fabry’s disease
α-Galactosidase
Cer—Glc—Gal—
Skin rash, kidney failure (full symptoms only in
:Gal
Globotriaosylceramide
males; X-linked recessive).
:
Metachromatic
Arylsulfatase A
Cer—Gal—
Mental retardation and psychologic disturbances in
:OSO3
leukodystrophy
3-Sulfogalactosylceramide
adults; demyelination.
:
Krabbe’s disease
β-Galactosidase
Cer—
Mental retardation; myelin almost absent.
:Gal
Galactosylceramide
:
Gaucher’s disease
β-Glucosidase
Cer—
Enlarged liver and spleen, erosion of long bones,
:Glc
Glucosylceramide
mental retardation in infants.
:
Niemann-Pick
Sphingomyelinase
Cer—
Enlarged liver and spleen, mental retardation; fatal in
:P—choline
disease
Sphingomyelin
early life.
:
Farber’s disease
Ceramidase
Acyl—
Hoarseness, dermatitis, skeletal deformation, mental
:Sphingosine
Ceramide
retardation; fatal in early life.
:
1NeuAc, N-acetylneuraminic acid; Cer, ceramide; Glc, glucose; Gal, galactose. —, site of deficient enzyme reaction.
:
204
/
CHAPTER 24
SUMMARY
(demyelination), and sphingolipidoses (inability to
break down sphingolipids in lysosomes due to inher-
•
Triacylglycerols are the major energy-storing lipids,
ited defects in hydrolase enzymes).
whereas phosphoglycerols, sphingomyelin, and gly-
cosphingolipids are amphipathic and have structural
functions in cell membranes as well as other special-
ized roles.
REFERENCES
•
Triacylglycerols and some phosphoglycerols are syn-
Griese M: Pulmonary surfactant in health and human lung dis-
thesized by progressive acylation of glycerol 3-phos-
eases: state of the art. Eur Respir J 1999;13:1455.
phate. The pathway bifurcates at phosphatidate,
Merrill AH, Sweeley CC: Sphingolipids: metabolism and cell sig-
forming inositol phospholipids and cardiolipin on
naling. In: Biochemistry of Lipids, Lipoproteins and Mem-
the one hand and triacylglycerol and choline and
branes. Vance DE, Vance JE (editors). Elsevier, 1996.
ethanolamine phospholipids on the other.
Prescott SM et al: Platelet-activating factor and related lipid media-
tors. Annu Rev Biochem 2000;69:419.
•
Plasmalogens and platelet-activating factor (PAF) are
Ruvolo PP: Ceramide regulates cellular homeostasis via diverse
ether phospholipids formed from dihydroxyacetone
stress signaling pathways. Leukemia 2001;15:1153.
phosphate.
Schuette CG et al: The glycosphingolipidoses—from disease to
•
Sphingolipids are formed from ceramide (N-acyl-
basic principles of metabolism. Biol Chem 1999;380:759.
sphingosine). Sphingomyelin is present in mem-
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
branes of organelles involved in secretory processes
herited Disease, 8th ed. McGraw-Hill, 2001.
(eg, Golgi apparatus). The simplest glycosphin-
Tijburg LBM, Geelen MJH, van Golde LMG: Regulation of the
golipids are a combination of ceramide plus a sugar
biosynthesis of triacylglycerol, phosphatidylcholine and phos-
residue
(eg, GalCer in myelin). Gangliosides are
phatidylethanolamine in the liver. Biochim Biophys Acta
1989;1004:1.
more complex glycosphingolipids containing more
sugar residues plus sialic acid. They are present in the
Vance DE: Glycerolipid biosynthesis in eukaryotes. In: Biochem-
istry of Lipids, Lipoproteins and Membranes. Vance DE, Vance
outer layer of the plasma membrane, where they con-
JE (editors). Elsevier, 1996.
tribute to the glycocalyx and are important as anti-
van Echten G, Sandhoff K: Ganglioside metabolism. Enzymology,
gens and cell receptors.
topology, and regulation. J Biol Chem 1993;268:5341.
•
Phospholipids and sphingolipids are involved in sev-
Waite M: Phospholipases. In: Biochemistry of Lipids, Lipoproteins
eral disease processes, including respiratory distress
and Membranes. Vance DE, Vance JE (editors). Elsevier,
syndrome (lack of lung surfactant), multiple sclerosis
1996.
Lipid Transport & Storage
25
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
Four Major Groups of Plasma Lipoproteins
Have Been Identified
Fat absorbed from the diet and lipids synthesized by the
liver and adipose tissue must be transported between
Because fat is less dense than water, the density of a
the various tissues and organs for utilization and stor-
lipoprotein decreases as the proportion of lipid to pro-
age. Since lipids are insoluble in water, the problem of
tein increases (Table 25-1). In addition to FFA, four
how to transport them in the aqueous blood plasma is
major groups of lipoproteins have been identified that
solved by associating nonpolar lipids
(triacylglycerol
are important physiologically and in clinical diagnosis.
and cholesteryl esters) with amphipathic lipids (phos-
These are (1) chylomicrons, derived from intestinal
pholipids and cholesterol) and proteins to make water-
absorption of triacylglycerol and other lipids; (2) very
miscible lipoproteins.
low density lipoproteins (VLDL, or pre-β-lipopro-
In a meal-eating omnivore such as the human, ex-
teins), derived from the liver for the export of triacyl-
cess calories are ingested in the anabolic phase of the
glycerol;
(3) low-density lipoproteins
(LDL, or β-
feeding cycle, followed by a period of negative caloric
lipoproteins), representing a final stage in the catabolism
balance when the organism draws upon its carbohy-
of VLDL; and (4) high-density lipoproteins (HDL, or
drate and fat stores. Lipoproteins mediate this cycle by
α-lipoproteins), involved in VLDL and chylomicron
transporting lipids from the intestines as chylomi-
metabolism and also in cholesterol transport. Triacyl-
crons—and from the liver as very low density lipopro-
glycerol is the predominant lipid in chylomicrons and
teins
(VLDL)—to most tissues for oxidation and to
VLDL, whereas cholesterol and phospholipid are the
adipose tissue for storage. Lipid is mobilized from adi-
predominant lipids in LDL and HDL, respectively
pose tissue as free fatty acids (FFA) attached to serum
(Table 25-1). Lipoproteins may be separated according
albumin. Abnormalities of lipoprotein metabolism
to their electrophoretic properties into
-,
-, and pre-
cause various hypo- or hyperlipoproteinemias. The
-lipoproteins.
most common of these is diabetes mellitus, where in-
sulin deficiency causes excessive mobilization of FFA
and underutilization of chylomicrons and VLDL, lead-
Lipoproteins Consist of a Nonpolar
ing to hypertriacylglycerolemia. Most other patho-
Core & a Single Surface Layer of
logic conditions affecting lipid transport are due pri-
Amphipathic Lipids
marily to inherited defects, some of which cause
The nonpolar lipid core consists of mainly triacylglyc-
hypercholesterolemia, and premature atherosclerosis.
erol and cholesteryl ester and is surrounded by a sin-
Obesity—particularly abdominal obesity—is a risk fac-
gle surface layer of amphipathic phospholipid and
tor for increased mortality, hypertension, type 2 dia-
cholesterol molecules (Figure 25-1). These are oriented
betes mellitus, hyperlipidemia, hyperglycemia, and vari-
so that their polar groups face outward to the aqueous
ous endocrine dysfunctions.
medium, as in the cell membrane (Chapter 14). The
protein moiety of a lipoprotein is known as an apo-
LIPIDS ARE TRANSPORTED IN THE
lipoprotein or apoprotein, constituting nearly 70% of
some HDL and as little as 1% of chylomicrons. Some
PLASMA AS LIPOPROTEINS
apolipoproteins are integral and cannot be removed,
Four Major Lipid Classes Are Present
whereas others are free to transfer to other lipoproteins.
in Lipoproteins
Plasma lipids consist of triacylglycerols (16%), phos-
The Distribution of Apolipoproteins
pholipids (30%), cholesterol (14%), and cholesteryl
Characterizes the Lipoprotein
esters (36%) and a much smaller fraction of unesteri-
fied long-chain fatty acids (free fatty acids) (4%). This
One or more apolipoproteins (proteins or polypeptides)
latter fraction, the free fatty acids (FFA), is metaboli-
are present in each lipoprotein. The major apolipopro-
cally the most active of the plasma lipids.
teins of HDL (α-lipoprotein) are designated A (Table
205
206
/
CHAPTER 25
Table 25-1.
Composition of the lipoproteins in plasma of humans.
Composition
Diameter
Density
Protein
Lipid
Main Lipid
Lipoprotein
Source
(nm)
(g/mL)
(%)
(%)
Components
Apolipoproteins
Chylomicrons
Intestine
90-1000
< 0.95
1-2
98-99
Triacylglycerol
A-I, A-II, A-IV,1 B-48, C-I, C-II, C-III,
E
Chylomicron
Chylomicrons
45-150
< 1.006
6-8
92-94
Triacylglycerol,
B-48, E
remnants
phospholipids,
cholesterol
VLDL
Liver (intestine)
30-90
0.95-1.006
7-10
90-93
Triacylglycerol
B-100, C-I, C-II, C-III
IDL
VLDL
25-35
1.006-1.019
11
89
Triacylglycerol,
B-100, E
cholesterol
LDL
VLDL
20-25
1.019-1.063
21
79
Cholesterol
B-100
HDL
Liver, intestine,
Phospholipids,
A-I, A-II, A-IV, C-I, C-II, C-III, D,2 E
HDL1
VLDL, chylo-
20-25
1.019-1.063
32
68
cholesterol
microns
HDL2
10-20
1.063-1.125
33
67
HDL3
5-10
1.125-1.210
57
43
Preβ-HDL3
< 5
> 1.210
A-I
Albumin/free
Adipose
> 1.281
99
1
Free fatty acids
fatty acids
tissue
Abbreviations: HDL, high-density lipoproteins; IDL, intermediate-density lipoproteins; LDL, low-density lipoproteins; VLDL, very low
density lipoproteins.
1Secreted with chylomicrons but transfers to HDL.
2Associated with HDL2 and HDL3 subfractions.
3Part of a minor fraction known as very high density lipoproteins (VHDL).
25-1). The main apolipoprotein of LDL (β-lipopro-
tein receptors in tissues, eg, apo B-100 and apo E for
tein) is apolipoprotein B (B-100) and is found also in
the LDL receptor, apo E for the LDL receptor-related
VLDL. Chylomicrons contain a truncated form of apo
protein (LRP), which has been identified as the rem-
B (B-48) that is synthesized in the intestine, while
nant receptor, and apo A-I for the HDL receptor. The
B-100 is synthesized in the liver. Apo B-100 is one of
functions of apo A-IV and apo D, however, are not yet
the longest single polypeptide chains known, having
clearly defined.
4536 amino acids and a molecular mass of 550,000 Da.
Apo B-48 (48% of B-100) is formed from the same
mRNA as apo B-100 after the introduction of a stop sig-
FREE FATTY ACIDS ARE
nal by an RNA editing enzyme. Apo C-I, C-II, and
RAPIDLY METABOLIZED
C-III are smaller polypeptides (molecular mass 7000-
9000 Da) freely transferable between several different
The free fatty acids (FFA, nonesterified fatty acids, un-
lipoproteins. Apo E is found in VLDL, HDL, chylomi-
esterified fatty acids) arise in the plasma from lipolysis
crons, and chylomicron remnants; it accounts for 5-
of triacylglycerol in adipose tissue or as a result of the
10% of total VLDL apolipoproteins in normal subjects.
action of lipoprotein lipase during uptake of plasma tri-
Apolipoproteins carry out several roles: (1) they can
acylglycerols into tissues. They are found in combina-
form part of the structure of the lipoprotein, eg, apo B;
tion with albumin, a very effective solubilizer, in con-
(2) they are enzyme cofactors, eg, C-II for lipoprotein
centrations varying between 0.1 and 2.0 µeq/mL of
lipase, A-I for lecithin:cholesterol acyltransferase, or en-
plasma. Levels are low in the fully fed condition and
zyme inhibitors, eg, apo A-II and apo C-III for lipopro-
rise to 0.7-0.8 µeq/mL in the starved state. In uncon-
tein lipase, apo C-I for cholesteryl ester transfer protein;
trolled diabetes mellitus, the level may rise to as much
and (3) they act as ligands for interaction with lipopro-
as 2 µeq/mL.
LIPID TRANSPORT & STORAGE
/
207
Peripheral apoprotein
are also to be found in chyle; however, most of the
(eg, apo C)
plasma VLDL are of hepatic origin. They are the vehi-
cles of transport of triacylglycerol from the liver to
the extrahepatic tissues.
Free
Phospholipid
cholesterol
There are striking similarities in the mechanisms of
Cholesteryl
formation of chylomicrons by intestinal cells and of
ester
VLDL by hepatic parenchymal cells (Figure 25-2), per-
haps because—apart from the mammary gland—the
Triacylglycerol
intestine and liver are the only tissues from which par-
ticulate lipid is secreted. Newly secreted or “nascent”
chylomicrons and VLDL contain only a small amount
of apolipoproteins C and E, and the full complement is
acquired from HDL in the circulation (Figures 25-3
Core of mainly
and 25-4). Apo B is essential for chylomicron and
nonpolar lipids
VLDL formation. In abetalipoproteinemia (a rare dis-
ease), lipoproteins containing apo B are not formed and
Integral
apoprotein
Monolayer of mainly
lipid droplets accumulate in the intestine and liver.
(eg, apo B)
amphipathic lipids
A more detailed account of the factors controlling
hepatic VLDL secretion is given below.
Figure 25-1. Generalized structure of a plasma
lipoprotein. The similarities with the structure of the
plasma membrane are to be noted. Small amounts of
CHYLOMICRONS & VERY LOW
cholesteryl ester and triacylglycerol are to be found in
DENSITY LIPOPROTEINS ARE
the surface layer and a little free cholesterol in the core.
RAPIDLY CATABOLIZED
The clearance of labeled chylomicrons from the blood
Free fatty acids are removed from the blood ex-
is rapid, the half-time of disappearance being under 1
tremely rapidly and oxidized (fulfilling 25-50% of en-
hour in humans. Larger particles are catabolized more
ergy requirements in starvation) or esterified to form
quickly than smaller ones. Fatty acids originating from
triacylglycerol in the tissues. In starvation, esterified
chylomicron triacylglycerol are delivered mainly to adi-
lipids from the circulation or in the tissues are oxidized
pose tissue, heart, and muscle (80%), while about 20%
as well, particularly in heart and skeletal muscle cells,
goes to the liver. However, the liver does not metabo-
where considerable stores of lipid are to be found.
lize native chylomicrons or VLDL significantly; thus,
The free fatty acid uptake by tissues is related di-
the fatty acids in the liver must be secondary to their
rectly to the plasma free fatty acid concentration, which
metabolism in extrahepatic tissues.
in turn is determined by the rate of lipolysis in adipose
tissue. After dissociation of the fatty acid-albumin com-
plex at the plasma membrane, fatty acids bind to a
Triacylglycerols of Chylomicrons & VLDL
membrane fatty acid transport protein that acts as a
Are Hydrolyzed by Lipoprotein Lipase
transmembrane cotransporter with Na+. On entering
Lipoprotein lipase is located on the walls of blood cap-
the cytosol, free fatty acids are bound by intracellular
illaries, anchored to the endothelium by negatively
fatty acid-binding proteins. The role of these proteins
charged proteoglycan chains of heparan sulfate. It has
in intracellular transport is thought to be similar to that
been found in heart, adipose tissue, spleen, lung, renal
of serum albumin in extracellular transport of long-
medulla, aorta, diaphragm, and lactating mammary
chain fatty acids.
gland, though it is not active in adult liver. It is not
normally found in blood; however, following injection
TRIACYLGLYCEROL IS TRANSPORTED
of heparin, lipoprotein lipase is released from its hep-
FROM THE INTESTINES IN
aran sulfate binding into the circulation. Hepatic li-
pase is bound to the sinusoidal surface of liver cells and
CHYLOMICRONS & FROM THE LIVER IN
is released by heparin. This enzyme, however, does not
VERY LOW DENSITY LIPOPROTEINS
react readily with chylomicrons or VLDL but is con-
By definition, chylomicrons are found in chyle formed
cerned with chylomicron remnant and HDL metabo-
only by the lymphatic system draining the intestine.
lism.
They are responsible for the transport of all dietary
Both phospholipids and apo C-II are required as
lipids into the circulation. Small quantities of VLDL
cofactors for lipoprotein lipase activity, while apo A-II
208
/
CHAPTER 25
A
Intestinal lumen
B
RER
•
G
RER
SER
N
SER
Bile
G
canaliculus
N
C
VLDL
Fenestra
SD
Endothelial
cell
Blood
Lymph vessel leading
E
capillary
to thoracic duct
Lumen of blood sinusoid
Figure 25-2. The formation and secretion of (A) chylomicrons by an intestinal cell and (B) very low density
lipoproteins by a hepatic cell. (RER, rough endoplasmic reticulum; SER, smooth endoplasmic reticulum; G, Golgi
apparatus; N, nucleus; C, chylomicrons; VLDL, very low density lipoproteins; E, endothelium; SD, space of Disse,
containing blood plasma.) Apolipoprotein B, synthesized in the RER, is incorporated into lipoproteins in the SER,
the main site of synthesis of triacylglycerol. After addition of carbohydrate residues in G, they are released from
the cell by reverse pinocytosis. Chylomicrons pass into the lymphatic system. VLDL are secreted into the space
of Disse and then into the hepatic sinusoids through fenestrae in the endothelial lining.
and apo C-III act as inhibitors. Hydrolysis takes place
The Action of Lipoprotein Lipase Forms
while the lipoproteins are attached to the enzyme on
Remnant Lipoproteins
the endothelium. Triacylglycerol is hydrolyzed progres-
Reaction with lipoprotein lipase results in the loss of
sively through a diacylglycerol to a monoacylglycerol
approximately 90% of the triacylglycerol of chylomi-
that is finally hydrolyzed to free fatty acid plus glycerol.
crons and in the loss of apo C (which returns to HDL)
Some of the released free fatty acids return to the circu-
but not apo E, which is retained. The resulting chy-
lation, attached to albumin, but the bulk is transported
lomicron remnant is about half the diameter of the
into the tissue (Figures 25-3 and 25-4). Heart lipopro-
parent chylomicron and is relatively enriched in choles-
tein lipase has a low Km for triacylglycerol, about one-
terol and cholesteryl esters because of the loss of triacyl-
tenth of that for the enzyme in adipose tissue. This en-
glycerol
(Figure
25-3). Similar changes occur to
ables the delivery of fatty acids from triacylglycerol to
VLDL, with the formation of VLDL remnants or IDL
be redirected from adipose tissue to the heart in the
(intermediate-density lipoprotein) (Figure 25-4).
starved state when the plasma triacylglycerol decreases.
A similar redirection to the mammary gland occurs
during lactation, allowing uptake of lipoprotein triacyl-
The Liver Is Responsible for the Uptake
glycerol fatty acid for milk fat synthesis. The VLDL re-
of Remnant Lipoproteins
ceptor plays an important part in the delivery of fatty
acids from VLDL triacylglycerol to adipocytes by bind-
Chylomicron remnants are taken up by the liver by re-
ing VLDL and bringing it into close contact with
ceptor-mediated endocytosis, and the cholesteryl esters
lipoprotein lipase. In adipose tissue, insulin enhances
and triacylglycerols are hydrolyzed and metabolized.
lipoprotein lipase synthesis in adipocytes and its
Uptake is mediated by a receptor specific for apo E
translocation to the luminal surface of the capillary en-
(Figure 25-3), and both the LDL (apo B-100, E) re-
dothelium.
ceptor and the LRP (LDL receptor-related protein)
LIPID TRANSPORT & STORAGE
/
209
Dietary TG
Nascent
chylomicron
B-48
SMALL
Lymphatics
INTESTINE
TG
C
Chylomicron
A
B-48
EXTRAHEPATIC
TG
TISSUES
E
LDL
A
C
(apo B-100, E)
C
E
PC
C
receptor
A
HDL
Cholesterol
LIPOPROTEIN LIPASE
p
Fatty acids
B-48
TG
Fatty acids
C
LIVER
E
Chylomicron
Glycerol
LRP receptor
remnant
Figure 25-3. Metabolic fate of chylomicrons. (A, apolipoprotein A; B-48, apolipoprotein B-48; C ,
apolipoprotein C; E, apolipoprotein E; HDL, high-density lipoprotein; TG, triacylglycerol; C, cholesterol and
cholesteryl ester; P, phospholipid; HL, hepatic lipase; LRP, LDL receptor-related protein.) Only the predominant
lipids are shown.
are believed to take part. Hepatic lipase has a dual role:
graded in extrahepatic tissues and 70% in the liver. A
(1) in acting as a ligand to the lipoprotein and (2) in
positive correlation exists between the incidence of
hydrolyzing its triacylglycerol and phospholipid.
coronary atherosclerosis and the plasma concentra-
VLDL is the precursor of IDL, which is then con-
tion of LDL cholesterol. For further discussion of the
verted to LDL. Only one molecule of apo B-100 is
regulation of the LDL receptor, see Chapter 26.
present in each of these lipoprotein particles, and this is
conserved during the transformations. Thus, each LDL
HDL TAKES PART IN BOTH
particle is derived from only one VLDL particle (Figure
LIPOPROTEIN TRIACYLGLYCEROL
25-4). Two possible fates await IDL. It can be taken up
& CHOLESTEROL METABOLISM
by the liver directly via the LDL (apo B-100, E) recep-
tor, or it is converted to LDL. In humans, a relatively
HDL is synthesized and secreted from both liver and
large proportion forms LDL, accounting for the in-
intestine (Figure 25-5). However, apo C and apo E are
creased concentrations of LDL in humans compared
synthesized in the liver and transferred from liver HDL
with many other mammals.
to intestinal HDL when the latter enters the plasma. A
major function of HDL is to act as a repository for the
apo C and apo E required in the metabolism of chy-
LDL IS METABOLIZED VIA
lomicrons and VLDL. Nascent HDL consists of discoid
THE LDL RECEPTOR
phospholipid bilayers containing apo A and free choles-
The liver and many extrahepatic tissues express the
terol. These lipoproteins are similar to the particles
LDL (B-100, E) receptor. It is so designated because it
found in the plasma of patients with a deficiency of the
is specific for apo B-100 but not B-48, which lacks the
plasma enzyme lecithin:cholesterol acyltransferase
carboxyl terminal domain of B-100 containing the
(LCAT) and in the plasma of patients with obstructive
LDL receptor ligand, and it also takes up lipoproteins
jaundice. LCAT—and the LCAT activator apo A-I—
rich in apo E. This receptor is defective in familial hy-
bind to the disk, and the surface phospholipid and free
percholesterolemia. Approximately 30% of LDL is de-
cholesterol are converted into cholesteryl esters and
210
/
CHAPTER 25
Nascent
VLDL
B-100
TG
C
VLDL
E
B-100
C
EXTRAHEPATIC
TG
TISSUES
E
LDL
A
C
(apo B-100, E)
C
E
PC C
receptor
HDL
Fatty acids
LIPOPROTEIN LIPASE
B-100
TG
Cholesterol
?
B-100
C
E
Fatty acids
C
IDL
LIVER
(VLDL remnant)
LDL
LDL
(apo B-100, E)
Glycerol
receptor
Final destruction in
liver, extrahepatic
tissues (eg, lympho-
EXTRAHEPATIC
cytes, fibroblasts)
TISSUES
via endocytosis
Figure 25-4. Metabolic fate of very low density lipoproteins (VLDL) and production of low-density
lipoproteins (LDL). (A, apolipoprotein A; B-100, apolipoprotein B-100; C , apolipoprotein C; E, apolipoprotein
E; HDL, high-density lipoprotein; TG, triacylglycerol; IDL, intermediate-density lipoprotein; C, cholesterol and
cholesteryl ester; P, phospholipid.) Only the predominant lipids are shown. It is possible that some IDL is also
metabolized via the LRP.
lysolecithin (Chapter 24). The nonpolar cholesteryl es-
then esterified by LCAT, increasing the size of the par-
ters move into the hydrophobic interior of the bilayer,
ticles to form the less dense HDL2. The cycle is com-
whereas lysolecithin is transferred to plasma albumin.
pleted by the re-formation of HDL3, either after selec-
Thus, a nonpolar core is generated, forming a spherical,
tive delivery of cholesteryl ester to the liver via the
pseudomicellar HDL covered by a surface film of polar
SR-B1 or by hydrolysis of HDL2 phospholipid and tri-
lipids and apolipoproteins. In this way, the LCAT sys-
acylglycerol by hepatic lipase. In addition, free apo A-I
tem is involved in the removal of excess unesterified
is released by these processes and forms pre
-HDL
cholesterol from lipoproteins and tissues. The class B
after associating with a minimum amount of phospho-
scavenger receptor B1
(SR-B1) has recently been
lipid and cholesterol. Preβ-HDL is the most potent
identified as an HDL receptor in the liver and in
form of HDL in inducing cholesterol efflux from the
steroidogenic tissues. HDL binds to the receptor via
tissues to form discoidal HDL. Surplus apo A-I is de-
apo A-I and cholesteryl ester is selectively delivered to
stroyed in the kidney.
the cells, but the particle itself, including apo A-I, is not
HDL concentrations vary reciprocally with plasma
taken up. The transport of cholesterol from the tissues
triacylglycerol concentrations and directly with the ac-
to the liver is known as reverse cholesterol transport
tivity of lipoprotein lipase. This may be due to surplus
and is mediated by an HDL cycle (Figure 25-5). The
surface constituents, eg, phospholipid and apo A-I
smaller HDL3 accepts cholesterol from the tissues via
being released during hydrolysis of chylomicrons and
the ATP-binding cassette transporter-1
(ABC-1).
VLDL and contributing toward the formation of preβ-
ABC-1 is a member of a family of transporter proteins
HDL and discoidal HDL. HDL2 concentrations are in-
that couple the hydrolysis of ATP to the binding of a
versely related to the incidence of coronary athero-
substrate, enabling it to be transported across the mem-
sclerosis, possibly because they reflect the efficiency of
brane. After being accepted by HDL3, the cholesterol is
reverse cholesterol transport. HDLc (HDL1) is found in
LIPID TRANSPORT & STORAGE
/
211
Bile C and
bile acids
A-1
SMALL INTESTINE
PL
LIVER
C
Synthesis
LCAT
Synthesis
C
Kidney
C CE PL
Discoidal
HDL
SR-B1
HEPATIC
A-1
LIPASE
PL C
A-1
Preβ-HDL
Phospholipid
bilayer
A-1
C
A-1
TISSUES
CE
C
LCAT
PL
CE
ABC-1
C
PL
HDL2
HDL3
Figure 25-5. Metabolism of high-density lipoprotein (HDL) in reverse cholesterol transport.
(LCAT, lecithin:cholesterol acyltransferase; C, cholesterol; CE, cholesteryl ester; PL, phospholipid;
A-I, apolipoprotein A-I; SR-B1, scavenger receptor B1; ABC-1, ATP binding cassette transporter 1.)
Preβ-HDL, HDL2, HDL3—see Table 25-1. Surplus surface constituents from the action of lipopro-
tein lipase on chylomicrons and VLDL are another source of preβ-HDL. Hepatic lipase activity is
increased by androgens and decreased by estrogens, which may account for higher concentra-
tions of plasma HDL2 in women.
the blood of diet-induced hypercholesterolemic ani-
Hepatic VLDL Secretion Is Related
mals. It is rich in cholesterol, and its sole apolipopro-
to Dietary & Hormonal Status
tein is apo E. It appears that all plasma lipoproteins are
The cellular events involved in VLDL formation and
interrelated components of one or more metabolic cy-
secretion have been described above. Hepatic triacyl-
cles that together are responsible for the complex
glycerol synthesis provides the immediate stimulus for
process of plasma lipid transport.
the formation and secretion of VLDL. The fatty acids
used are derived from two possible sources: (1) synthe-
sis within the liver from acetyl-CoA derived mainly
THE LIVER PLAYS A CENTRAL ROLE IN
from carbohydrate (perhaps not so important in hu-
LIPID TRANSPORT & METABOLISM
mans) and (2) uptake of free fatty acids from the circu-
The liver carries out the following major functions in
lation. The first source is predominant in the well-fed
lipid metabolism: (1) It facilitates the digestion and ab-
condition, when fatty acid synthesis is high and the
sorption of lipids by the production of bile, which con-
level of circulating free fatty acids is low. As triacylglyc-
tains cholesterol and bile salts synthesized within the
erol does not normally accumulate in the liver under
liver de novo or from uptake of lipoprotein cholesterol
this condition, it must be inferred that it is transported
(Chapter 26). (2) The liver has active enzyme systems
from the liver in VLDL as rapidly as it is synthesized
for synthesizing and oxidizing fatty acids (Chapters 21
and that the synthesis of apo B-100 is not rate-limiting.
and 22) and for synthesizing triacylglycerols and phos-
Free fatty acids from the circulation are the main source
pholipids (Chapter 24). (3) It converts fatty acids to ke-
during starvation, the feeding of high-fat diets, or in di-
tone bodies (ketogenesis) (Chapter 22). (4) It plays an
abetes mellitus, when hepatic lipogenesis is inhibited.
integral part in the synthesis and metabolism of plasma
Factors that enhance both the synthesis of triacylglyc-
lipoproteins (this chapter).
erol and the secretion of VLDL by the liver include (1)
212
/
CHAPTER 25
the fed state rather than the starved state; (2) the feed-
causing lipid peroxidation. Some protection against this
ing of diets high in carbohydrate (particularly if they
is provided by the antioxidant action of vitamin E-sup-
contain sucrose or fructose), leading to high rates of li-
plemented diets. The action of ethionine is thought to
pogenesis and esterification of fatty acids; (3) high lev-
be due to a reduction in availability of ATP due to its
els of circulating free fatty acids;
(4) ingestion of
replacing methionine in S-adenosylmethionine, trap-
ethanol; and (5) the presence of high concentrations of
ping available adenine and preventing synthesis of
insulin and low concentrations of glucagon, which en-
ATP. Orotic acid also causes fatty liver; it is believed to
hance fatty acid synthesis and esterification and inhibit
interfere with glycosylation of the lipoprotein, thus in-
their oxidation (Figure 25-6).
hibiting release, and may also impair the recruitment of
triacylglycerol to the particles. A deficiency of vitamin
E enhances the hepatic necrosis of the choline defi-
CLINICAL ASPECTS
ciency type of fatty liver. Added vitamin E or a source
of selenium has a protective effect by combating lipid
Imbalance in the Rate of Triacylglycerol
peroxidation. In addition to protein deficiency, essen-
Formation & Export Causes Fatty Liver
tial fatty acid and vitamin deficiencies (eg, linoleic acid,
For a variety of reasons, lipid—mainly as triacylglyc-
pyridoxine, and pantothenic acid) can cause fatty infil-
erol—can accumulate in the liver (Figure 25-6). Exten-
tration of the liver.
sive accumulation is regarded as a pathologic condition.
When accumulation of lipid in the liver becomes
Ethanol Also Causes Fatty Liver
chronic, fibrotic changes occur in the cells that progress
to cirrhosis and impaired liver function.
Alcoholism leads to fat accumulation in the liver, hy-
Fatty livers fall into two main categories. The first
perlipidemia, and ultimately cirrhosis. The exact
type is associated with raised levels of plasma free
mechanism of action of ethanol in the long term is still
fatty acids resulting from mobilization of fat from adi-
uncertain. Ethanol consumption over a long period
pose tissue or from the hydrolysis of lipoprotein triacyl-
leads to the accumulation of fatty acids in the liver that
glycerol by lipoprotein lipase in extrahepatic tissues.
are derived from endogenous synthesis rather than from
The production of VLDL does not keep pace with the
increased mobilization from adipose tissue. There is no
increasing influx and esterification of free fatty acids, al-
impairment of hepatic synthesis of protein after ethanol
lowing triacylglycerol to accumulate, causing a fatty
ingestion. Oxidation of ethanol by alcohol dehydrogen-
liver. This occurs during starvation and the feeding of
ase leads to excess production of NADH.
high-fat diets. The ability to secrete VLDL may also be
impaired (eg, in starvation). In uncontrolled diabetes
ALCOHOL
DEHYDROGENASE
mellitus, twin lamb disease, and ketosis in cattle,
fatty infiltration is sufficiently severe to cause visible
CH3
CH2
OH
CH3
CHO
pallor (fatty appearance) and enlargement of the liver
NAD+ NADH + H+
with possible liver dysfunction.
Ethanol
Acetaldehyde
The second type of fatty liver is usually due to a
metabolic block in the production of plasma lipo-
The NADH generated competes with reducing
proteins, thus allowing triacylglycerol to accumulate.
equivalents from other substrates, including fatty acids,
Theoretically, the lesion may be due to (1) a block in
for the respiratory chain, inhibiting their oxidation, and
apolipoprotein synthesis, (2) a block in the synthesis of
decreasing activity of the citric acid cycle. The net effect
the lipoprotein from lipid and apolipoprotein, (3) a
of inhibiting fatty acid oxidation is to cause increased
failure in provision of phospholipids that are found in
esterification of fatty acids in triacylglycerol, resulting
lipoproteins, or (4) a failure in the secretory mechanism
in the fatty liver. Oxidation of ethanol leads to the for-
itself.
mation of acetaldehyde, which is oxidized by aldehyde
One type of fatty liver that has been studied exten-
dehydrogenase, producing acetate. Other effects of
sively in rats is due to a deficiency of choline, which
ethanol may include increased lipogenesis and choles-
has therefore been called a lipotropic factor. The an-
terol synthesis from acetyl-CoA, and lipid peroxidation.
tibiotic puromycin, ethionine (α-amino-γ-mercaptobu-
The increased [NADH]/[NAD+] ratio also causes in-
tyric acid), carbon tetrachloride, chloroform, phospho-
creased
[lactate]/[pyruvate], resulting in hyperlactic-
rus, lead, and arsenic all cause fatty liver and a marked
acidemia, which decreases excretion of uric acid, aggra-
reduction in concentration of VLDL in rats. Choline
vating gout. Some metabolism of ethanol takes place
will not protect the organism against these agents but
via a cytochrome P450-dependent microsomal ethanol
appears to aid in recovery. The action of carbon tetra-
oxidizing system (MEOS) involving NADPH and O2.
chloride probably involves formation of free radicals
This system increases in activity in chronic alcoholism
LIPID TRANSPORT & STORAGE
/
213
VLDL
Apo C
HDL
Nascent
Apo E
BLOOD
VLDL
Nascent
LIVER
VLDL
-
HEPATOCYTE
Glycosyl
Golgi complex
residues
Smooth
Orotic acid
Carbon tetrachloride
endoplasmic
Destruction
Puromycin
of surplus apo B-100
Amino
reticulum
Ethionine
Carbon
acids
tetrachloride
Cholesterol
Apo B-100
Protein
Cholesteryl
Apo C
synthesis
ester
Apo E
M
-
Polyribosomes
M
Rough
-
Membrane
endoplasmic
synthesis
reticulum
Nascent
polypeptide
chains of
Triacylglycerol*
Phospholipid
apo B-100
Cholesterol feeding
EFA deficiency
-
Lipid
EFA
Choline
TRIACYLGLYCEROL
deficiency
1,2-Diacylglycerol
CDP-choline
Phosphocholine
Choline
Glucagon
-
+
Insulin
Insulin
Ethanol
Acyl-CoA
Oxidation
–
Insulin
FFA
Lipogenesis from
carbohydrate
Figure 25-6. The synthesis of very low density lipoprotein (VLDL) in the liver and the possible loci of action of
factors causing accumulation of triacylglycerol and a fatty liver. (EFA, essential fatty acids; FFA, free fatty acids;
HDL, high-density lipoproteins; Apo, apolipoprotein; M, microsomal triacylglycerol transfer protein.) The pathways
indicated form a basis for events depicted in Figure 25-2. The main
triacylglycerol
pool in liver is not on the direct
pathway of VLDL synthesis from acyl-CoA. Thus, FFA, insulin, and glucagon have immediate effects on VLDL secre-
tion as their effects impinge directly on the small triacylglycerol* precursor pool. In the fully fed state, apo B-100 is
synthesized in excess of requirements for VLDL secretion and the surplus is destroyed in the liver. During transla-
tion of apo B-100, microsomal transfer protein-mediated lipid transport enables lipid to become associated with
the nascent polypeptide chain. After release from the ribosomes, these particles fuse with more lipids from the
smooth endoplasmic reticulum, producing nascent VLDL.
214
/
CHAPTER 25
and may account for the increased metabolic clearance
Glucose
in this condition. Ethanol will also inhibit the metabo-
Insulin
+
lism of some drugs, eg, barbiturates, by competing for
BLOOD
cytochrome P450-dependent enzymes.
ADIPOSE TISSUE
Glucose 6-phosphate
MEOS
CH3
CH2
OH
+ NADPH + H+ + O2
Ethanol
CH3
CHO
+ NADP+ + 2H2O
Glycolysis
Acetaldehyde
CO2
PPP
Acetyl-CoA
In some Asian populations and Native Americans,
NADPH + H+
alcohol consumption results in increased adverse reac-
CO2
tions to acetaldehyde owing to a genetic defect of mito-
chondrial aldehyde dehydrogenase.
Glycerol
Acyl-CoA
3-phosphate
Esterification
ADIPOSE TISSUE IS THE MAIN STORE
OF TRIACYLGLYCEROL IN THE BODY
ATP
CoA
TG
The triacylglycerol stores in adipose tissue are continu-
ACYL-CoA
ally undergoing lipolysis (hydrolysis) and reesterifica-
SYNTHETASE
HORMONE-
tion (Figure 25-7). These two processes are entirely dif-
SENSITIVE
ferent pathways involving different reactants and
LIPASE
enzymes. This allows the processes of esterification or
lipolysis to be regulated separately by many nutritional,
Lipolysis
metabolic, and hormonal factors. The resultant of these
two processes determines the magnitude of the free
FFA
FFA
Glycerol
(pool 2)
(pool 1)
fatty acid pool in adipose tissue, which in turn deter-
mines the level of free fatty acids circulating in the
plasma. Since the latter has most profound effects upon
LIPOPROTEIN
the metabolism of other tissues, particularly liver and
LIPASE
muscle, the factors operating in adipose tissue that reg-
ulate the outflow of free fatty acids exert an influence
FFA
Glycerol
far beyond the tissue itself.
TG
BLOOD
(chylomicrons, VLDL)
The Provision of Glycerol 3-Phosphate
Regulates Esterification: Lipolysis Is
Controlled by Hormone-Sensitive Lipase
FFA
Glycerol
(Figure 25-7)
Figure 25-7. Metabolism of adipose tissue. Hor-
Triacylglycerol is synthesized from acyl-CoA and glyc-
mone-sensitive lipase is activated by ACTH, TSH,
erol 3-phosphate (Figure 24-2). Because the enzyme
glucagon, epinephrine, norepinephrine, and vaso-
glycerol kinase is not expressed in adipose tissue, glyc-
pressin and inhibited by insulin, prostaglandin E1, and
erol cannot be utilized for the provision of glycerol
nicotinic acid. Details of the formation of glycerol
3-phosphate, which must be supplied by glucose via
3-phosphate from intermediates of glycolysis are
glycolysis.
shown in Figure 24-2. (PPP, pentose phosphate path-
Triacylglycerol undergoes hydrolysis by a hormone-
way; TG, triacylglycerol; FFA, free fatty acids; VLDL, very
sensitive lipase to form free fatty acids and glycerol.
low density lipoprotein.)
This lipase is distinct from lipoprotein lipase that cat-
alyzes lipoprotein triacylglycerol hydrolysis before its
uptake into extrahepatic tissues (see above). Since glyc-
erol cannot be utilized, it diffuses into the blood,
whence it is utilized by tissues such as those of the liver
and kidney, which possess an active glycerol kinase.
LIPID TRANSPORT & STORAGE
/
215
The free fatty acids formed by lipolysis can be recon-
regulated in a coordinate manner by phosphorylation-
verted in the tissue to acyl-CoA by acyl-CoA syn-
dephosphorylation mechanisms.
thetase and reesterified with glycerol 3-phosphate to
A principal action of insulin in adipose tissue is to
form triacylglycerol. Thus, there is a continuous cycle
inhibit the activity of hormone-sensitive lipase, reduc-
of lipolysis and reesterification within the tissue.
ing the release not only of free fatty acids but of glycerol
However, when the rate of reesterification is not suffi-
as well. Adipose tissue is much more sensitive to insulin
cient to match the rate of lipolysis, free fatty acids accu-
than are many other tissues, which points to adipose
mulate and diffuse into the plasma, where they bind to
tissue as a major site of insulin action in vivo.
albumin and raise the concentration of plasma free fatty
acids.
Several Hormones Promote Lipolysis
Increased Glucose Metabolism Reduces
Other hormones accelerate the release of free fatty acids
the Output of Free Fatty Acids
from adipose tissue and raise the plasma free fatty acid
concentration by increasing the rate of lipolysis of the
When the utilization of glucose by adipose tissue is in-
triacylglycerol stores (Figure 25-8). These include epi-
creased, the free fatty acid outflow decreases. However,
nephrine, norepinephrine, glucagon, adrenocorticotro-
the release of glycerol continues, demonstrating that the
pic hormone (ACTH), α- and β-melanocyte-stimulat-
effect of glucose is not mediated by reducing the rate of
ing hormones (MSH), thyroid-stimulating hormone
lipolysis. The effect is due to the provision of glycerol
(TSH), growth hormone (GH), and vasopressin. Many
3-phosphate, which enhances esterification of free fatty
of these activate the hormone-sensitive lipase. For an
acids. Glucose can take several pathways in adipose tis-
optimal effect, most of these lipolytic processes require
sue, including oxidation to CO2 via the citric acid
the presence of glucocorticoids and thyroid hor-
cycle, oxidation in the pentose phosphate pathway,
mones. These hormones act in a facilitatory or per-
conversion to long-chain fatty acids, and formation of
missive capacity with respect to other lipolytic en-
acylglycerol via glycerol
3-phosphate (Figure
25-7).
docrine factors.
When glucose utilization is high, a larger proportion of
The hormones that act rapidly in promoting lipoly-
the uptake is oxidized to CO2 and converted to fatty
sis, ie, catecholamines, do so by stimulating the activity
acids. However, as total glucose utilization decreases,
of adenylyl cyclase, the enzyme that converts ATP to
the greater proportion of the glucose is directed to the
cAMP. The mechanism is analogous to that responsible
formation of glycerol 3-phosphate for the esterification
for hormonal stimulation of glycogenolysis
(Chap-
of acyl-CoA, which helps to minimize the efflux of free
ter 18). cAMP, by stimulating cAMP-dependent pro-
fatty acids.
tein kinase, activates hormone-sensitive lipase. Thus,
processes which destroy or preserve cAMP influence
HORMONES REGULATE
lipolysis. cAMP is degraded to 5′-AMP by the enzyme
FAT MOBILIZATION
cyclic 3 ,5 -nucleotide phosphodiesterase. This en-
zyme is inhibited by methylxanthines such as caffeine
Insulin Reduces the Output
and theophylline. Insulin antagonizes the effect of the
of Free Fatty Acids
lipolytic hormones. Lipolysis appears to be more sensi-
The rate of release of free fatty acids from adipose tissue
tive to changes in concentration of insulin than are glu-
is affected by many hormones that influence either the
cose utilization and esterification. The antilipolytic ef-
rate of esterification or the rate of lipolysis. Insulin in-
fects of insulin, nicotinic acid, and prostaglandin E1 are
hibits the release of free fatty acids from adipose tissue,
accounted for by inhibition of the synthesis of cAMP at
which is followed by a fall in circulating plasma free
the adenylyl cyclase site, acting through a Gi protein.
fatty acids. It enhances lipogenesis and the synthesis of
Insulin also stimulates phosphodiesterase and the lipase
acylglycerol and increases the oxidation of glucose to
phosphatase that inactivates hormone-sensitive lipase.
CO2 via the pentose phosphate pathway. All of these ef-
The effect of growth hormone in promoting lipolysis is
fects are dependent on the presence of glucose and can
dependent on synthesis of proteins involved in the for-
be explained, to a large extent, on the basis of the abil-
mation of cAMP. Glucocorticoids promote lipolysis via
ity of insulin to enhance the uptake of glucose into adi-
synthesis of new lipase protein by a cAMP-independent
pose cells via the GLUT 4 transporter. Insulin also in-
pathway, which may be inhibited by insulin, and also
creases the activity of pyruvate dehydrogenase, acetyl-
by promoting transcription of genes involved in the
CoA carboxylase, and glycerol phosphate acyltrans-
cAMP signal cascade. These findings help to explain
ferase, reinforcing the effects of increased glucose up-
the role of the pituitary gland and the adrenal cortex in
take on the enhancement of fatty acid and acylglycerol
enhancing fat mobilization. The recently discovered
synthesis. These three enzymes are now known to be
body weight regulatory hormone, leptin, stimulates
216
/
CHAPTER 25
Epinephrine,
ACTH,
Insulin, prostaglandin E1,
norepinephrine
TSH,
nicotinic acid
(
)
glucagon
β-Adrenergic
-
blockers
ATP
+
+
+
-
Thyroid hormone
Hormone-sensitive
ADENYLYL
-
lipase b
Insulin
GTP
FFA
CYCLASE
(inactive)
ATP
+
P
i
+
-
Growth hormone
-
cAMP-
-
PP
+
i
dependent
+
Lipase
Inhibitors of
cAMP
Mg2
protein
phosphatase
protein synthesis
Adenosine
kinase
TRIACYL-
Methyl-
ADP
GLYCEROL
xanthines
–
PHOSPHODI-
Hormone-sensitive
–
(eg, caffeine)
ESTERASE
lipase a
(active)
FFA +
+
P
Diacylglycerol
-
+
Thyroid hormone
Hormone-sensitive
lipase
5′ AMP
-
Insulin
FFA +
Insulin
2-Monoacylglycerol
2-Monoacylglycerol
-
lipase
Inhibitors of
Glucocorticoids
protein synthesis
FFA + glycerol
Figure 25-8.
Control of adipose tissue lipolysis. (TSH, thyroid-stimulating hormone; FFA, free fatty acids.)
Note the cascade sequence of reactions affording amplification at each step. The lipolytic stimulus is “switched
off” by removal of the stimulating hormone; the action of lipase phosphatase; the inhibition of the lipase and
adenylyl cyclase by high concentrations of FFA; the inhibition of adenylyl cyclase by adenosine; and the removal
of cAMP by the action of phosphodiesterase. ACTH, TSH, and glucagon may not activate adenylyl cyclase in vivo,
since the concentration of each hormone required in vitro is much higher than is found in the circulation. Posi-
tive ( + ) and negative ( − ) regulatory effects are represented by broken lines and substrate flow by solid lines.
lipolysis and inhibits lipogenesis by influencing the ac-
citrate lyase, a key enzyme in lipogenesis, does not ap-
tivity of the enzymes in the pathways for the break-
pear to be present, and other lipogenic enzymes—eg,
down and synthesis of fatty acids.
glucose-6-phosphate dehydrogenase and the malic en-
The sympathetic nervous system, through liberation
zyme—do not undergo adaptive changes. Indeed, it has
of norepinephrine in adipose tissue, plays a central role
been suggested that in humans there is a “carbohydrate
in the mobilization of free fatty acids. Thus, the in-
excess syndrome” due to a unique limitation in ability
creased lipolysis caused by many of the factors de-
to dispose of excess carbohydrate by lipogenesis. In
scribed above can be reduced or abolished by denerva-
birds, lipogenesis is confined to the liver, where it is
tion of adipose tissue or by ganglionic blockade.
particularly important in providing lipids for egg for-
mation, stimulated by estrogens. Human adipose tissue
is unresponsive to most of the lipolytic hormones apart
A Variety of Mechanisms Have Evolved for
from the catecholamines.
Fine Control of Adipose Tissue Metabolism
On consideration of the profound derangement of
Human adipose tissue may not be an important site of
metabolism in diabetes mellitus (due in large part to
lipogenesis. There is no significant incorporation of
increased release of free fatty acids from the depots) and
glucose or pyruvate into long-chain fatty acids; ATP-
the fact that insulin to a large extent corrects the condi-
LIPID TRANSPORT & STORAGE
/
217
INNER
tion, it must be concluded that insulin plays a promi-
OUTSIDE
MITOCHONDRIAL
INSIDE
nent role in the regulation of adipose tissue metabolism.
MEMBRANE
BROWN ADIPOSE TISSUE
Norepinephine
PROMOTES THERMOGENESIS
F0
+
F
1
Brown adipose tissue is involved in metabolism particu-
ATP
H+
larly at times when heat generation is necessary. Thus,
synthase
cAMP
the tissue is extremely active in some species in arousal
F0
from hibernation, in animals exposed to cold (nonshiv-
+
ering thermogenesis), and in heat production in the
Heat
newborn animal. Though not a prominent tissue in hu-
H+
Hormone-
mans, it is present in normal individuals, where it could
sensitive
lipase
be responsible for “diet-induced thermogenesis.” It is
noteworthy that brown adipose tissue is reduced or ab-
+
Respiratory
sent in obese persons. The tissue is characterized by a
chain
well-developed blood supply and a high content of mi-
Triacyl-
tochondria and cytochromes but low activity of ATP
glycerol
synthase. Metabolic emphasis is placed on oxidation of
H+
H+
both glucose and fatty acids. Norepinephrine liberated
FFA
from sympathetic nerve endings is important in increas-
ing lipolysis in the tissue and increasing synthesis of
Thermogenin
lipoprotein lipase to enhance utilization of triacylglyc-
+
erol-rich lipoproteins from the circulation. Oxidation
Acyl-CoA
Reducing
and phosphorylation are not coupled in mitochondria
+
equivalents
of this tissue, and the phosphorylation that does occur
is at the substrate level, eg, at the succinate thiokinase
-
Heat
step and in glycolysis. Thus, oxidation produces much
heat, and little free energy is trapped in ATP. A ther-
Purine
mogenic uncoupling protein, thermogenin, acts as a
nucleotides
proton conductance pathway dissipating the electro-
chemical potential across the mitochondrial membrane
(Figure 25-9).
Carnitine
transporter
SUMMARY
• Since nonpolar lipids are insoluble in water, for
transport between the tissues in the aqueous blood
plasma they are combined with amphipathic lipids
Figure 25-9. Thermogenesis in brown adipose tis-
and proteins to make water-miscible lipoproteins.
sue. Activity of the respiratory chain produces heat in
• Four major groups of lipoproteins are recognized:
addition to translocating protons (Chapter 12). These
Chylomicrons transport lipids resulting from diges-
protons dissipate more heat when returned to the
tion and absorption. Very low density lipoproteins
inner mitochondrial compartment via thermogenin in-
(VLDL) transport triacylglycerol from the liver. Low-
stead of generating ATP when returning via the F1 ATP
density lipoproteins (LDL) deliver cholesterol to the
synthase. The passage of H+ via thermogenin is inhib-
tissues, and high-density lipoproteins (HDL) remove
ited by purine nucleotides when brown adipose tissue
cholesterol from the tissues in the process known as
is unstimulated. Under the influence of norepinephrine,
reverse cholesterol transport.
the inhibition is removed by the production of free
• Chylomicrons and VLDL are metabolized by hydrol-
fatty acids (FFA) and acyl-CoA. Note the dual role of
ysis of their triacylglycerol, and lipoprotein remnants
acyl-CoA in both facilitating the action of thermogenin
are left in the circulation. These are taken up by liver,
and supplying reducing equivalents for the respiratory
but some of the remnants
(IDL) resulting from
chain.
+
and − signify positive or negative regulatory
VLDL form LDL which is taken up by the liver and
effects.
other tissues via the LDL receptor.
218
/
CHAPTER 25
• Apolipoproteins constitute the protein moiety of
REFERENCES
lipoproteins. They act as enzyme activators (eg, apo
Chappell DA, Medh JD: Receptor-mediated mechanisms of
C-II and apo A-I) or as ligands for cell receptors (eg,
lipoprotein remnant catabolism. Prog Lipid Res 1998;37:
apo A-I, apo E, and apo B-100).
393.
• Triacylglycerol is the main storage lipid in adipose
Eaton S et al: Multiple biochemical effects in the pathogenesis of
tissue. Upon mobilization, free fatty acids and glyc-
fatty liver. Eur J Clin Invest 1997;27:719.
erol are released. Free fatty acids are an important
Goldberg IJ, Merkel M: Lipoprotein lipase: physiology, biochem-
fuel source.
istry and molecular biology. Front Biosci 2001;6:D388.
• Brown adipose tissue is the site of
“nonshivering
Holm C et al: Molecular mechanisms regulating hormone sensitive
lipase and lipolysis. Annu Rev Nutr 2000;20:365.
thermogenesis.” It is found in hibernating and new-
Kaikans RM, Bass NM, Ockner RK: Functions of fatty acid bind-
born animals and is present in small quantity in hu-
ing proteins. Experientia 1990;46:617.
mans. Thermogenesis results from the presence of an
Lardy H, Shrago E: Biochemical aspects of obesity. Annu Rev
uncoupling protein, thermogenin, in the inner mito-
Biochem 1990;59:689.
chondrial membrane.
Rye K-A et al: Overview of plasma lipid transport. In: Plasma
Lipids and Their Role in Disease. Barter PJ, Rye K-A (editors).
Harwood Academic Publishers, 1999.
Shelness GS, Sellers JA: Very-low-density lipoprotein assembly and
secretion. Curr Opin Lipidol 2001;12:151.
Various authors: Biochemistry of Lipids, Lipoproteins and Mem-
branes. Vance DE, Vance JE (editors). Elsevier, 1996.
Various authors: Brown adipose tissue—role in nutritional energet-
ics. (Symposium.) Proc Nutr Soc 1989;48:165.
Cholesterol Synthesis,Transport,
26
& Excretion
Peter A. Mayes, PhD, DSc, & Kathleen M. Botham, PhD, DSc
BIOMEDICAL IMPORTANCE
from mevalonate by loss of CO2 (Figure 26-2). (3) Six
isoprenoid units condense to form squalene. (4) Squa-
Cholesterol is present in tissues and in plasma either as
lene cyclizes to give rise to the parent steroid, lanos-
free cholesterol or as a storage form, combined with a
terol. (5) Cholesterol is formed from lanosterol (Figure
long-chain fatty acid as cholesteryl ester. In plasma,
26-3).
both forms are transported in lipoproteins (Chapter
25). Cholesterol is an amphipathic lipid and as such is
Step 1—Biosynthesis of Mevalonate: HMG-CoA
an essential structural component of membranes and of
(3-hydroxy-3-methylglutaryl-CoA) is formed by the re-
the outer layer of plasma lipoproteins. It is synthesized
actions used in mitochondria to synthesize ketone bod-
in many tissues from acetyl-CoA and is the precursor of
ies (Figure 22-7). However, since cholesterol synthesis
all other steroids in the body such as corticosteroids, sex
is extramitochondrial, the two pathways are distinct.
hormones, bile acids, and vitamin D. As a typical prod-
Initially, two molecules of acetyl-CoA condense to
uct of animal metabolism, cholesterol occurs in foods
form acetoacetyl-CoA catalyzed by cytosolic thiolase.
of animal origin such as egg yolk, meat, liver, and
Acetoacetyl-CoA condenses with a further molecule of
brain. Plasma low-density lipoprotein (LDL) is the ve-
acetyl-CoA catalyzed by HMG-CoA synthase to form
hicle of uptake of cholesterol and cholesteryl ester into
HMG-CoA, which is reduced to mevalonate by
many tissues. Free cholesterol is removed from tissues
NADPH catalyzed by HMG-CoA reductase. This is
by plasma high-density lipoprotein (HDL) and trans-
the principal regulatory step in the pathway of choles-
ported to the liver, where it is eliminated from the body
terol synthesis and is the site of action of the most effec-
either unchanged or after conversion to bile acids in the
tive class of cholesterol-lowering drugs, the HMG-CoA
process known as reverse cholesterol transport. Cho-
reductase inhibitors (statins) (Figure 26-1).
lesterol is a major constituent of gallstones. However,
Step 2—Formation of Isoprenoid Units: Meval-
its chief role in pathologic processes is as a factor in the
onate is phosphorylated sequentially by ATP by three
genesis of atherosclerosis of vital arteries, causing cere-
kinases, and after decarboxylation (Figure 26-2) the ac-
brovascular, coronary, and peripheral vascular disease.
tive isoprenoid unit, isopentenyl diphosphate, is
formed.
CHOLESTEROL IS DERIVED
Step 3—Six Isoprenoid Units Form Squalene:
ABOUT EQUALLY FROM THE DIET
Isopentenyl diphosphate is isomerized by a shift of the
& FROM BIOSYNTHESIS
double bond to form dimethylallyl diphosphate, then
condensed with another molecule of isopentenyl
A little more than half the cholesterol of the body arises
diphosphate to form the ten-carbon intermediate ger-
by synthesis (about 700 mg/d), and the remainder is
anyl diphosphate (Figure 26-2). A further condensa-
provided by the average diet. The liver and intestine ac-
tion with isopentenyl diphosphate forms farnesyl
count for approximately 10% each of total synthesis in
diphosphate. Two molecules of farnesyl diphosphate
humans. Virtually all tissues containing nucleated cells
condense at the diphosphate end to form squalene. Ini-
are capable of cholesterol synthesis, which occurs in the
tially, inorganic pyrophosphate is eliminated, forming
endoplasmic reticulum and the cytosol.
presqualene diphosphate, which is then reduced by
NADPH with elimination of a further inorganic py-
Acetyl-CoA Is the Source of All Carbon
rophosphate molecule.
Atoms in Cholesterol
Step 4—Formation of Lanosterol: Squalene can
The biosynthesis of cholesterol may be divided into five
fold into a structure that closely resembles the steroid
steps: (1) Synthesis of mevalonate occurs from acetyl-
nucleus (Figure 26-3). Before ring closure occurs, squa-
CoA (Figure 26-1). (2) Isoprenoid units are formed
lene is converted to squalene 2,3-epoxide by a mixed-
219
220
/
CHAPTER 26
O
Farnesyl Diphosphate Gives Rise
CH3
C S CoA
to Dolichol & Ubiquinone
2 Acetyl-CoA
The polyisoprenoids dolichol
(Figure
14-20 and
Chapter 47) and ubiquinone (Figure 12-5) are formed
THIOLASE
from farnesyl diphosphate by the further addition of up
to
16
(dolichol) or
3-7
(ubiquinone) isopentenyl
CoA SH
diphosphate residues, respectively. Some GTP-binding
CH3
O
proteins in the cell membrane are prenylated with far-
C
CH2
C
S
CoA
nesyl or geranylgeranyl (20 carbon) residues. Protein
Acetoacetyl-CoA
O
prenylation is believed to facilitate the anchoring of
O
proteins into lipoid membranes and may also be in-
H2O
CH3
C S CoA
volved in protein-protein interactions and membrane-
Acetyl-CoA
HMG-CoA SYNTHASE
associated protein trafficking.
CoA SH
CH3
O
CHOLESTEROL SYNTHESIS IS
CONTROLLED BY REGULATION
-OOC
CH2
C
CH
2
C
S CoA
OF HMG-CoA REDUCTASE
OH
3-Hydroxy-3-methylglutaryl-CoA (HMG-CoA)
Regulation of cholesterol synthesis is exerted near the
Bile acid, cholesterol
beginning of the pathway, at the HMG-CoA reductase
2NADPH + 2H+
step. The reduced synthesis of cholesterol in starving
Statins, eg,
animals is accompanied by a decrease in the activity of
HMG-CoA REDUCTASE
simvastatin
the enzyme. However, it is only hepatic synthesis that is
2NADP+ + CoA SH
inhibited by dietary cholesterol. HMG-CoA reductase
Mevalonate
CH3
in liver is inhibited by mevalonate, the immediate prod-
-
uct of the pathway, and by cholesterol, the main prod-
OOC
CH
2
C CH2
CH2
OH
uct. Cholesterol (or a metabolite, eg, oxygenated sterol)
OH
represses transcription of the HMG-CoA reductase
Mevalonate
gene and is also believed to influence translation. A di-
urnal variation occurs in both cholesterol synthesis
Figure 26-1. Biosynthesis of mevalonate. HMG-CoA
and reductase activity. In addition to these mechanisms
reductase is inhibited by atorvastatin, pravastatin, and
regulating the rate of protein synthesis, the enzyme ac-
simvastatin. The open and solid circles indicate the fate
tivity is also modulated more rapidly by posttransla-
of each of the carbons in the acetyl moiety of acetyl-
tional modification (Figure 26-4). Insulin or thyroid
CoA.
hormone increases HMG-CoA reductase activity,
whereas glucagon or glucocorticoids decrease it. Activ-
ity is reversibly modified by phosphorylation-dephos-
function oxidase in the endoplasmic reticulum, squa-
phorylation mechanisms, some of which may be
lene epoxidase. The methyl group on C14 is transferred
cAMP-dependent and therefore immediately responsive
to C13 and that on C8 to C14 as cyclization occurs, cat-
to glucagon. Attempts to lower plasma cholesterol in
alyzed by oxidosqualene:lanosterol cyclase.
humans by reducing the amount of cholesterol in the
Step 5—Formation of Cholesterol: The forma-
diet produce variable results. Generally, a decrease of
tion of cholesterol from lanosterol takes place in the
100 mg in dietary cholesterol causes a decrease of ap-
membranes of the endoplasmic reticulum and involves
proximately 0.13 mmol/L of serum.
changes in the steroid nucleus and side chain (Figure
26-3). The methyl groups on C14 and C4 are removed
MANY FACTORS INFLUENCE THE
to form 14-desmethyl lanosterol and then zymosterol.
CHOLESTEROL BALANCE IN TISSUES
The double bond at C8-C9 is subsequently moved to
C5-C6 in two steps, forming desmosterol. Finally, the
In tissues, cholesterol balance is regulated as follows (Fig-
double bond of the side chain is reduced, producing
ure 26-5): Cell cholesterol increase is due to uptake of
cholesterol. The exact order in which the steps de-
cholesterol-containing lipoproteins by receptors, eg, the
scribed actually take place is not known with cer-
LDL receptor or the scavenger receptor; uptake of free
tainty.
cholesterol from cholesterol-rich lipoproteins to the cell
ATP
ADP
CH3
OH
CH3
OH
Mg2+
-OOC
C
CH2
–OOC
C
CH2
MEVALONATE
CH2
CH2
OH
CH2
CH2
O P
KINASE
Mevalonate
Mevalonate 5-phosphate
ATP
PHOSPHOMEVALONATE
Mg2+
KINASE
ADP
ADP
ATP
CH
3
O P
CH3
OH
Mg2+
-OOC
C
CH2
-OOC
C
CH2
DIPHOSPHOMEVALONATE
CH2
CH
O P P
CH2
CH2
O P
P
2
KINASE
Mevalonate 3-phospho-5-diphosphate
Mevalonate 5-diphosphate
CO2 + Pi
HMG-CoA
DIPHOSPHO-
MEVALONATE
trans -Methyl-
CH3
DECARBOXYLASE
CH3
glutaconate
shunt
C
CH2
C
CH2
ISOPENTENYL-
CH3
CH
O P P
CH2
CH2
O P P
DIPHOSPHATE
3,3-Dimethylallyl
ISOMERASE
Isopentenyl
diphosphate
diphosphate
Isopentenyl tRNA
CIS-PRENYL
TRANSFERASE
PP
i
CH3
CH3
C
CH2
C
CH2
Prenylated proteins
CH3
CH
CH2
CH
O P P
Geranyl diphosphate
CIS-PRENYL
TRANSFERASE
PPi
TRANS-PRENYL
CIS-PRENYL
TRANSFERASE
TRANSFERASE
Side chain of
CH2
Dolichol
ubiquinone
O P P
Farnesyl diphosphate
Heme a
NADPH + H+
SQUALENE SYNTHETASE
Mg2+, Mn2+
2PPi
NADP+
CH2
*CH2
Squalene
Figure 26-2. Biosynthesis of squalene, ubiquinone, dolichol, and other polyisoprene derivatives. (HMG,
3-hydroxy-3-methylglutaryl;
⋅⋅, cytokinin.) A farnesyl residue is present in heme a of cytochrome oxidase.
⋅
The carbon marked with asterisk becomes C11 or C12 in squalene. Squalene synthetase is a microsomal en-
zyme; all other enzymes indicated are soluble cytosolic proteins, and some are found in peroxisomes.
221
222
/
CHAPTER 26
O
CH3
CH3
CH3
C
S
CoA
-OOC
CH2
C
CH2
CH2OH
3
CH C
CH CH2-
OH
CO2
H2O
Acetyl-CoA
Mevalonate
Isoprenoid unit
CH3
CH2
CH3
CH2
C
CH2
C
CH2
12
12
24
CH
3
24
CH3
CH2
13
CH
HC C
CH2
13
CH
HC C
Squalene
11
CH3
11
CH3
epoxide
CH2
CH
CH2
CH2
CH
CH2
1
CH3
1
CH3
CH
CH
C
CH2
CH
CH
C
CH2
2
14
2
14
8
SQUALENE
8
H
2C
C
C
CH3
EPOXIDASE
H
2C
C
C
CH3
CH3
CH3
3
NADPH
3
X6
HC
CH
CH2
1/2 O2
HC
CH
CH2
FAD
C
CH2
C
CH2
Squalene
O
OXIDOSQUALENE:
CH3
CH3
CH3
LANOSTEROL
CH3
CYCLASE
H
COOH
2CO2
14
14
8
NADPH
O2, NADPH
8
O2
NAD+
4
HO
HO
HO
Lanosterol
14-Desmethyl
Zymosterol
lanosterol
ISOMERASE
21
22
18
20
23
26
24
25
24
24
17
NADPH
NADPH
19
1112
13
16
27
C
D
14
15
O2
1
9
∆24-REDUCTASE
2
10
8
A
B
3
5
7
3
7
4
6
5
HO
HO
HO
-
Cholesterol
Desmosterol
∆7,24-Cholestadienol
(24-dehydrocholesterol)
Triparanol
Figure 26-3. Biosynthesis of cholesterol. The numbered positions are those of the steroid nucleus and the
open and solid circles indicate the fate of each of the carbons in the acetyl moiety of acetyl-CoA. Asterisks: Refer
to labeling of squalene in Figure 26-2.
CHOLESTEROL SYNTHESIS, TRANSPORT, & EXCRETION
/
223
REDUCTASE
ATP
KINASE
P
i
(inactive)
+
REDUCTASE
Insulin
PROTEIN
KINASE
?
PHOSPHATASES
KINASE
P
-
REDUCTASE
Glucagon
ADP
KINASE
H2O
(active)
+
ATP
ADP
Inhibitor-1-
cAMP
phosphate*
+
HMG-CoA
HMG-CoA
HMG-CoA
LDL-cholesterol
REDUCTASE
P
REDUCTASE
(active)
(inactive)
Cholesterol
H2O
Insulin
?
P
i
+
-
PROTEIN
Oxysterols
PHOSPHATASES
-
Enzyme synthesis
Figure 26-4.
Possible mechanisms in the regulation of cholesterol synthesis by HMG-CoA reductase. Insulin
has a dominant role compared with glucagon. Asterisk: See Figure 18-6.
membrane; cholesterol synthesis; and hydrolysis of cho-
ity; and down-regulates synthesis of the LDL receptor.
lesteryl esters by the enzyme cholesteryl ester hydrolase.
Thus, the number of LDL receptors on the cell surface
Decrease is due to efflux of cholesterol from the mem-
is regulated by the cholesterol requirement for mem-
brane to HDL, promoted by LCAT (lecithin:cholesterol
branes, steroid hormones, or bile acid synthesis (Figure
acyltransferase) (Chapter 25); esterification of cholesterol
26-5). The apo B-100, E receptor is a “high-affinity”
by ACAT (acyl-CoA:cholesterol acyltransferase); and uti-
LDL receptor, which may be saturated under most cir-
lization of cholesterol for synthesis of other steroids, such
cumstances. Other “low-affinity” LDL receptors also
as hormones, or bile acids in the liver.
appear to be present in addition to a scavenger path-
way, which is not regulated.
The LDL Receptor Is Highly Regulated
LDL (apo B-100, E) receptors occur on the cell surface
CHOLESTEROL IS TRANSPORTED
in pits that are coated on the cytosolic side of the cell
BETWEEN TISSUES IN PLASMA
membrane with a protein called clathrin. The glycopro-
LIPOPROTEINS
tein receptor spans the membrane, the B-100 binding
(Figure 26-6)
region being at the exposed amino terminal end. After
binding, LDL is taken up intact by endocytosis. The
In Western countries, the total plasma cholesterol in
apoprotein and cholesteryl ester are then hydrolyzed in
humans is about 5.2 mmol/L, rising with age, though
the lysosomes, and cholesterol is translocated into the
there are wide variations between individuals. The
cell. The receptors are recycled to the cell surface. This
greater part is found in the esterified form. It is trans-
influx of cholesterol inhibits in a coordinated man-
ported in lipoproteins of the plasma, and the highest
ner HMG-CoA synthase, HMG-CoA reductase, and,
proportion of cholesterol is found in the LDL. Dietary
therefore, cholesterol synthesis; stimulates ACAT activ-
cholesterol equilibrates with plasma cholesterol in days
224
/
CHAPTER 26
CELL MEMBRANE
Recycling
Receptor
–
vesicle
LDL (apo B -100, E)
synthesis
receptors
Cholesterol
(in coated pits)
Lysosome
synthesis
CE
-
Endosome
ACAT
C
+
CE
LDL
CE
Unesterified
CE
cholesterol
CE
Coated
pool
vesicle
(mainly in membranes)
Scavenger receptor or
LDL
CE
C
nonregulated pathway
CE
Lysosome
HYDROLASE
LDL
C
Synthesis
VLDL
of steroids
ABC-1
CE A-1
A-1
LCAT
PL
PL C
Preβ-HDL
HDL3
Figure 26-5. Factors affecting cholesterol balance at the cellular level. Reverse cholesterol transport may
be initiated by preβ HDL binding to the ABC-1 transporter protein via apo A-I. Cholesterol is then moved out
of the cell via the transporter, lipidating the HDL, and the larger particles then dissociate from the ABC-1 mol-
ecule. (C, cholesterol; CE, cholesteryl ester; PL, phospholipid; ACAT, acyl-CoA:cholesterol acyltransferase; LCAT,
lecithin:cholesterol acyltransferase; A-I, apolipoprotein A-I; LDL, low-density lipoprotein; VLDL, very low den-
sity lipoprotein.) LDL and HDL are not shown to scale.
and with tissue cholesterol in weeks. Cholesteryl ester
ates a concentration gradient and draws in cholesterol
in the diet is hydrolyzed to cholesterol, which is then
from tissues and from other lipoproteins (Figures 26-5
absorbed by the intestine together with dietary unesteri-
and 26-6), thus enabling HDL to function in reverse
fied cholesterol and other lipids. With cholesterol syn-
cholesterol transport (Figure 25-5).
thesized in the intestines, it is then incorporated into
chylomicrons. Of the cholesterol absorbed, 80-90% is
esterified with long-chain fatty acids in the intestinal
Cholesteryl Ester Transfer Protein
mucosa. Ninety-five percent of the chylomicron choles-
Facilitates Transfer of Cholesteryl Ester
terol is delivered to the liver in chylomicron remnants,
From HDL to Other Lipoproteins
and most of the cholesterol secreted by the liver in
This protein is found in plasma of humans and many
VLDL is retained during the formation of IDL and ul-
timately LDL, which is taken up by the LDL receptor
other species, associated with HDL. It facilitates transfer
of cholesteryl ester from HDL to VLDL, IDL, and LDL
in liver and extrahepatic tissues (Chapter 25).
in exchange for triacylglycerol, relieving product inhibi-
tion of LCAT activity in HDL. Thus, in humans, much
Plasma LCAT Is Responsible for Virtually
of the cholesteryl ester formed by LCAT finds its way to
All Plasma Cholesteryl Ester in Humans
the liver via VLDL remnants (IDL) or LDL (Figure
LCAT activity is associated with HDL containing apo
26-6). The triacylglycerol-enriched HDL2 delivers its
A-I. As cholesterol in HDL becomes esterified, it cre-
cholesterol to the liver in the HDL cycle (Figure 25-5).
CHOLESTEROL SYNTHESIS, TRANSPORT, & EXCRETION
/
225
ENTEROHEPATIC CIRCULATION
HEPATIC PORTAL VEIN
Diet (0.4 g/d)
C
CE
GALL
BLADDER
Synthesis
–
-
Bile acids
(total pool, 3-5 g)
BILE DUCT
Unesterified
cholesterol
CE
pool
C
ACAT
CE
Bile
C
C
acids
HL
VLDL
CE
C
TG
Chylomicron
ILEUM
CE
LDL
C
(apo B-100, E)
TG
LIVER
receptor
CE
LDL
C
LRP receptor
CE
C
CE
C
C
Bile acids
TG
TG
A-I
(0.6 g/d)
(0.4 g/d)
CE
CE
IDL
Feces
C
C
HDL
(VLDL remnant)
Chylomicron
remnant
LPL
C
LDL
(apo B-100, E)
C
receptor
EXTRAHEPATIC
C
Synthesis
TISSUES
CE
Figure 26-6. Transport of cholesterol between the tissues in humans. (C, unesterified cholesterol; CE, cho-
lesteryl ester; TG, triacylglycerol; VLDL, very low density lipoprotein; IDL, intermediate-density lipoprotein; LDL,
low-density lipoprotein; HDL, high-density lipoprotein; ACAT, acyl-CoA:cholesterol acyltransferase; LCAT,
lecithin:cholesterol acyltransferase; A-I, apolipoprotein A-I; CETP, cholesteryl ester transfer protein; LPL, lipopro-
tein lipase; HL, hepatic lipase; LRP, LDL receptor-related protein.)
CHOLESTEROL IS EXCRETED FROM THE
feces; it is formed from cholesterol by the bacteria in
the lower intestine.
BODY IN THE BILE AS CHOLESTEROL OR
BILE ACIDS (SALTS)
Bile Acids Are Formed From Cholesterol
About 1 g of cholesterol is eliminated from the body
per day. Approximately half is excreted in the feces after
The primary bile acids are synthesized in the liver from
conversion to bile acids. The remainder is excreted as
cholesterol. These are cholic acid (found in the largest
cholesterol. Coprostanol is the principal sterol in the
amount) and chenodeoxycholic acid (Figure 26-7).
226
/
CHAPTER 26
Vitamin C
12
17
NADPH + H+
NADP+
O2
3
7
7
7α-HYDROXYLASE
HO
HO
OH
Cholesterol
7α-Hydroxycholesterol
12α-HYDROX-
Bile
YLASE
acids
Vitamin C
O2
O2
deficiency
NADPH + H+
NADPH + H+
(Several
2 CoA SH
steps)
2 CoA SH
Propionyl-CoA
Propionyl-CoA
C
S CoA
OH
H
O
C
N
(CH2)
SO3H
2
HO
OH
O
CoA SH
OH
H
Taurine
12
C
S CoA
Chenodeoxycholyl-CoA
HO
OH
H
O
Taurocholic acid
Glycine
(primary bile acid)
CoA SH
HO
OH
H
Tauro- and glyco-
Cholyl-CoA
chenodeoxycholic acid
(primary bile acids)
OH
H
Deconjugation
C
N
CH2COOH
+ 7α-dehydroxylation
O
OH
HO
OH
H
COOH
COOH
Glycocholic acid
(primary bile acid)
*
Deconjugation
HO
HO
H
H
+ 7α-dehydroxylation
Deoxycholic acid
Lithocholic acid
(secondary bile acid)
(secondary bile acid)
Figure 26-7. Biosynthesis and degradation of bile acids. A second pathway in mitochondria involves hy-
droxylation of cholesterol by sterol 27-hydroxylase. Asterisk: Catalyzed by microbial enzymes.
The 7α-hydroxylation of cholesterol is the first and
chenodeoxycholyl-CoA (Figure 26-7). A second path-
principal regulatory step in the biosynthesis of bile acids
way in mitochondria involving the 27-hydroxylation of
catalyzed by 7
-hydroxylase, a microsomal enzyme. A
cholesterol by sterol 27-hydroxylase as the first step is
typical monooxygenase, it requires oxygen, NADPH,
responsible for a significant proportion of the primary
and cytochrome P450. Subsequent hydroxylation steps
bile acids synthesized. The primary bile acids (Figure
are also catalyzed by monooxygenases. The pathway of
26-7) enter the bile as glycine or taurine conjugates.
bile acid biosynthesis divides early into one subpathway
Conjugation takes place in peroxisomes. In humans, the
leading to cholyl-CoA, characterized by an extra α-OH
ratio of the glycine to the taurine conjugates is normally
group on position 12, and another pathway leading to
3:1. In the alkaline bile, the bile acids and their conju-
CHOLESTEROL SYNTHESIS, TRANSPORT, & EXCRETION
/
227
gates are assumed to be in a salt form—hence the term
ized by the deposition of cholesterol and cholesteryl
“bile salts.”
ester from the plasma lipoproteins into the artery wall.
A portion of the primary bile acids in the intestine is
Diseases in which prolonged elevated levels of VLDL,
subjected to further changes by the activity of the in-
IDL, chylomicron remnants, or LDL occur in the
testinal bacteria. These include deconjugation and 7α-
blood (eg, diabetes mellitus, lipid nephrosis, hypothy-
dehydroxylation, which produce the secondary bile
roidism, and other conditions of hyperlipidemia) are
acids, deoxycholic acid and lithocholic acid.
often accompanied by premature or more severe ather-
osclerosis. There is also an inverse relationship between
HDL (HDL2) concentrations and coronary heart dis-
Most Bile Acids Return to the Liver
ease, and some consider that the most predictive rela-
in the Enterohepatic Circulation
tionship is the LDL:HDL cholesterol ratio. This is
Although products of fat digestion, including choles-
consistent with the function of HDL in reverse choles-
terol, are absorbed in the first 100 cm of small intestine,
terol transport. Susceptibility to atherosclerosis varies
the primary and secondary bile acids are absorbed al-
widely among species, and humans are one of the few
most exclusively in the ileum, and 98-99% are re-
in which the disease can be induced by diets high in
turned to the liver via the portal circulation. This is
cholesterol.
known as the enterohepatic circulation (Figure 26-6).
However, lithocholic acid, because of its insolubility, is
Diet Can Play an Important Role in
not reabsorbed to any significant extent. Only a small
Reducing Serum Cholesterol
fraction of the bile salts escapes absorption and is there-
fore eliminated in the feces. Nonetheless, this represents
Hereditary factors play the greatest role in determining
a major pathway for the elimination of cholesterol.
individual serum cholesterol concentrations; however,
Each day the small pool of bile acids (about 3-5 g) is
dietary and environmental factors also play a part, and
cycled through the intestine six to ten times and an
the most beneficial of these is the substitution in the
amount of bile acid equivalent to that lost in the feces is
diet of polyunsaturated and monounsaturated fatty
synthesized from cholesterol, so that a pool of bile acids
acids for saturated fatty acids. Plant oils such as corn oil
of constant size is maintained. This is accomplished by
and sunflower seed oil contain a high proportion of
a system of feedback controls.
polyunsaturated fatty acids, while olive oil contains a
high concentration of monounsaturated fatty acids. On
the other hand, butterfat, beef fat, and palm oil contain
Bile Acid Synthesis Is Regulated
a high proportion of saturated fatty acids. Sucrose and
at the 7
-Hydroxylase Step
fructose have a greater effect in raising blood lipids, par-
The principal rate-limiting step in the biosynthesis of
ticularly triacylglycerols, than do other carbohydrates.
bile acids is at the cholesterol 7
-hydroxylase reac-
The reason for the cholesterol-lowering effect of
tion (Figure 26-7). The activity of the enzyme is feed-
polyunsaturated fatty acids is still not fully understood.
back-regulated via the nuclear bile acid-binding recep-
It is clear, however, that one of the mechanisms in-
tor farnesoid X receptor (FXR). When the size of the
volved is the up-regulation of LDL receptors by poly-
bile acid pool in the enterohepatic circulation increases,
and monounsaturated as compared with saturated fatty
FXR is activated and transcription of the cholesterol
acids, causing an increase in the catabolic rate of LDL,
7α-hydroxylase gene is suppressed. Chenodeoxycholic
the main atherogenic lipoprotein. In addition, saturated
acid is particularly important in activating FXR. Cho-
fatty acids cause the formation of smaller VLDL parti-
lesterol
7α-hydroxylase activity is also enhanced by
cles that contain relatively more cholesterol, and they
cholesterol of endogenous and dietary origin and regu-
are utilized by extrahepatic tissues at a slower rate than
lated by insulin, glucagon, glucocorticoids, and thyroid
are larger particles—tendencies that may be regarded as
hormone.
atherogenic.
Lifestyle Affects the Serum
CLINICAL ASPECTS
Cholesterol Level
The Serum Cholesterol Is Correlated With
Additional factors considered to play a part in coronary
the Incidence of Atherosclerosis &
heart disease include high blood pressure, smoking,
Coronary Heart Disease
male gender, obesity (particularly abdominal obesity),
While cholesterol is believed to be chiefly concerned in
lack of exercise, and drinking soft as opposed to hard
the relationship, other serum lipids such as triacylglyc-
water. Factors associated with elevation of plasma FFA
erols may also play a role. Atherosclerosis is character-
followed by increased output of triacylglycerol and cho-
228
/
CHAPTER 26
lesterol into the circulation in VLDL include emotional
of coronary heart disease. This may be due to elevation
stress and coffee drinking. Premenopausal women ap-
of HDL concentrations resulting from increased syn-
pear to be protected against many of these deleterious
thesis of apo A-I and changes in activity of cholesteryl
factors, and this is thought to be related to the benefi-
ester transfer protein. It has been claimed that red wine
cial effects of estrogen. There is an association between
is particularly beneficial, perhaps because of its content
moderate alcohol consumption and a lower incidence
of antioxidants. Regular exercise lowers plasma LDL
Table 26-1. Primary disorders of plasma lipoproteins (dyslipoproteinemias).
Name
Defect
Remarks
Hypolipoproteinemias
No chylomicrons, VLDL, or LDL are
Rare; blood acylglycerols low; intestine and liver
Abetalipoproteinemia
formed because of defect in the
accumulate acylglycerols. Intestinal malabsorp-
loading of apo B with lipid.
tion. Early death avoidable by administration of
large doses of fat-soluble vitamins, particularly
vitamin E.
Familial alpha-lipoprotein deficiency
All have low or near absence of HDL.
Tendency toward hypertriacylglycerolemia as a
Tangier disease
result of absence of apo C-II, causing inactive
Fish-eye disease
LPL. Low LDL levels. Atherosclerosis in the el-
Apo-A-I deficiencies
derly.
Hyperlipoproteinemias
Hypertriacylglycerolemia due to de-
Slow clearance of chylomicrons and VLDL. Low
Familial lipoprotein lipase
ficiency of LPL, abnormal LPL, or apo
levels of LDL and HDL. No increased risk of coro-
deficiency (type I)
C-II deficiency causing inactive LPL.
nary disease.
Familial hypercholesterolemia
Defective LDL receptors or mutation
Elevated LDL levels and hypercholesterolemia,
(type IIa)
in ligand region of apo B-100.
resulting in atherosclerosis and coronary disease.
Familial type III hyperlipoprotein-
Deficiency in remnant clearance by
Increase in chylomicron and VLDL remnants of
emia (broad beta disease, rem-
the liver is due to abnormality in apo
density < 1.019 (β-VLDL). Causes hypercholes-
nant removal disease, familial
E. Patients lack isoforms E3 and E4
terolemia, xanthomas, and atherosclerosis.
dysbetalipoproteinemia)
and have only E2, which does not
react with the E receptor.1
Familial hypertriacylglycerolemia
Overproduction of VLDL often
Cholesterol levels rise with the VLDL concentra-
(type IV)
associated with glucose intolerance
tion. LDL and HDL tend to be subnormal. This
and hyperinsulinemia.
type of pattern is commonly associated with
coronary heart disease, type II diabetes mellitus,
obesity, alcoholism, and administration of
progestational hormones.
Familial hyperalphalipoproteinemia
Increased concentrations of HDL.
A rare condition apparently beneficial to health
and longevity.
Hepatic lipase deficiency
Deficiency of the enzyme leads to
Patients have xanthomas and coronary heart
accumulation of large triacylgly-
disease.
cerol-rich HDL and VLDL remnants.
Familial lecithin:cholesterol
Absence of LCAT leads to block in
Plasma concentrations of cholesteryl esters and
acyltransferase (LCAT) deficiency
reverse cholesterol transport. HDL
lysolecithin are low. Present is an abnormal LDL
remains as nascent disks incapable
fraction, lipoprotein X, found also in patients
of taking up and esterifying choles-
with cholestasis. VLDL is abnormal (β-VLDL).
terol.
Familial lipoprotein(a) excess
Lp(a) consists of 1 mol of LDL
Premature coronary heart disease due to athero-
attached to 1 mol of apo(a). Apo(a)
sclerosis, plus thrombosis due to inhibition of
shows structural homologies to plas-
fibrinolysis.
minogen.
1There is an association between patients possessing the apo E4 allele and the incidence of Alzheimer’s disease. Apparently, apo E4 binds
more avidly to β-amyloid found in neuritic plaques.
CHOLESTEROL SYNTHESIS, TRANSPORT, & EXCRETION
/
229
but raises HDL. Triacylglycerol concentrations are also
acids, and vitamin D. It also plays an important
reduced, due most likely to increased insulin sensitivity,
structural role in membranes and in the outer layer of
which enhances expression of lipoprotein lipase.
lipoproteins.
•
Cholesterol is synthesized in the body entirely from
When Diet Changes Fail, Hypolipidemic
acetyl-CoA. Three molecules of acetyl-CoA form
mevalonate via the important regulatory reaction for
Drugs Will Reduce Serum Cholesterol
the pathway, catalyzed by HMG-CoA reductase.
& Triacylglycerol
Next, a five-carbon isoprenoid unit is formed, and
Significant reductions of plasma cholesterol can be ef-
six of these condense to form squalene. Squalene un-
fected medically by the use of cholestyramine resin or
dergoes cyclization to form the parent steroid lanos-
surgically by the ileal exclusion operations. Both proce-
terol, which, after the loss of three methyl groups,
dures block the reabsorption of bile acids, causing in-
forms cholesterol.
creased bile acid synthesis in the liver. This increases
•
Cholesterol synthesis in the liver is regulated partly
cholesterol excretion and up-regulates LDL receptors,
by cholesterol in the diet. In tissues, cholesterol bal-
lowering plasma cholesterol. Sitosterol is a hypocholes-
ance is maintained between the factors causing gain
terolemic agent that acts by blocking the absorption of
of cholesterol (eg, synthesis, uptake via the LDL or
cholesterol from the gastrointestinal tract.
scavenger receptors) and the factors causing loss of
Several drugs are known to block the formation of
cholesterol (eg, steroid synthesis, cholesteryl ester for-
cholesterol at various stages in the biosynthetic path-
mation, excretion). The activity of the LDL receptor
way. The statins inhibit HMG-CoA reductase, thus
is modulated by cellular cholesterol levels to achieve
up-regulating LDL receptors. Statins currently in use
this balance. In reverse cholesterol transport, HDL
include atorvastatin, simvastatin, and pravastatin. Fi-
(preβ-HDL, discoidal, or HDL3) takes up cholesterol
brates such as clofibrate and gemfibrozil act mainly to
from the tissues and LCAT esterifies it and deposits
lower plasma triacylglycerols by decreasing the secretion
it in the core of HDL, which is converted to HDL2.
of triacylglycerol and cholesterol-containing VLDL by
The cholesteryl ester in HDL2 is taken up by the
the liver. In addition, they stimulate hydrolysis of
liver, either directly or after transfer to VLDL, IDL,
VLDL triacylglycerols by lipoprotein lipase. Probucol
or LDL via the cholesteryl ester transfer protein.
appears to increase LDL catabolism via receptor-
•
Excess cholesterol is excreted from the liver in the
independent pathways, but its antioxidant properties
bile as cholesterol or bile salts. A large proportion of
may be more important in preventing accumulation of
bile salts is absorbed into the portal circulation and
oxidized LDL, which has enhanced atherogenic proper-
returned to the liver as part of the enterohepatic cir-
ties, in arterial walls. Nicotinic acid reduces the flux of
culation.
FFA by inhibiting adipose tissue lipolysis, thereby in-
•
Elevated levels of cholesterol present in VLDL, IDL,
hibiting VLDL production by the liver.
or LDL are associated with atherosclerosis, whereas
high levels of HDL have a protective effect.
Primary Disorders of the Plasma
•
Inherited defects in lipoprotein metabolism lead to a
Lipoproteins (Dyslipoproteinemias)
primary condition of hypo- or hyperlipoproteinemia.
Are Inherited
Conditions such as diabetes mellitus, hypothy-
Inherited defects in lipoprotein metabolism lead to the
roidism, kidney disease, and atherosclerosis exhibit
secondary abnormal lipoprotein patterns that resem-
primary condition of either hypo- or hyperlipopro-
teinemia (Table 26-1). In addition, diseases such as
ble certain primary conditions.
diabetes mellitus, hypothyroidism, kidney disease
(nephrotic syndrome), and atherosclerosis are associ-
ated with secondary abnormal lipoprotein patterns that
REFERENCES
are very similar to one or another of the primary inher-
Illingworth DR: Management of hypercholesterolemia. Med Clin
ited conditions. Virtually all of the primary conditions
North Am 2000;84:23.
are due to a defect at a stage in lipoprotein formation,
Ness GC, Chambers CM: Feedback and hormonal regulation of
transport, or destruction (see Figures 25-4, 26-5, and
hepatic
3-hydroxy-3-methylglutaryl coenzyme A reductase:
26-6). Not all of the abnormalities are harmful.
the concept of cholesterol buffering capacity. Proc Soc Exp
Biol Med 2000;224:8.
SUMMARY
Parks DJ et al: Bile acids: natural ligands for a nuclear orphan re-
ceptor. Science 1999;284:1365.
• Cholesterol is the precursor of all other steroids in
Princen HMG: Regulation of bile acid synthesis. Curr Pharm De-
the body, eg, corticosteroids, sex hormones, bile
sign 1997;3:59.
230
/
CHAPTER 26
Russell DW: Cholesterol biosynthesis and metabolism. Cardiovas-
Various authors: The cholesterol facts. A summary of the evidence
cular Drugs Therap 1992;6:103.
relating dietary fats, serum cholesterol, and coronary heart
Spady DK, Woollett LA, Dietschy JM: Regulation of plasma LDL-
disease. Circulation 1990;81:1721.
cholesterol levels by dietary cholesterol and fatty acids. Annu
Zhang FL, Casey PJ: Protein prenylation: Molecular mechanisms
Rev Nutr 1993;13:355.
and functional consequences. Annu Rev Biochem
1996;
Tall A: Plasma lipid transfer proteins. Annu Rev Biochem 1995;
65:241.
64:235.
Various authors: Biochemistry of Lipids, Lipoproteins and Mem-
branes. Vance DE, Vance JE (editors). Elsevier, 1996.
Integration of Metabolism—
27
The Provision of Metabolic Fuels
David A Bender, PhD, & Peter A. Mayes, PhD, DSc
BIOMEDICAL IMPORTANCE
MANY METABOLIC FUELS
ARE INTERCONVERTIBLE
An adult human weighing 70 kg requires about 10-12
MJ (2400-2900 kcal) from metabolic fuels each day.
Carbohydrate in excess of immediate requirements as
This requirement is met from carbohydrates (40-60%),
fuel or for synthesis of glycogen in muscle and liver may
lipids
(mainly triacylglycerol, 30-40%), protein (10-
be used for lipogenesis (Chapter 21) and hence triacyl-
15%), and alcohol if consumed. The mix being oxi-
glycerol synthesis in both adipose tissue and liver
dized varies depending on whether the subject is in the
(whence it is exported in very low density lipoprotein).
fed or starving state and on the intensity of physical
The importance of lipogenesis in human beings is un-
work. The requirement for metabolic fuels is relatively
clear; in Western countries, dietary fat provides
constant throughout the day, since average physical ac-
35-45% of energy intake, while in less developed coun-
tivity only increases metabolic rate by about 40-50%
tries where carbohydrate may provide 60-75% of en-
over the basal metabolic rate. However, most people
ergy intake the total intake of food may be so low that
consume their daily intake of metabolic fuels in two or
there is little surplus for lipogenesis. A high intake of fat
three meals, so there is a need to form reserves of carbo-
inhibits lipogenesis.
hydrate (glycogen in liver and muscle) and lipid (tri-
Fatty acids (and ketone bodies formed from them)
acylglycerol in adipose tissue) for use between meals.
cannot be used for the synthesis of glucose. The reac-
If the intake of fuels is consistently greater than en-
tion of pyruvate dehydrogenase, forming acetyl-CoA, is
ergy expenditure, the surplus is stored, largely as fat,
irreversible, and for every two-carbon unit from acetyl-
leading to the development of obesity and its associated
CoA that enters the citric acid cycle there is a loss of
health hazards. If the intake of fuels is consistently
two carbon atoms as carbon dioxide before only one
lower than energy expenditure, there will be negligible
molecule of oxaloacetate is re-formed—ie, there is no
fat and carbohydrate reserves, and amino acids arising
net increase. This means that acetyl-CoA (and therefore
from protein turnover will be used for energy rather
any substrates that yield acetyl-CoA) can never be used
than replacement protein synthesis, leading to emacia-
for gluconeogenesis (Chapter 19). The (relatively rare)
tion and eventually death.
fatty acids with an odd number of carbon atoms yield
After a normal meal there is an ample supply of car-
propionyl-CoA as the product of the final cycle of β-
bohydrate, and the fuel for most tissues is glucose. In
oxidation (Chapter 22), and this can be a substrate for
the starving state, glucose must be spared for use by the
gluconeogenesis, as can the glycerol released by lipolysis
central nervous system (which is largely dependent on
of adipose tissue triacylglycerol reserves. Most of the
glucose) and the erythrocytes (which are wholly reliant
amino acids in excess of requirements for protein syn-
on glucose). Other tissues can utilize alternative fuels
thesis
(arising from the diet or from tissue protein
such as fatty acids and ketone bodies. As glycogen re-
turnover) yield pyruvate, or five- and four-carbon in-
serves become depleted, so amino acids arising from
termediates of the citric acid cycle. Pyruvate can be
protein turnover and glycerol arising from lipolysis are
carboxylated to oxaloacetate, which is the primary
used for gluconeogenesis. These events are largely con-
substrate for gluconeogenesis, and the five- and
trolled by the hormones insulin and glucagon. In dia-
four-carbon intermediates also result in a net increase in
betes mellitus there is either impaired synthesis and
the formation of oxaloacetate, which is then available
secretion of insulin (type 1 diabetes mellitus) or im-
for gluconeogenesis. These amino acids are classified as
paired sensitivity of tissues to insulin action (type 2 di-
glucogenic. Lysine and leucine yield only acetyl-CoA
abetes mellitus), leading to severe metabolic derange-
on oxidation and thus cannot be used for gluconeogen-
ment. In cattle the demands of heavy lactation can lead
esis, while phenylalanine, tyrosine, tryptophan, and
to ketosis, as can the demands of twin pregnancy in
isoleucine give rise to both acetyl-CoA and to interme-
sheep.
diates of the citric acid cycle that can be used for gluco-
231
232
/
CHAPTER 27
neogenesis. Those amino acids that give rise to acetyl-
rate of synthesis of glucose 6-phosphate. This is in ex-
CoA are classified as ketogenic because in the starving
cess of the liver’s requirement for energy and is used
state much of the acetyl-CoA will be used for synthesis
mainly for synthesis of glycogen. In both liver and
of ketone bodies in the liver.
skeletal muscle, insulin acts to stimulate glycogen syn-
thase and inhibit glycogen phosphorylase. Some of the
glucose entering the liver may also be used for lipogene-
sis and synthesis of triacylglycerol. In adipose tissue, in-
A SUPPLY OF METABOLIC FUELS
sulin stimulates glucose uptake, its conversion to fatty
IS PROVIDED IN BOTH THE FED
acids, and their esterification; and inhibits intracellular
& STARVING STATES
lipolysis and the release of free fatty acids.
(Figure 27-1)
The products of lipid digestion enter the circulation
as triacylglycerol-rich chylomicrons (Chapter 25). In
Glucose Is Always Required by the Central
adipose tissue and skeletal muscle, lipoprotein lipase is
Nervous System & Erythrocytes
activated in response to insulin; the resultant free fatty
Erythrocytes lack mitochondria and hence are wholly
acids are largely taken up to form triacylglycerol re-
reliant on glycolysis and the pentose phosphate path-
serves, while the glycerol remains in the blood stream
way. The brain can metabolize ketone bodies to meet
and is taken up by the liver and used for glycogen syn-
about 20% of its energy requirements; the remainder
thesis or lipogenesis. Free fatty acids remaining in the
must be supplied by glucose. The metabolic changes
blood stream are taken up by the liver and reesterified.
that occur in starvation are the consequences of the
The lipid-depleted chylomicron remnants are also
need to preserve glucose and the limited reserves of
cleared by the liver, and surplus liver triacylglycerol—
glycogen in liver for use by the brain and erythrocytes
including that from lipogenesis—is exported in very
and to ensure the provision of alternative fuels for other
low density lipoprotein.
tissues. The fetus and synthesis of lactose in milk also
Under normal feeding patterns the rate of tissue
require a significant amount of glucose.
protein catabolism is more or less constant throughout
the day; it is only in cachexia that there is an increased
rate of protein catabolism. There is net protein catabo-
lism in the postabsorptive phase of the feeding cycle
In the Fed State, Metabolic Fuel
and net protein synthesis in the absorptive phase, when
Reserves Are Laid Down
the rate of synthesis increases by about 20-25%. The
For several hours after a meal, while the products of di-
increased rate of protein synthesis is, again, a response
gestion are being absorbed, there is an abundant supply
to insulin action. Protein synthesis is an energy-expen-
of metabolic fuels. Under these conditions, glucose is
sive process, accounting for up to almost 20% of energy
the major fuel for oxidation in most tissues; this is ob-
expenditure in the fed state, when there is an ample
served as an increase in the respiratory quotient (the
supply of amino acids from the diet, but under 9% in
ratio of carbon dioxide produced to oxygen consumed)
the starved state.
from about 0.8 in the starved state to near 1 (Table
27-1).
Metabolic Fuel Reserves Are Mobilized
Glucose uptake into muscle and adipose tissue is
in the Starving State
controlled by insulin, which is secreted by the B islet
cells of the pancreas in response to an increased concen-
There is a small fall in plasma glucose upon starvation,
tration of glucose in the portal blood. An early response
then little change as starvation progresses (Table 27-2;
to insulin in muscle and adipose tissue is the migration
Figure
27-2). Plasma free fatty acids increase with
of glucose transporter vesicles to the cell surface, expos-
onset of starvation but then plateau. There is an initial
ing active glucose transporters (GLUT 4). These in-
delay in ketone body production, but as starvation pro-
sulin-sensitive tissues will only take up glucose from the
gresses the plasma concentration of ketone bodies in-
blood stream to any significant extent in the presence of
creases markedly.
the hormone. As insulin secretion falls in the starved
In the postabsorptive state, as the concentration of
state, so the transporters are internalized again, reduc-
glucose in the portal blood falls, so insulin secretion de-
ing glucose uptake.
creases, resulting in skeletal muscle and adipose tissue
The uptake of glucose into the liver is independent
taking up less glucose. The increase in secretion of
of insulin, but liver has an isoenzyme of hexokinase
glucagon from the A cells of the pancreas inhibits
(glucokinase) with a high Km, so that as the concentra-
glycogen synthase and activates glycogen phosphorylase
tion of glucose entering the liver increases, so does the
in liver. The resulting glucose 6-phosphate in liver is
INTEGRATION OF METABOLISM—THE PROVISION OF METABOLIC FUELS
/
233
Glucose 6-phosphate
Acyl-CoA
Glycerol 3-phosphate
ADIPOSE
TISSUE
TRIACYLGLYCEROL (TG)
cAMP
FFA
Glycerol
LPL
EXTRAHEPATIC
TISSUE (eg,
BLOOD
FFA
Glycerol
heart muscle)
Glycerol
Chylomicrons
GASTRO-
LPL
TG
INTESTINAL
(lipoproteins)
TRACT
FFA
FFA
Glucose
Glucose
Extra
glucose
drain (eg,
diabetes,
pregnancy,
VLDL
lactation)
Ketone bodies
FFA
TG
Glucose
LIVER
Acyl-CoA
Glycerol 3-phosphate
Acetyl-CoA
Glucose 6-phosphate
Citric
acid
2CO2
Amino acids,
Glycogen
cycle
lactate
Figure 27-1. Metabolic interrelationships between adipose tissue, the liver, and extrahepatic
tissues. In extrahepatic tissues such as heart, metabolic fuels are oxidized in the following order of
preference: (1) ketone bodies, (2) fatty acids, (3) glucose. (LPL, lipoprotein lipase; FFA, free fatty
acids; VLDL, very low density lipoproteins.)
234
/
CHAPTER 27
Table 27-1. Energy yields, oxygen consumption, and carbon
dioxide production in the oxidation of metabolic fuels.
Energy Yield
O2 Consumed
CO2 Produced
Oxygen
(kJ/g)
(L/g)
(L/g)
RQ
(kJ/L)
Carbohydrate
16
0.829
0.829
1.00
20
Protein
17
0.966
0.782
0.81
20
Fat
37
2.016
1.427
0.71
20
hydrolyzed by glucose-6-phosphatase, and glucose is re-
Although muscle takes up and preferentially oxidizes
leased into the blood stream for use by other tissues,
free fatty acids in the starving state, it cannot meet all of
particularly the brain and erythrocytes.
its energy requirements by β-oxidation. By contrast, the
Muscle glycogen cannot contribute directly to
liver has a greater capacity for β-oxidation than it re-
plasma glucose, since muscle lacks glucose-6-phos-
quires to meet its own energy needs and forms more
phatase, and the primary purpose of muscle glycogen is
acetyl-CoA than can be oxidized. This acetyl-CoA is
to provide a source of glucose 6-phosphate for energy-
used to synthesize ketone bodies (Chapter 22), which
yielding metabolism in the muscle itself. However,
are major metabolic fuels for skeletal and heart muscle
acetyl-CoA formed by oxidation of fatty acids in muscle
and can meet some of the brain’s energy needs. In pro-
inhibits pyruvate dehydrogenase and leads to citrate ac-
longed starvation, glucose may represent less than 10%
cumulation, which in turn inhibits phosphofructoki-
of whole body energy-yielding metabolism. Further-
nase and therefore glycolysis, thus sparing glucose. Any
more, as a result of protein catabolism, an increasing
accumulated pyruvate is transaminated to alanine at the
number of amino acids are released and utilized in the
expense of amino acids arising from breakdown of pro-
liver and kidneys for gluconeogenesis.
tein reserves. The alanine—and much of the keto acids
resulting from this transamination—are exported from
muscle and taken up by the liver, where the alanine is
Plasma glucagon
transaminated to yield pyruvate. The resultant amino
acids are largely exported back to muscle to provide
amino groups for formation of more alanine, while the
pyruvate is a major substrate for gluconeogenesis in the
liver.
In adipose tissue, the effect of the decrease in insulin
and increase in glucagon results in inhibition of lipo-
genesis, inactivation of lipoprotein lipase, and activa-
tion of hormone-sensitive lipase
(Chapter 25). This
leads to release of increased amounts of glycerol (a sub-
strate for gluconeogenesis in the liver) and free fatty
acids, which are used by skeletal muscle and liver as
their preferred metabolic fuels, so sparing glucose.
Blood glucose
Table 27-2. Plasma concentrations of metabolic
fuels (mmol/L) in the fed and starving states.
40 Hours
7 Days
Fed
Starvation
Starvation
0
12-24
Glucose
5.5
3.6
3.5
Hours of starvation
Free fatty acids
0.30
1.15
1.19
Figure 27-2. Relative changes in metabolic parame-
Ketone bodies
Negligible
2.9
4.5
ters during the onset of starvation.
236
/
CHAPTER 27
CLINICAL ASPECTS
A summary of the major and unique metabolic fea-
tures of the principal tissues is presented in Table 27-3.
In prolonged starvation, as adipose tissue reserves are
depleted there is a very considerable increase in the net
rate of protein catabolism to provide amino acids not
SUMMARY
only as substrates for gluconeogenesis but also as the
• The body can interconvert the majority of foodstuffs.
main metabolic fuel of the tissues. Death results when
essential tissue proteins are catabolized beyond the
However, there is no net conversion of most fatty
acids
(or other acetyl-CoA-forming substances) to
point at which they can sustain this metabolic drain. In
patients with cachexia as a result of release of cytokines
glucose. Most amino acids, arising from the diet or
from tissue protein, can be used for gluconeogenesis,
in response to tumors and a number of other patho-
logic conditions, there is an increase in the rate of tissue
as can the glycerol from triacylglycerol.
protein catabolism as well as a considerably increased
• In starvation, glucose must be provided for the brain
metabolic rate, resulting in a state of advanced starva-
and erythrocytes; initially, this is supplied from liver
tion. Again, death results when essential tissue proteins
glycogen reserves. To spare glucose, muscle and other
have been catabolized.
tissues reduce glucose uptake in response to lowered
The high demand for glucose by the fetus and for
insulin secretion; they also oxidize fatty acids and ke-
synthesis of lactose in lactation can lead to ketosis. This
tone bodies preferentially to glucose.
may be seen as mild ketosis with hypoglycemia in
• Adipose tissue releases free fatty acids in starvation,
women, but in lactating cattle and in ewes carrying
and these are used by many tissues as fuel. Further-
twins there may be very pronounced ketosis and pro-
more, in the liver they are the substrate for synthesis
found hypoglycemia.
of ketone bodies.
In poorly controlled type 1 diabetes mellitus, pa-
• Ketosis, a metabolic adaptation to starvation, is exac-
tients may become hyperglycemic, partly as a result of
erbated in pathologic conditions such as diabetes
lack of insulin to stimulate uptake and utilization of
mellitus and ruminant ketosis.
glucose and partly because of increased gluconeogenesis
from amino acids in the liver. At the same time, the
lack of insulin results in increased lipolysis in adipose
REFERENCES
tissue, and the resultant free fatty acids are substrates
for ketogenesis in the liver. It is possible that in very se-
Bender DA: Introduction to Nutrition and Metabolism, 3rd edition.
Taylor & Francis, 2002.
vere diabetes utilization of ketone bodies in muscle
Caprio S et al: Oxidative fuel metabolism during mild hypo-
(and other tissues) is impaired because of lack of ox-
glycemia: critical role of free fatty acids. Am J Physiol
aloacetate (most tissues have a requirement for some
1989;256:E413.
glucose metabolism to maintain an adequate amount of
Fell D: Understanding the Control of Metabolism. Portland Press,
oxaloacetate for citric acid cycle activity). In uncon-
1997.
trolled diabetes, the magnitude of ketosis may be such
Frayn KN: Metabolic Regulation—A Human Perspective. Portland
as to result in severe acidosis (ketoacidosis) since ace-
Press, 1996.
toacetic acid and 3-hydroxybutyric acid are relatively
McNamara JP: Role and regulation of metabolism in adipose tissue
strong acids. Coma results from both the acidosis and
during lactation. J Nutr Biochem 1995;6:120.
the considerably increased osmolarity of extracellular
Randle PJ: The glucose-fatty acid cycle—biochemical aspects. Ath-
fluid (mainly due to the hyperglycemia).
erosclerosis Rev 1991;22:183.
SECTION III
Metabolism of Proteins & Amino Acids
Biosynthesis of the Nutritionally
28
Nonessential Amino Acids
Victor W. Rodwell, PhD
BIOMEDICAL IMPORTANCE
those three enzymes is to transform ammonium ion
into the α-amino nitrogen of various amino acids.
All 20 of the amino acids present in proteins are essential
Glutamate and Glutamine. Reductive amination of
for health. While comparatively rare in the Western
α-ketoglutarate is catalyzed by glutamate dehydrogenase
world, amino acid deficiency states are endemic in cer-
(Figure 28-1). Amination of glutamate to glutamine is
tain regions of West Africa where the diet relies heavily
catalyzed by glutamine synthetase (Figure 28-2).
on grains that are poor sources of amino acids such as
Alanine. Transamination of pyruvate forms alanine
tryptophan and lysine. These disorders include kwash-
(Figure 28-3).
iorkor, which results when a child is weaned onto a
Aspartate and Asparagine. Transamination of
starchy diet poor in protein; and marasmus, in which
oxaloacetate forms aspartate. The conversion of aspartate
both caloric intake and specific amino acids are deficient.
Humans can synthesize 12 of the 20 common amino
acids from the amphibolic intermediates of glycolysis and
of the citric acid cycle (Table 28-1). While nutritionally
Table 28-1. Amino acid requirements
nonessential, these 12 amino acids are not “nonessential.”
of humans.
All 20 amino acids are biologically essential. Of the 12 nu-
tritionally nonessential amino acids, nine are formed from
Nutritionally Essential
Nutritionally Nonessential
amphibolic intermediates and three (cysteine, tyrosine
Arginine1
Alanine
and hydroxylysine) from nutritionally essential amino
Histidine
Asparagine
acids. Identification of the twelve amino acids that hu-
Isoleucine
Aspartate
mans can synthesize rested primarily on data derived from
Leucine
Cysteine
feeding diets in which purified amino acids replaced pro-
Lysine
Glutamate
tein. This chapter considers only the biosynthesis of the
Methionine
Glutamine
twelve amino acids that are synthesized in human tissues,
Phenylalanine
Glycine
not the other eight that are synthesized by plants.
Threonine
Hydroxyproline2
Tryptophan
Hydroxylysine2
NUTRITIONALLY NONESSENTIAL
Valine
Proline
Serine
AMINO ACIDS HAVE SHORT
Tyrosine
BIOSYNTHETIC PATHWAYS
1“Nutritionally semiessential.” Synthesized at rates inadequate
The enzymes glutamate dehydrogenase, glutamine syn-
to support growth of children.
thetase, and aminotransferases occupy central positions
2Not necessary for protein synthesis but formed during post-
in amino acid biosynthesis. The combined effect of
translational processing of collagen.
237
238
/
CHAPTER 28
+
+
O
NH3
O
NH3
-
-O
O
–O
O-
O-
O-
Pyruvate
Alanine
O
O
O
O
O
O
α-Ketoglutarate
L-Glutamate
NH4+
H2O
Glu or Asp
α-Ketoglutarate or oxaloacetate
+
NAD(P)H + H+
NAD(P)
Figure 28-3. Formation of alanine by transamina-
tion of pyruvate. The amino donor may be glutamate or
Figure 28-1. The glutamate dehydrogenase
aspartate. The other product thus is α-ketoglutarate or
reaction.
oxaloacetate.
to asparagine is catalyzed by asparagine synthetase (Fig-
ure 28-4), which resembles glutamine synthetase (Fig-
ure 28-2) except that glutamine, not ammonium ion,
+
provides the nitrogen. Bacterial asparagine synthetases
O NH3+
O NH
3
can, however, also use ammonium ion. Coupled hy-
O-
O-
-
O
H2N
drolysis of PPi to Pi by pyrophosphatase ensures that
O
O
the reaction is strongly favored.
Serine. Oxidation of the α-hydroxyl group of the
L-Aspartate
L-Asparagine
glycolytic intermediate 3-phosphoglycerate converts it
to an oxo acid, whose subsequent transamination and
Gln
Glu
dephosphorylation leads to serine (Figure 28-5).
Glycine. Glycine aminotransferases can catalyze the
synthesis of glycine from glyoxylate and glutamate or
Mg-ATP
Mg-AMP + PPi
alanine. Unlike most aminotransferase reactions, these
strongly favor glycine synthesis. Additional important
Figure 28-4. The asparagine synthetase reaction.
mammalian routes for glycine formation are from
Note similarities to and differences from the glutamine
choline (Figure 28-6) and from serine (Figure 28-7).
synthetase reaction (Figure 28-2).
Proline. Proline is formed from glutamate by rever-
sal of the reactions of proline catabolism (Figure 28-8).
Cysteine. Cysteine, while not nutritionally essen-
tial, is formed from methionine, which is nutritionally
essential. Following conversion of methionine to ho-
OH
O
O−
NADH
O−
P
O
O
P
O
O
D-3-Phosphoglycerate
Phosphohydroxy
pyruvate
+
+
α-AA
NH3
NH3
-O
O-
H2N
O-
α-KA
O
O
O
O
+
+
L-Glutamate
L-Glutamine
NH3
NH3
P
H2O
O−
i
O−
+
NH4
HO
O
P
O
O
L-Serine
Phospho-L-serine
Mg-ATP
Mg-ADP + Pi
Figure 28-5. Serine biosynthesis. (α-AA, α-amino
Figure 28-2. The glutamine synthetase reaction.
acids; α-KA, α-keto acids.)
BIOSYNTHESIS OF THE NUTRITIONALLY NONESSENTIAL AMINO ACIDS
/
239
CH3
2H
CH3
O
O
H3C
N+ CH3
H3C
N+ CH3
NADH
O−
O−
-O
O
NH3+
O
NH3+
Choline
Betaine aldehyde
H2O
OH
O
L-Glutamate
L-Glutamate-
γ-semialdehyde
NAD+
H2O
H3C
CH3
CH3
+
[CH3]
N
H
H3C
N+ CH3
O−
O
O
O−
Dimethylglycine O
Betaine
O−
NADH
O−
O
NH2+
NH+
[CH2O]
L-Proline
∆2-Pyrrolidine-
5-carboxylate
H
CH3
Figure 28-8. Biosynthesis of proline from glutamate
+
[CH2O]
NH3+
N
by reversal of reactions of proline catabolism.
H
O−
O−
Sarcosine
Glycine
O
O
Figure 28-6. Formation of glycine from choline.
NH3+
O−
mocysteine (see Chapter 30), homocysteine and serine
HO
O
form cysteine and homoserine (Figure 28-9).
+
L-Serine
Tyrosine. Phenylalanine hydroxylase converts
H3N+
S
H
phenylalanine to tyrosine
(Figure
28-10). Provided
H2O
that the diet contains adequate nutritionally essential
NH3+
phenylalanine, tyrosine is nutritionally nonessential.
O−
But since the reaction is irreversible, dietary tyrosine
L-Homocysteine
cannot replace phenylalanine. Catalysis by this mixed-
H3N+
S
O
function oxygenase incorporates one atom of O2 into
H2O
phenylalanine and reduces the other atom to water. Re-
O
ducing power, provided as tetrahydrobiopterin, derives
Cystathionine
ultimately from NADPH.
NH3+
O−
Methylene
H4 folate
H4 folate
HS
O
H3N+
OH
NH3+
L-Cysteine
NH3+
+
O-
O-
O
HO
O
O
L-Homoserine
Serine
Glycine
Figure 28-9. Conversion of homocysteine and ser-
Figure 28-7. The serine hydroxymethyltransferase
ine to homoserine and cysteine. The sulfur of cysteine
reaction. The reaction is freely reversible. (H4 folate,
derives from methionine and the carbon skeleton from
tetrahydrofolate.)
serine.
240
/
CHAPTER 28
NADP+ NADPH + H+
α-Ketoglutarate
[18O] Succinate
II
Fe2+
18O2
Tetrahydro-
Dihydro-
Ascorbate
18OH
biopterin
biopterin
Pro
Pro
I
O2
H2O
Figure 28-11. The prolyl hydroxylase reaction. The
CH2
CH
COO-
CH2
CH
COO−
substrate is a proline-rich peptide. During the course of
the reaction, molecular oxygen is incorporated into
NH3+
NH3+
both succinate and proline. Lysyl hydroxylase catalyzes
HO
an analogous reaction.
L-Phenylalanine
L-Tyrosine
H
H2N
N
N
amino acids, tissue aminotransferases reversibly inter-
HN
CH
CH
CH3
convert all three amino acids and their corresponding
N
H
α-keto acids. These α-keto acids thus can replace their
OH OH
amino acids in the diet.
Tetrahydrobiopterin
Selenocysteine. While not normally considered
an amino acid present in proteins, selenocysteine oc-
Figure 28-10. The phenylalanine hydroxylase reac-
curs at the active sites of several enzymes. Examples in-
tion. Two distinct enzymatic activities are involved. Ac-
clude the human enzymes thioredoxin reductase, glu-
tivity II catalyzes reduction of dihydrobiopterin by
tathione peroxidase, and the deiodinase that converts
NADPH, and activity I the reduction of O2 to H2O and of
thyroxine to triiodothyronine. Unlike hydroxyproline
phenylalanine to tyrosine. This reaction is associated
or hydroxylysine, selenocysteine arises co-translation-
with several defects of phenylalanine metabolism dis-
ally during its incorporation into peptides. The UGA
cussed in Chapter 30.
anticodon of the unusual tRNA designated tRNASec
normally signals STOP. The ability of the protein syn-
Hydroxyproline and Hydroxylysine. Hydroxy-
thetic apparatus to identify a selenocysteine-specific
proline and hydroxylysine are present principally in
UGA codon involves the selenocysteine insertion ele-
collagen. Since there is no tRNA for either hydroxy-
ment, a stem-loop structure in the untranslated region
lated amino acid, neither dietary hydroxyproline nor
of the mRNA. Selenocysteine-tRNASec is first charged
hydroxylysine is incorporated into protein. Both are
with serine by the ligase that charges tRNASer. Subse-
completely degraded (see Chapter 30). Hydroxyproline
quent replacement of the serine oxygen by selenium
and hydroxylysine arise from proline and lysine, but
involves selenophosphate formed by selenophosphate
only after these amino acids have been incorporated
synthase (Figure 28-12).
into peptides. Hydroxylation of peptide-bound prolyl
and lysyl residues is catalyzed by prolyl hydroxylase and
lysyl hydroxylase of tissues, including skin and skeletal
muscle, and of granulating wounds (Figure 28-11).
H
The hydroxylases are mixed-function oxygenases that
H Se
CH2
C
COO-
require substrate, molecular O2, ascorbate, Fe2+, and
α-ketoglutarate. For every mole of proline or lysine hy-
NH3+
droxylated, one mole of α-ketoglutarate is decarboxy-
O
lated to succinate. One atom of O2 is incorporated into
proline or lysine, the other into succinate
(Figure
Se + ATP
AMP + Pi
+ H Se
P
O-
28-11). A deficiency of the vitamin C required for
O-
these hydroxylases results in scurvy.
Valine, Leucine, and Isoleucine. While leucine,
Figure 28-12. Selenocysteine (top) and the reaction
valine, and isoleucine are all nutritionally essential
catalyzed by selenophosphate synthetase (bottom).
BIOSYNTHESIS OF THE NUTRITIONALLY NONESSENTIAL AMINO ACIDS
/
241
SUMMARY
• Selenocysteine, an essential active site residue in sev-
eral mammalian enzymes, arises by co-translational
• All vertebrates can form certain amino acids from
insertion of a previously modified tRNA.
amphibolic intermediates or from other dietary
amino acids. The intermediates and the amino acids
to which they give rise are α-ketoglutarate (Glu, Gln,
Pro, Hyp), oxaloacetate (Asp, Asn) and 3-phospho-
REFERENCES
glycerate (Ser, Gly).
Brown KM, Arthur JR: Selenium, selenoproteins and human
• Cysteine, tyrosine, and hydroxylysine are formed
health: a review. Public Health Nutr 2001;4:593.
from nutritionally essential amino acids. Serine pro-
Combs GF, Gray WP: Chemopreventive agents—selenium. Phar-
vides the carbon skeleton and homocysteine the sul-
macol Ther 1998;79:179.
fur for cysteine biosynthesis. Phenylalanine hydroxy-
Mercer LP, Dodds SJ, Smith DI: Dispensable, indispensable, and
lase converts phenylalanine to tyrosine.
conditionally indispensable amino acid ratios in the diet. In:
• Neither dietary hydroxyproline nor hydroxylysine is
Absorption and Utilization of Amino Acids. Friedman M (edi-
incorporated into proteins because no codon or
tor). CRC Press, 1989.
tRNA dictates their insertion into peptides.
Nordberg J et al: Mammalian thioredoxin reductase is irreversibly
inhibited by dinitrohalobenzenes by alkylation of both the
• Peptidyl hydroxyproline and hydroxylysine are
redox active selenocysteine and its neighboring cysteine
formed by hydroxylation of peptidyl proline or lysine
residue. J Biol Chem 1998;273:10835.
in reactions catalyzed by mixed-function oxidases
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
that require vitamin C as cofactor. The nutritional
herited Disease, 8th ed. McGraw-Hill, 2001.
disease scurvy reflects impaired hydroxylation due to
St Germain DL, Galton VA: The deiodinase family of selenopro-
a deficiency of vitamin C.
teins. Thyroid 1997;7:655.
Catabolism of Proteins
29
& of Amino Acid Nitrogen
Victor W. Rodwell, PhD
BIOMEDICAL IMPORTANCE
zymes have a t1/2 of 0.5-2 hours. PEST sequences, re-
gions rich in proline (P), glutamate (E), serine (S), and
This chapter describes how the nitrogen of amino acids is
threonine (T), target some proteins for rapid degrada-
converted to urea and the rare disorders that accompany
tion. Intracellular proteases hydrolyze internal peptide
defects in urea biosynthesis. In normal adults, nitrogen
bonds. The resulting peptides are then degraded to
intake matches nitrogen excreted. Positive nitrogen bal-
amino acids by endopeptidases that cleave internal
ance, an excess of ingested over excreted nitrogen, ac-
bonds and by aminopeptidases and carboxypeptidases
companies growth and pregnancy. Negative nitrogen
that remove amino acids sequentially from the amino
balance, where output exceeds intake, may follow
and carboxyl terminals, respectively. Degradation of
surgery, advanced cancer, and kwashiorkor or marasmus.
circulating peptides such as hormones follows loss of a
While ammonia, derived mainly from the α-amino
sialic acid moiety from the nonreducing ends of their
nitrogen of amino acids, is highly toxic, tissues convert
oligosaccharide chains. Asialoglycoproteins are internal-
ammonia to the amide nitrogen of nontoxic glutamine.
ized by liver cell asialoglycoprotein receptors and de-
Subsequent deamination of glutamine in the liver re-
graded by lysosomal proteases termed cathepsins.
leases ammonia, which is then converted to nontoxic
Extracellular, membrane-associated, and long-lived
urea. If liver function is compromised, as in cirrhosis or
intracellular proteins are degraded in lysosomes by
hepatitis, elevated blood ammonia levels generate clini-
ATP-independent processes. By contrast, degradation
cal signs and symptoms. Rare metabolic disorders in-
of abnormal and other short-lived proteins occurs in
volve each of the five urea cycle enzymes.
the cytosol and requires ATP and ubiquitin. Ubiquitin,
so named because it is present in all eukaryotic cells, is a
PROTEIN TURNOVER OCCURS
small (8.5 kDa) protein that targets many intracellular
proteins for degradation. The primary structure of
IN ALL FORMS OF LIFE
ubiquitin is highly conserved. Only 3 of 76 residues
The continuous degradation and synthesis of cellular
differ between yeast and human ubiquitin. Several mol-
proteins occur in all forms of life. Each day humans
ecules of ubiquitin are attached by non-α-peptide
turn over 1-2% of their total body protein, principally
bonds formed between the carboxyl terminal of ubiqui-
muscle protein. High rates of protein degradation occur
tin and the ε-amino groups of lysyl residues in the tar-
in tissues undergoing structural rearrangement—eg,
get protein (Figure 29-1). The residue present at its
uterine tissue during pregnancy, tadpole tail tissue dur-
amino terminal affects whether a protein is ubiquiti-
ing metamorphosis, or skeletal muscle in starvation. Of
nated. Amino terminal Met or Ser retards whereas Asp
the liberated amino acids, approximately 75% are reuti-
or Arg accelerates ubiquitination. Degradation occurs
lized. The excess nitrogen forms urea. Since excess
in a multicatalytic complex of proteases known as the
amino acids are not stored, those not immediately in-
proteasome.
corporated into new protein are rapidly degraded.
ANIMALS CONVERT
-AMINO NITROGEN
PROTEASES & PEPTIDASES DEGRADE
TO VARIED END PRODUCTS
PROTEINS TO AMINO ACIDS
Different animals excrete excess nitrogen as ammonia,
The susceptibility of a protein to degradation is ex-
uric acid, or urea. The aqueous environment of
pressed as its half-life (t1/2), the time required to lower
teleostean fish, which are ammonotelic (excrete ammo-
its concentration to half the initial value. Half-lives of
nia), compels them to excrete water continuously, facil-
liver proteins range from under 30 minutes to over 150
itating excretion of highly toxic ammonia. Birds, which
hours. Typical “housekeeping” enzymes have t1/2 values
must conserve water and maintain low weight, are uri-
of over 100 hours. By contrast, many key regulatory en-
cotelic and excrete uric acid as semisolid guano. Many
242
CATABOLISM OF PROTEINS & OF AMINO ACID NITROGEN
/
243
O
O
1.
UB C O- + E1 SH + ATP
AMP + PPi + UB C S E1
O
O
2.
UB C S E1 + E2 SH
E1 SH + UB C S E2
O
O
H
E3
3. UB C S E2 + H2N ε Protein
E2 SH + UB C N ε Protein
Figure 29-1. Partial reactions in the attachment of ubiquitin (UB) to
proteins. (1) The terminal COOH of ubiquitin forms a thioester bond with
an -SH of E1 in a reaction driven by conversion of ATP to AMP and PPi. Sub-
sequent hydrolysis of PPi by pyrophosphatase ensures that reaction 1 will
proceed readily. (2) A thioester exchange reaction transfers activated ubiq-
uitin to E2. (3) E3 catalyzes transfer of ubiquitin to ε-amino groups of lysyl
residues of target proteins.
land animals, including humans, are ureotelic and ex-
acids except lysine, threonine, proline, and hydroxypro-
crete nontoxic, water-soluble urea. High blood urea lev-
line participate in transamination. Transamination is
els in renal disease are a consequence—not a cause—of
readily reversible, and aminotransferases also function
impaired renal function.
in amino acid biosynthesis. The coenzyme pyridoxal
phosphate
(PLP) is present at the catalytic site of
aminotransferases and of many other enzymes that act
BIOSYNTHESIS OF UREA
on amino acids. PLP, a derivative of vitamin B6, forms
Urea biosynthesis occurs in four stages: (1) transamina-
an enzyme-bound Schiff base intermediate that can re-
tion, (2) oxidative deamination of glutamate, (3) am-
arrange in various ways. During transamination, bound
monia transport, and (4) reactions of the urea cycle
PLP serves as a carrier of amino groups. Rearrangement
(Figure 29-2).
forms an α-keto acid and enzyme-bound pyridoxamine
phosphate, which forms a Schiff base with a second
keto acid. Following removal of α-amino nitrogen by
Transamination Transfers α-Amino
transamination, the remaining carbon “skeleton” is de-
Nitrogen to α-Ketoglutarate,
graded by pathways discussed in Chapter 30.
Forming Glutamate
Alanine-pyruvate aminotransferase (alanine amino-
Transamination interconverts pairs of α-amino acids
transferase) and glutamate-α-ketoglutarate aminotrans-
and α-keto acids (Figure 29-3). All the protein amino
ferase (glutamate aminotransferase) catalyze the transfer
α-Amino acid
α-Keto acid
TRANSAMINATION
NH+
O
α-Ketoglutarate
L-Glutamate
CH
O-
C
O-
R1
C
R1
C
OXIDATIVE
O
O
DEAMINATION
NH3
CO2
O
NH+
-
UREA CYCLE
C
O
CH
R
C
R2
C O-
2
Urea
O
O
Figure 29-2. Overall flow of nitrogen in amino acid
Figure 29-3. Transamination. The reaction is freely
catabolism.
reversible with an equilibrium constant close to unity.
244
/
CHAPTER 29
of amino groups to pyruvate (forming alanine) or to α-
NAD(P)+
NAD(P)H + H+
ketoglutarate (forming glutamate) (Figure 29-4). Each
aminotransferase is specific for one pair of substrates
NH3
but nonspecific for the other pair. Since alanine is also a
substrate for glutamate aminotransferase, all the amino
L-Glutamate
α-Ketoglutarate
nitrogen from amino acids that undergo transamina-
tion can be concentrated in glutamate. This is impor-
Figure 29-5. The L-glutamate dehydrogenase reac-
tant because L-glutamate is the only amino acid that
tion. NAD(P)+ means that either NAD+ or NADP+ can
undergoes oxidative deamination at an appreciable rate
serve as co-substrate. The reaction is reversible but fa-
in mammalian tissues. The formation of ammonia
vors glutamate formation.
from α-amino groups thus occurs mainly via the α-
amino nitrogen of L-glutamate.
Transamination is not restricted to α-amino groups.
drogen peroxide (H2O2), which then is split to O2 and
The δ-amino group of ornithine—but not the ε-amino
H2O by catalase.
group of lysine—readily undergoes transamination.
Serum levels of aminotransferases are elevated in some
Ammonia Intoxication Is Life-Threatening
disease states (see Figure 7-11).
The ammonia produced by enteric bacteria and ab-
sorbed into portal venous blood and the ammonia pro-
L-GLUTAMATE DEHYDROGENASE
duced by tissues are rapidly removed from circulation
OCCUPIES A CENTRAL POSITION
by the liver and converted to urea. Only traces (10-20
µg/dL) thus normally are present in peripheral blood.
IN NITROGEN METABOLISM
This is essential, since ammonia is toxic to the central
Transfer of amino nitrogen to α-ketoglutarate forms L-
nervous system. Should portal blood bypass the liver,
glutamate. Release of this nitrogen as ammonia is then
systemic blood ammonia levels may rise to toxic levels.
catalyzed by hepatic L-glutamate dehydrogenase
This occurs in severely impaired hepatic function or the
(GDH), which can use either NAD+ or NADP+ (Fig-
development of collateral links between the portal and
ure 29-5). Conversion of α-amino nitrogen to ammo-
systemic veins in cirrhosis. Symptoms of ammonia in-
nia by the concerted action of glutamate aminotrans-
toxication include tremor, slurred speech, blurred vi-
ferase and GDH is often termed “transdeamination.”
sion, coma, and ultimately death. Ammonia may be
Liver GDH activity is allosterically inhibited by ATP,
toxic to the brain in part because it reacts with α-keto-
GTP, and NADH and activated by ADP. The reaction
glutarate to form glutamate. The resulting depleted lev-
catalyzed by GDH is freely reversible and functions also
els of α-ketoglutarate then impair function of the tri-
in amino acid biosynthesis (see Figure 28-1).
carboxylic acid (TCA) cycle in neurons.
Amino Acid Oxidases Also Remove
+
+
NH3
NH2
Nitrogen as Ammonia
AMINO ACID
C
O-
OXIDASE
C
O-
R
H
C
R
C
While their physiologic role is uncertain, L-amino acid
oxidases of liver and kidney convert amino acids to an
O
O
α-imino acid that decomposes to an α-keto acid with
α-Amino acid
Flavin
Flavin-H2
α-Imino acid
release of ammonium ion (Figure 29-6). The reduced
H2O
flavin is reoxidized by molecular oxygen, forming hy-
NH4+
H2O2
O2
O
Pyruvate
α-Amino acid
C
O-
CATALASE
R
C
L-Alanine
α-Keto acid
1/2O2
O
H
2O
α-Keto acid
α-Ketoglutarate
α-Amino acid
Figure 29-6. Oxidative deamination catalyzed by
L-Glutamate
α-Keto acid
L-amino acid oxidase (L-α-amino acid:O2 oxidoreduc-
Figure 29-4. Alanine aminotransferase (top) and
tase). The α-imino acid, shown in brackets, is not a
glutamate aminotransferase (bottom).
stable intermediate.
CATABOLISM OF PROTEINS & OF AMINO ACID NITROGEN
/
245
Glutamine Synthase Fixes Ammonia
NH+
as Glutamine
H2N
CH2
CH
O-
C
CH2
C
Formation of glutamine is catalyzed by mitochondrial
glutamine synthase (Figure 29-7). Since amide bond
O
O
synthesis is coupled to the hydrolysis of ATP to ADP
L-Glutamine
and Pi, the reaction strongly favors glutamine synthesis.
H2O
One function of glutamine is to sequester ammonia in
a nontoxic form.
GLUTAMINASE
NH+
Glutaminase & Asparaginase Deamidate
NH+
Glutamine & Asparagine
O
CH2
CH
O-
Hydrolytic release of the amide nitrogen of glutamine
C
CH2
C
as ammonia, catalyzed by glutaminase (Figure 29-8),
O
O
strongly favors glutamate formation. The concerted ac-
L-Glutamate
tion of glutamine synthase and glutaminase thus cat-
alyzes the interconversion of free ammonium ion and
Figure 29-8. The glutaminase reaction proceeds es-
glutamine. An analogous reaction is catalyzed by L-as-
sentially irreversibly in the direction of glutamate and
paraginase.
NH4+ formation. Note that the amide nitrogen, not the
α-amino nitrogen, is removed.
Formation & Secretion of Ammonia
Maintains Acid-Base Balance
Excretion into urine of ammonia produced by renal tubu-
reactions of Figure
29-9. Of the six participating
lar cells facilitates cation conservation and regulation of
amino acids, N-acetylglutamate functions solely as an
acid-base balance. Ammonia production from intracellu-
enzyme activator. The others serve as carriers of the
lar renal amino acids, especially glutamine, increases in
atoms that ultimately become urea. The major meta-
metabolic acidosis and decreases in metabolic alkalosis.
bolic role of ornithine, citrulline, and argininosucci-
nate in mammals is urea synthesis. Urea synthesis is a
cyclic process. Since the ornithine consumed in reac-
UREA IS THE MAJOR END PRODUCT OF
tion 2 is regenerated in reaction 5, there is no net loss
NITROGEN CATABOLISM IN HUMANS
or gain of ornithine, citrulline, argininosuccinate, or
Synthesis of 1 mol of urea requires 3 mol of ATP plus
arginine. Ammonium ion, CO2, ATP, and aspartate
1 mol each of ammonium ion and of the α-amino nitro-
are, however, consumed. Some reactions of urea syn-
gen of aspartate. Five enzymes catalyze the numbered
thesis occur in the matrix of the mitochondrion, other
reactions in the cytosol (Figure 29-9).
NH+
Carbamoyl Phosphate Synthase I
O
CH2
CH
O-
C
CH2
C
Initiates Urea Biosynthesis
O
O
Condensation of CO2, ammonia, and ATP to form
L-Glutamate
carbamoyl phosphate is catalyzed by mitochondrial
Mg-ATP
NH+
carbamoyl phosphate synthase I (reaction 1, Figure
29-9). A cytosolic form of this enzyme, carbamoyl
GLUTAMINE
phosphate synthase II, uses glutamine rather than am-
SYNTHASE
monia as the nitrogen donor and functions in pyrimi-
Mg-ADP
H2O
dine biosynthesis (see Chapter 34). Carbamoyl phos-
+ Pi
NH+
phate synthase I, the rate-limiting enzyme of the urea
cycle, is active only in the presence of its allosteric acti-
H2
N
CH2
CH
O-
C
CH2
C
vator N-acetylglutamate, which enhances the affinity
of the synthase for ATP. Formation of carbamoyl phos-
O
O
phate requires 2 mol of ATP, one of which serves as a
L-Glutamine
phosphate donor. Conversion of the second ATP to
Figure 29-7. The glutamine synthase reaction
AMP and pyrophosphate, coupled to the hydrolysis of
strongly favors glutamine synthesis.
pyrophosphate to orthophosphate, provides the driving
246
/
CHAPTER 29
CO2
NH4+
CO2 + NH4+
NH2
Urea
C O
CARBAMOYL
NH2
H2O
2Mg-ATP
NH3+
PHOSPHATE
5
SYNTHASE I
C
NH
N-Acetyl-
1
+
glutamate
CH2NH3
ARGINASE
CH2 NH
CH2
CH2
2Mg-ADP + Pi
CH2
CH2
+
+
H
C
NH3
H
C
NH3
COO−
COO−
L-Ornithine
L-Arginine
HC COO−
O
O
−
OOC
CH
H2N C
O
P
O−
ORNITHINE
TRANSCARBAMOYLASE
4
Fumarate
O−
Carbamoyl
phosphate
2
ARGININOSUCCINASE
P
i
NH2
NH
COO−
C
O
C
NH CH
CH2
NH
CH2 NH
CH2
CH2
CH2
COO−
CH2
3
CH2
Argininosuccinate
+
+
H
C
NH3
H
C
NH3
ARGININOSUCCINIC ACID
COO−
SYNTHASE
COO−
L-Citrulline
Mg-ATP
AMP + Mg-PPi
COO−
H2N C
H
CH2
COO−
L-Aspartate
Figure 29-9. Reactions and intermediates of urea biosynthesis. The nitrogen-containing groups that
contribute to the formation of urea are shaded. Reactions 1 and 2
occur in the matrix of liver mitochon-
dria and reactions
3,
4 , and
5
in liver cytosol. CO2 (as bicarbonate), ammonium ion, ornithine, and cit-
rulline enter the mitochondrial matrix via specific carriers (see heavy dots) present in the inner membrane of
liver mitochondria.
force for synthesis of the amide bond and the mixed
Carbamoyl Phosphate Plus Ornithine
acid anhydride bond of carbamoyl phosphate. The con-
Forms Citrulline
certed action of GDH and carbamoyl phosphate syn-
thase I thus shuttles nitrogen into carbamoyl phos-
L-Ornithine transcarbamoylase catalyzes transfer
of
phate, a compound with high group transfer potential.
the carbamoyl group of carbamoyl phosphate to or-
The reaction proceeds stepwise. Reaction of bicarbo-
nithine, forming citrulline and orthophosphate (reac-
nate with ATP forms carbonyl phosphate and ADP.
tion 2, Figure 29-9). While the reaction occurs in the
Ammonia then displaces ADP, forming carbamate and
mitochondrial matrix, both the formation of ornithine
orthophosphate. Phosphorylation of carbamate by the
and the subsequent metabolism of citrulline take place
second ATP then forms carbamoyl phosphate.
in the cytosol. Entry of ornithine into mitochondria
CATABOLISM OF PROTEINS & OF AMINO ACID NITROGEN
/
247
and exodus of citrulline from mitochondria therefore
of ammonia that accompanies enhanced protein degra-
involve mitochondrial inner membrane transport sys-
dation.
tems (Figure 29-9).
METABOLIC DISORDERS ARE
Citrulline Plus Aspartate
ASSOCIATED WITH EACH REACTION
Forms Argininosuccinate
OF THE UREA CYCLE
Argininosuccinate synthase links aspartate and cit-
Metabolic disorders of urea biosynthesis, while ex-
rulline via the amino group of aspartate (reaction 3,
tremely rare, illustrate four important principles: (1)
Figure 29-9) and provides the second nitrogen of urea.
Defects in any of several enzymes of a metabolic path-
The reaction requires ATP and involves intermediate
way enzyme can result in similar clinical signs and
formation of citrullyl-AMP. Subsequent displacement
symptoms. (2) The identification of intermediates and
of AMP by aspartate then forms citrulline.
of ancillary products that accumulate prior to a meta-
bolic block provides insight into the reaction that is im-
Cleavage of Argininosuccinate
paired. (3) Precise diagnosis requires quantitative assay
Forms Arginine & Fumarate
of the activity of the enzyme thought to be defective.
Cleavage of argininosuccinate, catalyzed by argini-
(4) Rational therapy must be based on an understand-
nosuccinase, proceeds with retention of nitrogen in
ing of the underlying biochemical reactions in normal
arginine and release of the aspartate skeleton as fu-
and impaired individuals.
marate (reaction 4, Figure 29-9). Addition of water to
All defects in urea synthesis result in ammonia in-
fumarate forms L-malate, and subsequent NAD+-
toxication. Intoxication is more severe when the meta-
dependent oxidation of malate forms oxaloacetate.
bolic block occurs at reactions 1 or 2 since some cova-
These two reactions are analogous to reactions of the
lent linking of ammonia to carbon has already occurred
citric acid cycle (see Figure 16-3) but are catalyzed by
if citrulline can be synthesized. Clinical symptoms
cytosolic fumarase and malate dehydrogenase. Transami-
common to all urea cycle disorders include vomiting,
nation of oxaloacetate by glutamate aminotransferase
avoidance of high-protein foods, intermittent ataxia, ir-
then re-forms aspartate. The carbon skeleton of aspartate-
ritability, lethargy, and mental retardation. The clinical
fumarate thus acts as a carrier of the nitrogen of gluta-
features and treatment of all five disorders discussed
mate into a precursor of urea.
below are similar. Significant improvement and mini-
mization of brain damage accompany a low-protein
Cleavage of Arginine Releases Urea
diet ingested as frequent small meals to avoid sudden
& Re-forms Ornithine
increases in blood ammonia levels.
Hyperammonemia Type 1. A consequence of
Hydrolytic cleavage of the guanidino group of arginine,
carbamoyl phosphate synthase I deficiency
(reac-
catalyzed by liver arginase, releases urea (reaction 5,
tion 1, Figure 29-9), this relatively infrequent condition
Figure 29-9). The other product, ornithine, reenters
(estimated frequency 1:62,000) probably is familial.
liver mitochondria for additional rounds of urea syn-
Hyperammonemia Type 2. A deficiency of or-
thesis. Ornithine and lysine are potent inhibitors of
nithine transcarbamoylase (reaction 2, Figure 29-9)
arginase, competitive with arginine. Arginine also serves
produces this X chromosome-linked deficiency. The
as the precursor of the potent muscle relaxant nitric
mothers also exhibit hyperammonemia and an aversion
oxide (NO) in a Ca2+-dependent reaction catalyzed by
to high-protein foods. Levels of glutamine are elevated
NO synthase (see Figure 49-15).
in blood, cerebrospinal fluid, and urine, probably due
to enhanced glutamine synthesis in response to elevated
Carbamoyl Phosphate Synthase I Is the
levels of tissue ammonia.
Pacemaker Enzyme of the Urea Cycle
Citrullinemia. In this rare disorder, plasma and
The activity of carbamoyl phosphate synthase I is deter-
cerebrospinal fluid citrulline levels are elevated and
mined by N-acetylglutamate, whose steady-state level is
1-2 g of citrulline are excreted daily. One patient lacked
dictated by its rate of synthesis from acetyl-CoA and
detectable argininosuccinate synthase activity
(reac-
glutamate and its rate of hydrolysis to acetate and glu-
tion 3, Figure 29-9). In another, the Km for citrulline
tamate. These reactions are catalyzed by N-acetylglu-
was 25 times higher than normal. Citrulline and argini-
tamate synthase and N-acetylglutamate hydrolase, re-
nosuccinate, which contain nitrogen destined for urea
spectively. Major changes in diet can increase the
synthesis, serve as alternative carriers of excess nitrogen.
concentrations of individual urea cycle enzymes 10-fold
Feeding arginine enhanced excretion of citrulline in these
to 20-fold. Starvation, for example, elevates enzyme lev-
patients. Similarly, feeding benzoate diverts ammonia
els, presumably to cope with the increased production
nitrogen to hippurate via glycine (see Figure 31-1).
248
/
CHAPTER 29
Argininosuccinicaciduria. A rare disease charac-
• Transamination channels α-amino acid nitrogen into
terized by elevated levels of argininosuccinate in blood,
glutamate. L-Glutamate dehydrogenase (GDH) oc-
cerebrospinal fluid, and urine is associated with friable,
cupies a central position in nitrogen metabolism.
tufted hair (trichorrhexis nodosa). Both early-onset and
• Glutamine synthase converts NH3 to nontoxic gluta-
late-onset types are known. The metabolic defect is
mine. Glutaminase releases NH3 for use in urea syn-
the absence of argininosuccinase (reaction 4, Figure
thesis.
29-9). Diagnosis by measurement of erythrocyte argini-
• NH3, CO2, and the amide nitrogen of aspartate pro-
nosuccinase activity can be performed on umbilical
vide the atoms of urea.
cord blood or amniotic fluid cells. As for citrullinemia,
• Hepatic urea synthesis takes place in part in the mi-
feeding arginine and benzoate promotes nitrogen excre-
tochondrial matrix and in part in the cytosol. Inborn
tion.
errors of metabolism are associated with each reac-
Hyperargininemia. This defect is characterized by
tion of the urea cycle.
elevated blood and cerebrospinal fluid arginine levels,
low erythrocyte levels of arginase (reaction 5, Figure
• Changes in enzyme levels and allosteric regulation of
carbamoyl phosphate synthase by N-acetylglutamate
29-9), and a urinary amino acid pattern resembling
that of lysine-cystinuria. This pattern may reflect com-
regulate urea biosynthesis.
petition by arginine with lysine and cystine for reab-
sorption in the renal tubule. A low-protein diet lowers
REFERENCES
plasma ammonia levels and abolishes lysine-cystinuria.
Brooks P et al: Subcellular localization of proteasomes and their
regulatory complexes in mammalian cells. Biochem J 2000;
Gene Therapy Offers Promise for
346:155.
Curthoys NP, Watford M: Regulation of glutaminase activity and
Correcting Defects in Urea Biosynthesis
glutamine metabolism. Annu Rev Nutr 1995;15:133.
Gene therapy for rectification of defects in the enzymes
Hershko A, Ciechanover A: The ubiquitin system. Annu Rev
of the urea cycle is an area of active investigation. En-
Biochem 1998;67:425.
couraging preliminary results have been obtained, for
Iyer R et al: The human arginases and arginase deficiency. J Inherit
example, in animal models using an adenoviral vector
Metab Dis 1998;21:86.
to treat citrullinemia.
Lim CB et al: Reduction in the rates of protein and amino acid ca-
tabolism to slow down the accumulation of endogenous am-
monia: a strategy potentially adopted by mudskippers during
SUMMARY
aerial exposure in constant darkness. J Exp Biol
2001;
204:1605.
• Human subjects degrade 1-2% of their body protein
Patejunas G et al: Evaluation of gene therapy for citrullinaemia
daily at rates that vary widely between proteins and
using murine and bovine models. J Inherit Metab Dis
with physiologic state. Key regulatory enzymes often
1998;21:138.
have short half-lives.
Pickart CM. Mechanisms underlying ubiquitination. Annu Rev
Biochem 2001;70:503.
• Proteins are degraded by both ATP-dependent and
ATP-independent pathways. Ubiquitin targets many
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
herited Disease, 8th ed. McGraw-Hill, 2001.
intracellular proteins for degradation. Liver cell sur-
Tuchman M et al: The biochemical and molecular spectrum of or-
face receptors bind and internalize circulating asialo-
nithine transcarbamoylase deficiency. J Inherit Metab Dis
glycoproteins destined for lysosomal degradation.
1998;21:40.
• Ammonia is highly toxic. Fish excrete NH3 directly;
Turner MA et al: Human argininosuccinate lyase: a structural basis
birds convert NH3 to uric acid. Higher vertebrates
for intragenic complementation. Proc Natl Acad Sci U S A
convert NH3 to urea.
1997;94:9063.
Catabolism of the Carbon Skeletons
30
of Amino Acids
Victor W. Rodwell, PhD
BIOMEDICAL IMPORTANCE
TRANSAMINATION TYPICALLY INITIATES
AMINO ACID CATABOLISM
This chapter considers conversion of the carbon skele-
tons of the common L-amino acids to amphibolic inter-
Removal of α-amino nitrogen by transamination (see
mediates and the metabolic diseases or “inborn errors of
Figure 28-3) is the first catabolic reaction of amino
metabolism” associated with these processes. Left un-
acids except in the case of proline, hydroxyproline,
treated, they can result in irreversible brain damage and
threonine, and lysine. The residual hydrocarbon skele-
early mortality. Prenatal or early postnatal detection
ton is then degraded to amphibolic intermediates as
and timely initiation of treatment thus are essential.
outlined in Figure 30-1.
Many of the enzymes concerned can be detected in cul-
Asparagine, Aspartate, Glutamine, and Gluta-
tured amniotic fluid cells, which facilitates early diag-
mate. All four carbons of asparagine and aspartate
nosis by amniocentesis. Treatment consists primarily of
form oxaloacetate (Figure 30-2, top). Analogous reac-
feeding diets low in the amino acids whose catabolism
tions convert glutamine and glutamate to
-ketoglu-
is impaired. While many changes in the primary struc-
tarate (Figure 30-2, bottom). Since the enzymes also
ture of enzymes have no adverse effects, others modify
fulfill anabolic functions, no metabolic defects are asso-
the three-dimensional structure of catalytic or regula-
ciated with the catabolism of these four amino acids.
tory sites, lower catalytic efficiency (lower Vmax or ele-
Proline. Proline forms dehydroproline, glutamate-
vate Km), or alter the affinity for an allosteric regulator
γ-semialdehyde, glutamate, and, ultimately,
-ketoglu-
of activity. A variety of mutations thus may give rise to
tarate (Figure 30-3, top). The metabolic block in type
the same clinical signs and symptoms.
I hyperprolinemia is at proline dehydrogenase.
Ala
Cys
Arg
Gly
His
α-Ketoglutarate
Glutamate
Hyp
Gln
Ser
Pro
Thr
lle
Leu
lle
Citrate
Succinyl-CoA
Met
Trp
Val
Pyruvate
Citrate
cycle
Acetyl-CoA
Acetoacetyl-CoA
Tyr
Oxaloacetate
Fumarate
Phe
Leu, Lys,
Phe, Trp,
Tyr
Aspartate
Asn
Figure 30-1. Amphibolic intermediates formed from the carbon skeletons
of amino acids.
249
250
/
CHAPTER 30
O
H2O
NH4+
O
O
PYR
ALA
C
C
C
NH2
O-
O-
CH2
CH2
CH2
H
C NH2
ASPARAGINASE
H
C NH3+
TRANSAMINASE
C
O
COO-
COO
COO-
L-Asparagine
L-Aspartate
Oxaloacetate
O
O
O
C
+
C
C
H2O
NH4
PYR
ALA
NH2
O-
O-
Figure 30-2. Catabolism of L-as-
CH2
CH2
CH2
paragine (top) and of L-glutamine
CH2
CH2
CH2
(bottom) to amphibolic intermedi-
GLUTAMINASE
TRANSAMINASE
+
+
ates. (PYR, pyruvate; ALA, L-alanine.)
H C NH
3
H
C NH3
C
O
In this and subsequent figures, color
COO-
COO-
COO-
highlights portions of the molecules
L-Glutamine
L-Glutamate
α-Ketoglutarate
undergoing chemical change.
There is no associated impairment of hydroxyproline
Glycine. The glycine synthase complex of liver mi-
catabolism. The metabolic block in type II hyperpro-
tochondria splits glycine to CO2 and NH4+ and forms
linemia is at glutamate-
-semialdehyde dehydroge-
N5,N10-methylene tetrahydrofolate (Figure 30-5).
nase, which also functions in hydroxyproline catabo-
Glycinuria results from a defect in renal tubular re-
lism. Both proline and hydroxyproline catabolism thus
absorption. The defect in primary hyperoxaluria is the
are affected and ∆1-pyrroline-3-hydroxy-5-carboxylate
failure to catabolize glyoxylate formed by deamination
(see Figure 30-10) is excreted.
of glycine. Subsequent oxidation of glyoxylate to ox-
Arginine and Ornithine. Arginine is converted to
alate results in urolithiasis, nephrocalcinosis, and early
ornithine, glutamate γ-semialdehyde, and then
-ke-
mortality from renal failure or hypertension.
toglutarate (Figure 30-3, bottom). Mutations in or-
Serine. Following conversion to glycine, catalyzed
nithine
-aminotransferase elevate plasma and urinary
by serine hydroxymethyltransferase (Figure 30-5),
ornithine and cause gyrate atrophy of the retina.
serine catabolism merges with that of glycine (Figure
Treatment involves restricting dietary arginine. In hy-
30-6).
perornithinemia-hyperammonemia syndrome, a de-
Alanine. Transamination of alanine forms pyru-
fective mitochondrial ornithine-citrulline antiporter
vate. Perhaps for the reason advanced under glutamate
(see Figure 29-9) impairs transport of ornithine into
and aspartate catabolism, there is no known metabolic
mitochondria for use in urea synthesis.
defect of alanine catabolism. Cysteine. Cystine is first
Histidine. Catabolism of histidine proceeds via
reduced to cysteine by cystine reductase
(Figure
urocanate,
4-imidazolone-5-propionate, and N-for-
30-7). Two different pathways then convert cysteine to
miminoglutamate (Figlu). Formimino group transfer to
pyruvate (Figure 30-8).
tetrahydrofolate forms glutamate, then
-ketoglu-
There are numerous abnormalities of cysteine me-
tarate (Figure 30-4). In folic acid deficiency, group
tabolism. Cystine, lysine, arginine, and ornithine are
transfer is impaired and Figlu is excreted. Excretion of
excreted in cystine-lysinuria (cystinuria), a defect in
Figlu following a dose of histidine thus has been used
renal reabsorption. Apart from cystine calculi, cystin-
to detect folic acid deficiency. Benign disorders of histi-
uria is benign. The mixed disulfide of L-cysteine and
dine catabolism include histidinemia and urocanic
L-homocysteine (Figure 30-9) excreted by cystinuric
aciduria associated with impaired histidase.
patients is more soluble than cystine and reduces for-
mation of cystine calculi. Several metabolic defects
result in vitamin B6-responsive or -unresponsive ho-
SIX AMINO ACIDS FORM PYRUVATE
mocystinurias. Defective carrier-mediated transport
All of the carbons of glycine, serine, alanine, and cys-
of cystine results in cystinosis (cystine storage dis-
teine and two carbons of threonine form pyruvate and
ease) with deposition of cystine crystals in tissues
subsequently acetyl-CoA.
and early mortality from acute renal failure. Despite
H
H
Figure 30-3. Top: Catabolism of proline. Numerals indicate
H
N
sites of the metabolic defects in 1
type I and 2
type II hyper-
+
H
prolinemias. Bottom: Catabolism of arginine. Glutamate-γ-
O−
C
semialdehyde forms α-ketoglutarate as shown above. 3 , site of
the metabolic defect in hyperargininemia.
O
L-Proline
NAD+
PROLINE
1
H3N+
+
DEHYDROGENASE
HN
NH
NADH + H+
-O
CH
C
C
H
NH+
O
O−
L-Histidine
C
H2O
O
HISTIDASE
NH4+
NH3+
+
H
HN
NH
CH
2
CH
O−
-O
C
HC
CH2
C
C
C
H
O
O
O
L-Glutamate-γ-semialdehyde
Urocanate
H2O
NAD+
GLUTAMATE SEMIALDEHYDE
UROCANASE
2
DEHYDROGENASE
NADH + H+
+
HN
NH
L-Glutamate
-O
CH2
C
CH2
O
α-Ketoglutarate
O
4-Imidazolone-5-propionate
NH3+
H
H2O
−
H2N
N
CH
2
CH
O
IMIDAZOLONE PROPIONATE
C
CH2
CH2
C
HYDROLASE
NH
O
+
+
L-Arginine
HN
NH2
H2O
–O
CH
2
CH
O-
3
ARGINASE
C
CH2
C
Urea
O
O
NH3+
N -Formiminoglutamate (Figlu)
CH
2
CH
O−
H4 folate
CH2
CH2
C
GLUTAMATE FORMIMINO
TRANSFERASE
NH3+
O
N
5-Formimino
H4 folate
L-Ornithine
L-Glutamate
α-KG
α-Ketoglutarate
Glu
Figure 30-4. Catabolism of L-histidine to α-ketoglu-
tarate. (H4 folate, tetrahydrofolate.) Histidase is the
L-Glutamate-γ-semialdehyde
probable site of the metabolic defect in histidinemia.
251
252
/
CHAPTER 30
+
Methylene
NH3
H4 folate
H4 folate
+
CH
O−
NH3+
NH
3
Cysteine
H2C
C
CH
O-
CH2
O-
HS
O
H2C
C
C
HO
O
O
[O]
L-Serine
Glycine
CYSTEINE
DIOXYGENASE
Figure 30-5. Interconversion of serine and glycine
catalyzed by serine hydroxymethyltransferase. (H4 fo-
+
NH3
late, tetrahydrofolate.)
CH
O−
H
2
C
C
Cysteine sulfinate
−O2S
O
NH3+
α-Keto acid
CH2
O- + NAD+
TRANSAMINASE
C
α-Amino acid
O
O
Glycine
H4 folate
C
O−
Sulfinylpyruvate
H2C
C
N5,N10-CH2-H4 folate
–O2S
O
+ + NADH + H+
CO2 + NH4
DESULFINASE
SO32−
Figure 30-6. Reversible cleavage of glycine by the
mitochondrial glycine synthase complex. (PLP, pyri-
Pyruvate
doxal phosphate.)
CYSTEINE
α-KA
NH3+
TRANSAMINASE
α-AA
CH
O−
H2C
C
O
S
O
3-Mercaptopyruvate
−
C
O
O
S
(thiolpyruvate)
H2C
C
C
CH2
HS O
−O
CH
NADH
+
2H
NH3
L-Cystine
+ H+
NADH + H+
+
H2S NAD
CYSTINE
Pyruvate
H
OH
−
REDUCTASE
C
O
NAD+
H2C
C
HS
O
NH3+
3-Mercaptolactate
CH
O−
2
CH2
C
Figure 30-8. Catabolism of L-cysteine via the cys-
teine sulfinate pathway (top) and by the 3-mercaptopy-
SH
O
L-Cysteine
ruvate pathway (bottom).
Figure 30-7. The cystine reductase reaction.
CATABOLISM OF THE CARBON SKELETONS OF AMINO ACIDS
/
253
CH2
S S
CH2
HH
H
+
N
H
H C
NH3+
CH2
OH
O-
COO-
H C
NH3+
C
COO-
O
(Cysteine)
(Homocysteine)
4-Hydroxy-L-proline
Figure 30-9.
Mixed disulfide of cysteine and homo-
1
HYDROXYPROLINE
cysteine.
DEHYDROGENASE
2H
NH3+
NH+
OH
H3C
CH
O−
O-
CH
C
C
OH
O
O
L-∆1-Pyrroline-3-hydroxy-5-carboxylate
L-Threonine
O
H2
THREONINE
NONENZYMATIC
ALDOLASE
Glycine
OH
NH3+
H3C
CH
CH
CH
O-
HC
CH2
C
O
O
O
Acetaldehyde
γ-Hydroxy-L-glutamate-γ-semialdehyde
+
H
2O
NAD
+
NAD
H2O
ALDEHYDE
+
2
DEHYDROGENASE
DEHYDROGENASE
NADH+H
NADH + H+
H3C
O−
+
C
OH
NH3
O
–O
CH
CH
O-
C
CH2
C
Acetate
O
O
CoASH
Mg-ATP
Erythro-γ-hydroxy-L-glutamate
ACETATE
α-KA
THIOKINASE
TRANSAMINASE
H2O
Mg-ADP
α-AA
OH
O
-O
CH
C
O-
H3C
S CoA
C
CH2
C
C
O
O
O
α-Keto-γ-hydroxyglutarate
Acetyl-CoA
AN ALDOLASE
Figure 30-10. Conversion of threonine to glycine
(see Figure 30-6) and acetyl-CoA.
O
O
–O
CH
C
O-
C
H3C
C
Figure 30-11. Intermediates in L-hydroxyproline catabolism. (α-KA,
O
O
α-keto acid; α-AA, α-amino acid.) Numerals identify sites of metabolic
Glyoxylate
Pyruvate
defects in 1
hyperhydroxyprolinemia and 2
type II hyperprolinemia.
NH3+
O
2
CH
1
O-
1
C
1
O-
3
2
α-KG
Glu
3
2
[O]
Ascorbate
1CO2
CH2
C
CH2
C
OH
PLP
Cu2+
3
4
4
4
O
O
9
CH2
2
O-
5
9
5
9
5
C
6
8
6
8
6
8
O
7
TYROSINE
7
p-HYDROXYPHENYLPYRUVATE
7
TRANSAMINASE
HYDROXYLASE
OH
OH
OH
L-Tyrosine
p-Hydroxyphenylpyruvate
Homogentisate
3
O
O
[O]
O
3
C
4
CH2
2
O-
C
Fe2+
Glutathione
6
8
3
5
O-
C
C
5
4
O-
9
C
CH
7
CH2
9
CH2
2
O-
=
8
3
4
HC
C
C
C
6
8
O
6
CH2
9
CH2
2
O-
5
7
7
C
C
C
MALEYLACETOACETATE
HOMOGENTISATE
O
O
O
O
CIS, TRANS ISOMERASE
OXIDASE
O
O
O
Maleylacetoacetate
Maleylacetoacetate
Fumarylacetoacetate
(rewritten)
6
C
CH
O-
4
CH
C
5
7
H2O
4
O
Fumarate
+
FUMARYLACETOACETATE
CoASH
HYDROLASE
H3C
CH2
O-
H3C
S CoA
H3C
O-
C
C
C
C
+
O
O
β-KETOTHIOLASE
O
O
Acetoacetate
Acetyl-CoA
Acetate
Figure 30-12. Intermediates in tyrosine catabolism. Carbons are numbered to emphasize their ultimate fate. (α-KG, α-ketoglutarate;
Glu, glutamate; PLP, pyridoxal phosphate.) Circled numerals represent the probable sites of the metabolic defects in 1
type II tyrosinemia;
2
neonatal tyrosinemia; 3
alkaptonuria; and 4
type I tyrosinemia, or tyrosinosis.
CATABOLISM OF THE CARBON SKELETONS OF AMINO ACIDS
/
255
epidemiologic data suggesting a relationship between
those of tyrosine
(Figure
30-12). Hyperphenylala-
plasma homocysteine and cardiovascular disease,
ninemias arise from defects in phenylalanine hydroxy-
whether homocysteine represents a causal cardiovas-
lase itself (type I, classic phenylketonuria or PKU), in
cular risk factor remains controversial.
dihydrobiopterin reductase (types II and III), or in di-
Threonine. Threonine is cleaved to acetaldehyde
hydrobiopterin biosynthesis (types IV and V) (Figure
and glycine. Oxidation of acetaldehyde to acetate is fol-
28-10). Alternative catabolites are excreted
(Figure
lowed by formation of acetyl-CoA (Figure 30-10). Ca-
30-13). DNA probes facilitate prenatal diagnosis of de-
tabolism of glycine is discussed above.
fects in phenylalanine hydroxylase or dihydrobiopterin
4-Hydroxyproline. Catabolism of 4-hydroxy-L-pro-
reductase. A diet low in phenylalanine can prevent the
line forms, successively, L-∆1-pyrroline-3-hydroxy-5-car-
mental retardation of PKU
(frequency
1:10,000
boxylate, γ-hydroxy-L-glutamate-γ-semialdehyde, erythro-
γ-hydroxy-L-glutamate, and α-keto-γ-hydroxyglutarate.
An aldol-type cleavage then forms glyoxylate plus pyru-
CH2
COO-
vate (Figure 30-11). A defect in 4-hydroxyproline de-
CH
hydrogenase results in hyperhydroxyprolinemia,
+
which is benign. There is no associated impairment of
NH3
proline catabolism.
L-Phenylalanine
α-Ketoglutarate
TWELVE AMINO ACIDS FORM
TRANSAMINASE
ACETYL-CoA
L-Glutamate
Tyrosine. Figure 30-12 diagrams the conversion of
CH2
COO-
tyrosine to amphibolic intermediates. Since ascorbate is
C
the reductant for conversion of p-hydroxyphenylpyru-
O
vate to homogentisate, scorbutic patients excrete in-
Phenylpyruvate
completely oxidized products of tyrosine catabolism.
Subsequent catabolism forms maleylacetoacetate, fu-
NAD+
NADH + H+
marylacetoacetate, fumarate, acetoacetate, and ulti-
mately acetyl-CoA.
H2O
The probable metabolic defect in type I tyrosine-
NADH + H+
NAD+
mia (tyrosinosis) is at fumarylacetoacetate hydrolase
CO2
(reaction 4, Figure 30-12). Therapy employs a diet low
in tyrosine and phenylalanine. Untreated acute and
CH2
CH2
COO-
chronic tyrosinosis leads to death from liver failure. Al-
COO-
CH
ternate metabolites of tyrosine are also excreted in type
II tyrosinemia (Richner-Hanhart syndrome), a de-
OH
fect in tyrosine aminotransferase (reaction 1, Figure
Phenylacetate
Phenyllactate
30-12), and in neonatal tyrosinemia, due to lowered
L-Glutamine
p-hydroxyphenylpyruvate hydroxylase activity (reaction
2, Figure
30-12). Therapy employs a diet low in
protein.
H2O
Alkaptonuria was first described in the 16th cen-
COO-
tury. Characterized in 1859, it provided the basis for
H
CH2
N C H
Garrod’s classic ideas concerning heritable metabolic
C
disorders. The defect is lack of homogentisate oxidase
CH2
O
(reaction 3, Figure 30-12). The urine darkens on expo-
CH2
sure to air due to oxidation of excreted homogentisate.
Phenylacetylglutamine
Late in the disease, there is arthritis and connective tis-
CONH2
sue pigmentation (ochronosis) due to oxidation of ho-
mogentisate to benzoquinone acetate, which polymer-
Figure 30-13. Alternative pathways of phenylala-
izes and binds to connective tissue.
nine catabolism in phenylketonuria. The reactions also
Phenylalanine. Phenylalanine is first converted to
occur in normal liver tissue but are of minor signifi-
tyrosine (see Figure 28-10). Subsequent reactions are
cance.
258
/
CHAPTER 30
O
of newborn infants is compulsory in the United States
and many other countries.
C
Lysine. Figure 30-14 summarizes the catabolism of
CH2
lysine. Lysine first forms a Schiff base with α-ketoglu-
tarate, which is reduced to saccharopine. In one form
CH
O-
N
N
C
of periodic hyperlysinemia, elevated lysine competi-
H2
H3+
tively inhibits liver arginase (see Figure 29-9), causing
HO
O
hyperammonemia. Restricting dietary lysine relieves the
3-Hydroxykynurenine
ammonemia, whereas ingestion of a lysine load precipi-
tates severe crises and coma. In a different periodic hy-
perlysinemia, lysine catabolites accumulate, but even a
NH4+
lysine load does not trigger hyperammonemia. In addi-
tion to impaired synthesis of saccharopine, some pa-
tients cannot cleave saccharopine.
Tryptophan. Tryptophan is degraded to amphi-
bolic intermediates via the kynurenine-anthranilate
pathway
(Figure
30-15). Tryptophan oxygenase
O-
(tryptophan pyrrolase) opens the indole ring, incor-
C
N
porates molecular oxygen, and forms N-formylkynure-
HO
O
nine. An iron porphyrin metalloprotein that is in-
Xanthurenate
ducible in liver by adrenal corticosteroids and by
tryptophan, tryptophan oxygenase is feedback-
Figure 30-16. Formation of xanthurenate in vitamin
inhibited by nicotinic acid derivatives, including
B6 deficiency. Conversion of the tryptophan metabolite
NADPH. Hydrolytic removal of the formyl group of
3-hydroxykynurenine to 3-hydroxyanthranilate is im-
N-formylkynurenine, catalyzed by kynurenine formy-
paired (see Figure 30-15). A large portion is therefore
lase, produces kynurenine. Since kynureninase re-
converted to xanthurenate.
quires pyridoxal phosphate, excretion of xanthurenate
(Figure 30-16) in response to a tryptophan load is di-
agnostic of vitamin B6 deficiency. Hartnup disease re-
births). Elevated blood phenylalanine may not be de-
flects impaired intestinal and renal transport of trypto-
tectable until 3-4 days postpartum. False-positives in
phan and other neutral amino acids. Indole derivatives
premature infants may reflect delayed maturation of en-
of unabsorbed tryptophan formed by intestinal bacteria
zymes of phenylalanine catabolism. A less reliable
are excreted. The defect limits tryptophan availability
screening test employs FeCl3
to detect urinary
for niacin biosynthesis and accounts for the pellagra-
phenylpyruvate. FeCl3 screening for PKU of the urine
like signs and symptoms.
COO-
COO-
+H3N
C H
+H3N
C
H
CH2
CH2
H2O
P
+ PP
CH2
P P P
i
i
CH2
S
+
CH2
Adenine
+S
CH
2
Adenine
O
O
CH3
L-METHIONINE
CH3
ADENOSYLTRANSFERASE
Ribose
Ribose
HO OH
HO OH
L-Methionine
ATP
S-Adenosyl-L-methionine
(“active methionine”)
Figure 30-17. Formation of S-adenosylmethionine. ~CH3 represents the high
group transfer potential of “active methionine.”
CATABOLISM OF THE CARBON SKELETONS OF AMINO ACIDS
/
259
NH3+
Methionine. Methionine reacts with ATP forming
S-adenosylmethionine,
“active methionine”
(Figure
H3C
CH
2
CH
O-
S
CH2
C
30-17). Subsequent reactions form propionyl-CoA
(Figure 30-18) and ultimately succinyl-CoA (see Fig-
O
L-Methionine
ure 19-2).
ATP
THE INITIAL REACTIONS ARE COMMON
Pi + PPi
TO ALL THREE BRANCHED-CHAIN
S-Adenosyl-L-methionine
AMINO ACIDS
Acceptor
Reactions 1-3 of Figure 30-19 are analogous to those
of fatty acid catabolism. Following transamination, all
CH3-Acceptor
three α-keto acids undergo oxidative decarboxylation
catalyzed by mitochondrial branched-chain
-keto
S-Adenosyl-L-homocysteine
acid dehydrogenase. This multimeric enzyme complex
H2O
of a decarboxylase, a transacylase, and a dihydrolipoyl
dehydrogenase closely resembles pyruvate dehydroge-
Adenosine
nase (see Figure 17-5). Its regulation also parallels that
of pyruvate dehydrogenase, being inactivated by phos-
NH3+
phorylation and reactivated by dephosphorylation (see
CH
2
CH
O-
Figure 17-6).
H
S
CH2
C
Reaction 3 is analogous to the dehydrogenation of
+
O
fatty acyl-CoA thioesters (see Figure 22-3). In isova-
O
OH
L-Homocysteine
leric acidemia, ingestion of protein-rich foods ele-
C
CH2
CYSTATHIONINE
vates isovalerate, the deacylation product of isovaleryl-
–
O
CH
β-SYNTHASE
CoA. Figures 30-20, 30-21, and 30-22 illustrate the
NH3
+
subsequent reactions unique to each amino acid skele-
H2O
L-Serine
ton.
NH3+
-
METABOLIC DISORDERS OF BRANCHED-
CH
2
CH
O
O
S
CH2
C
CHAIN AMINO ACID CATABOLISM
C
CH2
O
–
As the name implies, the odor of urine in maple syrup
O
CH
Cystathionine
urine disease
(branched-chain ketonuria) suggests
+
NH3
maple syrup or burnt sugar. The biochemical defect in-
H2O
volves the
-keto acid decarboxylase complex (reac-
O
SH
O
tion 2, Figure 30-19). Plasma and urinary levels of
leucine, isoleucine, valine, α-keto acids, and α-hydroxy
C
CH2
H3C
C
O-
acids (reduced α-keto acids) are elevated. The mecha-
-O
CH
CH2
C
+
nism of toxicity is unknown. Early diagnosis, especially
+
NH4
NH
O
3
prior to 1 week of age, employs enzymatic analysis.
L-Cysteine
α-Ketobutyrate
Prompt replacement of dietary protein by an amino
+
acid mixture that lacks leucine, isoleucine, and valine
CoASH
NAD
averts brain damage and early mortality.
Mutation of the dihydrolipoate reductase compo-
+
CO2
NADH + H
nent impairs decarboxylation of branched-chain α-
O
keto acids, of pyruvate, and of α-ketoglutarate. In in-
termittent branched-chain ketonuria, the α-keto
H3C
C
acid decarboxylase retains some activity, and symp-
CH2
S
CoA
toms occur later in life. The impaired enzyme in iso-
Propionyl-CoA
valeric acidemia is isovaleryl-CoA dehydrogenase
Figure 30-18. Conversion of methionine to propi-
(reaction 3, Figure 30-19). Vomiting, acidosis, and
onyl-CoA.
coma follow ingestion of excess protein. Accumulated
260
/
CHAPTER 30
+
+
CH3
NH3
NH3
NH3+
CH
CH
O
H
3
C
CH
O
CH
2
CH
O-
H3C
CH2
C
CH
C
H
3C
CH
C
O
CH3
O
CH3
O
L-Leucine
L-Valine
L-Isoleucine
α-Keto acid
α-Keto acid
α-Keto acid
1
1
1
α-Amino acid
α-Amino acid
α-Amino acid
CH3
O
O
O
CH
C
O-
H3C
C
O-
CH2
C
O-
H3C
CH2
C
CH
C
H
3C
CH
C
O
CH3
O
CH3
O
α-Ketoisocaproate
α-Ketoisovalerate
α-Keto-β-methylvalerate
CoASH
CoASH
CoASH
2
2
2
CO2
CO2
CO2
CH3
O
O
O
CH
C
H3C
C
CH
C
H3C
CH2
S
CoA
CH
S
CoA
H
3C
CH
S
CoA
Isovaleryl-CoA
CH3
CH3
Isobutyryl-CoA
α-Methylbutyryl-CoA
3
3
3
[2H]
[2H]
[2H]
CH3
O
O
O
H
C
C
H2C
C
C
C
H
3
C
CH
S
CoA
C
S
CoA
H
3C
C
S
CoA
β-Methylcrotonyl-CoA
CH
3
CH3
Methacrylyl-CoA
Tiglyl-CoA
Figure 30-19. The analogous first three reactions in the catabolism of leucine, valine, and
isoleucine. Note also the analogy of reactions 2
and 3 to reactions of the catabolism of fatty
acids (see Figure 22-3). The analogy to fatty acid catabolism continues, as shown in subsequent
figures.
CATABOLISM OF THE CARBON SKELETONS OF AMINO ACIDS
/
261
CH3
O
O
C
C
CH
C
H
CH
S
CoA
H3C
C
S
CoA
3C
β-Methylcrotonyl-CoA
CH
3
Biotinyl-*CO2
Tiglyl-CoA
H2O
4L
4I
Biotin
O H
O
O
CH3
O
H
C
C
C*
C
C
H3C
CH
S
CoA
−O
CH
2
CH
S
CoA
CH3
β-Methylglutaconyl-CoA
α-Methyl-β-hydroxybutyryl-CoA
H2O
5L
5I
[2H]
O
O
C OH
O
O
C*H3
C
−O
CH2
CH2
S
CoA
C
C
β-Hydroxy-β-methylglutaryl-CoA
H3C
CH
S
CoA
O
O
O
CH3
6L
α-Methylacetoacetyl-CoA
C*
C
C
−O
CH2
CH3
H3C
S
CoA
Acetoacetate
Acetyl-CoA
CoASH
6I
Figure 30-20. Catabolism of the β-methylcrotonyl-
O
O
CoA formed from L-leucine. Asterisks indicate carbon
C
C
atoms derived from CO2.
H3C
S
CoA
+
CH2
S
CoA
CH3
Acetyl-CoA
Propionyl-CoA
Figure 30-21. Subsequent catabolism of the tiglyl-
CoA formed from L-isoleucine.
262
/
CHAPTER 30
O
isovaleryl-CoA is hydrolyzed to isovalerate and ex-
creted.
H2C
C
C
S CoA
CH3
SUMMARY
Methacrylyl-CoA
•
Excess amino acids are catabolized to amphibolic in-
4V
H2O
termediates used as sources of energy or for carbohy-
drate and lipid biosynthesis.
HO
O
•
Transamination is the most common initial reaction
H2C
C
of amino acid catabolism. Subsequent reactions re-
CH
S CoA
move any additional nitrogen and restructure the hy-
CH3
drocarbon skeleton for conversion to oxaloacetate,
β-Hydroxyisobutyryl-CoA
α-ketoglutarate, pyruvate, and acetyl-CoA.
H2O
•
Metabolic diseases associated with glycine catabolism
5V
include glycinuria and primary hyperoxaluria.
CoASH
•
Two distinct pathways convert cysteine to pyruvate.
HO
O
Metabolic disorders of cysteine catabolism include
H2C
C
cystine-lysinuria, cystine storage disease, and the ho-
CH
O
-
mocystinurias.
CH3
•
Threonine catabolism merges with that of glycine
β-Hydroxyisobutyrate
after threonine aldolase cleaves threonine to glycine
NAD+
and acetaldehyde.
6V
NADH + H
+
•
Following transamination, the carbon skeleton of ty-
rosine is degraded to fumarate and acetoacetate.
O
O
Metabolic diseases of tyrosine catabolism include ty-
HC
C
rosinosis, Richner-Hanhart syndrome, neonatal ty-
–
CH
O
rosinemia, and alkaptonuria.
CH3
•
Metabolic disorders of phenylalanine catabolism in-
Methylmalonate semialdehyde
clude phenylketonuria
(PKU) and several hyper-
α-AA
phenylalaninemias.
CoASH
NAD+
7V
•
Neither nitrogen of lysine undergoes transamination.
8V
α-KA
Metabolic diseases of lysine catabolism include peri-
NADH + H+
odic and persistent forms of hyperlysinemia-
+
O
O
NH
3
O
ammonemia.
C
C
H2C
C
•
The catabolism of leucine, valine, and isoleucine pre-
-
-
O
CH
S CoA
CH
O
sents many analogies to fatty acid catabolism. Meta-
CH
3
CH3
bolic disorders of branched-chain amino acid catabo-
Methylmalonyl-CoA
β-Aminoisobutyrate
lism include hypervalinemia, maple syrup urine
disease, intermittent branched-chain ketonuria, iso-
9V
B12 COENZYME
valeric acidemia, and methylmalonic aciduria.
O
C
REFERENCES
-
O
CH2
Blacher J, Safar ME: Homocysteine, folic acid, B vitamins and car-
H2 C
S CoA
diovascular risk. J Nutr Health Aging 2001;5:196.
C
Cooper AJL: Biochemistry of the sulfur-containing amino acids.
O
Annu Rev Biochem 1983;52:187.
Succinyl-CoA
Gjetting T et al: A phenylalanine hydroxylase amino acid polymor-
phism with implications for molecular diagnostics. Mol
Figure 30-22. Subsequent catabolism of the
Genet Metab 2001;73:280.
methacrylyl-CoA formed from L-valine (see Figure
Harris RA et al: Molecular cloning of the branched-chain α-ke-
30-19). (α-KA, α-keto acid; α-AA, α-amino acid.)
toacid dehydrogenase kinase and the CoA-dependent methyl-
CATABOLISM OF THE CARBON SKELETONS OF AMINO ACIDS
/
263
malonate semialdehyde dehydrogenase. Adv Enzyme Regul
Waters PJ, Scriver CR, Parniak MA: Homomeric and heteromeric
1993;33:255.
interactions between wild-type and mutant phenylalanine hy-
Scriver CR: Garrod’s foresight; our hindsight. J Inherit Metab Dis
droxylase subunits: evaluation of two-hybrid approaches for
2001;24:93.
functional analysis of mutations causing hyperphenylalanine-
mia. Mol Genet Metab 2001;73:230.
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
herited Disease, 8th ed. McGraw-Hill, 2001.
Conversion of Amino Acids
31
to Specialized Products
Victor W. Rodwell, PhD
BIOMEDICAL IMPORTANCE
β-aminoisobutyrate are elevated in the rare metabolic
disorder hyperbeta-alaninemia.
Important products derived from amino acids include
heme, purines, pyrimidines, hormones, neurotransmit-
-Alanyl Dipeptides
ters, and biologically active peptides. In addition, many
proteins contain amino acids that have been modified
The β-alanyl dipeptides carnosine and anserine
for a specific function such as binding calcium or as in-
(N-methylcarnosine)
(Figure
31-2) activate myosin
termediates that serve to stabilize proteins—generally
ATPase, chelate copper, and enhance copper uptake.
structural proteins—by subsequent covalent cross-link-
β-Alanyl-imidazole buffers the pH of anaerobically
ing. The amino acid residues in those proteins serve as
contracting skeletal muscle. Biosynthesis of carnosine is
precursors for these modified residues. Small peptides
catalyzed by carnosine synthetase in a two-stage reac-
or peptide-like molecules not synthesized on ribosomes
tion that involves initial formation of an enzyme-bound
fulfill specific functions in cells. Histamine plays a cen-
acyl-adenylate of β-alanine and subsequent transfer of
tral role in many allergic reactions. Neurotransmitters
the β-alanyl moiety to L-histidine.
derived from amino acids include γ-aminobutyrate,
5-hydroxytryptamine
(serotonin), dopamine, norepi-
ATP+
β
-Alanine
→
β
-Alanyl−AMP →+PP
i
nephrine, and epinephrine. Many drugs used to treat
Alanyl−AMP
Histidine
Carnosine+AMP
L
β-
+
-
→
neurologic and psychiatric conditions affect the metab-
olism of these neurotransmitters.
Hydrolysis of carnosine to β-alanine and L-histidine is
catalyzed by carnosinase. The heritable disorder
carnosinase deficiency is characterized by carnosinuria.
Glycine
Homocarnosine (Figure 31-2), present in human
Metabolites and pharmaceuticals excreted as water-
brain at higher levels than carnosine, is synthesized in
soluble glycine conjugates include glycocholic acid
brain tissue by carnosine synthetase. Serum carnosinase
(Chapter 24) and hippuric acid formed from the food
does not hydrolyze homocarnosine. Homocarnosinosis,
additive benzoate
(Figure
31-1). Many drugs, drug
a rare genetic disorder, is associated with progressive
metabolites, and other compounds with carboxyl
spastic paraplegia and mental retardation.
groups are excreted in the urine as glycine conjugates.
Glycine is incorporated into creatine (see Figure 31-6),
Phosphorylated Serine, Threonine,
the nitrogen and α-carbon of glycine are incorporated
& Tyrosine
into the pyrrole rings and the methylene bridge carbons
of heme (Chapter 32), and the entire glycine molecule
The phosphorylation and dephosphorylation of seryl,
becomes atoms 4, 5, and 7 of purines (Figure 34-1).
threonyl, and tyrosyl residues regulate the activity of
certain enzymes of lipid and carbohydrate metabolism
and the properties of proteins that participate in signal
transduction cascades.
-Alanine
β-Alanine, a metabolite of cysteine (Figure 34-9), is
Methionine
present in coenzyme A and as β-alanyl dipeptides, prin-
cipally carnosine (see below). Mammalian tissues form
S-Adenosylmethionine, the principal source of methyl
β-alanine from cytosine (Figure 34-9), carnosine, and
groups in the body, also contributes its carbon skeleton
anserine (Figure 31-2). Mammalian tissues transami-
for the biosynthesis of the 3-diaminopropane portions
nate β-alanine, forming malonate semialdehyde. Body
of the polyamines spermine and spermidine (Figure
fluid and tissue levels of β-alanine, taurine, and
31-4).
264
CONVERSION OF AMINO ACIDS TO SPECIALIZED PRODUCTS
/
265
O
SH
+
C
N
(CH3)3
N
NH2+
O-
CH
O-
CH2
C
Benzoate
O
Ergothioneine
ATP
CoASH
O
CH2
NH3+
AMP + PPi
C
CH
2
O
+
NH
N
NH2
C
CH
O-
S CoA
CH2
C
O
Benzoyl-CoA
Carnosine
Glycine
O
CH2
NH3+
C
CH2
CoASH
+
CH3
N
N
NH
O
H
-
CH
O
C
CH2
O-
CH2
C
N
C
H
O
O
Anserine
Hippurate
Figure 31-1. Biosynthesis of hippurate. Analogous
O
CH2
CH2
reactions occur with many acidic drugs and catabolites.
C
CH2
NH
+
3
NH
N
NH2+
Cysteine
CH
O-
CH2
C
L-Cysteine is a precursor of the thioethanolamine por-
tion of coenzyme A and of the taurine that conjugates
O
with bile acids such as taurocholic acid (Chapter 26).
Homocarnosine
Figure 31-2.
Compounds related to histidine. The
Histidine
boxes surround the components not derived from histi-
Decarboxylation of histidine to histamine is catalyzed by
dine. The SH group of ergothioneine derives from cys-
a broad-specificity aromatic L-amino acid decarboxylase
teine.
that also catalyzes the decarboxylation of dopa, 5-hy-
droxytryptophan, phenylalanine, tyrosine, and trypto-
phan. α-Methyl amino acids, which inhibit decarboxy-
tric oxide
(NO) that serves as a neurotransmitter,
lase activity, find application as antihypertensive agents.
smooth muscle relaxant, and vasodilator. Synthesis of
Histidine compounds present in the human body in-
NO, catalyzed by NO synthase, involves the NADPH-
clude ergothioneine, carnosine, and dietary anserine
dependent reaction of L-arginine with O2 to yield L-cit-
(Figure 31-2). Urinary levels of 3-methylhistidine are
rulline and NO.
unusually low in patients with Wilson’s disease.
Polyamines
Ornithine & Arginine
The polyamines spermidine and spermine
(Figure
Arginine is the formamidine donor for creatine synthe-
31-4) function in cell proliferation and growth, are
sis (Figure 31-6) and via ornithine to putrescine, sper-
growth factors for cultured mammalian cells, and stabi-
mine, and spermidine (Figure 31-3) Arginine is also
lize intact cells, subcellular organelles, and membranes.
the precursor of the intercellular signaling molecule ni-
Pharmacologic doses of polyamines are hypothermic
266
/
CHAPTER 31
PROTEINS
NITRIC OXIDE
UREA
PROTEINS
CREATINE
ARGININE
PHOSPHATE,
CREATININE
PROLINE
ORNITHINE
ARGININE
PHOSPHATE
Glutamate-γ-
PUTRESCINE,
semialdehyde
SPERMIDINE,
SPERMINE
GLUTAMATE
Figure 31-3. Arginine, ornithine, and proline metabolism. Reactions with solid ar-
rows all occur in mammalian tissues. Putrescine and spermine synthesis occurs in
both mammals and bacteria. Arginine phosphate of invertebrate muscle functions
as a phosphagen analogous to creatine phosphate of mammalian muscle (see
Figure 31-6).
and hypotensive. Since they bear multiple positive
mine), a potent vasoconstrictor and stimulator of
charges, polyamines associate readily with DNA and
smooth muscle contraction. Catabolism of serotonin is
RNA. Figure 31-4 summarizes polyamine biosynthesis.
initiated by monoamine oxidase-catalyzed oxidative
deamination to 5-hydroxyindoleacetate. The psychic
stimulation that follows administration of iproniazid
Tryptophan
results from its ability to prolong the action of sero-
Following hydroxylation of tryptophan to 5-hydroxy-
tonin by inhibiting monoamine oxidase. In carcinoid
tryptophan by liver tyrosine hydroxylase, subsequent
(argentaffinoma), tumor cells overproduce serotonin.
decarboxylation forms serotonin
(5-hydroxytrypta-
Urinary metabolites of serotonin in patients with carci-
H2
+H3N
+N
NH3+
Spermidine
Decarboxylated
S -adenosylmethionine
SPERMINE
SYNTHASE
Methylthio-
adenosine
Figure 31-4. Conversion of spermidine to spermine.
H2
Spermidine formed from putrescine (decarboxylated
+H3N
+N
N
+
L-ornithine) by transfer of a propylamine moiety from
NH3
+
H2
decarboxylated S-adenosylmethionine accepts a
Spermine
second propylamine moiety to form spermidine.
CONVERSION OF AMINO ACIDS TO SPECIALIZED PRODUCTS
/
267
HO
+
noid include N-acetylserotonin glucuronide and the
NH3
glycine conjugate of 5-hydroxyindoleacetate. Serotonin
CH
O-
and 5-methoxytryptamine are metabolized to the corre-
CH2
C
sponding acids by monoamine oxidase. N-Acetylation
O
of serotonin, followed by O-methylation in the pineal
L-Tyrosine
body, forms melatonin. Circulating melatonin is taken
H4 • biopterin
up by all tissues, including brain, but is rapidly metabo-
TYROSINE
lized by hydroxylation followed by conjugation with
HYDROXYLASE
sulfate or with glucuronic acid.
H2 • biopterin
Kidney tissue, liver tissue, and fecal bacteria all con-
OH
vert tryptophan to tryptamine, then to indole 3-acetate.
HO
+
NH3
The principal normal urinary catabolites of tryptophan
CH
O-
are 5-hydroxyindoleacetate and indole 3-acetate.
CH2
C
O
Tyrosine
Dopa
Neural cells convert tyrosine to epinephrine and norepi-
PLP
nephrine (Figure 31-5). While dopa is also an interme-
DOPA
DECARBOXYLASE
diate in the formation of melanin, different enzymes
CO2
hydroxylate tyrosine in melanocytes. Dopa decarboxy-
OH
lase, a pyridoxal phosphate-dependent enzyme, forms
HO
dopamine. Subsequent hydroxylation by dopamine
β-oxidase then forms norepinephrine. In the adrenal
CH2
medulla, phenylethanolamine-N-methyltransferase uti-
+
CH2
NH3
lizes S-adenosylmethionine to methylate the primary
Dopamine
amine of norepinephrine, forming epinephrine (Figure
31-5). Tyrosine is also a precursor of triiodothyronine
O2
DOPAMINE
and thyroxine (Chapter 42).
β-OXIDASE
Cu2+
Vitamin C
Creatinine
OH
HO
Creatinine is formed in muscle from creatine phosphate
by irreversible, nonenzymatic dehydration and loss of
CH2
phosphate (Figure 31-6). The 24-hour urinary excre-
CH
NH3+
tion of creatinine is proportionate to muscle mass.
OH
Glycine, arginine, and methionine all participate in cre-
Norepinephrine
atine biosynthesis. Synthesis of creatine is completed by
methylation of guanidoacetate by S-adenosylmethio-
S-Adenosylmethionine
PHENYLETHANOL-
nine (Figure 31-6).
AMINE N-METHYL-
TRANSFERASE
S-Adenosylhomocysteine
-Aminobutyrate
OH
γ-Aminobutyrate (GABA) functions in brain tissue as
HO
an inhibitory neurotransmitter by altering transmem-
brane potential differences. It is formed by decarboxyla-
CH2
CH3
CH
N
tion of L-glutamate, a reaction catalyzed by L-glutamate
+
H2
decarboxylase
(Figure
31-7). Transamination of γ-
OH
aminobutyrate forms succinate semialdehyde (Figure
Epinephrine
31-7), which may then undergo reduction to γ-hydroxy-
Figure 31-5. Conversion of tyrosine to epinephrine
butyrate, a reaction catalyzed by L-lactate dehydro-
genase, or oxidation to succinate and thence via the cit-
and norepinephrine in neuronal and adrenal cells. (PLP,
ric acid cycle to CO2 and H2O. A rare genetic disorder
pyridoxal phosphate.)
of GABA metabolism involves a defective GABA amino-
transferase, an enzyme that participates in the catabo-
lism of GABA subsequent to its postsynaptic release in
brain tissue.
NH2
+
H2N
C
(Kidney)
NH
ARGININE-GLYCINE
TRANSAMIDINASE
NH2
CH2
+
H2N
C
CH2
HN CH2
COO-
CH2
+H3N
CH
2
COO-
Ornithine
Glycocyamine
+
(guanidoacetate)
H
C NH
Glycine
3
(Liver)
COO-
S-Adenosyl-
ATP
L
-Arginine
methionine
GUANIDOACETATE
METHYLTRANSFERASE
S-Adenosyl-
ADP
homocysteine
O
H
N
C
NONENZYMATIC
IN MUSCLE
NH
P
HN
C
HN C
N
CH2
N CH2
COO-
CH3
Pi + H2O
CH3
Creatinine
Creatine phosphate
Figure 31-6. Biosynthesis and metabolism of creatine and creatinine.
COO-
H
C NH3+
α-KA
CH2
L-GLUTAMATE
DECARBOXYLASE
TRANSAMINASE
CH2
α-AA
COO-
PLP
L-Glutamate
CO2
CH2OH
COO-
CH
2
C O
+H
CH2
CH2
CH2
COO-
CH2
CH2
3N
γ-Aminobutyrate
COO-
CH
2
γ-Hydroxybutyrate
-
[O]
COO
α-Ketoglutarate
NAD+
PLP
LACTATE
DEHYDROGENASE
[NH4+]
NADH + H+
CO
2
O
SUCCINIC
SEMIALDEHYDE
-
C H
COO
DEHYDROGENASE
CH2
CH2
CH
2
CH2
+
H
2
O
NAD+
NADH + H
COO-
COO-
Succinate semialdehyde
Succinate
Figure 31-7. Metabolism of γ-aminobutyrate. (α-KA, α-keto acids; α-AA, α-amino acids; PLP, pyri-
doxal phosphate.)
268
CONVERSION OF AMINO ACIDS TO SPECIALIZED PRODUCTS
/
269
SUMMARY
• Decarboxylation of histidine forms histamine, and
several dipeptides are derived from histidine and
• In addition to their roles in proteins and polypep-
β-alanine.
tides, amino acids participate in a wide variety of ad-
• Arginine serves as the formamidine donor for crea-
ditional biosynthetic processes.
tine biosynthesis, participates in polyamine biosyn-
• Glycine participates in the biosynthesis of heme,
thesis, and provides the nitrogen of nitric oxide
purines, and creatine and is conjugated to bile acids
(NO).
and to the urinary metabolites of many drugs.
• Important tryptophan metabolites include serotonin,
• In addition to its roles in phospholipid and sphingo-
melanin, and melatonin.
sine biosynthesis, serine provides carbons 2 and 8 of
• Tyrosine forms both epinephrine and norepineph-
purines and the methyl group of thymine.
rine, and its iodination forms thyroid hormone.
• S-Adenosylmethionine, the methyl group donor for
many biosynthetic processes, also participates directly
in spermine and spermidine biosynthesis.
• Glutamate and ornithine form the neurotransmitter
γ-aminobutyrate (GABA).
REFERENCE
• The thioethanolamine of coenzyme A and the tau-
Scriver CR et al (editors): The Metabolic and Molecular Bases of
rine of taurocholic acid arise from cysteine.
Inherited Disease, 8th ed. McGraw-Hill, 2001.
Porphyrins & Bile Pigments
32
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
substituent positions numbered as shown in Figure
32-2. Various porphyrins are represented in Figures
The biochemistry of the porphyrins and of the bile pig-
32-2, 32-3, and 32-4.
ments is presented in this chapter. These topics are
The arrangement of the acetate (A) and propionate
closely related, because heme is synthesized from por-
(P) substituents in the uroporphyrin shown in Figure
phyrins and iron, and the products of degradation of
32-2 is asymmetric (in ring IV, the expected order of
heme are the bile pigments and iron.
the A and P substituents is reversed). A porphyrin with
Knowledge of the biochemistry of the porphyrins
this type of asymmetric substitution is classified as a
and of heme is basic to understanding the varied func-
type III porphyrin. A porphyrin with a completely sym-
tions of hemoproteins (see below) in the body. The
metric arrangement of the substituents is classified as a
porphyrias are a group of diseases caused by abnormal-
type I porphyrin. Only types I and III are found in na-
ities in the pathway of biosynthesis of the various por-
ture, and the type III series is far more abundant (Figure
phyrins. Although porphyrias are not very prevalent,
32-3)—and more important because it includes heme.
physicians must be aware of them. A much more preva-
Heme and its immediate precursor, protoporphyrin
lent clinical condition is jaundice, due to elevation of
IX (Figure 32-4), are both type III porphyrins (ie, the
bilirubin in the plasma. This elevation is due to over-
methyl groups are asymmetrically distributed, as in type
production of bilirubin or to failure of its excretion and
III coproporphyrin). However, they are sometimes
is seen in numerous diseases ranging from hemolytic
identified as belonging to series IX, because they were
anemias to viral hepatitis and to cancer of the pancreas.
designated ninth in a series of isomers postulated by
Hans Fischer, the pioneer worker in the field of por-
phyrin chemistry.
METALLOPORPHYRINS
& HEMOPROTEINS ARE
IMPORTANT IN NATURE
HEME IS SYNTHESIZED FROM
Porphyrins are cyclic compounds formed by the linkage
SUCCINYL-COA & GLYCINE
of four pyrrole rings through
HC methenyl
Heme is synthesized in living cells by a pathway that has
bridges (Figure 32-1). A characteristic property of the
been much studied. The two starting materials are suc-
porphyrins is the formation of complexes with metal
cinyl-CoA, derived from the citric acid cycle in mito-
ions bound to the nitrogen atom of the pyrrole rings.
chondria, and the amino acid glycine. Pyridoxal phos-
Examples are the iron porphyrins such as heme of he-
phate is also necessary in this reaction to
“activate”
moglobin and the magnesium-containing porphyrin
glycine. The product of the condensation reaction be-
chlorophyll, the photosynthetic pigment of plants.
tween succinyl-CoA and glycine is α-amino-β-ketoadipic
Proteins that contain heme
(hemoproteins) are
acid, which is rapidly decarboxylated to form α-amino-
widely distributed in nature. Examples of their impor-
levulinate (ALA) (Figure 32-5). This reaction sequence
tance in humans and animals are listed in Table 32-1.
is catalyzed by ALA synthase, the rate-controlling en-
zyme in porphyrin biosynthesis in mammalian liver.
Synthesis of ALA occurs in mitochondria. In the cy-
Natural Porphyrins Have Substituent Side
tosol, two molecules of ALA are condensed by the en-
Chains on the Porphin Nucleus
zyme ALA dehydratase to form two molecules of water
The porphyrins found in nature are compounds in
and one of porphobilinogen (PBG) (Figure 32-5). ALA
which various side chains are substituted for the eight
dehydratase is a zinc-containing enzyme and is sensitive
hydrogen atoms numbered in the porphin nucleus
to inhibition by lead, as can occur in lead poisoning.
shown in Figure 32-1. As a simple means of showing
The formation of a cyclic tetrapyrrole—ie, a por-
these substitutions, Fischer proposed a shorthand for-
phyrin—occurs by condensation of four molecules of
mula in which the methenyl bridges are omitted and
PBG (Figure 32-6). These four molecules condense in a
each pyrrole ring is shown as indicated with the eight
head-to-tail manner to form a linear tetrapyrrole, hy-
270
PORPHYRINS & BILE PIGMENTS
/
271
HC
CH
1
2
A
P
HC
CH
I
I
8
3
A
A
N
N
H
IV
II
IV
II
Pyrrole
7
III
4
P
III
P
1
2
6
5
P
A
H
H
C
C
Figure 32-2. Uroporphyrin III. A (acetate) =
δ
I
α
HC
C
C CH
CH2COOH; P (propionate) = CH2CH2COOH.
N
3
8 HC
C
C
CH
IV NH
HN
(CH2 ), which do not form a conjugated ring sys-
7
HC
C
C
CH
tem. Thus, these compounds are colorless (as are all
4
porphyrinogens). However, the porphyrinogens are
HC
C
C
CH
γ
III
β
readily auto-oxidized to their respective colored por-
C
C
phyrins. These oxidations are catalyzed by light and by
H
H
6
5
the porphyrins that are formed.
Uroporphyrinogen III is converted to copropor-
Porphin
phyrinogen III by decarboxylation of all of the acetate
(C20H14N4)
(A) groups, which changes them to methyl (M) sub-
Figure 32-1. The porphin molecule. Rings are la-
stituents. The reaction is catalyzed by uroporphyrino-
gen decarboxylase, which is also capable of converting
beled I, II, III, and IV. Substituent positions on the rings
uroporphyrinogen I to coproporphyrinogen I (Figure
are labeled 1, 2, 3, 4, 5, 6, 7, and 8. The methenyl
32-7). Coproporphyrinogen III then enters the mito-
bridges ( HC) are labeled α, β, γ, and δ.
chondria, where it is converted to protoporphyrinogen
III and then to protoporphyrin III. Several steps are
droxymethylbilane (HMB). The reaction is catalyzed by
involved in this conversion. The mitochondrial enzyme
uroporphyrinogen I synthase, also named PBG deami-
coproporphyrinogen oxidase catalyzes the decarboxy-
nase or HMB synthase. HMB cyclizes spontaneously to
lation and oxidation of two propionic side chains to
form uroporphyrinogen I (left-hand side of Figure
form protoporphyrinogen. This enzyme is able to act
32-6) or is converted to uroporphyrinogen III by the
only on type III coproporphyrinogen, which would ex-
action of uroporphyrinogen III synthase (right-hand side
plain why type I protoporphyrins do not generally occur
of Figure 32-6). Under normal conditions, the uropor-
in nature. The oxidation of protoporphyrinogen to pro-
phyrinogen formed is almost exclusively the III isomer,
toporphyrin is catalyzed by another mitochondrial en-
but in certain of the porphyrias (discussed below), the
zyme, protoporphyrinogen oxidase. In mammalian
type I isomers of porphyrinogens are formed in excess.
liver, the conversion of coproporphyrinogen to proto-
Note that both of these uroporphyrinogens have
porphyrin requires molecular oxygen.
the pyrrole rings connected by methylene bridges
Formation of Heme Involves Incorporation
of Iron Into Protoporphyrin
Table 32-1. Examples of some important human
The final step in heme synthesis involves the incorpora-
and animal hemoproteins.1
tion of ferrous iron into protoporphyrin in a reaction
catalyzed by ferrochelatase (heme synthase), another
Protein
Function
mitochondrial enzyme (Figure 32-4).
Hemoglobin
Transport of oxygen in blood
A summary of the steps in the biosynthesis of the
Myoglobin
Storage of oxygen in muscle
porphyrin derivatives from PBG is given in Figure
Cytochrome c
Involvement in electron transport chain
32-8. The last three enzymes in the pathway and ALA
Cytochrome P450
Hydroxylation of xenobiotics
synthase are located in the mitochondrion, whereas the
Catalase
Degradation of hydrogen peroxide
other enzymes are cytosolic. Both erythroid and non-
Tryptophan
Oxidation of trypotophan
erythroid (“housekeeping”) forms of the first four en-
pyrrolase
zymes are found. Heme biosynthesis occurs in most
1The functions of the above proteins are described in various
mammalian cells with the exception of mature erythro-
chapters of this text.
cytes, which do not contain mitochondria. However,
272
/
CHAPTER 32
A
P
A
P
P
A
A
A
Uroporphyrins were first
found in the urine, but they
are not restricted to urine.
A
P
P
P
P
A
P
A
Uroporphyrin I
Uroporphyrin III
M
P
M
P
P
M
M
M
Coproporphyrins were first
isolated from feces, but they
are also found in urine.
M
P
P
P
P
M
P
M
Coproporphyrin I
Coproporphyrin III
Figure 32-3. Uroporphyrins and coproporphyrins. A (acetate); P (propionate); M
(methyl) = CH3; V (vinyl) = CHCH2.
approximately 85% of heme synthesis occurs in eryth-
ALAS1. This repression-derepression mechanism is de-
roid precursor cells in the bone marrow and the major-
picted diagrammatically in Figure 32-9. Thus, the rate
ity of the remainder in hepatocytes.
of synthesis of ALAS1 increases greatly in the absence
The porphyrinogens described above are colorless,
of heme and is diminished in its presence. The turnover
containing six extra hydrogen atoms as compared with
rate of ALAS1 in rat liver is normally rapid (half-life
the corresponding colored porphyrins. These reduced
about 1 hour), a common feature of an enzyme catalyz-
porphyrins (the porphyrinogens) and not the corre-
ing a rate-limiting reaction. Heme also affects transla-
sponding porphyrins are the actual intermediates in the
tion of the enzyme and its transfer from the cytosol to
biosynthesis of protoporphyrin and of heme.
the mitochondrion.
Many drugs when administered to humans can re-
sult in a marked increase in ALAS1. Most of these
ALA Synthase Is the Key Regulatory
drugs are metabolized by a system in the liver that uti-
Enzyme in Hepatic Biosynthesis of Heme
lizes a specific hemoprotein, cytochrome P450 (see
ALA synthase occurs in both hepatic (ALAS1) and ery-
Chapter 53). During their metabolism, the utilization
throid (ALAS2) forms. The rate-limiting reaction in the
of heme by cytochrome P450 is greatly increased,
synthesis of heme in liver is that catalyzed by ALAS1
which in turn diminishes the intracellular heme con-
(Figure
32-5), a regulatory enzyme. It appears that
centration. This latter event effects a derepression of
heme, probably acting through an aporepressor mole-
ALAS1 with a corresponding increased rate of heme
cule, acts as a negative regulator of the synthesis of
synthesis to meet the needs of the cells.
M
V
M
V
M
M
Fe2+
M
M
Fe2+
FERROCHELATASE
P
V
P
V
P
M
P
M
Protoporphyrin III (IX)
Heme
(parent porphyrin of heme)
(prosthetic group of hemoglobin)
Figure 32-4. Addition of iron to protoporphyrin to form heme.
PORPHYRINS & BILE PIGMENTS
/
273
COOH
COOH
COOH
CH2
ALA
CH2
ALA
CH2
SYNTHASE
SYNTHASE
Succinyl-CoA
CH2
CH2
CH2
(“active”
CoA • SH
CO2
succinate)
C O
C O
C O
S CoA
H
C NH2
H
C NH2
+
Pyridoxal
H
phosphate
COOH
H
Glycine
H
C NH2
α-Amino-β-ketoadipate
δ-Aminolevulinate (ALA)
COOH
COOH
COOH
COOH
CH2
COOH
CH2
2H2O
CH2
CH2
CH2
CH2
CH2
O
C
C
C
ALA
C O
H
C
H
C
CH
DEHYDRATASE
CH2
H
CH2
N
NH
H
NH2
NH2
Two molecules of
Porphobilinogen
δ-aminolevulinate
(first precursor pyrrole)
Figure 32-5. Biosynthesis of porphobilinogen. ALA synthase occurs in the mitochon-
dria, whereas ALA dehydratase is present in the cytosol.
Several factors affect drug-mediated derepression of
side chains present. This band is termed the Soret band
ALAS1 in liver—eg, the administration of glucose can
after its discoverer, the French physicist Charles Soret.
prevent it, as can the administration of hematin (an ox-
When porphyrins dissolved in strong mineral acids
idized form of heme).
or in organic solvents are illuminated by ultraviolet
The importance of some of these regulatory mecha-
light, they emit a strong red fluorescence. This fluores-
nisms is further discussed below when the porphyrias
cence is so characteristic that it is often used to detect
are described.
small amounts of free porphyrins. The double bonds
Regulation of the erythroid form of ALAS (ALAS2)
joining the pyrrole rings in the porphyrins are responsi-
differs from that of ALAS1. For instance, it is not in-
ble for the characteristic absorption and fluorescence of
duced by the drugs that affect ALAS1, and it does not
these compounds; these double bonds are absent in the
undergo feedback regulation by heme.
porphyrinogens.
An interesting application of the photodynamic
properties of porphyrins is their possible use in the
PORPHYRINS ARE COLORED
treatment of certain types of cancer, a procedure called
& FLUORESCE
cancer phototherapy. Tumors often take up more por-
phyrins than do normal tissues. Thus, hematopor-
The various porphyrinogens are colorless, whereas the
phyrin or other related compounds are administered to
various porphyrins are all colored. In the study of por-
a patient with an appropriate tumor. The tumor is then
phyrins or porphyrin derivatives, the characteristic ab-
exposed to an argon laser, which excites the porphyrins,
sorption spectrum that each exhibits—in both the visible
producing cytotoxic effects.
and the ultraviolet regions of the spectrum—is of great
value. An example is the absorption curve for a solution
Spectrophotometry Is Used to Test
of porphyrin in 5% hydrochloric acid (Figure 32-10).
for Porphyrins & Their Precursors
Note particularly the sharp absorption band near 400
nm. This is a distinguishing feature of the porphin ring
Coproporphyrins and uroporphyrins are of clinical in-
and is characteristic of all porphyrins regardless of the
terest because they are excreted in increased amounts in
274
/
CHAPTER 32
HOOC
COOH
pain and of a variety of neuropsychiatric findings); oth-
A
H2C
CH2
P
erwise, patients will be subjected to inappropriate treat-
CH2
ments. It has been speculated that King George III had
a type of porphyria, which may account for his periodic
C
C
confinements in Windsor Castle and perhaps for some
C
CH
of his views regarding American colonists. Also, the
H2C
N
photosensitivity (favoring nocturnal activities) and se-
H
NH2
vere disfigurement exhibited by some victims of con-
genital erythropoietic porphyria have led to the sugges-
Four molecules of
porphobilinogen
tion that these individuals may have been the
prototypes of so-called werewolves. No evidence to sup-
UROPORPHYRINOGEN I
4NH3
SYNTHASE
port this notion has been adduced.
Hydroxymethylbilane
(linear tetrapyrrole)
Biochemistry Underlies the
SPONTANEOUS
UROPORPHYRINOGEN III
Causes, Diagnoses, & Treatments
CYCLIZATION
SYNTHASE
of the Porphyrias
Six major types of porphyria have been described, re-
A
P
A
P
A
P
A
P
sulting from depressions in the activities of enzymes 3
through 8 shown in Figure 32-9 (see also Table 32-2).
C
C
H2
C
C
C
C
H2
C
C
C
C
Assay of the activity of one or more of these enzymes
I
II
I
II
C
C
C
C
C
C
C
C
using an appropriate source (eg, red blood cells) is thus
N
N
N
N
important in making a definitive diagnosis in a sus-
H
H
H
H
CH2
CH2
CH2
CH2
pected case of porphyria. Individuals with low activities
H
H
H
H
N
N
N
N
of enzyme 1 (ALAS2) develop anemia, not porphyria
(see Table 32-2). Patients with low activities of enzyme
C
C
C
C
C
C
C
C
IV
III
IV
III
C
C
2 (ALA dehydratase) have been reported, but very
C
C
H2
C
C
C
C
H2
C
C
rarely; the resulting condition is called ALA dehy-
P
A
P
A
A
P
P
A
dratase-deficient porphyria.
Type I
Type III
In general, the porphyrias described are inherited in
uroporphyrinogen
uroporphyrinogen
an autosomal dominant manner, with the exception of
congenital erythropoietic porphyria, which is inherited
Figure 32-6. Conversion of porphobilinogen to uro-
in a recessive mode. The precise abnormalities in the
porphyrinogens. Uroporphyrinogen synthase I is also
genes directing synthesis of the enzymes involved in
called porphobilinogen (PBG) deaminase or hydroxy-
heme biosynthesis have been determined in some in-
methylbilane (HMB) synthase.
stances. Thus, the use of appropriate gene probes has
made possible the prenatal diagnosis of some of the
porphyrias.
the porphyrias. These compounds, when present in
As is true of most inborn errors, the signs and symp-
urine or feces, can be separated from each other by ex-
toms of porphyria result from either a deficiency of
traction with appropriate solvent mixtures. They can
metabolic products beyond the enzymatic block or
then be identified and quantified using spectrophoto-
from an accumulation of metabolites behind the block.
metric methods.
If the enzyme lesion occurs early in the pathway
ALA and PBG can also be measured in urine by ap-
prior to the formation of porphyrinogens (eg, enzyme 3
propriate colorimetric tests.
of Figure 32-9, which is affected in acute intermittent
porphyria), ALA and PBG will accumulate in body tis-
sues and fluids
(Figure
32-11). Clinically, patients
THE PORPHYRIAS ARE GENETIC
complain of abdominal pain and neuropsychiatric
DISORDERS OF HEME METABOLISM
symptoms. The precise biochemical cause of these
The porphyrias are a group of disorders due to abnor-
symptoms has not been determined but may relate to
malities in the pathway of biosynthesis of heme; they
elevated levels of ALA or PBG or to a deficiency of
can be genetic or acquired. They are not prevalent, but
heme.
it is important to consider them in certain circum-
On the other hand, enzyme blocks later in the path-
stances (eg, in the differential diagnosis of abdominal
way result in the accumulation of the porphyrinogens
PORPHYRINS & BILE PIGMENTS
/
275
A
P
M
P
4CO2
I
I
P
A
P
M
IV
II
IV
II
A
III
P
M
III
P
P A
P M
Uroporphyrinogen I
Coproporphyrinogen I
UROPORPHYRINOGEN
DECARBOXYLASE
A
P
M
P
I
I
A
A
M
M
IV
II
IV
II
P
III
P
P
III
P
Figure 32-7. Decarboxylation of uropor-
4CO2
P A
P M
phyrinogens to coproporphyrinogens in cy-
Uroporphyrinogen III
Coproporphyrinogen III
tosol. (A, acetyl; M, methyl; P, propionyl.)
Porphobilinogen
UROPORPHYRINOGEN I
SYNTHASE
Hydroxymethylbilane
UROPORPHYRINOGEN III
SYNTHASE
SPONTANEOUS
6H
6H
Uroporphyrin
Uroporphyrinogen
Uroporphyrinogen
Uroporphyrin
III
Light
III
I
Light
I
UROPORPHYRINOGEN
6H
DECARBOXYLASE
6H
4CO2
4CO2
Coproporphyrin
Coproporphyrinogen
Coproporphyrinogen
Coproporphyrin
III
Light
III
I
Light
I
COPROPORPHYRINOGEN
OXIDASE
Protoporphyrinogen III
Or light in vitro
PROTOPORPHYRINOGEN
OXIDASE
6H
Protoporphyrin III
Fe2+
FERROCHELATASE
Heme
Figure 32-8. Steps in the biosynthesis of the porphyrin derivatives from porphobilinogen. Uropor-
phyrinogen I synthase is also called porphobilinogen deaminase or hydroxymethylbilane synthase.
276
/
CHAPTER 32
Hemoproteins
Proteins
Heme
Aporepressor
8. FERROCHELATASE
Fe2+
Protoporphyrin III
7. PROTOPORPHYRINOGEN
OXIDASE
Protoporphyrinogen III
6. COPROPORPHYRINOGEN
OXIDASE
Coproporphyrinogen III
5. UROPORPHYRINOGEN
DECARBOXYLASE
Uroporphyrinogen III
4. UROPORPHYRINOGEN III
SYNTHASE
Hydroxymethylbilane
3. UROPORPHYRINOGEN I
SYNTHASE
Porphobilinogen
2. ALA
DEHYDRATASE
ALA
1. ALA
SYNTHASE
Succinyl-CoA + Glycine
Figure 32-9. Intermediates, enzymes, and regulation of heme syn-
thesis. The enzyme numbers are those referred to in column 1 of Table
32-2. Enzymes 1, 6, 7, and 8 are located in mitochondria, the others in
the cytosol. Mutations in the gene encoding enzyme 1 causes X-linked
sideroblastic anemia. Mutations in the genes encoding enzymes 2-8
cause the porphyrias, though only a few cases due to deficiency of en-
zyme 2 have been reported. Regulation of hepatic heme synthesis oc-
curs at ALA synthase (ALAS1) by a repression-derepression mecha-
nism mediated by heme and its hypothetical aporepressor. The
dotted lines indicate the negative ( − ) regulation by repression. En-
zyme 3 is also called porphobilinogen deaminase or hydroxymethyl-
bilane synthase.
PORPHYRINS & BILE PIGMENTS
/
277
Mutations in DNA
5
4
Abnormalities of the
enzymes of heme synthesis
3
2
Accumulation of
Accumulation of
ALA and PBG and/or
porphyrinogens in skin
decrease in heme in
1
and tissues
cells and body fluids
300
400
500
600
700
Spontaneous oxidation
Wavelength (nm)
Neuropsychiatric signs
of porphyrinogens to
and symptoms
porphyrins
Figure 32-10. Absorption spectrum of hematopor-
phyrin (0.01% solution in 5% HCl).
Photosensitivity
Figure 32-11. Biochemical causes of the major
signs and symptoms of the porphyrias.
Table 32-2. Summary of major findings in the porphyrias.1
Enzyme Involved2
Type, Class, and MIM Number
Major Signs and Symptoms
Results of Laboratory Tests
1. ALA synthase
X-linked sideroblastic anemia3
Anemia
Red cell counts and hemoglobin
(erythroid form)
(erythropoietic) (MIM
decreased
201300)
2. ALA dehydratase
ALA dehydratase deficiency
Abdominal pain, neuropsychiatric
Urinary δ-aminolevulinic acid
(hepatic) (MIM 125270)
symptoms
3. Uroporphyrinogen I
Acute intermittent porphyria
Abdominal pain, neuropsychiatric
Urinary porphobilinogen positive,
synthase4
(hepatic) (MIM 176000)
symptoms
uroporphyrin positive
4. Uroporphyrinogen III
Congenital erythropoietic
No photosensitivity
Uroporphyrin positive, porpho-
synthase
(erythropoietic) (MIM
bilinogen negative
263700)
5. Uroporphyrinogen
Porphyria cutanea tarda (he-
Photosensitivity
Uroporphyrin positive, porpho-
decarboxylase
patic) (MIM 176100)
bilinogen negative
6. Coproporphyrinogen
Hereditary coproporphyria
Photosensitivity, abdominal pain,
Urinary porphobilinogen posi-
oxidase
(hepatic) (MIM 121300)
neuropsychiatric symptoms
tive, urinary uroporphyrin
positive, fecal protopor-
phyrin positive
7. Protoporphyrinogen
Variegate porphyria (hepatic)
Photosensitivity, abdominal pain,
Urinary porphobilinogen posi-
oxidase
(MIM 176200)
neuropsychiatric symptoms
tive, fecal protoporphyrin
positive
8. Ferrochelatase
Protoporphyria (erythropoietic)
Photosensitivity
Fecal protoporphyrin posi-
`
(MIM 177000)
tive, red cell protoporphyrin
positive
1Only the biochemical findings in the active stages of these diseases are listed. Certain biochemical abnormalities are detectable in the la-
tent stages of some of the above conditions. Conditions 3, 5, and 8 are generally the most prevalent porphyrias.
2The numbering of the enzymes in this table corresponds to that used in Figure 32-9.
3X-linked sideroblastic anemia is not a porphyria but is included here because δ−aminolevulinic acid synthase is involved.
4This enzyme is also called porphobilinogen deaminase or hydroxymethylbilane synthase.
278
/
CHAPTER 32
indicated in Figures 32-9 and 32-11. Their oxidation
chrome P450. Ingestion of large amounts of carbohy-
products, the corresponding porphyrin derivatives,
drates (glucose loading) or administration of hematin (a
cause photosensitivity, a reaction to visible light of
hydroxide of heme) may repress ALAS1, resulting in di-
about 400 nm. The porphyrins, when exposed to light
minished production of harmful heme precursors. Pa-
of this wavelength, are thought to become “excited”
tients exhibiting photosensitivity may benefit from ad-
and then react with molecular oxygen to form oxygen
ministration of β-carotene; this compound appears to
radicals. These latter species injure lysosomes and other
lessen production of free radicals, thus diminishing
organelles. Damaged lysosomes release their degradative
photosensitivity. Sunscreens that filter out visible light
enzymes, causing variable degrees of skin damage, in-
can also be helpful to such patients.
cluding scarring.
The porphyrias can be classified on the basis of the
organs or cells that are most affected. These are gener-
CATABOLISM OF HEME
ally organs or cells in which synthesis of heme is partic-
PRODUCES BILIRUBIN
ularly active. The bone marrow synthesizes considerable
hemoglobin, and the liver is active in the synthesis of
Under physiologic conditions in the human adult, 1-2
another hemoprotein, cytochrome P450. Thus, one
× 108 erythrocytes are destroyed per hour. Thus, in 1
classification of the porphyrias is to designate them as
day, a 70-kg human turns over approximately 6 g of he-
predominantly either erythropoietic or hepatic; the
moglobin. When hemoglobin is destroyed in the body,
types of porphyrias that fall into these two classes are so
globin is degraded to its constituent amino acids,
characterized in Table 32-2. Porphyrias can also be
which are reused, and the iron of heme enters the iron
classified as acute or cutaneous on the basis of their
pool, also for reuse. The iron-free porphyrin portion of
clinical features. Why do specific types of porphyria af-
heme is also degraded, mainly in the reticuloendothelial
fect certain organs more markedly than others? A par-
cells of the liver, spleen, and bone marrow.
tial answer is that the levels of metabolites that cause
The catabolism of heme from all of the heme pro-
damage (eg, ALA, PBG, specific porphyrins, or lack of
teins appears to be carried out in the microsomal frac-
heme) can vary markedly in different organs or cells de-
tions of cells by a complex enzyme system called heme
pending upon the differing activities of their heme-
oxygenase. By the time the heme derived from heme
forming enzymes.
proteins reaches the oxygenase system, the iron has usu-
As described above, ALAS1 is the key regulatory en-
ally been oxidized to the ferric form, constituting
zyme of the heme biosynthetic pathway in liver. A large
hemin. The heme oxygenase system is substrate-in-
number of drugs (eg, barbiturates, griseofulvin) induce
ducible. As depicted in Figure 32-12, the hemin is re-
the enzyme. Most of these drugs do so by inducing cy-
duced to heme with NADPH, and, with the aid of
tochrome P450 (see Chapter 53), which uses up heme
more NADPH, oxygen is added to the α-methenyl
and thus derepresses (induces) ALAS1. In patients with
bridge between pyrroles I and II of the porphyrin. The
porphyria, increased activities of ALAS1 result in in-
ferrous iron is again oxidized to the ferric form. With
creased levels of potentially harmful heme precursors
the further addition of oxygen, ferric ion is released,
prior to the metabolic block. Thus, taking drugs that
carbon monoxide is produced, and an equimolar
cause induction of cytochrome P450 (so-called micro-
quantity of biliverdin results from the splitting of the
somal inducers) can precipitate attacks of porphyria.
tetrapyrrole ring.
The diagnosis of a specific type of porphyria can
In birds and amphibia, the green biliverdin IX is ex-
generally be established by consideration of the clinical
creted; in mammals, a soluble enzyme called biliverdin
and family history, the physical examination, and ap-
reductase reduces the methenyl bridge between pyrrole
propriate laboratory tests. The major findings in the six
III and pyrrole IV to a methylene group to produce
principal types of porphyria are listed in Table 32-2.
bilirubin, a yellow pigment (Figure 32-12).
High levels of lead can affect heme metabolism by
It is estimated that 1 g of hemoglobin yields 35 mg
combining with SH groups in enzymes such as fer-
of bilirubin. The daily bilirubin formation in human
rochelatase and ALA dehydratase. This affects por-
adults is approximately 250-350 mg, deriving mainly
phyrin metabolism. Elevated levels of protoporphyrin
from hemoglobin but also from ineffective erythro-
are found in red blood cells, and elevated levels of ALA
poiesis and from various other heme proteins such as
and of coproporphyrin are found in urine.
cytochrome P450.
It is hoped that treatment of the porphyrias at the
The chemical conversion of heme to bilirubin by
gene level will become possible. In the meantime, treat-
reticuloendothelial cells can be observed in vivo as the
ment is essentially symptomatic. It is important for pa-
purple color of the heme in a hematoma is slowly con-
tients to avoid drugs that cause induction of cyto-
verted to the yellow pigment of bilirubin.
PORPHYRINS & BILE PIGMENTS
/
279
Heme
O
I
α
Hemin
HN
N
IV
N
+
N II
Fe3
HN
P
N
H
III
P
P
H
P
NADPH
HN
NADP
HN
Bilirubin
I
α
Heme
O
N
NADP
IV
N
+
N II
Fe2
NADPH
P
N
III
O
P
HN
II
NADPH
O
2
NADP
HN
III
3+
Fe
(reutilized)
OH
CO (exhaled)
P
I
P
N
N
IV
O2
IV
N
Fe3
+
N II
P
N
III
HN
I
Biliverdin
P
O
Figure 32-12. Schematic representation of the microsomal heme oxygenase system. (Modified from
Schmid R, McDonough AF in: The Porphyrins. Dolphin D [editor]. Academic Press, 1978.)
280
/
CHAPTER 32
Bilirubin formed in peripheral tissues is transported
converted to water-soluble derivatives by conjugation in
to the liver by plasma albumin. The further metabolism
preparation for excretion (see Chapter 53).
of bilirubin occurs primarily in the liver. It can be di-
The conjugation of bilirubin is catalyzed by a spe-
vided into three processes: (1) uptake of bilirubin by
cific glucuronosyltransferase. The enzyme is mainly
liver parenchymal cells,
(2) conjugation of bilirubin
located in the endoplasmic reticulum, uses UDP-
with glucuronate in the endoplasmic reticulum, and (3)
glucuronic acid as the glucuronosyl donor, and is re-
secretion of conjugated bilirubin into the bile. Each of
ferred to as bilirubin-UGT. Bilirubin monoglucuronide
these processes will be considered separately.
is an intermediate and is subsequently converted to the
diglucuronide (Figures 32-13 and 32-14). Most of the
bilirubin excreted in the bile of mammals is in the form
THE LIVER TAKES UP BILIRUBIN
of bilirubin diglucuronide. However, when bilirubin
conjugates exist abnormally in human plasma (eg, in
Bilirubin is only sparingly soluble in water, but its solu-
obstructive jaundice), they are predominantly mono-
bility in plasma is increased by noncovalent binding to
glucuronides. Bilirubin-UGT activity can be induced
albumin. Each molecule of albumin appears to have
by a number of clinically useful drugs, including phe-
one high-affinity site and one low-affinity site for
nobarbital. More information about glucuronosylation
bilirubin. In 100 mL of plasma, approximately 25 mg
is presented below in the discussion of inherited disor-
of bilirubin can be tightly bound to albumin at its high-
ders of bilirubin conjugation.
affinity site. Bilirubin in excess of this quantity can be
bound only loosely and thus can easily be detached and
diffuse into tissues. A number of compounds such as
Bilirubin Is Secreted Into Bile
antibiotics and other drugs compete with bilirubin for
Secretion of conjugated bilirubin into the bile occurs by
the high-affinity binding site on albumin. Thus, these
an active transport mechanism, which is probably rate-
compounds can displace bilirubin from albumin and
limiting for the entire process of hepatic bilirubin me-
have significant clinical effects.
tabolism. The protein involved is MRP-2 (multidrug
In the liver, the bilirubin is removed from albumin
resistance-like protein 2), also called multispecific or-
and taken up at the sinusoidal surface of the hepato-
ganic anion transporter (MOAT). It is located in the
cytes by a carrier-mediated saturable system. This facil-
plasma membrane of the bile canalicular membrane
itated transport system has a very large capacity, so
and handles a number of organic anions. It is a member
that even under pathologic conditions the system does
of the family of ATP-binding cassette (ABC) trans-
not appear to be rate-limiting in the metabolism of
porters. The hepatic transport of conjugated bilirubin
bilirubin.
into the bile is inducible by those same drugs that are
Since this facilitated transport system allows the
capable of inducing the conjugation of bilirubin. Thus,
equilibrium of bilirubin across the sinusoidal mem-
the conjugation and excretion systems for bilirubin be-
brane of the hepatocyte, the net uptake of bilirubin will
have as a coordinated functional unit.
be dependent upon the removal of bilirubin via subse-
Figure 32-15 summarizes the three major processes
quent metabolic pathways.
involved in the transfer of bilirubin from blood to bile.
Once bilirubin enters the hepatocytes, it can bind to
Sites that are affected in a number of conditions caus-
certain cytosolic proteins, which help to keep it solubi-
ing jaundice (see below) are also indicated.
lized prior to conjugation. Ligandin (a family of glu-
tathione S-transferases) and protein Y are the involved
proteins. They may also help to prevent efflux of biliru-
bin back into the blood stream.
O O
-OOC(CH2O)4C
O
C
C
O
C(CH2O)4COO-
Conjugation of Bilirubin With Glucuronic
H2C
CH2
Acid Occurs in the Liver
H2C
CH
2
M V M
M
M
V
Bilirubin is nonpolar and would persist in cells (eg,
II
III
IV
I
bound to lipids) if not rendered water-soluble. Hepato-
O
C
C
C
O
cytes convert bilirubin to a polar form, which is readily
excreted in the bile, by adding glucuronic acid mole-
Figure 32-13. Structure of bilirubin diglucuronide
cules to it. This process is called conjugation and can
(conjugated, “direct-reacting” bilirubin). Glucuronic
employ polar molecules other than glucuronic acid (eg,
acid is attached via ester linkage to the two propionic
sulfate). Many steroid hormones and drugs are also
acid groups of bilirubin to form an acylglucuronide.
PORPHYRINS & BILE PIGMENTS
/
281
UDP-GLUCOSE
DEHYDROGENASE
UDP-Glucose
UDP-Glucuronic acid
2NAD+
2NADH + 2H+
UDP-GLUCURONOSYL-
TRANSFERASE
UDP-Glucuronic acid
Bilirubin monoglucuronide
+
+
Bilirubin
UDP
Figure 32-14. Conjugation of bilirubin
with glucuronic acid. The glucuronate donor,
UDP-GLUCURONOSYL-
UDP-glucuronic acid, is formed from UDP-
TRANSFERASE
UDP-Glucuronic acid
Bilirubin diglucuronide
glucose as depicted. The UDP-glucuronosyl-
+
+
Bilirubin monoglucuronide
UDP
transferase is also called bilirubin-UGT.
Conjugated Bilirubin Is Reduced to
Urobilinogen by Intestinal Bacteria
As the conjugated bilirubin reaches the terminal ileum
and the large intestine, the glucuronides are removed by
BLOOD
specific bacterial enzymes (
-glucuronidases), and the
Bilirubin • Albumin
pigment is subsequently reduced by the fecal flora to a
group of colorless tetrapyrrolic compounds called uro-
1. UPTAKE
bilinogens (Figure 32-16). In the terminal ileum and
large intestine, a small fraction of the urobilinogens is re-
absorbed and reexcreted through the liver to constitute
HEPATOCYTE
the enterohepatic urobilinogen cycle. Under abnormal
Bilirubin
conditions, particularly when excessive bile pigment is
2. CONJUGATION
formed or liver disease interferes with this intrahepatic
Neonatal jaundice
cycle, urobilinogen may also be excreted in the urine.
UDP-GlcUA
“Toxic” jaundice
Normally, most of the colorless urobilinogens
UDP-GlcUA
Crigler-Najjar syndrome
formed in the colon by the fecal flora are oxidized there
Gilbert syndrome
to urobilins (colored compounds) and are excreted in
the feces
(Figure
32-16). Darkening of feces upon
Bilirubin diglucuronide
standing in air is due to the oxidation of residual uro-
bilinogens to urobilins.
3. SECRETION
Dubin-Johnson syndrome
HYPERBILIRUBINEMIA CAUSES
JAUNDICE
BILE DUCTULE
When bilirubin in the blood exceeds 1 mg/dL (17.1
Bilirubin diglucuronide
µmol/L), hyperbilirubinemia exists. Hyperbilirubine-
mia may be due to the production of more bilirubin
Figure 32-15.
Diagrammatic representation of the
than the normal liver can excrete, or it may result from
three major processes (uptake, conjugation, and secre-
the failure of a damaged liver to excrete bilirubin pro-
tion) involved in the transfer of bilirubin from blood to
duced in normal amounts. In the absence of hepatic
bile. Certain proteins of hepatocytes, such as ligandin (a
damage, obstruction of the excretory ducts of the
family of glutathione S-transferase) and Y protein, bind
liver—by preventing the excretion of bilirubin—will
intracellular bilirubin and may prevent its efflux into the
also cause hyperbilirubinemia. In all these situations,
blood stream. The process affected in a number of con-
bilirubin accumulates in the blood, and when it reaches
ditions causing jaundice is also shown.
a certain concentration (approximately 2-2.5 mg/dL),
282
/
CHAPTER 32
M
E
M
E
M
E
H
H
I
I
H
I
H
H2C
OH
H2C
OH
H2C
OH
N
OH
N
OH
H
N
OH
H
M
M
M
M
M
M
H
H
H
H
H
NH
HN
NH
HN
N
HN
H
H
H
P
E
P
E
P
E
N
N
N
H2C
CH2
H2C
CH2
HC
CH2
P
M
P
M
P
M
Mesobilirubinogen
Stercobilinogen
Stercobilin
(C33H44O6N4)
(L-Urobilinogen)
(L-Urobilin)
Figure 32-16. Structure of some bile pigments.
it diffuses into the tissues, which then become yellow.
soluble, can react directly with the diazo reagent, so
That condition is called jaundice or icterus.
that the “direct bilirubin” of van den Bergh is actually a
In clinical studies of jaundice, measurement of
bilirubin conjugate (bilirubin glucuronide).
bilirubin in the serum is of great value. A method for
Depending on the type of bilirubin present in
quantitatively assaying the bilirubin content of the
plasma—ie, unconjugated or conjugated—hyperbiliru-
serum was first devised by van den Bergh by application
binemia may be classified as retention hyperbiliru-
of Ehrlich’s test for bilirubin in urine. The Ehrlich reac-
binemia, due to overproduction, or regurgitation hy-
tion is based on the coupling of diazotized sulfanilic
perbilirubinemia, due to reflux into the bloodstream
acid (Ehrlich’s diazo reagent) and bilirubin to produce
because of biliary obstruction.
a reddish-purple azo compound. In the original proce-
Because of its hydrophobicity, only unconjugated
dure as described by Ehrlich, methanol was used to
bilirubin can cross the blood-brain barrier into the cen-
provide a solution in which both bilirubin and the
tral nervous system; thus, encephalopathy due to hyper-
diazo regent were soluble. Van den Bergh inadvertently
bilirubinemia (kernicterus) can occur only in connec-
omitted the methanol on an occasion when assay of bile
tion with unconjugated bilirubin, as found in retention
pigment in human bile was being attempted. To his
hyperbilirubinemia. On the other hand, because of its
surprise, normal development of the color occurred “di-
water-solubility, only conjugated bilirubin can appear
rectly.” This form of bilirubin that would react without
in urine. Accordingly, choluric jaundice (choluria is
the addition of methanol was thus termed “direct-
the presence of bile pigments in the urine) occurs only
reacting.” It was then found that this same direct reac-
in regurgitation hyperbilirubinemia, and acholuric
tion would also occur in serum from cases of jaundice
jaundice occurs only in the presence of an excess of un-
due to biliary obstruction. However, it was still neces-
conjugated bilirubin.
sary to add methanol to detect bilirubin in normal
serum or that which was present in excess in serum
Elevated Amounts of Unconjugated
from cases of hemolytic jaundice where no evidence of
Bilirubin in Blood Occur in a Number
obstruction was to be found. To that form of bilirubin
of Conditions
which could be measured only after the addition of
methanol, the term “indirect-reacting” was applied.
A. HEMOLYTIC ANEMIAS
It was subsequently discovered that the indirect
Hemolytic anemias are important causes of unconju-
bilirubin is “free” (unconjugated) bilirubin en route to
gated hyperbilirubinemia, though unconjugated hyper-
the liver from the reticuloendothelial tissues, where the
bilirubinemia is usually only slight (< 4 mg/dL; < 68.4
bilirubin was originally produced by the breakdown of
µmol/L) even in the event of extensive hemolysis be-
heme porphyrins. Since this bilirubin is not water-solu-
cause of the healthy liver’s large capacity for handling
ble, it requires methanol to initiate coupling with the
bilirubin.
diazo reagent. In the liver, the free bilirubin becomes
conjugated with glucuronic acid, and the conjugate,
B. NEONATAL “PHYSIOLOGIC JAUNDICE”
bilirubin glucuronide, can then be excreted into the
This transient condition is the most common cause of
bile. Furthermore, conjugated bilirubin, being water-
unconjugated hyperbilirubinemia. It results from an ac-
PORPHYRINS & BILE PIGMENTS
/
283
celerated hemolysis around the time of birth and an im-
mushroom poisoning. These acquired disorders are due
mature hepatic system for the uptake, conjugation, and
to hepatic parenchymal cell damage, which impairs
secretion of bilirubin. Not only is the bilirubin-UGT
conjugation.
activity reduced, but there probably is reduced synthesis
of the substrate for that enzyme, UDP-glucuronic acid.
Obstruction in the Biliary Tree Is the
Since the increased amount of bilirubin is unconju-
Commonest Cause of Conjugated
gated, it is capable of penetrating the blood-brain bar-
rier when its concentration in plasma exceeds that
Hyperbilirubinemia
which can be tightly bound by albumin
(20-25
A. OBSTRUCTION OF THE BILIARY TREE
mg/dL). This can result in a hyperbilirubinemic toxic
Conjugated hyperbilirubinemia commonly results from
encephalopathy, or kernicterus, which can cause men-
blockage of the hepatic or common bile ducts, most
tal retardation. Because of the recognized inducibility
often due to a gallstone or to cancer of the head of the
of this bilirubin-metabolizing system, phenobarbital
pancreas. Because of the obstruction, bilirubin diglu-
has been administered to jaundiced neonates and is ef-
curonide cannot be excreted. It thus regurgitates into
fective in this disorder. In addition, exposure to blue
the hepatic veins and lymphatics, and conjugated
light (phototherapy) promotes the hepatic excretion of
bilirubin appears in the blood and urine (choluric jaun-
unconjugated bilirubin by converting some of the
dice).
bilirubin to other derivatives such as maleimide frag-
The term cholestatic jaundice is used to include all
ments and geometric isomers that are excreted in the
cases of extrahepatic obstructive jaundice. It also covers
bile.
those cases of jaundice that exhibit conjugated hyper-
C. CRIGLER-NAJJAR SYNDROME, TYPE I;
bilirubinemia due to micro-obstruction of intrahepatic
CONGENITAL NONHEMOLYTIC JAUNDICE
biliary ductules by swollen, damaged hepatocytes (eg, as
Type I Crigler-Najjar syndrome is a rare autosomal re-
may occur in infectious hepatitis).
cessive disorder. It is characterized by severe congenital
jaundice (serum bilirubin usually exceeds 20 mg/dL)
B. DUBIN-JOHNSON SYNDROME
due to mutations in the gene encoding bilirubin-UGT
This benign autosomal recessive disorder consists of
activity in hepatic tissues. The disease is often fatal
conjugated hyperbilirubinemia in childhood or during
within the first 15 months of life. Children with this
adult life. The hyperbilirubinemia is caused by muta-
condition have been treated with phototherapy, result-
tions in the gene encoding MRP-2 (see above), the pro-
ing in some reduction in plasma bilirubin levels. Phe-
tein involved in the secretion of conjugated bilirubin
nobarbital has no effect on the formation of bilirubin
into bile. The centrilobular hepatocytes contain an ab-
glucuronides in patients with type I Crigler-Najjar syn-
normal black pigment that may be derived from epi-
drome. A liver transplant may be curative.
nephrine.
D. CRIGLER-NAJJAR SYNDROME, TYPE II
C. ROTOR SYNDROME
This rare inherited disorder also results from mutations
This is a rare benign condition characterized by chronic
in the gene encoding bilirubin-UGT, but some activity
conjugated hyperbilirubinemia and normal liver histol-
of the enzyme is retained and the condition has a more
ogy. Its precise cause has not been identified, but it is
benign course than type I. Serum bilirubin concentra-
thought to be due to an abnormality in hepatic storage.
tions usually do not exceed 20 mg/dL. Patients with
this condition can respond to treatment with large
doses of phenobarbital.
Some Conjugated Bilirubin Can Bind
Covalently to Albumin
E. GILBERT SYNDROME
Again, this is caused by mutations in the gene encoding
When levels of conjugated bilirubin remain high in
bilirubin-UGT, but approximately
30% of the en-
plasma, a fraction can bind covalently to albumin (delta
zyme’s activity is preserved and the condition is entirely
bilirubin). Because it is bound covalently to albumin,
harmless.
this fraction has a longer half-life in plasma than does
conventional conjugated bilirubin. Thus, it remains ele-
F. TOXIC HYPERBILIRUBINEMIA
vated during the recovery phase of obstructive jaundice
Unconjugated hyperbilirubinemia can result from
after the remainder of the conjugated bilirubin has de-
toxin-induced liver dysfunction such as that caused by
clined to normal levels; this explains why some patients
chloroform, arsphenamines, carbon tetrachloride, ace-
continue to appear jaundiced after conjugated bilirubin
taminophen, hepatitis virus, cirrhosis, and Amanita
levels have returned to normal.
284
/
CHAPTER 32
Table 32-3. Laboratory results in normal patients and patients with three different causes of jaundice.
Condition
Serum Bilirubin
Urine Urobilinogen
Urine Bilirubin
Fecal Urobilinogen
Normal
Direct: 0.1-0.4 mg/dL
0-4 mg/24 h
Absent
40-280 mg/24 h
Indirect: 0.2-0.7 mg/dL
Hemolytic anemia
↑ Indirect
Increased
Absent
Increased
Hepatitis
↑ Direct and indirect
Decreased if micro-
Present if micro-
Decreased
obstruction is
obstruction occurs
present
Obstructive jaundice1
↑ Direct
Absent
Present
Trace to absent
1The commonest causes of obstructive (posthepatic) jaundice are cancer of the head of the pancreas and a gallstone lodged in the com-
mon bile duct. The presence of bilirubin in the urine is sometimes referred to as choluria—therefore, hepatitis and obstruction of the
common bile duct cause choluric jaundice, whereas the jaundice of hemolytic anemia is referred to as acholuric. The laboratory results in
patients with hepatitis are variable, depending on the extent of damage to parenchymal cells and the extent of micro-obstruction to bile
ductules. Serum levels of ALT and AST are usually markedly elevated in hepatitis, whereas serum levels of alkaline phosphatase are ele-
vated in obstructive liver disease.
Urobilinogen & Bilirubin in Urine
which four pyrrole rings are joined by methenyl
Are Clinical Indicators
bridges. The eight side groups (methyl, vinyl, and
propionyl substituents) on the four pyrrole rings of
Normally, there are mere traces of urobilinogen in the
heme are arranged in a specific sequence.
urine. In complete obstruction of the bile duct, no
•
Biosynthesis of the heme ring occurs in mitochondria
urobilinogen is found in the urine, since bilirubin has
and cytosol via eight enzymatic steps. It commences
no access to the intestine, where it can be converted to
with formation of δ-aminolevulinate
(ALA) from
urobilinogen. In this case, the presence of bilirubin
succinyl-CoA and glycine in a reaction catalyzed by
(conjugated) in the urine without urobilinogen suggests
ALA synthase, the regulatory enzyme of the pathway.
obstructive jaundice, either intrahepatic or posthepatic.
•
Genetically determined abnormalities of seven of the
In jaundice secondary to hemolysis, the increased
eight enzymes involved in heme biosynthesis result in
production of bilirubin leads to increased production of
the inherited porphyrias. Red blood cells and liver
urobilinogen, which appears in the urine in large
are the major sites of metabolic expression of the por-
amounts. Bilirubin is not usually found in the urine in
phyrias. Photosensitivity and neurologic problems
hemolytic jaundice
(because unconjugated bilirubin
are common complaints. Intake of certain com-
does not pass into the urine), so that the combination
pounds (such as lead) can cause acquired porphyrias.
of increased urobilinogen and absence of bilirubin is
Increased amounts of porphyrins or their precursors
suggestive of hemolytic jaundice. Increased blood de-
can be detected in blood and urine, facilitating diag-
struction from any cause brings about an increase in
nosis.
urine urobilinogen.
Table 32-3 summarizes laboratory results obtained
•
Catabolism of the heme ring is initiated by the en-
on patients with three different causes of jaundice—he-
zyme heme oxygenase, producing a linear tetrapyr-
molytic anemia (a prehepatic cause), hepatitis (a hepatic
role.
cause), and obstruction of the common bile duct (a
•
Biliverdin is an early product of catabolism and on
posthepatic cause). Laboratory tests on blood (evalua-
reduction yields bilirubin. The latter is transported
tion of the possibility of a hemolytic anemia and mea-
by albumin from peripheral tissues to the liver, where
surement of prothrombin time) and on serum (eg, elec-
it is taken up by hepatocytes. The iron of heme and
trophoresis of proteins; activities of the enzymes ALT,
the amino acids of globin are conserved and reuti-
AST, and alkaline phosphatase) are also important in
lized.
helping to distinguish between prehepatic, hepatic, and
•
In the liver, bilirubin is made water-soluble by conju-
posthepatic causes of jaundice.
gation with two molecules of glucuronic acid and is
secreted into the bile. The action of bacterial en-
SUMMARY
zymes in the gut produces urobilinogen and urobilin,
which are excreted in the feces and urine.
• Hemoproteins, such as hemoglobin and the cy-
tochromes, contain heme. Heme is an iron-por-
•
Jaundice is due to elevation of the level of bilirubin
phyrin compound
(Fe2+-protoporphyrin IX) in
in the blood. The causes of jaundice can be classified
PORPHYRINS & BILE PIGMENTS
/
285
as prehepatic (eg, hemolytic anemias), hepatic (eg,
Berk PD, Wolkoff AW: Bilirubin metabolism and the hyperbiliru-
binemias. In: Harrison’s Principles of Internal Medicine, 15th
hepatitis), and posthepatic
(eg, obstruction of the
ed. Braunwald E et al (editors). McGraw-Hill, 2001.
common bile duct). Measurements of plasma total
Chowdhury JR et al: Hereditary jaundice and disorders of bilirubin
and nonconjugated bilirubin, of urinary urobilino-
metabolism. In: The Metabolic and Molecular Bases of Inher-
gen and bilirubin, and of certain serum enzymes as
ited Disease, 8th ed. Scriver CR et al (editors). McGraw-Hill,
well as inspection of stool samples help distinguish
2001.
between these causes.
Desnick RJ: The porphyrias. In: Harrison’s Principles of Internal
Medicine, 15th ed. Braunwald E et al (editors). McGraw-Hill,
2001.
REFERENCES
Elder GH: Haem synthesis and the porphyrias. In: Scientific Foun-
Anderson KE et al: Disorders of heme biosynthesis: X-linked sid-
dations of Biochemistry in Clinical Practice, 2nd ed. Williams
eroblastic anemia and the porphyrias. In: The Metabolic and
DL, Marks V (editors). Butterworth-Heinemann, 1994.
Molecular Bases of Inherited Disease, 8th ed. Scriver CR et al
(editors). McGraw-Hill, 2001.
SECTION IV
Structure, Function, & Replication
of Informational Macromolecules
Nucleotides
33
Victor W. Rodwell, PhD
BIOMEDICAL IMPORTANCE
H
H
6
7
4
C
5
N
C
5
Nucleotides—the monomer units or building blocks of
1
3
N
C
8
N
CH
nucleic acids—serve multiple additional functions. They
CH
2C
C
HC
CH
form a part of many coenzymes and serve as donors of
H
N
4
N9
2
N
6
3
H
1
phosphoryl groups (eg, ATP or GTP), of sugars (eg,
UDP- or GDP-sugars), or of lipid (eg, CDP-acylglyc-
Purine
Pyrimidine
erol). Regulatory nucleotides include the second mes-
Figure 33-1. Purine and pyrimidine. The atoms are
sengers cAMP and cGMP, the control by ADP of ox-
numbered according to the international system.
idative phosphorylation, and allosteric regulation of
enzyme activity by ATP, AMP, and CTP. Synthetic
purine and pyrimidine analogs that contain halogens,
thiols, or additional nitrogen are employed for chemo-
Nucleosides & Nucleotides
therapy of cancer and AIDS and as suppressors of the
immune response during organ transplantation.
Nucleosides are derivatives of purines and pyrimidines
that have a sugar linked to a ring nitrogen. Numerals
PURINES, PYRIMIDINES, NUCLEOSIDES,
with a prime (eg, 2′ or 3′) distinguish atoms of the
sugar from those of the heterocyclic base. The sugar in
& NUCLEOTIDES
ribonucleosides is D-ribose, and in deoxyribonucleo-
Purines and pyrimidines are nitrogen-containing hete-
sides it is 2-deoxy-D-ribose. The sugar is linked to the
rocycles, cyclic compounds whose rings contain both
heterocyclic base via a
-N-glycosidic bond, almost al-
carbon and other elements (hetero atoms). Note that
ways to N-1 of a pyrimidine or to N-9 of a purine (Fig-
the smaller pyrimidine has the longer name and the
ure 33-3).
larger purine the shorter name and that their six-atom
rings are numbered in opposite directions
(Figure
33-1). The planar character of purines and pyrimidines
facilitates their close association, or “stacking,” which
NH
2
NH
O
OH
stabilizes double-stranded DNA (Chapter 36). The oxo
and amino groups of purines and pyrimidines exhibit
keto-enol and amine-imine tautomerism (Figure 33-2),
but physiologic conditions strongly favor the amino
Figure 33-2. Tautomerism of the oxo and amino
and oxo forms.
functional groups of purines and pyrimidines.
286
NUCLEOTIDES
/
287
NH2
O
NH2
O
N
N
N
HN
N
HN
9
1
9
1
O
O
N
N
H2N
N
N
N
N
HO
HO
HO
HO
O
O
O
O
OH
OH
OH
OH
OH
OH
OH
OH
Adenosine
Cytidine
Guanosine
Uridine
Figure 33-3. Ribonucleosides, drawn as the syn conformers.
Mononucleotides are nucleosides with a phosphoryl
cleotides. Both therefore exist as syn or anti conformers
group esterified to a hydroxyl group of the sugar. The
(Figure 33-5). While both conformers occur in nature,
3′- and 5′-nucleotides are nucleosides with a phospho-
anti conformers predominate. Table
33-1 lists the
ryl group on the 3′- or 5′-hydroxyl group of the sugar,
major purines and pyrimidines and their nucleoside
respectively. Since most nucleotides are 5′-, the prefix
and nucleotide derivatives. Single-letter abbreviations
“5′-” is usually omitted when naming them. UMP and
are used to identify adenine (A), guanine (G), cytosine
dAMP thus represent nucleotides with a phosphoryl
(C), thymine (T), and uracil (U), whether free or pre-
group on C-5 of the pentose. Additional phosphoryl
sent in nucleosides or nucleotides. The prefix
“d”
groups linked by acid anhydride bonds to the phos-
(deoxy) indicates that the sugar is
2′-deoxy-D-ribose
phoryl group of a mononucleotide form nucleoside
(eg, dGTP) (Figure 33-6).
diphosphates and triphosphates (Figure 33-4).
Steric hindrance by the base restricts rotation about
Nucleic Acids Also Contain
the β-N-glycosidic bond of nucleosides and nu-
Additional Bases
Small quantities of additional purines and pyrimidines
NH
occur in DNA and RNAs. Examples include 5-methyl-
2
cytosine of bacterial and human DNA,
5-hydroxy-
N
N
Adenine
methylcytosine of bacterial and viral nucleic acids, and
mono- and di-N-methylated adenine and guanine of
N
N
CH2
O
NH
NH2
2
O
Ribose
O-
O
O-
N
N
N
N
HO
P
O
P O
P
HO OH
O
O-
O
N
N
N
N
HO
HO
Adenosine 5′-monophosphate (AMP)
O
O
Adenosine 5′-diphosphate (ADP)
Syn
Anti
OH
OH
OH
OH
Adenosine 5′-triphosphate (ATP)
Figure 33-5.
The syn and anti conformers of adeno-
Figure 33-4. ATP, its diphosphate, and its
sine differ with respect to orientation about the N-gly-
monophosphate.
cosidic bond.
288
/
CHAPTER 33
Table 33-1. Bases, nucleosides, and nucleotides.
Nucleoside
Base
Base
X = Ribose or
Nucleotide, Where
Formula
X = H
Deoxyribose
X = Ribose Phosphate
NH2
N
N
Adenine
Adenosine
Adenosine monophosphate
A
A
AMP
N N
X
O
H
N
N
Guanine
Guanosine
Guanosine monophosphate
G
G
GMP
H2N
N
N
X
NH2
N
Cytosine
Cytidine
Cytidine monophosphate
C
C
CMP
O
N
X
O
H
N
Uracil
Uridine
Uridine monophosphate
U
U
UMP
O
N
X
O
H
CH3
N
Thymine
Thymidine
Thymidine monophosphate
T
T
TMP
O
N
dX
NH2
NH2
O
O
N
N
CH3
N
N
HN
HN
N
N
O
O
N
N
N
N
O
O
O
O
O
O
O
O
O
O
O
O
P
P
P
P
-O
O-
-O
O-
-O
O-
-O O-
OH
OH
OH
H
OH
OH
OH
H
AMP
dAMP
UMP
TMP
Figure 33-6. AMP, dAMP, UMP, and TMP.
NUCLEOTIDES
/
289
NH2
NH2
NH
2
O
CH3
CH2OH
N
N
N
N
N
HN
N
H2N
N
O
O
N
N
N
N
H
H
O
CH
2
O
CH2
5-Methylcytosine
5-Hydroxymethylcytosine
O
O
-
O
P
O
-O
P
O
H3C
CH3
O
O
N
O
CH3
OH
OH
N
N
Figure 33-9. cAMP, 3′,5′-cyclic AMP, and cGMP.
N
HN
7
N
H2N
N
N
N
H
Nucleotides Serve Diverse
Dimethylaminoadenine
7-Methylguanine
Physiologic Functions
Figure 33-7. Four uncommon naturally occurring
Nucleotides participate in reactions that fulfill physio-
pyrimidines and purines.
logic functions as diverse as protein synthesis, nucleic
acid synthesis, regulatory cascades, and signal transduc-
tion pathways.
mammalian messenger RNAs (Figure 33-7). These
atypical bases function in oligonucleotide recognition
Nucleoside Triphosphates Have High
and in regulating the half-lives of RNAs. Free nu-
Group Transfer Potential
cleotides include hypoxanthine, xanthine, and uric acid
(see Figure 34-8), intermediates in the catabolism of
Acid anhydrides, unlike phosphate esters, have high
adenine and guanine. Methylated heterocyclic bases of
group transfer potential. ∆0′ for the hydrolysis of each
plants include the xanthine derivatives caffeine of cof-
of the terminal phosphates of nucleoside triphosphates
fee, theophylline of tea, and theobromine of cocoa (Fig-
is about −7 kcal/mol (−30 kJ/mol). The high group
ure 33-8).
transfer potential of purine and pyrimidine nucleoside
Posttranslational modification of preformed polynu-
triphosphates permits them to function as group trans-
cleotides can generate additional bases such as
fer reagents. Cleavage of an acid anhydride bond typi-
pseudouridine, in which D-ribose is linked to C-5 of
cally is coupled with a highly endergonic process such
uracil by a carbon-to-carbon bond rather than by a
as covalent bond synthesis—eg, polymerization of nu-
β-N-glycosidic bond. The nucleotide pseudouridylic
cleoside triphosphates to form a nucleic acid.
acid Ψ arises by rearrangement of UMP of a preformed
In addition to their roles as precursors of nucleic
tRNA. Similarly, methylation by S-adenosylmethionine
acids, ATP, GTP, UTP, CTP, and their derivatives
of a UMP of preformed tRNA forms TMP (thymidine
each serve unique physiologic functions discussed in
monophosphate), which contains ribose rather than de-
other chapters. Selected examples include the role of
oxyribose.
ATP as the principal biologic transducer of free energy;
the second messenger cAMP (Figure 33-9); adenosine
3′-phosphate-5′-phosphosulfate
(Figure
33-10), the
O
CH
3
sulfate donor for sulfated proteoglycans (Chapter 48)
H
3C
N
and for sulfate conjugates of drugs; and the methyl
N
group donor S-adenosylmethionine
(Figure
33-11).
N
O
N
CH3
P
Figure 33-8. Caffeine, a trimethylxanthine. The di-
2-
Adenine Ribose
P
O SO3
methylxanthines theobromine and theophylline are
similar but lack the methyl group at N-1 and at N-7, re-
Figure 33-10. Adenosine 3′-phosphate-5′-phos-
spectively.
phosulfate.
290
/
CHAPTER 33
NH2
Table 33-2. Many coenzymes and related
N
compounds are derivatives of adenosine
N
monophosphate.
N
NH2
N
COO-
CH3
CH2
N
N
Adenine
O
CH
CH2
CH2
S
+
N N
O
NH3+
R
O
P O
CH2
HO OH
O-
n
O
Adenosine
Methionine
R'' O OR'
Figure 33-11. S-Adenosylmethionine.
D-Ribose
Coenzyme
R
R
R
n
GTP serves as an allosteric regulator and as an energy
Active methionine
Methionine*
H
H
0
source for protein synthesis, and cGMP (Figure 33-9)
Amino acid adenylates
Amino acid
H
H
1
serves as a second messenger in response to nitric oxide
Active sulfate
SO32−
H
PO32−
1
(NO) during relaxation of smooth muscle (Chapter
3′,5′-Cyclic AMP
H
PO32−
1
†
48). UDP-sugar derivatives participate in sugar epimer-
NAD*
H
H
2
†
izations and in biosynthesis of glycogen, glucosyl disac-
NADP*
PO32−
H
2
†
charides, and the oligosaccharides of glycoproteins and
FAD
H
H
2
†
proteoglycans (Chapters 47 and 48). UDP-glucuronic
CoASH
H
PO32−
2
acid forms the urinary glucuronide conjugates of biliru-
*Replaces phosphoryl group.
bin (Chapter 32) and of drugs such as aspirin. CTP
†R is a B vitamin derivative.
participates in biosynthesis of phosphoglycerides,
sphingomyelin, and other substituted sphingosines
(Chapter 24). Finally, many coenzymes incorporate nu-
cleotides as well as structures similar to purine and
nucleic acids thus often is expressed in terms of “ab-
pyrimidine nucleotides (see Table 33-2).
sorbance at 260 nm.”
Nucleotides Are Polyfunctional Acids
SYNTHETIC NUCLEOTIDE ANALOGS
ARE USED IN CHEMOTHERAPY
Nucleosides or free purine or pyrimidine bases are un-
charged at physiologic pH. By contrast, the primary
Synthetic analogs of purines, pyrimidines, nucleosides,
phosphoryl groups (pK about 1.0) and secondary phos-
and nucleotides altered in either the heterocyclic ring or
phoryl groups (pK about 6.2) of nucleotides ensure that
the sugar moiety have numerous applications in clinical
they bear a negative charge at physiologic pH. Nu-
medicine. Their toxic effects reflect either inhibition of
cleotides can, however, act as proton donors or accep-
enzymes essential for nucleic acid synthesis or their in-
tors at pH values two or more units above or below
corporation into nucleic acids with resulting disruption
neutrality.
of base-pairing. Oncologists employ
5-fluoro- or
5-
iodouracil, 3-deoxyuridine, 6-thioguanine and 6-mer-
captopurine, 5- or 6-azauridine, 5- or 6-azacytidine,
Nucleotides Absorb Ultraviolet Light
and 8-azaguanine (Figure 33-12), which are incorpo-
The conjugated double bonds of purine and pyrimidine
rated into DNA prior to cell division. The purine ana-
derivatives absorb ultraviolet light. The mutagenic ef-
log allopurinol, used in treatment of hyperuricemia and
fect of ultraviolet light results from its absorption by
gout, inhibits purine biosynthesis and xanthine oxidase
nucleotides in DNA with accompanying chemical
activity. Cytarabine is used in chemotherapy of cancer.
changes. While spectra are pH-dependent, at pH 7.0 all
Finally, azathioprine, which is catabolized to 6-mercap-
the common nucleotides absorb light at a wavelength
topurine, is employed during organ transplantation to
close to 260 nm. The concentration of nucleotides and
suppress immunologic rejection.
NUCLEOTIDES
/
291
O
O
I
HN
5
HN
6N
O
O
N
N
HO
HO
O
O
O
N
F
HN
O
HN
5
8N
H2N
N
2′
O
N
N
H
H
HO OH
HO H
5-Iodo-2′-deoxyuridine
5-Fluorouracil
6-Azauridine
8-Azaguanine
SH
SH
OH
6
N
6
N
6
N
N
N
1
5
N
2
4
3
N
H2N
N
N
N
N
N
H
H
H
6-Mercaptopurine
6-Thioguanine
Alloburinol
Figure 33-12. Selected synthetic pyrimidine and purine analogs.
Nonhydrolyzable Nucleoside
(absent from DNA) functions as a nucleophile during
Triphosphate Analogs Serve as
hydrolysis of the 3′,5′-phosphodiester bond.
Research Tools
Synthetic nonhydrolyzable analogs of nucleoside
Polynucleotides Are Directional
triphosphates (Figure 33-13) allow investigators to dis-
Macromolecules
tinguish the effects of nucleotides due to phosphoryl
Phosphodiester bonds link the 3′- and 5′-carbons of ad-
transfer from effects mediated by occupancy of al-
jacent monomers. Each end of a nucleotide polymer
losteric nucleotide-binding sites on regulated enzymes.
thus is distinct. We therefore refer to the “5′- end” or
the “3′- end” of polynucleotides, the 5′- end being the
POLYNUCLEOTIDES
one with a free or phosphorylated 5′-hydroxyl.
The 5′-phosphoryl group of a mononucleotide can es-
terify a second OH group, forming a phosphodi-
Polynucleotides Have Primary Structure
ester. Most commonly, this second OH group is the
3′-OH of the pentose of a second nucleotide. This
The base sequence or primary structure of a polynu-
forms a dinucleotide in which the pentose moieties are
cleotide can be represented as shown below. The phos-
linked by a 3′ → 5′ phosphodiester bond to form the
phodiester bond is represented by P or p, bases by a sin-
“backbone” of RNA and DNA.
gle letter, and pentoses by a vertical line.
While formation of a dinucleotide may be repre-
A
T
C
A
sented as the elimination of water between two
monomers, the reaction in fact strongly favors phos-
phodiester hydrolysis. Phosphodiesterases rapidly cat-
alyze the hydrolysis of phosphodiester bonds whose
spontaneous hydrolysis is an extremely slow process.
Consequently, DNA persists for considerable periods
P
P
P
P
OH
and has been detected even in fossils. RNAs are far less
stable than DNA since the 2′-hydroxyl group of RNA
292
/
CHAPTER 33
O
O
O
SUMMARY
Pu /Py R O
P
O
P
O
P
O-
•
Under physiologic conditions, the amino and oxo
tautomers of purines, pyrimidines, and their deriva-
O-
O-
O-
tives predominate.
Parent nucleoside triphosphate
•
Nucleic acids contain, in addition to A, G, C, T, and
O
O
O
U, traces of 5-methylcytosine, 5-hydroxymethylcyto-
sine, pseudouridine (Ψ), or N-methylated bases.
Pu /Py R O
P
O
P
CH2
P
O-
•
Most nucleosides contain D-ribose or
2-deoxy-D-
O-
O-
O-
ribose linked to N-1 of a pyrimidine or to N-9 of a
β,γ-Methylene derivative
purine by a β-glycosidic bond whose syn conformers
predominate.
O
O
O
H
•
A primed numeral locates the position of the phos-
Pu /Py R O
P
O
P
N
P
O-
phate on the sugars of mononucleotides
(eg,
3′-
GMP, 5′-dCMP). Additional phosphoryl groups
O-
O-
O-
linked to the first by acid anhydride bonds form nu-
β,γ-Imino derivative
cleoside diphosphates and triphosphates.
Figure 33-13. Synthetic derivatives of nucleoside
•
Nucleoside triphosphates have high group transfer
triphosphates incapable of undergoing hydrolytic re-
potential and participate in covalent bond syntheses.
lease of the terminal phosphoryl group. (Pu/Py, a
The cyclic phosphodiesters cAMP and cGMP func-
purine or pyrimidine base; R, ribose or deoxyribose.)
tion as intracellular second messengers.
Shown are the parent (hydrolyzable) nucleoside
•
Mononucleotides linked by 3′ → 5′-phosphodiester
triphosphate (top) and the unhydrolyzable β-methyl-
bonds form polynucleotides, directional macromole-
ene (center) and γ-imino derivatives (bottom).
cules with distinct 3′- and 5′- ends. For pTpGpTp or
TGCATCA, the 5′- end is at the left, and all phos-
phodiester bonds are 3′ → 5′.
Where all the phosphodiester bonds are 5′ → 3′, a
•
Synthetic analogs of purine and pyrimidine bases and
more compact notation is possible:
their derivatives serve as anticancer drugs either by
inhibiting an enzyme of nucleotide biosynthesis or
pGpGpApTpCpA
by being incorporated into DNA or RNA.
This representation indicates that the 5′-hydroxyl—
REFERENCES
but not the 3′-hydroxyl—is phosphorylated.
The most compact representation shows only the
Adams RLP, Knowler JT, Leader DP: The Biochemistry of the Nu-
base sequence with the 5′- end on the left and the 3′-
cleic Acids, 11th ed. Chapman & Hall, 1992.
end on the right. The phosphoryl groups are assumed
Blackburn GM, Gait MJ: Nucleic Acids in Chemistry & Biology. IRL
but not shown:
Press, 1990.
Bugg CE, Carson WM, Montgomery JA: Drugs by design. Sci Am
GGATCA
1992;269(6):92.
Metabolism of Purine &
34
Pyrimidine Nucleotides
Victor W. Rodwell, PhD
BIOMEDICAL IMPORTANCE
(synthesis de novo), (2) phosphoribosylation of purines,
and (3) phosphorylation of purine nucleosides.
The biosynthesis of purines and pyrimidines is strin-
gently regulated and coordinated by feedback mecha-
nisms that ensure their production in quantities and at
INOSINE MONOPHOSPHATE (IMP)
times appropriate to varying physiologic demand. Ge-
IS SYNTHESIZED FROM AMPHIBOLIC
netic diseases of purine metabolism include gout,
Lesch-Nyhan syndrome, adenosine deaminase defi-
INTERMEDIATES
ciency, and purine nucleoside phosphorylase deficiency.
Figure 34-2 illustrates the intermediates and reactions
By contrast, apart from the orotic acidurias, there are
for conversion of α-D-ribose 5-phosphate to inosine
few clinically significant disorders of pyrimidine catab-
monophosphate (IMP). Separate branches then lead to
olism.
AMP and GMP (Figure 34-3). Subsequent phosphoryl
transfer from ATP converts AMP and GMP to ADP
and GDP. Conversion of GDP to GTP involves a sec-
PURINES & PYRIMIDINES ARE
ond phosphoryl transfer from ATP, whereas conversion
DIETARILY NONESSENTIAL
of ADP to ATP is achieved primarily by oxidative
Human tissues can synthesize purines and pyrimidines
phosphorylation (see Chapter 12).
from amphibolic intermediates. Ingested nucleic acids
and nucleotides, which therefore are dietarily nonessen-
tial, are degraded in the intestinal tract to mononu-
Multifunctional Catalysts Participate in
cleotides, which may be absorbed or converted to
Purine Nucleotide Biosynthesis
purine and pyrimidine bases. The purine bases are then
In prokaryotes, each reaction of Figure 34-2 is cat-
oxidized to uric acid, which may be absorbed and ex-
alyzed by a different polypeptide. By contrast, in eu-
creted in the urine. While little or no dietary purine or
karyotes, the enzymes are polypeptides with multiple
pyrimidine is incorporated into tissue nucleic acids, in-
catalytic activities whose adjacent catalytic sites facili-
jected compounds are incorporated. The incorporation
tate channeling of intermediates between sites. Three
of injected [3H]thymidine into newly synthesized DNA
distinct multifunctional enzymes catalyze reactions 3,
thus is used to measure the rate of DNA synthesis.
4, and 6, reactions 7 and 8, and reactions 10 and 11 of
Figure 34-2.
BIOSYNTHESIS OF PURINE NUCLEOTIDES
Purine and pyrimidine nucleotides are synthesized in
Antifolate Drugs or Glutamine Analogs
vivo at rates consistent with physiologic need. Intracel-
Block Purine Nucleotide Biosynthesis
lular mechanisms sense and regulate the pool sizes of
nucleotide triphosphates
(NTPs), which rise during
The carbons added in reactions 4 and 5 of Figure 34-2
growth or tissue regeneration when cells are rapidly di-
are contributed by derivatives of tetrahydrofolate.
viding. Early investigations of nucleotide biosynthesis
Purine deficiency states, which are rare in humans, gen-
employed birds, and later ones used Escherichia coli.
erally reflect a deficiency of folic acid. Compounds that
Isotopic precursors fed to pigeons established the source
inhibit formation of tetrahydrofolates and therefore
of each atom of a purine base (Figure 34-1) and initi-
block purine synthesis have been used in cancer
ated study of the intermediates of purine biosynthesis.
chemotherapy. Inhibitory compounds and the reactions
Three processes contribute to purine nucleotide
they inhibit include azaserine (reaction 5, Figure 34-2),
biosynthesis. These are, in order of decreasing impor-
diazanorleucine (reaction 2), 6-mercaptopurine (reac-
tance:
(1) synthesis from amphibolic intermediates
tions 13 and 14), and mycophenolic acid (reaction 14).
293
294
/
CHAPTER 34
Respiratory CO2
and therefore utilize exogenous purines to form nu-
Glycine
cleotides.
Aspartate
C
N
AMP & GMP Feedback-Regulate PRPP
6
7
N1
5C
Glutamyl Amidotransferase
8
C
C2
4
C
9
N5,N10 -Methenyl-
Since biosynthesis of IMP consumes glycine, gluta-
3
N
tetrahydrofolate
N10 -Formyl-
N
mine, tetrahydrofolate derivatives, aspartate, and ATP,
H
tetrahydro-
it is advantageous to regulate purine biosynthesis. The
folate
major determinant of the rate of de novo purine nu-
cleotide biosynthesis is the concentration of PRPP,
whose pool size depends on its rates of synthesis, uti-
Amide nitrogen of glutamine
lization, and degradation. The rate of PRPP synthesis
depends on the availability of ribose 5-phosphate and
Figure 34-1. Sources of the nitrogen and carbon
on the activity of PRPP synthase, an enzyme sensitive
atoms of the purine ring. Atoms 4, 5, and 7 (shaded) de-
to feedback inhibition by AMP, ADP, GMP, and
rive from glycine.
GDP.
AMP & GMP Feedback-Regulate
“SALVAGE REACTIONS” CONVERT
Their Formation From IMP
PURINES & THEIR NUCLEOSIDES TO
Two mechanisms regulate conversion of IMP to GMP
MONONUCLEOTIDES
and AMP. AMP and GMP feedback-inhibit adenylo-
Conversion of purines, their ribonucleosides, and their
succinate synthase and IMP dehydrogenase (reactions
deoxyribonucleosides to mononucleotides involves so-
12 and 14, Figure 34-3), respectively. Furthermore,
called “salvage reactions” that require far less energy
conversion of IMP to adenylosuccinate en route to
than de novo synthesis. The more important mecha-
AMP requires GTP, and conversion of xanthinylate
nism involves phosphoribosylation by PRPP (structure
(XMP) to GMP requires ATP. This cross-regulation
II, Figure 34-2) of a free purine (Pu) to form a purine
between the pathways of IMP metabolism thus serves
5′-mononucleotide (Pu-RP).
to decrease synthesis of one purine nucleotide when
there is a deficiency of the other nucleotide. AMP and
GMP also inhibit hypoxanthine-guanine phosphoribo-
Pu+PR−PP→PRP+PP
i
syltransferase, which converts hypoxanthine and gua-
nine to IMP and GMP (Figure 34-4), and GMP feed-
back-inhibits PRPP glutamyl amidotransferase (reaction
Two phosphoribosyl transferases then convert adenine
2, Figure 34-2).
to AMP and hypoxanthine and guanine to IMP or
GMP (Figure 34-4). A second salvage mechanism in-
volves phosphoryl transfer from ATP to a purine ri-
REDUCTION OF RIBONUCLEOSIDE
bonucleoside (PuR):
DIPHOSPHATES FORMS
DEOXYRIBONUCLEOSIDE
PuR + ATP → PuR − P + ADP
DIPHOSPHATES
Reduction of the 2′-hydroxyl of purine and pyrimidine
Adenosine kinase catalyzes phosphorylation of adeno-
ribonucleotides, catalyzed by the ribonucleotide re-
sine and deoxyadenosine to AMP and dAMP, and de-
ductase complex (Figure 34-5), forms deoxyribonu-
oxycytidine kinase phosphorylates deoxycytidine and
cleoside diphosphates (dNDPs). The enzyme complex
2′-deoxyguanosine to dCMP and dGMP.
is active only when cells are actively synthesizing DNA.
Liver, the major site of purine nucleotide biosynthe-
Reduction requires thioredoxin, thioredoxin reductase,
sis, provides purines and purine nucleosides for salvage
and NADPH. The immediate reductant, reduced
and utilization by tissues incapable of their biosynthe-
thioredoxin, is produced by NADPH:thioredoxin re-
sis. For example, human brain has a low level of PRPP
ductase
(Figure
34-5). Reduction of ribonucleoside
amidotransferase (reaction 2, Figure 34-2) and hence
diphosphates (NDPs) to deoxyribonucleoside diphos-
depends in part on exogenous purines. Erythrocytes
phates (dNDPs) is subject to complex regulatory con-
and polymorphonuclear leukocytes cannot synthesize
trols that achieve balanced production of deoxyribonu-
5-phosphoribosylamine
(structure III, Figure
34-2)
cleotides for synthesis of DNA (Figure 34-6).
296
/
CHAPTER 34
H
H
-
-
-
OOC C C COO
OOC
C
C COO-
H2
H2
H
H
O
+
-
-
NH
H2O
NH
OOC
C
C COO
NH2
3
N
N
N
HN
12
N
13
N
2+
GTP, Mg
N
N
N
N
ADENYLOSUCCINASE
N
N
ADENYLOSUCCINATE
R-5- P
SYNTHASE
R-5-
P
R-5- P
Inosine monophosphate
Adenylosuccinate
Adenosine monophosphate
(IMP)
(AMPS)
(AMP)
+
NAD
H2O
14
IMP DEHYDROGENASE
NADH
+ H
+
O
O
Glutamine
Glutamate
N
N
HN
15
HN
ATP
O
N
N
H2 N
N
N
H
TRANSAMIDINASE
R-5-
P
R-5-
P
Xanthosine monophosphate
Guanosine monophosphate
(XMP)
(GMP)
Figure 34-3. Conversion of IMP to AMP and GMP.
BIOSYNTHESIS OF PYRIMIDINE
THE DEOXYRIBONUCLEOSIDES OF
NUCLEOTIDES
URACIL & CYTOSINE ARE SALVAGED
Figure 34-7 summarizes the roles of the intermediates
While mammalian cells reutilize few free pyrimidines,
and enzymes of pyrimidine nucleotide biosynthesis.
“salvage reactions” convert the ribonucleosides uridine
The catalyst for the initial reaction is cytosolic carbamoyl
and cytidine and the deoxyribonucleosides thymidine
phosphate synthase II, a different enzyme from the mi-
and deoxycytidine to their respective nucleotides. ATP-
tochondrial carbamoyl phosphate synthase I of urea syn-
dependent phosphoryltransferases (kinases) catalyze the
thesis (Figure 29-9). Compartmentation thus provides
phosphorylation of the nucleoside diphosphates 2′-de-
two independent pools of carbamoyl phosphate. PRPP,
oxycytidine, 2′-deoxyguanosine, and 2′-deoxyadenosine
an early participant in purine nucleotide synthesis (Fig-
to their corresponding nucleoside triphosphates. In ad-
ure 34-2), is a much later participant in pyrimidine
dition, orotate phosphoribosyltransferase
(reaction
5,
biosynthesis.
Figure 34-7), an enzyme of pyrimidine nucleotide syn-
thesis, salvages orotic acid by converting it to orotidine
Multifunctional Proteins
monophosphate (OMP).
Catalyze the Early Reactions
of Pyrimidine Biosynthesis
Methotrexate Blocks Reduction
of Dihydrofolate
Five of the first six enzyme activities of pyrimidine
biosynthesis reside on multifunctional polypeptides.
Reaction 12 of Figure 34-7 is the only reaction of pyrimi-
One such polypeptide catalyzes the first three reactions
dine nucleotide biosynthesis that requires a tetrahydrofo-
of Figure 34-2 and ensures efficient channeling of car-
late derivative. The methylene group of N5,N10-methyl-
bamoyl phosphate to pyrimidine biosynthesis. A second
ene-tetrahydrofolate is reduced to the methyl group that
bifunctional enzyme catalyzes reactions 5 and 6.
is transferred, and tetrahydrofolate is oxidized to dihydro-
METABOLISM OF PURINE & PYRIMIDINE NUCLEOTIDES
/
297
NH2
NH2
RIBONUCLEOTIDE
PRPP
PPi
REDUCTASE
N
N
N
N
Ribonucleoside
2 ′-Deoxyribonucleoside
diphosphate
diphosphate
N
N
N
N
H
P
O
H
2C
Adenine
O
Reduced
Oxidized
ADENINE
PHOSPHORIBOSYL
thioredoxin
thioredoxin
TRANSFERASE
H
H
THIOREDOXIN
H
H
REDUCTASE
OH
OH
AMP
NADP+
NADPH + H+
Figure 34-5. Reduction of ribonucleoside diphos-
O
O
PRPP
PPi
phates to 2′-deoxyribonucleoside diphosphates.
N
N
HN
HN
N
N
N
N
a nucleotide in which the ribosyl phosphate is attached
H
P
O
H
to N-1 of the pyrimidine ring. The anticancer drug
Hypoxanthine
2C
5-fluorouracil (Figure 33-12) is also phosphoribosy-
O
lated by orotate phosphoribosyl transferase.
H
H
H
H
HYPOXANTHINE-GUANINE
PHOSPHORIBOSYLTRANSFERASE
OH
OH
REGULATION OF PYRIMIDINE
IMP
NUCLEOTIDE BIOSYNTHESIS
O
O
Gene Expression & Enzyme Activity
N
N
Both Are Regulated
HN
HN
The activities of the first and second enzymes of pyrim-
H2N
N
H2N
N
N
N
H
idine nucleotide biosynthesis are controlled by allosteric
Guanine
PRPP
PPi
P
O
H
2C
CDP
2′dCDP
2′dCTP
O
-
-
-
+
H H
H
H
ATP
OH OH
GMP
+
UDP
2′dUDP
2′dTTP
Figure 34-4. Phosphoribosylation of adenine, hy-
poxanthine, and guanine to form AMP, IMP, and GMP,
-
-
-
respectively.
+
folate. For further pyrimidine synthesis to occur, dihydro-
folate must be reduced back to tetrahydrofolate, a reac-
GDP
2′dGDP
2′dGTP
tion catalyzed by dihydrofolate reductase. Dividing cells,
–
which must generate TMP and dihydrofolate, thus are es-
pecially sensitive to inhibitors of dihydrofolate reductase
+
such as the anticancer drug methotrexate.
ADP
2′dADP
2′dATP
Certain Pyrimidine Analogs Are
Figure 34-6. Regulation of the reduction of purine
Substrates for Enzymes of Pyrimidine
and pyrimidine ribonucleotides to their respective
Nucleotide Biosynthesis
2′-deoxyribonucleotides. Solid lines represent chemical
Orotate phosphoribosyltransferase (reaction 5, Figure
flow. Broken lines show negative ( - ) or positive ( + )
34-7) converts the drug allopurinol (Figure 33-12) to
feedback regulation.
298
/
CHAPTER 34
CO2 + Glutamine + ATP
CARBAMOYL
PHOSPHATE
1
SYNTHASE II
O
ASPARTATE
O
O
–
4
TRANSCAR-
-
DIHYDRO-
+
O
C
O
C
C
H3 N3
BAMOYLASE
OROTASE
5
CH
4
CH
CH
+
2
H2 N
3
5
2
HN
2
2
C
H
6C
2
6
O
1
2
C
1
CH
3
C
CH
O
+
H3N
N
P
COO-
O
N
O
COO-
COO-
H
Pi
H
H2O
Carbamoyl
Aspartic
Carbamoyl
Dihydroorotic
phosphate
acid
aspartic acid
acid (DHOA)
NAD+
(CAP)
(CAA)
DIHYDROOROTATE
DEHYDROGENASE
4
NADH + H+
O
O
O
CO
2
PPi
PRPP
4
6
5
HN
3
5
HN
HN
2
6
1
O
N
OROTIDYLIC ACID
O
N
COO-
OROTATE
O
N
COO-
DECARBOXYLASE
PHOSPHORIBOSYL-
H
R-5-
P
R-5- P
TRANSFERASE
Orotic acid
UMP
OMP
(OA)
ATP
7
NADPH + H+ NADP+
ADP
10
UDP
dUDP (deoxyuridine diphosphate)
H2O
ATP
RIBONUCLEOTIDE
REDUCTASE
8
11
ADP
Pi
UTP
dUMP
ATP
N5,N10 -Methylene H4 folate
Glutamine
THYMIDYLATE
12
CTP
SYNTHASE
SYNTHASE
9
H2 folate
NH
2
O
CH3
N
HN
O
N
O N
R-5-
P
-
P
-
P
dR-5-
P
CTP
TMP
Figure 34-7.
The biosynthetic pathway for pyrimidine nucleotides.
METABOLISM OF PURINE & PYRIMIDINE NUCLEOTIDES
/
299
regulation. Carbamoyl phosphate synthase II (reaction
NH2
1, Figure 34-7) is inhibited by UTP and purine nu-
N
cleotides but activated by PRPP. Aspartate transcar-
N
bamoylase (reaction 2, Figure 34-7) is inhibited by
N N
CTP but activated by ATP. In addition, the first three
HO
H
and the last two enzymes of the pathway are regulated
2C
by coordinate repression and derepression.
O
H
H
H
H
Purine & Pyrimidine Nucleotide
OH
OH
Biosynthesis Are Coordinately Regulated
Adenosine
Purine and pyrimidine biosynthesis parallel one an-
H2O
other mole for mole, suggesting coordinated control of
their biosynthesis. Several sites of cross-regulation char-
NH+
acterize purine and pyrimidine nucleotide biosynthesis.
4
The PRPP synthase reaction (reaction 1, Figure 34-2),
O
O
which forms a precursor essential for both processes, is
N
feedback-inhibited by both purine and pyrimidine nu-
N
HN
HN
cleotides.
H2N
N
N
N N
HO
H
HO
H
2C
2C
HUMANS CATABOLIZE PURINES
O
O
TO URIC ACID
H
H
H
H
H
H
H
H
Humans convert adenosine and guanosine to uric acid
OH
OH
OH
OH
(Figure 34-8). Adenosine is first converted to inosine
Inosine
Guanosine
by adenosine deaminase. In mammals other than
higher primates, uricase converts uric acid to the water-
Pi
Pi
soluble product allantoin. However, since humans lack
uricase, the end product of purine catabolism in hu-
Ribose 1-phosphate
mans is uric acid.
O
O
N
N
HN
HN
GOUT IS A METABOLIC DISORDER
OF PURINE CATABOLISM
H2N
N NH
N NH
Hypoxanthine
Guanine
Various genetic defects in PRPP synthetase (reaction 1,
Figure 34-2) present clinically as gout. Each defect—
H2O + O2
eg, an elevated Vmax, increased affinity for ribose 5-
phosphate, or resistance to feedback inhibition—results
O
HN3
in overproduction and overexcretion of purine catabo-
H2O
2
N
lites. When serum urate levels exceed the solubility
HN
limit, sodium urate crystalizes in soft tissues and joints
and causes an inflammatory reaction, gouty arthritis.
O
NH
NH
However, most cases of gout reflect abnormalities in
Xanthine
renal handling of uric acid.
H2O + O2
H2O2
O
Figure 34-8. Formation of uric acid from purine nucleosides
HN
by way of the purine bases hypoxanthine, xanthine, and gua-
HN
7
1
O
nine. Purine deoxyribonucleosides are degraded by the same
9
O
3
NH
catabolic pathway and enzymes, all of which exist in the mucosa
NH
of the mammalian gastrointestinal tract.
Uric acid
300
/
CHAPTER 34
OTHER DISORDERS OF
CATABOLISM OF PYRIMIDINES
PURINE CATABOLISM
PRODUCES WATER-SOLUBLE
METABOLITES
While purine deficiency states are rare in human sub-
jects, there are numerous genetic disorders of purine ca-
Unlike the end products of purine catabolism, those
tabolism. Hyperuricemias may be differentiated based
of pyrimidine catabolism are highly water-soluble:
on whether patients excrete normal or excessive quanti-
CO2, NH3, β-alanine, and β-aminoisobutyrate (Figure
ties of total urates. Some hyperuricemias reflect specific
34-9). Excretion of β-aminoisobutyrate increases in
enzyme defects. Others are secondary to diseases such
leukemia and severe x-ray radiation exposure due to in-
as cancer or psoriasis that enhance tissue turnover.
creased destruction of DNA. However, many persons
of Chinese or Japanese ancestry routinely excrete
β-aminoisobutyrate. Humans probably transaminate
Lesch-Nyhan Syndrome
β-aminoisobutyrate to methylmalonate semialdehyde,
Lesch-Nyhan syndrome, an overproduction hyper-
which then forms succinyl-CoA (Figure 19-2).
uricemia characterized by frequent episodes of uric acid
lithiasis and a bizarre syndrome of self-mutilation, re-
Pseudouridine Is Excreted Unchanged
flects a defect in hypoxanthine-guanine phosphoribo-
Since no human enzyme catalyzes hydrolysis or phos-
syl transferase, an enzyme of purine salvage (Figure
phorolysis of pseudouridine, this unusual nucleoside is
34-4). The accompanying rise in intracellular PRPP re-
excreted unchanged in the urine of normal subjects.
sults in purine overproduction. Mutations that decrease
or abolish hypoxanthine-guanine phosphoribosyltrans-
ferase activity include deletions, frameshift mutations,
OVERPRODUCTION OF PYRIMIDINE
base substitutions, and aberrant mRNA splicing.
CATABOLITES IS ONLY RARELY
ASSOCIATED WITH CLINICALLY
Von Gierke’s Disease
SIGNIFICANT ABNORMALITIES
Purine overproduction and hyperuricemia in von
Since the end products of pyrimidine catabolism are
Gierke’s disease
(glucose-6-phosphatase deficiency)
highly water-soluble, pyrimidine overproduction results
occurs secondary to enhanced generation of the PRPP
in few clinical signs or symptoms. In hyperuricemia as-
precursor ribose 5-phosphate. An associated lactic aci-
sociated with severe overproduction of PRPP, there is
dosis elevates the renal threshold for urate, elevating
overproduction of pyrimidine nucleotides and in-
total body urates.
creased excretion of β-alanine. Since N5,N10-methyl-
ene-tetrahydrofolate is required for thymidylate synthe-
sis, disorders of folate and vitamin B12
metabolism
Hypouricemia
result in deficiencies of TMP.
Hypouricemia and increased excretion of hypoxanthine
and xanthine are associated with xanthine oxidase de-
Orotic Acidurias
ficiency due to a genetic defect or to severe liver dam-
The orotic aciduria that accompanies Reye’s syndrome
age. Patients with a severe enzyme deficiency may ex-
probably is a consequence of the inability of severely
hibit xanthinuria and xanthine lithiasis.
damaged mitochondria to utilize carbamoyl phosphate,
which then becomes available for cytosolic overproduc-
Adenosine Deaminase & Purine
tion of orotic acid. Type I orotic aciduria reflects a de-
ficiency of both orotate phosphoribosyltransferase and
Nucleoside Phosphorylase Deficiency
orotidylate decarboxylase
(reactions
5 and 6, Figure
Adenosine deaminase deficiency is associated with an
34-7); the rarer type II orotic aciduria is due to a defi-
immunodeficiency disease in which both thymus-
ciency only of orotidylate decarboxylase (reaction 6,
derived lymphocytes (T cells) and bone marrow-de-
Figure 34-7).
rived lymphocytes (B cells) are sparse and dysfunc-
tional. Purine nucleoside phosphorylase deficiency is
Deficiency of a Urea Cycle Enzyme Results
associated with a severe deficiency of T cells but appar-
in Excretion of Pyrimidine Precursors
ently normal B cell function. Immune dysfunctions ap-
pear to result from accumulation of dGTP and dATP,
Increased excretion of orotic acid, uracil, and uridine
which inhibit ribonucleotide reductase and thereby de-
accompanies a deficiency in liver mitochondrial or-
plete cells of DNA precursors.
nithine transcarbamoylase (reaction 2, Figure 29-9).
METABOLISM OF PURINE & PYRIMIDINE NUCLEOTIDES
/
301
Excess carbamoyl phosphate exits to the cytosol, where
NH2
it stimulates pyrimidine nucleotide biosynthesis. The
N
resulting mild orotic aciduria is increased by high-
nitrogen foods.
O
N
H
Cytosine
Drugs May Precipitate Orotic Aciduria
1/2
O2
Allopurinol (Figure 33-12), an alternative substrate for
orotate phosphoribosyltransferase
(reaction
5, Figure
34-7), competes with orotic acid. The resulting nu-
NH
3
cleotide product also inhibits orotidylate decarboxylase
O
(reaction 6, Figure 34-7), resulting in orotic aciduria
O
HN
CH3
and orotidinuria. 6-Azauridine, following conversion
HN
to 6-azauridylate, also competitively inhibits orotidylate
O N
decarboxylase (reaction 6, Figure 34-7), enhancing ex-
O
N
H
cretion of orotic acid and orotidine.
H
Thymine
Uracil
NADPH + H+
SUMMARY
•
Ingested nucleic acids are degraded to purines and
pyrimidines. New purines and pyrimidines are
NADP+
formed from amphibolic intermediates and thus are
O
dietarily nonessential.
O
CH3
•
Several reactions of IMP biosynthesis require folate
H
HN
HN
H
derivatives and glutamine. Consequently, antifolate
H
H
H
drugs and glutamine analogs inhibit purine biosyn-
H
O
N
H
O N
thesis.
H
H
Dihydrouracil
Dihydrothymine
•
Oxidation and amination of IMP forms AMP and
GMP, and subsequent phosphoryl transfer from
H2O
H2O
ATP forms ADP and GDP. Further phosphoryl
transfer from ATP to GDP forms GTP. ADP is con-
verted to ATP by oxidative phosphorylation. Reduc-
tion of NDPs forms dNDPs.
COO−
COO−
CH3
H2N
CH2
•
Hepatic purine nucleotide biosynthesis is stringently
H2N
C
H
C
CH2
regulated by the pool size of PRPP and by feedback
C
CH2
N
N
inhibition of PRPP-glutamyl amidotransferase by
O
O
H
H
AMP and GMP.
β -Ureidopropionate
β -Ureidoisobutyrate
•
Coordinated regulation of purine and pyrimidine
(N -carbamoyl-β -alanine)
(N -carbamoyl-β -amino-
isobutyrate)
nucleotide biosynthesis ensures their presence in pro-
portions appropriate for nucleic acid biosynthesis
and other metabolic needs.
•
Humans catabolize purines to uric acid (pKa 5.8),
CO2
+ NH3
present as the relatively insoluble acid at acidic pH or
as its more soluble sodium urate salt at a pH near
H
3N+
CH2
CH2
COO−
H3N+ CH2
CH COO−
neutrality. Urate crystals are diagnostic of gout.
β-Alanine
CH3
Other disorders of purine catabolism include Lesch-
β
-Aminoisobutyrate
Nyhan syndrome, von Gierke’s disease, and hypo-
uricemias.
Figure 34-9. Catabolism of pyrimidines.
•
Since pyrimidine catabolites are water-soluble, their
overproduction does not result in clinical abnormali-
ties. Excretion of pyrimidine precursors can, how-
ever, result from a deficiency of ornithine transcar-
bamoylase because excess carbamoyl phosphate is
available for pyrimidine biosynthesis.
302
/
CHAPTER 34
Martinez J et al: Human genetic disorders, a phylogenetic perspec-
REFERENCES
tive. J Mol Biol 2001;308:587.
Benkovic SJ: The transformylase enzymes in de novo purine
Puig JG et al: Gout: new questions for an ancient disease. Adv Exp
biosynthesis. Trends Biochem Sci 1994;9:320.
Med Biol 1998;431:1.
Brooks EM et al: Molecular description of three macro-deletions
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
and an Alu-Alu recombination-mediated duplication in the
herited Disease, 8th ed. McGraw-Hill, 2001.
HPRT gene in four patients with Lesch-Nyhan disease.
Tvrdik T et al: Molecular characterization of two deletion events
Mutat Res 2001;476:43.
involving Alu-sequences, one novel base substitution and two
Curto R, Voit EO, Cascante M: Analysis of abnormalities in purine
tentative hotspot mutations in the hypoxanthine phosphori-
metabolism leading to gout and to neurological dysfunctions
bosyltransferase gene in five patients with Lesch-Nyhan-
in man. Biochem J 1998;329:477.
syndrome. Hum Genet 1998;103:311.
Harris MD, Siegel LB, Alloway JA: Gout and hyperuricemia. Am
Zalkin H, Dixon JE: De novo purine nucleotide synthesis. Prog
Family Physician 1999;59:925.
Nucleic Acid Res Mol Biol 1992;42:259.
Lipkowitz MS et al: Functional reconstitution, membrane target-
ing, genomic structure, and chromosomal localization of a
human urate transporter. J Clin Invest 2001;107:1103.
Nucleic Acid Structure & Function
35
Daryl K. Granner, MD
BIOMEDICAL IMPORTANCE
The informational content of DNA (the genetic code)
resides in the sequence in which these monomers—
The discovery that genetic information is coded along
purine and pyrimidine deoxyribonucleotides—are or-
the length of a polymeric molecule composed of only
dered. The polymer as depicted possesses a polarity;
four types of monomeric units was one of the major sci-
one end has a 5′-hydroxyl or phosphate terminal while
entific achievements of the twentieth century. This
the other has a 3′-phosphate or hydroxyl terminal. The
polymeric molecule, DNA, is the chemical basis of
importance of this polarity will become evident. Since
heredity and is organized into genes, the fundamental
the genetic information resides in the order of the
units of genetic information. The basic information
monomeric units within the polymers, there must exist
pathway—ie, DNA directs the synthesis of RNA,
a mechanism of reproducing or replicating this specific
which in turn directs protein synthesis—has been eluci-
information with a high degree of fidelity. That re-
dated. Genes do not function autonomously; their
quirement, together with x-ray diffraction data from
replication and function are controlled by various gene
the DNA molecule and the observation of Chargaff
products, often in collaboration with components of
that in DNA molecules the concentration of de-
various signal transduction pathways. Knowledge of the
oxyadenosine (A) nucleotides equals that of thymidine
structure and function of nucleic acids is essential in
(T) nucleotides (A = T), while the concentration of de-
understanding genetics and many aspects of pathophys-
oxyguanosine (G) nucleotides equals that of deoxycyti-
iology as well as the genetic basis of disease.
dine (C) nucleotides (G = C), led Watson, Crick, and
Wilkins to propose in the early 1950s a model of a dou-
ble-stranded DNA molecule. The model they proposed
DNA CONTAINS THE
is depicted in Figure 35-2. The two strands of this
GENETIC INFORMATION
double-stranded helix are held in register by hydrogen
The demonstration that DNA contained the genetic in-
bonds between the purine and pyrimidine bases of the
formation was first made in 1944 in a series of experi-
respective linear molecules. The pairings between the
ments by Avery, MacLeod, and McCarty. They showed
purine and pyrimidine nucleotides on the opposite
that the genetic determination of the character (type) of
strands are very specific and are dependent upon hydro-
the capsule of a specific pneumococcus could be trans-
gen bonding of A with T and G with C (Figure 35-3).
mitted to another of a different capsular type by intro-
This common form of DNA is said to be right-
ducing purified DNA from the former coccus into the
handed because as one looks down the double helix the
latter. These authors referred to the agent (later shown
base residues form a spiral in a clockwise direction. In
to be DNA) accomplishing the change as “transforming
the double-stranded molecule, restrictions imposed by
factor.” Subsequently, this type of genetic manipulation
the rotation about the phosphodiester bond, the fa-
has become commonplace. Similar experiments have
vored anti configuration of the glycosidic bond (Figure
recently been performed utilizing yeast, cultured mam-
33-8), and the predominant tautomers
(see Figure
malian cells, and insect and mammalian embryos as re-
33-3) of the four bases (A, G, T, and C) allow A to pair
cipients and cloned DNA as the donor of genetic infor-
only with T and G only with C, as depicted in Figure
mation.
35-3. This base-pairing restriction explains the earlier
observation that in a double-stranded DNA molecule
the content of A equals that of T and the content of G
DNA Contains Four Deoxynucleotides
equals that of C. The two strands of the double-helical
The chemical nature of the monomeric deoxynucleo-
molecule, each of which possesses a polarity, are an-
tide units of DNA—deoxyadenylate, deoxyguanylate,
tiparallel; ie, one strand runs in the 5′ to 3′ direction
deoxycytidylate, and thymidylate—is described in
and the other in the 3′ to 5′ direction. This is analogous
Chapter 33. These monomeric units of DNA are held
to two parallel streets, each running one way but carry-
in polymeric form by 3′,5′-phosphodiester bridges con-
ing traffic in opposite directions. In the double-
stituting a single strand, as depicted in Figure 35-1.
stranded DNA molecules, the genetic information re-
303
304
/
CHAPTER 35
O
N
NH
G
5′
CH2
N
NH2
N
NH2
N
O
O
C
P
O
O
H
H
CH2
N
H
H
H3C
O
NH
O
H
T
P
O
H
CH2
N
NH2
H H
H
O
N
O
N
O
H
A
P
H
CH2
N
N
H H
H
O
O
O
H
P
H
H H
H
O
3′
H
P
O
Figure 35-1.
A segment of one strand of a DNA molecule in which the purine and pyrimidine bases guanine
(G), cytosine (C), thymine (T), and adenine (A) are held together by a phosphodiester backbone between 2′-de-
oxyribosyl moieties attached to the nucleobases by an N-glycosidic bond. Note that the backbone has a polarity
(ie, a direction). Convention dictates that a single-stranded DNA sequence is written in the 5′ to 3′ direction (ie,
pGpCpTpA, where G, C, T, and A represent the four bases and p represents the interconnecting phosphates).
sides in the sequence of nucleotides on one strand, the
dine nucleotide, whereas the other pair, the A-T pair, is
template strand. This is the strand of DNA that is
held together by two hydrogen bonds. Thus, the G-C
copied during nucleic acid synthesis. It is sometimes re-
bonds are much more resistant to denaturation, or
ferred to as the noncoding strand. The opposite strand
“melting,” than A-T-rich regions.
is considered the coding strand because it matches the
RNA transcript that encodes the protein.
The Denaturation (Melting) of DNA
The two strands, in which opposing bases are held
Is Used to Analyze Its Structure
together by hydrogen bonds, wind around a central axis
in the form of a double helix. Double-stranded DNA
The double-stranded structure of DNA can be sepa-
exists in at least six forms (A-E and Z). The B form is
rated into two component strands (melted) in solution
usually found under physiologic conditions (low salt,
by increasing the temperature or decreasing the salt
high degree of hydration). A single turn of B-DNA
concentration. Not only do the two stacks of bases pull
about the axis of the molecule contains ten base pairs.
apart but the bases themselves unstack while still con-
The distance spanned by one turn of B-DNA is 3.4
nected in the polymer by the phosphodiester backbone.
nm. The width (helical diameter) of the double helix in
Concomitant with this denaturation of the DNA mole-
B-DNA is 2 nm.
cule is an increase in the optical absorbance of the
As depicted in Figure 35-3, three hydrogen bonds
purine and pyrimidine bases—a phenomenon referred
hold the deoxyguanosine nucleotide to the deoxycyti-
to as hyperchromicity of denaturation. Because of the
NUCLEIC ACID STRUCTURE & FUNCTION
/
305
CH3
O
H
H
N
N
N
H
N
O
N
Thymidine
Minor groove
N
N
S
A
T
S
o
Adenosine
P
P
34 A
S
T
A
S
P
P
S
C
G
S
H
P
P
S
G C
S
N
Major groove
H
N
N
O
H
N
N
O
H
Cytosine
N
N
N
H Guanosine
20 Ao
Figure 35-3. Base pairing between deoxyadenosine
Figure 35-2.
A diagrammatic representation of the
and thymidine involves the formation of two hydrogen
Watson and Crick model of the double-helical structure
bonds. Three such bonds form between deoxycytidine
of the B form of DNA. The horizontal arrow indicates
and deoxyguanosine. The broken lines represent hy-
the width of the double helix (20 Å), and the vertical
drogen bonds.
arrow indicates the distance spanned by one complete
turn of the double helix (34 Å). One turn of B-DNA in-
cludes ten base pairs (bp), so the rise is 3.4 Å per bp.
or DNA-RNA hybrids to be separated at much lower
temperatures and minimizes the phosphodiester bond
The central axis of the double helix is indicated by the
breakage that occurs at high temperatures.
vertical rod. The short arrows designate the polarity of
the antiparallel strands. The major and minor grooves
are depicted. (A, adenine; C, cytosine; G, guanine;
Renaturation of DNA Requires
T, thymine; P, phosphate; S, sugar [deoxyribose].)
Base Pair Matching
Separated strands of DNA will renature or reassociate
when appropriate physiologic temperature and salt con-
stacking of the bases and the hydrogen bonding be-
ditions are achieved. The rate of reassociation depends
tween the stacks, the double-stranded DNA molecule
upon the concentration of the complementary strands.
exhibits properties of a rigid rod and in solution is a vis-
Reassociation of the two complementary DNA strands
cous material that loses its viscosity upon denaturation.
of a chromosome after DNA replication is a physiologic
The strands of a given molecule of DNA separate
example of renaturation (see below). At a given temper-
over a temperature range. The midpoint is called the
ature and salt concentration, a particular nucleic acid
melting temperature, or Tm. The Tm is influenced by
strand will associate tightly only with a complementary
the base composition of the DNA and by the salt con-
strand. Hybrid molecules will also form under appro-
centration of the solution. DNA rich in G-C pairs,
priate conditions. For example, DNA will form a hy-
which have three hydrogen bonds, melts at a higher tem-
brid with a complementary DNA (cDNA) or with a
perature than that rich in A-T pairs, which have two hy-
cognate messenger RNA (mRNA; see below). When
drogen bonds. A tenfold increase of monovalent cation
combined with gel electrophoresis techniques that sepa-
concentration increases the Tm by 16.6 °C. Formamide,
rate hybrid molecules by size and radioactive labeling to
which is commonly used in recombinant DNA experi-
provide a detectable signal, the resulting analytic tech-
ments, destabilizes hydrogen bonding between bases,
niques are called Southern (DNA/cDNA) and North-
thereby lowering the Tm. This allows the strands of DNA
ern blotting (DNA/RNA), respectively. These proce-
306
/
CHAPTER 35
dures allow for very specific identification of hybrids
the cell and organism, and it provides the information
from mixtures of DNA or RNA (see Chapter 40).
inherited by daughter cells or offspring. Both of these
functions require that the DNA molecule serve as a
There Are Grooves in the DNA Molecule
template—in the first case for the transcription of the
information into RNA and in the second case for the
Careful examination of the model depicted in Figure
replication of the information into daughter DNA mol-
35-2 reveals a major groove and a minor groove wind-
ecules.
ing along the molecule parallel to the phosphodiester
The complementarity of the Watson and Crick dou-
backbones. In these grooves, proteins can interact specif-
ble-stranded model of DNA strongly suggests that
ically with exposed atoms of the nucleotides (usually by
replication of the DNA molecule occurs in a semicon-
H bonding) and thereby recognize and bind to specific
servative manner. Thus, when each strand of the dou-
nucleotide sequences without disrupting the base pair-
ble-stranded parental DNA molecule separates from its
ing of the double-helical DNA molecule. As discussed in
complement during replication, each serves as a tem-
Chapters 37 and 39, regulatory proteins control the ex-
plate on which a new complementary strand is synthe-
pression of specific genes via such interactions.
sized (Figure 35-4). The two newly formed double-
stranded daughter DNA molecules, each containing
DNA Exists in Relaxed
one strand (but complementary rather than identical)
& Supercoiled Forms
from the parent double-stranded DNA molecule, are
then sorted between the two daughter cells (Figure
In some organisms such as bacteria, bacteriophages, and
35-5). Each daughter cell contains DNA molecules
many DNA-containing animal viruses, the ends of the
with information identical to that which the parent
DNA molecules are joined to create a closed circle with
possessed; yet in each daughter cell the DNA molecule
no covalently free ends. This of course does not destroy
of the parent cell has been only semiconserved.
the polarity of the molecules, but it eliminates all free 3′
and 5′ hydroxyl and phosphoryl groups. Closed circles
exist in relaxed or supercoiled forms. Supercoils are intro-
THE CHEMICAL NATURE OF RNA DIFFERS
duced when a closed circle is twisted around its own axis
FROM THAT OF DNA
or when a linear piece of duplex DNA, whose ends are
fixed, is twisted. This energy-requiring process puts the
Ribonucleic acid (RNA) is a polymer of purine and
molecule under stress, and the greater the number of su-
pyrimidine ribonucleotides linked together by
3′,5′-
percoils, the greater the stress or torsion (test this by
phosphodiester bridges analogous to those in DNA
twisting a rubber band). Negative supercoils are formed
(Figure 35-6). Although sharing many features with
when the molecule is twisted in the direction opposite
DNA, RNA possesses several specific differences:
from the clockwise turns of the right-handed double
helix found in B-DNA. Such DNA is said to be under-
(1) In RNA, the sugar moiety to which the phos-
wound. The energy required to achieve this state is, in a
phates and purine and pyrimidine bases are attached is
sense, stored in the supercoils. The transition to another
ribose rather than the 2′-deoxyribose of DNA.
form that requires energy is thereby facilitated by the un-
(2) The pyrimidine components of RNA differ from
derwinding. One such transition is strand separation,
those of DNA. Although RNA contains the ribonu-
which is a prerequisite for DNA replication and tran-
cleotides of adenine, guanine, and cytosine, it does not
scription. Supercoiled DNA is therefore a preferred form
possess thymine except in the rare case mentioned
in biologic systems. Enzymes that catalyze topologic
below. Instead of thymine, RNA contains the ribonu-
changes of DNA are called topoisomerases. Topoisom-
cleotide of uracil.
erases can relax or insert supercoils. The best-character-
(3) RNA exists as a single strand, whereas DNA ex-
ized example is bacterial gyrase, which induces negative
ists as a double-stranded helical molecule. However,
supercoiling in DNA using ATP as energy source. Ho-
given the proper complementary base sequence with
mologs of this enzyme exist in all organisms and are im-
opposite polarity, the single strand of RNA—as
portant targets for cancer chemotherapy.
demonstrated in Figure 35-7—is capable of folding
back on itself like a hairpin and thus acquiring double-
stranded characteristics.
DNA PROVIDES A TEMPLATE FOR
(4) Since the RNA molecule is a single strand com-
REPLICATION & TRANSCRIPTION
plementary to only one of the two strands of a gene, its
The genetic information stored in the nucleotide se-
guanine content does not necessarily equal its cytosine
quence of DNA serves two purposes. It is the source of
content, nor does its adenine content necessarily equal
information for the synthesis of all protein molecules of
its uracil content.
NUCLEIC ACID STRUCTURE & FUNCTION
/
307
OLD
OLD
5′
3′
G
C
Original
C
G
parent molecule
A
T
A
A
T
G
C
G
C
A
First-generation
A
T
daughter molecules
T
A
G
C
G
C
3′
5′
T
A
T
A
C
G
C
G
C
C
Second-generation
daughter molecules
A
T
T
A
A
T
A
T
T
A
T
A
G C
G
C
Figure 35-5. DNA replication is semiconservative.
G
G
During a round of replication, each of the two strands
A
T
A
T
of DNA is used as a template for synthesis of a new,
complementary strand.
3′
5′
3′
5′
OLD
NEW
NEW
OLD
Figure 35-4.
The double-stranded structure of DNA
and the template function of each old strand (dark
complementarity, an RNA molecule can bind specifi-
shading) on which a new (light shading) complemen-
cally via the base-pairing rules to its template DNA
tary strand is synthesized.
strand; it will not bind (“hybridize”) with the other
(coding) strand of its gene. The sequence of the RNA
molecule (except for U replacing T) is the same as that
of the coding strand of the gene (Figure 35-8).
(5) RNA can be hydrolyzed by alkali to 2′,3′ cyclic
diesters of the mononucleotides, compounds that can-
Nearly All of the Several Species of RNA
not be formed from alkali-treated DNA because of the
Are Involved in Some Aspect of Protein
absence of a 2′-hydroxyl group. The alkali lability of
Synthesis
RNA is useful both diagnostically and analytically.
Those cytoplasmic RNA molecules that serve as tem-
Information within the single strand of RNA is con-
plates for protein synthesis (ie, that transfer genetic in-
tained in its sequence (“primary structure”) of purine
formation from DNA to the protein-synthesizing ma-
and pyrimidine nucleotides within the polymer. The
chinery) are designated messenger RNAs, or mRNAs.
sequence is complementary to the template strand of
Many other cytoplasmic RNA molecules (ribosomal
the gene from which it was transcribed. Because of this
RNAs; rRNAs) have structural roles wherein they con-
308
/
CHAPTER 35
O
N
NH
G
5′
NH2
CH2
N
NH2
N
N
O
O
C
P
O
O
CH2
N
H H
H
H
O
NH
HO
O
U
P
O
H
CH
2
N
NH2
H H
H
O
N
O
N
O
HO
A
P
H
CH2
N
N
H H
H
O
O
O
HO
P
H
H H
H
O
3′
HO
P
O
Figure 35-6.
A segment of a ribonucleic acid (RNA) molecule in which the purine and pyrimidine bases—
guanine (G), cytosine (C), uracil (U), and adenine (A)—are held together by phosphodiester bonds between ribo-
syl moieties attached to the nucleobases by N-glycosidic bonds. Note that the polymer has a polarity as indi-
cated by the labeled 3′- and 5′-attached phosphates.
tribute to the formation and function of ribosomes (the
The genetic material for some animal and plant
organellar machinery for protein synthesis) or serve as
viruses is RNA rather than DNA. Although some RNA
adapter molecules
(transfer RNAs; tRNAs) for the
viruses never have their information transcribed into a
translation of RNA information into specific sequences
DNA molecule, many animal RNA viruses—specifi-
of polymerized amino acids.
cally, the retroviruses (the HIV virus, for example)—are
Some RNA molecules have intrinsic catalytic activ-
transcribed by an RNA-dependent DNA polymerase,
ity. The activity of these ribozymes often involves the
the so-called reverse transcriptase, to produce a dou-
cleavage of a nucleic acid. An example is the role of
ble-stranded DNA copy of their RNA genome. In
RNA in catalyzing the processing of the primary tran-
many cases, the resulting double-stranded DNA tran-
script of a gene into mature messenger RNA.
script is integrated into the host genome and subse-
Much of the RNA synthesized from DNA templates
quently serves as a template for gene expression and
in eukaryotic cells, including mammalian cells, is de-
from which new viral RNA genomes can be tran-
graded within the nucleus, and it never serves as either a
scribed.
structural or an informational entity within the cellular
cytoplasm.
RNA Is Organized in Several
In all eukaryotic cells there are small nuclear RNA
Unique Structures
(snRNA) species that are not directly involved in pro-
tein synthesis but play pivotal roles in RNA processing.
In all prokaryotic and eukaryotic organisms, three main
These relatively small molecules vary in size from 90 to
classes of RNA molecules exist: messenger RNA
about 300 nucleotides (Table 35-1).
(mRNA), transfer RNA (tRNA), and ribosomal RNA
NUCLEIC ACID STRUCTURE & FUNCTION
/
309
Table 35-1. Some of the species of small stable
RNAs found in mammalian cells.
Loop
Length
Molecules
C
G
Name
(nucleotides)
per Cell
Localization
C
G
U1
165
1 × 106
Nucleoplasm/hnRNA
G
C
U2
188
5 × 105
Nucleoplasm
A
U
U3
216
3 × 105
Nucleolus
A
U
U4
139
1 × 105
Nucleoplasm
A
U
U5
118
2 × 105
Nucleoplasm
U
G
U6
106
3 × 105
Perichromatin granules
U
G
Stem
4.5S
91-95
3 x 105
Nucleus and cytoplasm
C
C
7S
280
5 × 105
Nucleus and cytoplasm
G
C
7-2
290
1 × 105
Nucleus and cytoplasm
U
A
7-3
300
2 × 105
Nucleus
U
A
U
C
U
A
Messenger RNAs, particularly in eukaryotes, have
C
G
some unique chemical characteristics. The 5′ terminal
G
C
of mRNA is “capped” by a 7-methylguanosine triphos-
phate that is linked to an adjacent 2′-O-methyl ribonu-
cleoside at its 5′-hydroxyl through the three phosphates
5′
3′
(Figure 35-10). The mRNA molecules frequently con-
tain internal 6-methyladenylates and other 2′-O-ribose
Figure 35-7.
Diagrammatic representation of the
methylated nucleotides. The cap is involved in the
secondary structure of a single-stranded RNA molecule
recognition of mRNA by the translating machinery,
in which a stem loop, or “hairpin,” has been formed and
and it probably helps stabilize the mRNA by preventing
is dependent upon the intramolecular base pairing.
the attack of 5′-exonucleases. The protein-synthesizing
Note that A forms hydrogen bonds with U in RNA.
machinery begins translating the mRNA into proteins
beginning downstream of the 5′ or capped terminal.
The other end of most mRNA molecules, the 3′-hy-
(rRNA). Each differs from the others by size, function,
droxyl terminal, has an attached polymer of adenylate
and general stability.
residues
20-250 nucleotides in length. The specific
function of the poly(A) “tail” at the 3′-hydroxyl termi-
nal of mRNAs is not fully understood, but it seems that
A. MESSENGER RNA (MRNA)
it maintains the intracellular stability of the specific
This class is the most heterogeneous in size and stabil-
mRNA by preventing the attack of 3′-exonucleases.
ity. All members of the class function as messengers
Some mRNAs, including those for some histones, do
conveying the information in a gene to the protein-
not contain poly(A). The poly(A) tail, because it will
synthesizing machinery, where each serves as a template
form a base pair with oligodeoxythymidine polymers
on which a specific sequence of amino acids is polymer-
attached to a solid substrate like cellulose, can be used
ized to form a specific protein molecule, the ultimate
to separate mRNA from other species of RNA, includ-
gene product (Figure 35-9).
ing mRNA molecules that lack this tail.
DNA strands:
Coding
5′ —
TGGAAT TGTGAGCGGATAACAAT T TCACACAGGAAACAGCTATGACCATG
— 3′
Template
3′ —
ACCT TAACACTCGCCTAT TGT TAAAGTGTGTCCT T TGTCGATACTGGTAC
— 5′
RNA
5′
p AUUGUGAGCGGAUAACAAUUUCACACAGGAAACAGCUAUGACCAUG
3′
transcript
Figure 35-8. The relationship between the sequences of an RNA transcript and its gene, in which the cod-
ing and template strands are shown with their polarities. The RNA transcript with a 5′ to 3′ polarity is comple-
mentary to the template strand with its 3′ to 5′ polarity. Note that the sequence in the RNA transcript and its
polarity is the same as that in the coding strand, except that the U of the transcript replaces the T of the gene.
310
/
CHAPTER 35
DNA
5′
3′
3′
5′
mRNA
5′
3′
Protein synthesis on mRNA template
3′
5′
Figure 35-9. The expression of genetic in-
Completed
Ribosome
protein
formation in DNA into the form of an mRNA
molecule
transcript. This is subsequently translated by
ribosomes into a specific protein molecule.
In mammalian cells, including cells of humans, the
The D, T C, and extra arms help define a specific
mRNA molecules present in the cytoplasm are not the
tRNA.
RNA products immediately synthesized from the DNA
Although tRNAs are quite stable in prokaryotes, they
template but must be formed by processing from a pre-
are somewhat less stable in eukaryotes. The opposite is
cursor molecule before entering the cytoplasm. Thus,
true for mRNAs, which are quite unstable in prokary-
in mammalian nuclei, the immediate products of gene
otes but generally stable in eukaryotic organisms.
transcription constitute a fourth class of RNA mole-
C. RIBOSOMAL RNA (RRNA)
cules. These nuclear RNA molecules are very heteroge-
neous in size and are quite large. The heterogeneous
A ribosome is a cytoplasmic nucleoprotein structure
nuclear RNA (hnRNA) molecules may have a molecu-
that acts as the machinery for the synthesis of proteins
lar weight in excess of
107, whereas the molecular
from the mRNA templates. On the ribosomes, the
weight of mRNA molecules is generally less than 2 ×
mRNA and tRNA molecules interact to translate into a
106. As discussed in Chapter 37, hnRNA molecules are
specific protein molecule information transcribed from
processed to generate the mRNA molecules which then
the gene. In active protein synthesis, many ribosomes
enter the cytoplasm to serve as templates for protein
are associated with an mRNA molecule in an assembly
synthesis.
called the polysome.
The components of the mammalian ribosome,
B. TRANSFER RNA (TRNA)
which has a molecular weight of about 4.2 × 106 and a
tRNA molecules vary in length from 74 to 95 nu-
sedimentation velocity of
80S (Svedberg units), are
cleotides. They also are generated by nuclear processing
shown in Table 35-2. The mammalian ribosome con-
of a precursor molecule (Chapter 37). The tRNA mole-
tains two major nucleoprotein subunits—a larger one
cules serve as adapters for the translation of the infor-
with a molecular weight of 2.8 × 106 (60S) and a
mation in the sequence of nucleotides of the mRNA
smaller subunit with a molecular weight of 1.4 × 106
into specific amino acids. There are at least 20 species
(40S). The 60S subunit contains a 5S ribosomal RNA
of tRNA molecules in every cell, at least one (and often
(rRNA), a 5.8S rRNA, and a 28S rRNA; there are also
several) corresponding to each of the 20 amino acids re-
probably more than 50 specific polypeptides. The 40S
quired for protein synthesis. Although each specific
subunit is smaller and contains a single 18S rRNA and
tRNA differs from the others in its sequence of nu-
approximately 30 distinct polypeptide chains. All of the
cleotides, the tRNA molecules as a class have many fea-
ribosomal RNA molecules except the 5S rRNA are
tures in common. The primary structure—ie, the nu-
processed from a single 45S precursor RNA molecule in
cleotide sequence—of all tRNA molecules allows
the nucleolus (Chapter 37). 5S rRNA is independently
extensive folding and intrastrand complementarity to
transcribed. The highly methylated ribosomal RNA
generate a secondary structure that appears like a
molecules are packaged in the nucleolus with the spe-
cloverleaf (Figure 35-11).
cific ribosomal proteins. In the cytoplasm, the ribo-
All tRNA molecules contain four main arms. The
somes remain quite stable and capable of many transla-
acceptor arm terminates in the nucleotides CpCpAOH.
tion cycles. The functions of the ribosomal RNA
These three nucleotides are added posttranscription-
molecules in the ribosomal particle are not fully under-
ally. The tRNA-appropriate amino acid is attached to
stood, but they are necessary for ribosomal assembly
the 3′-OH group of the A moiety of the acceptor arm.
and seem to play key roles in the binding of mRNA to
NUCLEIC ACID STRUCTURE & FUNCTION
/
311
OH OH
C C
H H
HC
CH
O
H2N
N
N
H2C
5′
O-
NH2
O
N
HN
N
N
O-
O
5′
O
CH3
CH
N
O-
2
N
O
O
O
CAP
HC
CH
H H
2′
3′
C C
O
OCH3
NH
5′
O
O
CH2
N
O
O
mRNA
O-
HC
CH
H H
3′ C
C
OH
O
O
O-
Figure 35-10.
The cap structure attached to the 5′ terminal of most eukaryotic messen-
ger RNA molecules. A 7-methylguanosine triphosphate (black) is attached at the 5′ terminal
of the mRNA (shown in blue), which usually contains a 2′-O-methylpurine nucleotide.
These modifications (the cap and methyl group) are added after the mRNA is transcribed
from DNA.
ribosomes
and its translation. Recent studies suggest
size from 90 to 300 nucleotides and are
present
in
that an rRNA component performs the peptidyl trans-
100,000-1,000,000 copies per cell.
ferase activity and thus is an enzyme (a ribozyme).
Small nuclear RNAs (snRNAs), a subset of these
RNAs, are significantly involved in mRNA processing
and gene regulation. Of the several snRNAs, U1, U2,
D. SMALL STABLE RNA
U4, U5, and U6 are involved in intron removal and the
A large number of discrete, highly conserved, and small
processing of hnRNA into mRNA (Chapter 37). The
stable RNA species are found in eukaryotic cells. The
U7 snRNA may be involved in production of the cor-
majority of these molecules are complexed with pro-
rect 3′ ends of histone mRNA—which lacks a poly(A)
teins to form ribonucleoproteins and are distributed in
tail. The U4 and U6 snRNAs may also be required for
the nucleus, in the cytoplasm, or in both. They range in
poly(A) processing.
312
/
CHAPTER 35
aa
SPECIFIC NUCLEASES DIGEST
NUCLEIC ACIDS
Enzymes capable of degrading nucleic acids have been
3′
A
recognized for many years. These nucleases can be clas-
C
Acceptor
sified in several ways. Those which exhibit specificity
arm
C
for deoxyribonucleic acid are referred to as deoxyri-
bonucleases. Those which specifically hydrolyze ri-
5′P
bonucleic acids are ribonucleases. Within both of
these classes are enzymes capable of cleaving internal
Region of hydrogen
phosphodiester bonds to produce either 3′-hydroxyl
bonding between
base pairs
and 5′-phosphoryl terminals or
5′-hydroxyl and
3′-
phosphoryl terminals. These are referred to as endonu-
TψC arm
cleases. Some are capable of hydrolyzing both strands
of a double-stranded molecule, whereas others can
only cleave single strands of nucleic acids. Some nucle-
G
ases can hydrolyze only unpaired single strands, while
G
others are capable of hydrolyzing single strands partici-
T
C
pating in the formation of a double-stranded molecule.
D arm
ψ
There exist classes of endonucleases that recognize spe-
Extra arm
cific sequences in DNA; the majority of these are the
restriction endonucleases, which have in recent years
become important tools in molecular genetics and med-
U
Alkylated purine
ical sciences. A list of some currently recognized restric-
tion endonucleases is presented in Table 40-2.
Anticodon arm
Some nucleases are capable of hydrolyzing a nu-
cleotide only when it is present at a terminal of a mole-
Figure 35-11. Typical aminoacyl tRNA in which the
cule; these are referred to as exonucleases. Exonucle-
amino acid (aa) is attached to the 3′ CCA terminal. The
ases act in one direction (3′ → 5′ or 5′ → 3′) only. In
anticodon, TΨC, and dihydrouracil (D) arms are indi-
bacteria, a 3′ → 5′ exonuclease is an integral part of the
cated, as are the positions of the intramolecular hydro-
DNA replication machinery and there serves to edit—
gen bonding between these base pairs. (From Watson
or proofread—the most recently added deoxynucleo-
JD: Molecular Biology of the Gene, 3rd ed. Copyright ©
tide for base-pairing errors.
1976, 1970, 1965, by W.A. Benjamin, Inc., Menlo Park, Cali-
fornia.)
Table 35-2. Components of mammalian ribosomes.1
Mass
Protein
RNA
Component
(mw)
Number Mass
Size Mass Bases
40S subunit
1.4 × 106
~35
7 × 105
18S
7 × 105
1900
60S subunit
2.8 × 106
~50
1 × 106
5S
35,000
120
5.8S
45,000
160
28S
1.6 × 106
4700
1The ribosomal subunits are defined according to their sedimentation ve-
locity in Svedberg units (40S or 60S). This table illustrates the total mass
(MW) of each. The number of unique proteins and their total mass (MW) and
the RNA components of each subunit in size (Svedberg units), mass, and
number of bases are listed.
NUCLEIC ACID STRUCTURE & FUNCTION
/
313
SUMMARY
sis. The linear array of nucleotides in RNA consists
of A, G, C, and U, and the sugar moiety is ribose.
• DNA consists of four bases—A, G, C, and T—
• The major forms of RNA include messenger RNA
which are held in linear array by phosphodiester
(mRNA), ribosomal RNA
(rRNA), and transfer
bonds through the 3′ and 5′ positions of adjacent de-
RNA (tRNA). Certain RNA molecules act as cata-
oxyribose moieties.
lysts (ribozymes).
• DNA is organized into two strands by the pairing of
bases A to T and G to C on complementary strands.
These strands form a double helix around a central
axis.
REFERENCES
• The 3 × 109 base pairs of DNA in humans are orga-
Green R, Noller HF: Ribosomes and translation. Annu Rev Bio-
nized into the haploid complement of 23 chromo-
chem 1997;66:689.
somes. The exact sequence of these 3 billion nu-
Guthrie C, Patterson B: Spliceosomal snRNAs. Ann Rev Genet
cleotides defines the uniqueness of each individual.
1988;22:387.
• DNA provides a template for its own replication and
Hunt T: DNA Makes RNA Makes Protein. Elsevier, 1983.
thus maintenance of the genotype and for the tran-
Watson JD, Crick FHC: Molecular structure of nucleic acids. Na-
scription of the 30,000-50,000 genes into a variety
ture 1953;171:737.
of RNA molecules.
Watson JD: The Double Helix. Atheneum, 1968.
• RNA exists in several different single-stranded struc-
Watson JD et al: Molecular Biology of the Gene, 5th ed. Benjamin-
tures, most of which are involved in protein synthe-
Cummings, 2000.
DNA Organization, Replication,
36
& Repair
Daryl K. Granner, MD, & P. Anthony Weil, PhD
BIOMEDICAL IMPORTANCE*
larger than histones) and a small quantity of RNA. The
nonhistone proteins include enzymes involved in DNA
The genetic information in the DNA of a chromosome
replication, such as DNA topoisomerases. Also in-
can be transmitted by exact replication or it can be ex-
cluded are proteins involved in transcription, such as
changed by a number of processes, including crossing
the RNA polymerase complex. The double-stranded
over, recombination, transposition, and conversion.
DNA helix in each chromosome has a length that is
These provide a means of ensuring adaptability and di-
thousands of times the diameter of the cell nucleus.
versity for the organism but, when these processes go
One purpose of the molecules that comprise chro-
awry, can also result in disease. A number of enzyme
matin, particularly the histones, is to condense the
systems are involved in DNA replication, alteration,
DNA. Electron microscopic studies of chromatin have
and repair. Mutations are due to a change in the base
demonstrated dense spherical particles called nucleo-
sequence of DNA and may result from the faulty repli-
somes, which are approximately 10 nm in diameter
cation, movement, or repair of DNA and occur with a
and connected by DNA filaments (Figure 36-1). Nu-
frequency of about one in every 106 cell divisions. Ab-
cleosomes are composed of DNA wound around a col-
normalities in gene products (either in protein function
lection of histone molecules.
or amount) can be the result of mutations that occur in
coding or regulatory-region DNA. A mutation in a
Histones Are the Most Abundant
germ cell will be transmitted to offspring (so-called ver-
Chromatin Proteins
tical transmission of hereditary disease). A number of
factors, including viruses, chemicals, ultraviolet light,
The histones are a small family of closely related basic
and ionizing radiation, increase the rate of mutation.
proteins. H1 histones are the ones least tightly bound
Mutations often affect somatic cells and so are passed
to chromatin Figure 36-1) and are, therefore, easily re-
on to successive generations of cells, but only within an
moved with a salt solution, after which chromatin be-
organism. It is becoming apparent that a number of
comes soluble. The organizational unit of this soluble
diseases—and perhaps most cancers—are due to the
chromatin is the nucleosome. Nucleosomes contain
combined effects of vertical transmission of mutations
four classes of histones: H2A, H2B, H3, and H4. The
as well as horizontal transmission of induced mutations.
structures of all four histones—H2A, H2B, H3, and
H4, the so-called core histones forming the nucleo-
CHROMATIN IS THE CHROMOSOMAL
some—have been highly conserved between species.
This extreme conservation implies that the function of
MATERIAL EXTRACTED FROM NUCLEI
histones is identical in all eukaryotes and that the entire
OF CELLS OF EUKARYOTIC ORGANISMS
molecule is involved quite specifically in carrying out
Chromatin consists of very long double-stranded DNA
this function. The carboxyl terminal two-thirds of the
molecules and a nearly equal mass of rather small basic
molecules have a typical random amino acid composi-
proteins termed histones as well as a smaller amount of
tion, while their amino terminal thirds are particularly
nonhistone proteins (most of which are acidic and
rich in basic amino acids. These four core histones are
subject to at least five types of covalent modifica-
tion: acetylation, methylation, phosphorylation, ADP-
ribosylation, and covalent linkage (H2A only) to ubiq-
*So far as is possible, the discussion in this chapter and in Chapters
uitin. These histone modifications probably play an
37, 38, and 39 will pertain to mammalian organisms, which are, of
important role in chromatin structure and function as
course, among the higher eukaryotes. At times it will be necessary
illustrated in Table 36-1.
to refer to observations in prokaryotic organisms such as bacteria
and viruses, but in such cases the information will be of a kind that
The histones interact with each other in very specific
can be extrapolated to mammalian organisms.
ways. H3 and H4 form a tetramer containing two mol-
314
DNA ORGANIZATION, REPLICATION, & REPAIR
/
315
In the nucleosome, the DNA is supercoiled in a left-
handed helix over the surface of the disk-shaped histone
octamer (Figure 36-2). The majority of core histone
proteins interact with the DNA on the inside of the su-
percoil without protruding, though the amino terminal
tails of all the histones probably protrude outside of this
structure and are available for regulatory covalent mod-
ifications (see Table 36-1).
The (H3/H4)2 tetramer itself can confer nucleo-
some-like properties on DNA and thus has a central
role in the formation of the nucleosome. The addition
of two H2A-H2B dimers stabilizes the primary particle
and firmly binds two additional half-turns of DNA pre-
viously bound only loosely to the (H3/H4)2. Thus,
1.75 superhelical turns of DNA are wrapped around
the surface of the histone octamer, protecting 146 base
pairs of DNA and forming the nucleosome core particle
Figure 36-1. Electron micrograph of nucleosomes
(Figure 36-2). The core particles are separated by an
attached by strands of nucleic acid. (The bar represents
about 30-bp linker region of DNA. Most of the DNA
2.5 µm.) (Reproduced, with permission, from Oudet P,
is in a repeating series of these structures, giving the so-
Gross-Bellard M, Chambon P: Electron microscopic and
called “beads-on-a-string” appearance when examined
biochemical evidence that chromatin structure is a re-
by electron microscopy (see Figure 36-1).
peating unit. Cell 1975;4:281.)
The assembly of nucleosomes is mediated by one of
several chromatin assembly factors facilitated by histone
chaperones, proteins such as the anionic nuclear protein
ecules of each (H3/H4)2, while H2A and H2B form
nucleoplasmin. As the nucleosome is assembled, his-
dimers
(H2A-H2B). Under physiologic conditions,
tones are released from the histone chaperones. Nucleo-
these histone oligomers associate to form the histone oc-
somes appear to exhibit preference for certain regions on
tamer of the composition (H3/H4)2-(H2A-H2B)2.
specific DNA molecules, but the basis for this nonran-
dom distribution, termed phasing, is not completely
The Nucleosome Contains Histone & DNA
When the histone octamer is mixed with purified, dou-
ble-stranded DNA, the same x-ray diffraction pattern is
formed as that observed in freshly isolated chromatin.
Electron microscopic studies confirm the existence of
Histone octamer
reconstituted nucleosomes. Furthermore, the reconsti-
tution of nucleosomes from DNA and histones H2A,
H2B, H3, and H4 is independent of the organismal or
cellular origin of the various components. The histone
H1 and the nonhistone proteins are not necessary for
the reconstitution of the nucleosome core.
Histone
DNA
Table 36-1. Possible roles of modified histones.
H1
Figure 36-2. Model for the structure of the nucleo-
1. Acetylation of histones H3 and H4 is associated with the ac-
some, in which DNA is wrapped around the surface of a
tivation or inactivation of gene transcription (Chapter 37).
flat protein cylinder consisting of two each of histones
2. Acetylation of core histones is associated with chromoso-
H2A, H2B, H3, and H4 that form the histone octamer.
mal assembly during DNA replication.
3. Phosphorylation of histone H1 is associated with the con-
The 146 base pairs of DNA, consisting of 1.75 superheli-
densation of chromosomes during the replication cycle.
cal turns, are in contact with the histone octamer. This
4. ADP-ribosylation of histones is associated with DNA repair.
protects the DNA from digestion by a nuclease. The po-
5. Methylation of histones is correlated with activation and
sition of histone H1, when it is present, is indicated by
repression of gene transcription.
the dashed outline at the bottom of the figure.
316
/
CHAPTER 36
understood. It is probably related to the relative physical
tion by a nuclease such as DNase I. DNase I makes sin-
flexibility of certain nucleotide sequences that are able to
gle-strand cuts in any segment of DNA (no sequence
accommodate the regions of kinking within the super-
specificity). It will digest DNA not protected by protein
coil as well as the presence of other DNA-bound factors
into its component deoxynucleotides. The sensitivity to
that limit the sites of nucleosome deposition.
DNase I of chromatin regions being actively transcribed
The super-packing of nucleosomes in nuclei is seem-
reflects only a potential for transcription rather than
ingly dependent upon the interaction of the H1 his-
transcription itself and in several systems can be corre-
tones with adjacent nucleosomes.
lated with a relative lack of 5-methyldeoxycytidine in the
DNA and particular histone covalent modifications
(phosphorylation, acetylation, etc; see Table 36-1).
HIGHER-ORDER STRUCTURES PROVIDE
Within the large regions of active chromatin there
FOR THE COMPACTION OF CHROMATIN
exist shorter stretches of 100-300 nucleotides that ex-
Electron microscopy of chromatin reveals two higher
hibit an even greater
(another tenfold) sensitivity to
orders of structure—the 10-nm fibril and the 30-nm
DNase I. These hypersensitive sites probably result
chromatin fiber—beyond that of the nucleosome itself.
from a structural conformation that favors access of the
The disk-like nucleosome structure has a 10-nm diame-
nuclease to the DNA. These regions are often located
ter and a height of 5 nm. The 10-nm fibril consists of
immediately upstream from the active gene and are the
nucleosomes arranged with their edges separated by a
location of interrupted nucleosomal structure caused by
small distance (30 bp of DNA) with their flat faces par-
the binding of nonhistone regulatory transcription factor
allel with the fibril axis (Figure 36-3). The 10-nm fibril
proteins. (See Chapters 37 and 39.) In many cases, it
is probably further supercoiled with six or seven nucleo-
seems that if a gene is capable of being transcribed, it
somes per turn to form the 30-nm chromatin fiber
very often has a DNase-hypersensitive site(s) in the chro-
(Figure 36-3). Each turn of the supercoil is relatively
matin immediately upstream. As noted above, nonhis-
flat, and the faces of the nucleosomes of successive
tone regulatory proteins involved in transcription control
turns would be nearly parallel to each other. H1 his-
and those involved in maintaining access to the template
tones appear to stabilize the 30-nm fiber, but their posi-
strand lead to the formation of hypersensitive sites. Hy-
tion and that of the variable length spacer DNA are not
persensitive sites often provide the first clue about the
clear. It is probable that nucleosomes can form a variety
presence and location of a transcription control element.
of packed structures. In order to form a mitotic chro-
Transcriptionally inactive chromatin is densely
mosome, the 30-nm fiber must be compacted in length
packed during interphase as observed by electron mi-
another 100-fold (see below).
croscopic studies and is referred to as heterochro-
In interphase chromosomes, chromatin fibers ap-
matin; transcriptionally active chromatin stains less
pear to be organized into 30,000-100,000 bp loops or
densely and is referred to as euchromatin. Generally,
domains anchored in a scaffolding (or supporting ma-
euchromatin is replicated earlier than heterochromatin
trix) within the nucleus. Within these domains, some
in the mammalian cell cycle (see below).
DNA sequences may be located nonrandomly. It has
There are two types of heterochromatin: constitutive
been suggested that each looped domain of chromatin
and facultative. Constitutive heterochromatin is al-
corresponds to one or more separate genetic functions,
ways condensed and thus inactive. It is found in the
containing both coding and noncoding regions of the
regions near the chromosomal centromere and at chro-
cognate gene or genes.
mosomal ends (telomeres). Facultative heterochro-
matin is at times condensed, but at other times it is ac-
tively transcribed and, thus, uncondensed and appears
SOME REGIONS OF CHROMATIN ARE
as euchromatin. Of the two members of the X chromo-
“ACTIVE” & OTHERS ARE “INACTIVE”
some pair in mammalian females, one X chromosome is
Generally, every cell of an individual metazoan organism
almost completely inactive transcriptionally and is hete-
contains the same genetic information. Thus, the differ-
rochromatic. However, the heterochromatic X chromo-
ences between cell types within an organism must be ex-
some decondenses during gametogenesis and becomes
plained by differential expression of the common genetic
transcriptionally active during early embryogenesis—
information. Chromatin containing active genes
(ie,
thus, it is facultative heterochromatin.
transcriptionally active chromatin) has been shown to
Certain cells of insects, eg, Chironomus, contain
differ in several ways from that of inactive regions. The
giant chromosomes that have been replicated for ten
nucleosome structure of active chromatin appears to be
cycles without separation of daughter chromatids.
altered, sometimes quite extensively, in highly active re-
These copies of DNA line up side by side in precise reg-
gions. DNA in active chromatin contains large regions
ister and produce a banded chromosome containing re-
(about 100,000 bases long) that are sensitive to diges-
gions of condensed chromatin and lighter bands of
Metaphase
chromosome
1400 nm
Condensed
loops
700 nm
Nuclear-scaffold
associated
form
Chromosome
scaffold
Non-condensed
loops
300 nm
30-nm
chromatin fibril
30 nm
composed of
nucleosomes
H1
H1
Oct
“Beads-
on-a-string”
10 nm
Oct
Oct
10-nm
chromatin
H1
fibril
Naked
double-helical
2 nm
DNA
Figure 36-3. Shown is the extent of DNA packaging in metaphase chromosomes (top) to noted duplex DNA (bot-
tom). Chromosomal DNA is packaged and organized at several levels as shown (see Table 36-2). Each phase of con-
densation or compaction and organization (bottom to top) decreases overall DNA accessibility to an extent that the
DNA sequences in metaphase chromosomes are almost totally transcriptionally inert. In toto, these five levels of
DNA compaction result in nearly a 104-fold linear decrease in end-to-end DNA length. Complete condensation and
decondensation of the linear DNA in chromosomes occur in the space of hours during the normal replicative cell
cycle (see Figure 36-20).
317
318
/
CHAPTER 36
more extended chromatin. Transcriptionally active re-
sition of which is characteristic for a given chromosome
gions of these polytene chromosomes are especially
(Figure 36-5). The centromere is an adenine-thymine
decondensed into “puffs” that can be shown to contain
(A-T) rich region ranging in size from 102 (brewers’
the enzymes responsible for transcription and to be the
yeast) to 106 (mammals) base pairs. It binds several pro-
sites of RNA synthesis (Figure 36-4).
teins with high affinity. This complex, called the kine-
tochore, provides the anchor for the mitotic spindle. It
thus is an essential structure for chromosomal segrega-
DNA IS ORGANIZED
tion during mitosis.
INTO CHROMOSOMES
The ends of each chromosome contain structures
At metaphase, mammalian chromosomes possess a
called telomeres. Telomeres consist of short, repeat
twofold symmetry, with the identical duplicated sister
TG-rich sequences. Human telomeres have a variable
chromatids connected at a centromere, the relative po-
number of repeats of the sequence 5′-TTAGGG-3′,
which can extend for several kilobases. Telomerase, a
multisubunit RNA-containing complex related to viral
RNA-dependent DNA polymerases (reverse transcrip-
tases), is the enzyme responsible for telomere synthesis
and thus for maintaining the length of the telomere.
Since telomere shortening has been associated with
both malignant transformation and aging, telomerase
has become an attractive target for cancer chemother-
apy and drug development. Each sister chromatid con-
tains one double-stranded DNA molecule. During in-
terphase, the packing of the DNA molecule is less dense
than it is in the condensed chromosome during
metaphase. Metaphase chromosomes are nearly com-
pletely transcriptionally inactive.
The human haploid genome consists of about
3 × 109 bp and about 1.7 × 107 nucleosomes. Thus, each
of the 23 chromatids in the human haploid genome
would contain on the average 1.3 × 108 nucleotides in
one double-stranded DNA molecule. The length of
each DNA molecule must be compressed about 8000-
fold to generate the structure of a condensed metaphase
5C
chromosome! In metaphase chromosomes, the 30-nm
5C
BR3
BR3
chromatin fibers are also folded into a series of looped
domains, the proximal portions of which are anchored
A
B
to a nonhistone proteinaceous scaffolding within the
nucleus (Figure 36-3). The packing ratios of each of
Figure 36-4.
Illustration of the tight correlation be-
the orders of DNA structure are summarized in Table
tween the presence of RNA polymerase II and RNA syn-
36-2.
thesis. A number of genes are activated when Chirono-
The packaging of nucleoproteins within chromatids
mus tentans larvae are subjected to heat shock (39 °C
is not random, as evidenced by the characteristic pat-
for 30 minutes). A: Distribution of RNA polymerase II
terns observed when chromosomes are stained with spe-
(also called type B) in isolated chromosome IV from the
cific dyes such as quinacrine or Giemsa’s stain (Figure
salivary gland (at arrows). The enzyme was detected by
36-6).
immunofluorescence using an antibody directed
From individual to individual within a single
against the polymerase. The 5C and BR3 are specific
species, the pattern of staining (banding) of the entire
bands of chromosome IV, and the arrows indicate puffs.
chromosome complement is highly reproducible; none-
B: Autoradiogram of a chromosome IV that was incu-
theless, it differs significantly from other species, even
bated in 3H-uridine to label the RNA. Note the corre-
those closely related. Thus, the packaging of the nucleo-
spondence of the immunofluorescence and presence
proteins in chromosomes of higher eukaryotes must in
of the radioactive RNA (black dots). Bar = 7 µm. (Repro-
some way be dependent upon species-specific character-
duced, with permission, from Sass H: RNA polymerase B in
istics of the DNA molecules.
polytene chromosomes. Cell 1982;28:274. Copyright ©
A combination of specialized staining techniques
1982 by the Massachusetts Institute of Technology.)
and high-resolution microscopy has allowed geneticists
DNA ORGANIZATION, REPLICATION, & REPAIR
/
319
Sister chromatid No. 2
Sister chromatid No. 1
Centromere
Figure 36-5. The two sister chromatids of
human chromosome 12 (× 27,850). The location
of the A+T-rich centromeric region connecting
sister chromatids is indicated, as are two of the
four telomeres residing at the very ends of the
chromatids that are attached one to the other at
the centromere. (Modified and reproduced, with
Telomeres
permission, from DuPraw EJ: DNA and Chromo-
(TTAGG)
n
somes. Holt, Rinehart, and Winston, 1970.)
to quite precisely map thousands of genes to specific re-
nonprotein coding DNA. Accordingly, the primary
gions of mouse and human chromosomes. With the re-
transcripts of DNA
(mRNA precursors, originally
cent elucidation of the human and mouse genome se-
termed hnRNA because this species of RNA was quite
quences, it has become clear that many of these visual
heterogeneous in size [length] and mostly restricted to
mapping methods were remarkably accurate.
the nucleus), contain noncoding intervening sequences
of RNA that must be removed in a process which also
joins together the appropriate coding segments to form
Coding Regions Are Often Interrupted
the mature mRNA. Most coding sequences for a single
by Intervening Sequences
mRNA are interrupted in the genome (and thus in the
primary transcript) by at least one—and in some cases
The protein coding regions of DNA, the transcripts
as many as 50—noncoding intervening sequences (in-
of which ultimately appear in the cytoplasm as single
trons). In most cases, the introns are much longer than
mRNA molecules, are usually interrupted in the eu-
the continuous coding regions (exons). The processing
karyotic genome by large intervening sequences of
of the primary transcript, which involves removal of in-
trons and splicing of adjacent exons, is described in de-
tail in Chapter 37.
The function of the intervening sequences, or in-
Table 36-2. The packing ratios of each of the
trons, is not clear. They may serve to separate func-
orders of DNA structure.
tional domains (exons) of coding information in a form
that permits genetic rearrangement by recombination
to occur more rapidly than if all coding regions for a
Chromatin Form
Packing Ratio
given genetic function were contiguous. Such an en-
Naked double-helical DNA
~1.0
hanced rate of genetic rearrangement of functional do-
10-nm fibril of nucleosomes
7-10
mains might allow more rapid evolution of biologic
25- to 30-nm chromatin fiber of superheli-
40-60
function. The relationships among chromosomal
cal nucleosomes
DNA, gene clusters on the chromosome, the exon-
Condensed metaphase chromosome of
8000
intron structure of genes, and the final mRNA product
loops
are illustrated in Figure 36-7.
320
/
CHAPTER 36
1
2
3
4
5
6
7
8
9
10
11
12
18
13
14
15
16
17
19
20
21
22
XY
Figure 36-6. A human karyotype (of a man with a normal 46,XY constitution), in
which the metaphase chromosomes have been stained by the Giemsa method and
aligned according to the Paris Convention. (Courtesy of H Lawce and F Conte.)
MUCH OF THE MAMMALIAN GENOME
The DNA in a eukaryotic genome can be divided
into different
“sequence classes.” These are unique-
IS REDUNDANT & MUCH IS
sequence, or nonrepetitive, DNA and repetitive-
NOT TRANSCRIBED
sequence DNA. In the haploid genome, unique-se-
The haploid genome of each human cell consists of
quence DNA generally includes the single copy genes
3 × 109 base pairs of DNA subdivided into 23 chromo-
that code for proteins. The repetitive DNA in the hap-
somes. The entire haploid genome contains sufficient
loid genome includes sequences that vary in copy num-
DNA to code for nearly 1.5 million average-sized
ber from two to as many as 107 copies per cell.
genes. However, studies of mutation rates and of the
complexities of the genomes of higher organisms
strongly suggest that humans have < 100,000 proteins
More Than Half the DNA in Eukaryotic
encoded by the ~1.1% of the human genome that is
Organisms Is in Unique or
composed of exonic DNA. This implies that most of
Nonrepetitive Sequences
the DNA is noncoding—ie, its information is never
translated into an amino acid sequence of a protein
This estimation
(and the distribution of repetitive-
molecule. Certainly, some of the excess DNA sequences
sequence DNA) is based on a variety of DNA-RNA hy-
serve to regulate the expression of genes during devel-
bridization techniques and, more recently, on direct
opment, differentiation, and adaptation to the environ-
DNA sequencing. Similar techniques are used to esti-
ment. Some excess clearly makes up the intervening se-
mate the number of active genes in a population of
quences or introns (24% of the total human genome)
unique-sequence DNA. In brewers’ yeast
(Saccha-
that split the coding regions of genes, but much of the
romyces cerevisiae, a lower eukaryote), about two thirds
excess appears to be composed of many families of re-
of its 6200 genes are expressed. In typical tissues in a
peated sequences for which no functions have been
higher eukaryote (eg, mammalian liver and kidney), be-
clearly defined. A summary of the salient features of the
tween 10,000 and 15,000 genes are expressed. Differ-
human genome is presented in Chapter 40.
ent combinations of genes are expressed in each tissue,
DNA ORGANIZATION, REPLICATION, & REPAIR
/
321
Chromosome
1.5 × 108 bp
(1-2 × 103 genes)
Gene cluster
(~20 genes)
1.5 × 106 bp
Gene
2 × 104
bp
Primary transcript
8 × 103
nt
mRNA
2 × 103 nt
Figure 36-7. The relationship between chromosomal DNA and mRNA. The human
haploid DNA complement of 3 × 109 base pairs (bp) is distributed between 23 chromo-
somes. Genes are clustered on these chromosomes. An average gene is 2 × 104 bp in
length, including the regulatory region (hatched area), which is usually located at the 5′
end of the gene. The regulatory region is shown here as being adjacent to the transcrip-
tion initiation site (arrow). Most eukaryotic genes have alternating exons and introns. In
this example, there are nine exons (dark blue areas) and eight introns (light blue areas).
The introns are removed from the primary transcript by the processing reaction, and the
exons are ligated together in sequence to form the mature mRNA. (nt, nucleotides.)
of course, and how this is accomplished is one of the
Depending on their length, moderately repetitive
major unanswered questions in biology.
sequences are classified as long interspersed repeat
sequences
(LINEs) or short interspersed repeat
sequences
(SINEs). Both types appear to be
In Human DNA, at Least 30% of the
retroposons, ie, they arose from movement from one
Genome Consists of Repetitive Sequences
location to another (transposition) through an RNA
Repetitive-sequence DNA can be broadly classified as
intermediate by the action of reverse transcriptase that
moderately repetitive or as highly repetitive. The highly
transcribes an RNA template into DNA. Mammalian
repetitive sequences consist of 5-500 base pair lengths
genomes contain 20-50 thousand copies of the 6-7 kb
repeated many times in tandem. These sequences are
LINEs. These represent species-specific families of re-
usually clustered in centromeres and telomeres of the
peat elements. SINEs are shorter (70-300 bp), and
chromosome and are present in about 1-10 million
there may be more than 100,000 copies per genome.
copies per haploid genome. These sequences are tran-
Of the SINEs in the human genome, one family, the
scriptionally inactive and may play a structural role in
Alu family, is present in about 500,000 copies per hap-
the chromosome (see Chapter 40).
loid genome and accounts for at least 5-6% of the
The moderately repetitive sequences, which are de-
human genome. Members of the human Alu family
fined as being present in numbers of less than 106
and their closely related analogs in other animals are
copies per haploid genome, are not clustered but are in-
transcribed as integral components of hnRNA or as dis-
terspersed with unique sequences. In many cases, these
crete RNA molecules, including the well-studied 4.5S
long interspersed repeats are transcribed by RNA poly-
RNA and 7S RNA. These particular family members
merase II and contain caps indistinguishable from those
are highly conserved within a species as well as between
on mRNA.
mammalian species. Components of the short inter-
322
/
CHAPTER 36
spersed repeats, including the members of the Alu fam-
with a gene in affected family members—and the lack
ily, may be mobile elements, capable of jumping into
of this association in unaffected members—may be the
and out of various sites within the genome (see below).
first clue about the genetic basis of a disease.
This can have disastrous results, as exemplified by the
Trinucleotide sequences that increase in number
insertion of Alu sequences into a gene, which, when so
(microsatellite instability) can cause disease. The unsta-
mutated, causes neurofibromatosis.
ble p(CGG)n repeat sequence is associated with the
fragile X syndrome. Other trinucleotide repeats that
undergo dynamic mutation (usually an increase) are
Microsatellite Repeat Sequences
associated with Huntington’s chorea (CAG), myotonic
One category of repeat sequences exists as both dis-
dystrophy (CTG), spinobulbar muscular atrophy (CAG),
persed and grouped tandem arrays. The sequences con-
and Kennedy’s disease (CAG).
sist of
2-6 bp repeated up to 50 times. These mi-
crosatellite sequences most commonly are found as
ONE PERCENT OF CELLULAR DNA
dinucleotide repeats of AC on one strand and TG on
IS IN MITOCHONDRIA
the opposite strand, but several other forms occur, in-
cluding CG, AT, and CA. The AC repeat sequences are
The majority of the peptides in mitochondria (about
estimated to occur at 50,000-100,000 locations in the
54 out of 67) are coded by nuclear genes. The rest are
genome. At any locus, the number of these repeats may
coded by genes found in mitochondrial (mt) DNA.
vary on the two chromosomes, thus providing heterozy-
Human mitochondria contain two to ten copies of a
gosity of the number of copies of a particular mi-
small circular double-stranded DNA molecule that
crosatellite number in an individual. This is a heritable
makes up approximately 1% of total cellular DNA.
trait, and, because of their number and the ease of de-
This mtDNA codes for mt ribosomal and transfer
tecting them using the polymerase chain reaction
RNAs and for 13 proteins that play key roles in the res-
(PCR) (Chapter 40), AC repeats are very useful in con-
piratory chain. The linearized structural map of the
structing genetic linkage maps. Most genes are associ-
human mitochondrial genes is shown in Figure 36-8.
ated with one or more microsatellite markers, so the rel-
Some of the features of mtDNA are shown in Table
ative position of genes on chromosomes can be
36-3.
assessed, as can the association of a gene with a disease.
An important feature of human mitochondrial
Using PCR, a large number of family members can be
mtDNA is that—because all mitochondria are con-
rapidly screened for a certain microsatellite polymor-
tributed by the ovum during zygote formation—it is
phism. The association of a specific polymorphism
transmitted by maternal nonmendelian inheritance.
kb
2
4
6
8
10
12
14
16
PH1
OH
OH
PH2
ATPase
8
6
12S
16S
ND1
ND2
CO1
CO2
CO3
ND4
ND5
CYT B
ND6
PL
OL
Figure 36-8. Maps of human mitochondrial genes. The maps represent the heavy (upper strand) and light
(lower map) strands of linearized mitochondrial (mt) DNA, showing the genes for the subunits of NADH-
coenzyme Q oxidoreductase (ND1 through ND6), cytochrome c oxidase (CO1 through CO3), cytochrome b
(CYT B), and ATP synthase (ATPase 8 and 6) and for the 12S and 16S ribosomal mt rRNAs. The transfer RNAs are de-
noted by small open boxes. The origin of heavy-strand (OH) and light-strand (OL) replication and the promoters
for the initiation of heavy-strand (PH1 and PH2) and light-strand (PL) transcription are indicated by arrows.
(Reproduced, with permission, from Moraes CT et al: Mitochondrial DNA deletions in progressive external ophthal-
moplegia and Kearns-Sayre syndrome. N Engl J Med 1989;320:1293.)
DNA ORGANIZATION, REPLICATION, & REPAIR
/
323
Table 36-3. Some major features of the structure
crossing over occurs as shown in Figure 36-9. This
and function of human mitochondrial DNA.1
usually results in an equal and reciprocal exchange of
genetic information between homologous chromo-
somes. If the homologous chromosomes possess differ-
• Is circular, double-stranded, and composed of heavy (H)
ent alleles of the same genes, the crossover may produce
and a light (L) chains or strands.
noticeable and heritable genetic linkage differences. In
• Contains 16,569 bp.
the rare case where the alignment of homologous chro-
• Encodes 13 protein subunits of the respiratory chain (of a
mosomes is not exact, the crossing over or recombina-
total of about 67):
Seven subunits of NADH dehydrogenase (complex I)
tion event may result in an unequal exchange of infor-
Cytochrome b of complex III
mation. One chromosome may receive less genetic
Three subunits of cytochrome oxidase (complex IV)
material and thus a deletion, while the other partner of
Two subunits of ATP synthase
the chromosome pair receives more genetic material
• Encodes large (16s) and small (12s) mt ribosomal RNAs.
and thus an insertion or duplication (Figure 36-9).
• Encodes 22 mt tRNA molecules.
Unequal crossing over does occur in humans, as evi-
• Genetic code differs slightly from the standard code:
denced by the existence of hemoglobins designated
UGA (standard stop codon) is read as Trp.
Lepore and anti-Lepore (Figure 36-10). The farther
AGA and AGG (standard codons for Arg) are read as stop
apart two sequences are on an individual chromosome,
codons.
the greater the likelihood of a crossover recombination
• Contains very few untranslated sequences.
• High mutation rate (five to ten times that of nuclear DNA).
• Comparisons of mtDNA sequences provide evidence about
evolutionary origins of primates and other species.
1Adapted from Harding AE: Neurological disease and mitochon-
drial genes. Trends Neurol Sci 1991;14:132.
Thus, in diseases resulting from mutations of mtDNA,
an affected mother would in theory pass the disease to
all of her children but only her daughters would trans-
mit the trait. However, in some cases, deletions in
mtDNA occur during oogenesis and thus are not inher-
ited from the mother. A number of diseases have now
been shown to be due to mutations of mtDNA. These
include a variety of myopathies, neurologic disorders,
and some cases of diabetes mellitus.
GENETIC MATERIAL CAN BE ALTERED
& REARRANGED
An alteration in the sequence of purine and pyrimidine
bases in a gene due to a change—a removal or an inser-
tion—of one or more bases may result in an altered
gene product. Such alteration in the genetic material re-
sults in a mutation whose consequences are discussed
in detail in Chapter 38.
Chromosomal Recombination Is One Way
of Rearranging Genetic Material
Genetic information can be exchanged between similar
or homologous chromosomes. The exchange or recom-
bination event occurs primarily during meiosis in
mammalian cells and requires alignment of homolo-
Figure 36-9.
The process of crossing-over between
gous metaphase chromosomes, an alignment that al-
homologous metaphase chromosomes to generate re-
most always occurs with great exactness. A process of
combinant chromosomes. See also Figure 36-12.
324
/
CHAPTER 36
Gγ
Aγ
δ
βδ
β
Figure 36-10. The process of unequal cross-
Anti-
Lepore
over in the region of the mammalian genome
that harbors the structural genes encoding he-
Gγ
Aγ
δ
β
moglobins and the generation of the unequal
recombinant products hemoglobin delta-beta
Lepore and beta-delta anti-Lepore. The exam-
Gγ
Aγ
ples given show the locations of the crossover
regions between amino acid residues. (Redrawn
and reproduced, with permission, from Clegg JB,
Gγ
Aγ
δβ
Weatherall DJ: β0 Thalassemia: Time for a reap-
Lepore
praisal? Lancet 1974;2:133.)
event. This is the basis for genetic mapping methods.
DNA polymerase, or reverse transcriptase—can be inte-
Unequal crossover affects tandem arrays of repeated
grated into chromosomes of the mammalian cell. The
DNAs whether they are related globin genes, as in Fig-
integration of the animal virus DNA into the animal
ure 36-10, or more abundant repetitive DNA. Un-
genome generally is not “site-specific” but does display
equal crossover through slippage in the pairing can re-
site preferences.
sult in expansion or contraction in the copy number of
the repeat family and may contribute to the expansion
Transposition Can Produce
and fixation of variant members throughout the array.
Processed Genes
In eukaryotic cells, small DNA elements that clearly are
Chromosomal Integration Occurs
not viruses are capable of transposing themselves in and
With Some Viruses
Some bacterial viruses (bacteriophages) are capable of
recombining with the DNA of a bacterial host in such a
way that the genetic information of the bacteriophage is
B
incorporated in a linear fashion into the genetic infor-
mation of the host. This integration, which is a form of
recombination, occurs by the mechanism illustrated in
Figure 36-11. The backbone of the circular bacterio-
A
C
phage genome is broken, as is that of the DNA mole-
cule of the host; the appropriate ends are resealed with
1
2
the proper polarity. The bacteriophage DNA is figura-
B
tively straightened out (“linearized”) as it is integrated
into the bacterial DNA molecule—frequently a closed
circle as well. The site at which the bacteriophage
A
C
genome integrates or recombines with the bacterial
genome is chosen by one of two mechanisms. If the
1
B
2
bacteriophage contains a DNA sequence homologous
to a sequence in the host DNA molecule, then a recom-
C
A
bination event analogous to that occurring between ho-
mologous chromosomes can occur. However, some
1
2
bacteriophages synthesize proteins that bind specific
sites on bacterial chromosomes to a nonhomologous
C
B
A
site characteristic of the bacteriophage DNA molecule.
Integration occurs at the site and is said to be “site-
1
2
specific.”
Many animal viruses, particularly the oncogenic
Figure 36-11.
The integration of a circular genome
viruses—either directly or, in the case of RNA viruses
from a virus (with genes A, B, and C) into the DNA mole-
such as HIV that causes AIDS, their DNA transcripts
cule of a host (with genes 1 and 2) and the consequent
generated by the action of the viral RNA-dependent
ordering of the genes.
DNA ORGANIZATION, REPLICATION, & REPAIR
/
325
out of the host genome in ways that affect the function
chromatids contains identical genetic information since
of neighboring DNA sequences. These mobile ele-
each is a product of the semiconservative replication of
ments, sometimes called “jumping DNA,” can carry
the original parent DNA molecule of that chromosome.
flanking regions of DNA and, therefore, profoundly af-
Crossing over occurs between these genetically identical
fect evolution. As mentioned above, the Alu family of
sister chromatids. Of course, these sister chromatid ex-
moderately repeated DNA sequences has structural
changes (Figure 36-12) have no genetic consequence as
characteristics similar to the termini of retroviruses,
long as the exchange is the result of an equal crossover.
which would account for the ability of the latter to
move into and out of the mammalian genome.
Immunoglobulin Genes Rearrange
Direct evidence for the transposition of other small
In mammalian cells, some interesting gene rearrange-
DNA elements into the human genome has been pro-
ments occur normally during development and differen-
vided by the discovery of “processed genes” for im-
munoglobulin molecules, α-globin molecules, and sev-
tiation. For example, in mice the V
L and CL genes for a
single immunoglobulin molecule (see Chapter 39) are
eral others. These processed genes consist of DNA
widely separated in the germ line DNA. In the DNA of a
sequences identical or nearly identical to those of the
differentiated immunoglobulin-producing (plasma) cell,
messenger RNA for the appropriate gene product. That
is, the
5′ nontranscribed region, the coding region
the same V
L and CL genes have been moved physically
closer together in the genome and into the same tran-
without intron representation, and the 3′ poly(A) tail
scription unit. However, even then, this rearrangement
are all present contiguously. This particular DNA se-
quence arrangement must have resulted from the re-
of DNA during differentiation does not bring the V
L and
verse transcription of an appropriately processed mes-
C
L genes into contiguity in the DNA. Instead, the DNA
senger RNA molecule from which the intron regions
had been removed and the poly(A) tail added. The only
recognized mechanism this reverse transcript could
have used to integrate into the genome would have
been a transposition event. In fact, these “processed
genes” have short terminal repeats at each end, as do
known transposed sequences in lower organisms. In the
absence of their transcription and thus genetic selection
for function, many of the processed genes have been
randomly altered through evolution so that they now
contain nonsense codons which preclude their ability to
encode a functional, intact protein (see Chapter 38).
Thus, they are referred to as “pseudogenes.”
Gene Conversion Produces
Rearrangements
Besides unequal crossover and transposition, a third
mechanism can effect rapid changes in the genetic ma-
terial. Similar sequences on homologous or nonhomol-
ogous chromosomes may occasionally pair up and elim-
inate any mismatched sequences between them. This
may lead to the accidental fixation of one variant or an-
other throughout a family of repeated sequences and
thereby homogenize the sequences of the members of
repetitive DNA families. This latter process is referred
to as gene conversion.
Figure 36-12.
Sister chromatid exchanges between
Sister Chromatids Exchange
human chromosomes. These are detectable by Giemsa
In diploid eukaryotic organisms such as humans, after
staining of the chromosomes of cells replicated for two
cells progress through the S phase they contain a
cycles in the presence of bromodeoxyuridine. The ar-
tetraploid content of DNA. This is in the form of sister
rows indicate some regions of exchange. (Courtesy of
chromatids of chromosome pairs. Each of these sister
S Wolff and J Bodycote.)
326
/
CHAPTER 36
contains an interspersed or interruption sequence of
Table 36-4. Steps involved in DNA replication
about 1200 base pairs at or near the junction of the V
in eukaryotes.
and C regions. The interspersed sequence is transcribed
genes, and the inter-
into RNA along with the V
L and CL
1. Identification of the origins of replication.
spersed information is removed from the RNA during its
2. Unwinding (denaturation) of dsDNA to provide an ssDNA
nuclear processing (Chapters 37 and 39).
template.
3. Formation of the replication fork.
DNA SYNTHESIS & REPLICATION
4. Initiation of DNA synthesis and elongation.
5. Formation of replication bubbles with ligation of the newly
ARE RIGIDLY CONTROLLED
synthesized DNA segments.
The primary function of DNA replication is under-
6. Reconstitution of chromatin structure.
stood to be the provision of progeny with the genetic
information possessed by the parent. Thus, the replica-
tion of DNA must be complete and carried out in such
a series of direct repeat DNA sequences. In bacterio-
a way as to maintain genetic stability within the organ-
phage λ, the oriλ is bound by the λ-encoded O protein
ism and the species. The process of DNA replication is
to four adjacent sites. In E coli, the oriC is bound by
complex and involves many cellular functions and sev-
the protein dnaA. In both cases, a complex is formed
eral verification procedures to ensure fidelity in replica-
consisting of 150-250 bp of DNA and multimers of
tion. About 30 proteins are involved in the replication
the DNA-binding protein. This leads to the local de-
of the E coli chromosome, and this process is almost
naturation and unwinding of an adjacent A+T-rich re-
certainly more complex in eukaryotic organisms. The
gion of DNA. Functionally similar autonomously
first enzymologic observations on DNA replication
replicating sequences (ARS) have been identified in
were made in E coli by Kornberg, who described in that
yeast cells. The ARS contains a somewhat degenerate
organism the existence of an enzyme now called DNA
11-bp sequence called the origin replication element
polymerase I. This enzyme has multiple catalytic activi-
(ORE). The ORE binds a set of proteins, analogous to
ties, a complex structure, and a requirement for the
the dnaA protein of E coli, which is collectively called
triphosphates of the four deoxyribonucleosides of ade-
the origin recognition complex (ORC). The ORE is
nine, guanine, cytosine, and thymine. The polymeriza-
located adjacent to an approximately 80-bp A+T-rich
tion reaction catalyzed by DNA polymerase I of E coli
sequence that is easy to unwind. This is called the DNA
has served as a prototype for all DNA polymerases of
unwinding element (DUE). The DUE is the origin of
both prokaryotes and eukaryotes, even though it is now
replication in yeast.
recognized that the major role of this polymerase is to
Consensus sequences similar to ori or ARS in struc-
complete replication on the lagging strand.
ture or function have not been precisely defined in
In all cells, replication can occur only from a single-
mammalian cells, though several of the proteins that
stranded DNA (ssDNA) template. Mechanisms must
participate in ori recognition and function have been
exist to target the site of initiation of replication and to
identified and appear quite similar to their yeast coun-
unwind the double-stranded DNA (dsDNA) in that re-
terparts in both amino acid sequence and function.
gion. The replication complex must then form. After
replication is complete in an area, the parent and daughter
Unwinding of DNA
strands must re-form dsDNA. In eukaryotic cells, an ad-
ditional step must occur. The dsDNA must precisely re-
The interaction of proteins with ori defines the start site
form the chromatin structure, including nucleosomes,
of replication and provides a short region of ssDNA es-
that existed prior to the onset of replication. Although this
sential for initiation of synthesis of the nascent DNA
entire process is not well understood in eukaryotic cells,
strand. This process requires the formation of a number
replication has been quite precisely described in prokary-
of protein-protein and protein-DNA interactions. A
otic cells, and the general principles are thought to be the
critical step is provided by a DNA helicase that allows
same in both. The major steps are listed in Table 36-4, il-
for processive unwinding of DNA. In uninfected E coli,
lustrated in Figure 36-13, and discussed, in sequence,
this function is provided by a complex of dnaB helicase
below. A number of proteins, most with specific enzy-
and the dnaC protein. Single-stranded DNA-binding
matic action, are involved in this process (Table 36-5).
proteins
(SSBs) stabilize this complex. In λ phage-
infected E coli, the phage protein P binds to dnaB and
the P/dnaB complex binds to oriλ by interacting with
The Origin of Replication
the O protein. dnaB is not an active helicase when in the
At the origin of replication (ori), there is an associa-
P/dnaB/O complex. Three E coli heat shock proteins
tion of sequence-specific dsDNA-binding proteins with
(dnaK, dnaJ, and GrpE) cooperate to remove the P
DNA ORGANIZATION, REPLICATION, & REPAIR
/
327
Ori
A + T region
A + T - rich
Ori-binding
Denaturation
region
protein
Binding
(
)
of SSB ( )
Binding of
factors,
formation of
3′
replication fork,
5′
initiation of
replication
Leading
strand
Polymerase
Helicase
3′
Primase
5′
SSB
5′
3′
3′
Lagging
= Ori-binding protein
strand
5′
= Polymerase
= Nascent DNA
= RNA primer
Replication fork
= Helicase
= Primase
= SSB
Figure 36-13. Steps involved in DNA replication. This figure describes DNA replication in an E coli cell, but the
general steps are similar in eukaryotes. A specific interaction of a protein (the O protein) to the origin of replication
(ori) results in local unwinding of DNA at an adjacent A+T-rich region. The DNA in this area is maintained in the
single-strand conformation (ssDNA) by single-strand-binding proteins (SSBs). This allows a variety of proteins, includ-
ing helicase, primase, and DNA polymerase, to bind and to initiate DNA synthesis. The replication fork proceeds as
DNA synthesis occurs continuously (long arrow) on the leading strand and discontinuously (short arrows) on the lag-
ging strand. The nascent DNA is always synthesized in the 5′ to 3′ direction, as DNA polymerases can add a nucleotide
only to the 3′ end of a DNA strand.
protein and activate the dnaB helicase. In cooperation
(2) a primase initiates synthesis of an RNA molecule
with SSB, this leads to DNA unwinding and active
that is essential for priming DNA synthesis; (3) the
replication. In this way, the replication of the λ phage is
DNA polymerase initiates nascent, daughter strand
accomplished at the expense of replication of the host
synthesis; and (4) SSBs bind to ssDNA and prevent
E coli cell.
premature reannealing of ssDNA to dsDNA. These re-
actions are illustrated in Figure 36-13.
The polymerase III holoenzyme (the dnaE gene
Formation of the Replication Fork
product in E coli) binds to template DNA as part of a
A replication fork consists of four components that
multiprotein complex that consists of several polym-
form in the following sequence: (1) the DNA helicase
erase accessory factors (β, γ, δ, δ′, and τ). DNA polym-
unwinds a short segment of the parental duplex DNA;
erases only synthesize DNA in the 5′ to 3′ direction,
328
/
CHAPTER 36
Table 36-5. Classes of proteins involved
replication fork. Of all polymerases, it catalyzes the
in replication.
highest rate of chain elongation and is the most proces-
sive. It is capable of polymerizing 0.5 Mb of DNA dur-
ing one cycle on the leading strand. Pol III is a large
Protein
Function
(> 1 MDa), ten-subunit protein complex in E coli. The
DNA polymerases
Deoxynucleotide polymerization
two identical β subunits of pol III encircle the DNA
template in a sliding “clamp,” which accounts for the
Helicases
Processive unwinding of DNA
stability of the complex and for the high degree of pro-
Topoisomerases
Relieve torsional strain that results
cessivity the enzyme exhibits.
from helicase-induced unwinding
Polymerase II (pol II) is mostly involved in proof-
DNA primase
Initiates synthesis of RNA primers
reading and DNA repair. Polymerase I (pol I) com-
pletes chain synthesis between Okazaki fragments on
Single-strand binding
Prevent premature reannealing of
the lagging strand. Eukaryotic cells have counterparts
proteins
dsDNA
for each of these enzymes plus some additional ones. A
DNA ligase
Seals the single strand nick between
comparison is shown in Table 36-6.
the nascent chain and Okazaki frag-
In mammalian cells, the polymerase is capable of
ments on lagging strand
polymerizing about 100 nucleotides per second, a rate
at least tenfold slower than the rate of polymerization of
deoxynucleotides by the bacterial DNA polymerase
complex. This reduced rate may result from interfer-
and only one of the several different types of polym-
ence by nucleosomes. It is not known how the replica-
erases is involved at the replication fork. Because the
tion complex negotiates nucleosomes.
DNA strands are antiparallel (Chapter 35), the polym-
erase functions asymmetrically. On the leading (for-
ward) strand, the DNA is synthesized continuously.
Initiation & Elongation of DNA Synthesis
On the lagging (retrograde) strand, the DNA is syn-
The initiation of DNA synthesis (Figure 36-14) re-
thesized in short (1-5 kb; see Figure 36-16) fragments,
quires priming by a short length of RNA, about
the so-called Okazaki fragments. Several Okazaki frag-
10-200 nucleotides long. This priming process involves
ments (up to 250) must be synthesized, in sequence, for
the nucleophilic attack by the 3′-hydroxyl group of the
each replication fork. To ensure that this happens, the
RNA primer on the α phosphate of the first entering
helicase acts on the lagging strand to unwind dsDNA in
deoxynucleoside triphosphate
(N in Figure
36-14)
a 5′ to 3′ direction. The helicase associates with the pri-
with the splitting off of pyrophosphate. The 3′-hy-
mase to afford the latter proper access to the template.
droxyl group of the recently attached deoxyribonu-
This allows the RNA primer to be made and, in turn,
cleoside monophosphate is then free to carry out a
the polymerase to begin replicating the DNA. This is
nucleophilic attack on the next entering deoxyribonu-
an important reaction sequence since DNA poly-
cleoside triphosphate (N + 1 in Figure 36-14), again at
merases cannot initiate DNA synthesis de novo. The
its α phosphate moiety, with the splitting off of py-
mobile complex between helicase and primase has been
rophosphate. Of course, selection of the proper de-
called a primosome. As the synthesis of an Okazaki
oxyribonucleotide whose terminal 3′-hydroxyl group is
fragment is completed and the polymerase is released, a
to be attacked is dependent upon proper base pairing
new primer has been synthesized. The same polymerase
molecule remains associated with the replication fork
and proceeds to synthesize the next Okazaki fragment.
Table 36-6. A comparison of prokaryotic and
eukaryotic DNA polymerases.
The DNA Polymerase Complex
A number of different DNA polymerase molecules en-
E coli
Mammalian
Function
gage in DNA replication. These share three important
properties: (1) chain elongation, (2) processivity, and
I
α
Gap filling and synthesis of lagging
(3) proofreading. Chain elongation accounts for the
strand
rate (in nucleotides per second) at which polymeriza-
II
ε
DNA proofreading and repair
tion occurs. Processivity is an expression of the number
β
DNA repair
of nucleotides added to the nascent chain before the
polymerase disengages from the template. The proof-
γ
Mitochondrial DNA synthesis
reading function identifies copying errors and corrects
III
δ
Processive, leading strand synthesis
them. In E coli, polymerase III (pol III) functions at the
X1
C
O
H H
H
H
X2
HO
C
O
RNA primer
O
P
O
H H
H
H
X3
HO
C
O
O
P
O
H H
H
H
X4
HO
C
O
O
P
O
H H
H
H
OH
N
OH
C
O
O
O
P
First entering dNTP
O
O
O-
H
H
P
H
H
O-
O
O-
P
OH
H
O
O-
X4
C
O
O
P
O
H H
H
H
N
C
HO
O
O
P
O
H
H
H
H
H
N+1
OH
C
O
O
O
P
Second entering dNTP
O
O
O-
H
H
P
H
H
O-
O O-
P
OH
H
O O-
Figure 36-14.
The initiation of DNA synthesis upon a primer of RNA and the sub-
sequent attachment of the second deoxyribonucleoside triphosphate.
329
330
/
CHAPTER 36
with the other strand of the DNA molecule according
of newly synthesized DNA by enzymes referred to as
to the rules proposed originally by Watson and Crick
DNA ligases.
(Figure 36-15). When an adenine deoxyribonucleoside
monophosphoryl moiety is in the template position, a
Replication Exhibits Polarity
thymidine triphosphate will enter and its α phosphate
will be attacked by the
3′-hydroxyl group of the
As has already been noted, DNA molecules are double-
deoxyribonucleoside monophosphoryl most recently
stranded and the two strands are antiparallel, ie, run-
added to the polymer. By this stepwise process, the
ning in opposite directions. The replication of DNA in
template dictates which deoxyribonucleoside triphos-
prokaryotes and eukaryotes occurs on both strands si-
phate is complementary and by hydrogen bonding
multaneously. However, an enzyme capable of poly-
holds it in place while the 3′-hydroxyl group of the
merizing DNA in the 3′ to 5′ direction does not exist in
growing strand attacks and incorporates the new nu-
any organism, so that both of the newly replicated
cleotide into the polymer. These segments of DNA
DNA strands cannot grow in the same direction simul-
attached to an RNA initiator component are the
taneously. Nevertheless, the same enzyme does replicate
Okazaki fragments (Figure 36-16). In mammals, after
both strands at the same time. The single enzyme repli-
many Okazaki fragments are generated, the replication
cates one strand
(“leading strand”) in a continuous
complex begins to remove the RNA primers, to fill in
manner in the 5′ to 3′ direction, with the same overall
the gaps left by their removal with the proper base-
forward direction. It replicates the other strand (“lag-
paired deoxynucleotide, and then to seal the fragments
ging strand”) discontinuously while polymerizing the
RNA primer
DNA template
Growing DNA polymer
Entering TTP
Figure 36-15. The RNA-primed synthesis of DNA demonstrating the template function of the
complementary strand of parental DNA.
DNA ORGANIZATION, REPLICATION, & REPAIR
/
331
DNA template
3′
5′
5′
3′
Newly synthesized
RNA
10 bp
10 bp
DNA strand
primer
100 bp
Okazaki fragments
Figure 36-16. The discontinuous polymerization of deoxyribonucleotides on the lagging
strand; formation of Okazaki fragments during lagging strand DNA synthesis is illustrated.
Okazaki fragments are 100-250 nt long in eukaryotes, 1000-2000 bp in prokaryotes.
nucleotides in short spurts of
150-250 nucleotides,
hours! Metazoan organisms get around this problem
again in the 5′ to 3′ direction, but at the same time it
using two strategies. First, replication is bidirectional.
faces toward the back end of the preceding RNA
Second, replication proceeds from multiple origins in
primer rather than toward the unreplicated portion.
each chromosome (a total of as many as 100 in hu-
This process of semidiscontinuous DNA synthesis is
mans). Thus, replication occurs in both directions
shown diagrammatically in Figures 36-13 and 36-16.
along all of the chromosomes, and both strands are
In the mammalian nuclear genome, most of the
replicated simultaneously. This replication process gen-
RNA primers are eventually removed as part of the
erates “replication bubbles” (Figure 36-17).
replication process, whereas after replication of the mi-
The multiple sites that serve as origins for DNA
tochondrial genome the small piece of RNA remains as
replication in eukaryotes are poorly defined except in a
an integral part of the closed circular DNA structure.
few animal viruses and in yeast. However, it is clear that
initiation is regulated both spatially and temporally,
since clusters of adjacent sites initiate replication syn-
Formation of Replication Bubbles
chronously. There are suggestions that functional do-
Replication proceeds from a single ori in the circular
mains of chromatin replicate as intact units, implying
bacterial chromosome, composed of roughly 6 × 106 bp
that the origins of replication are specifically located
of DNA. This process is completed in about 30 min-
with respect to transcription units.
utes, a replication rate of 3 × 105 bp/min. The entire
During the replication of DNA, there must be a sep-
mammalian genome replicates in approximately
9
aration of the two strands to allow each to serve as a
hours, the average period required for formation of a
template by hydrogen bonding its nucleotide bases to
tetraploid genome from a diploid genome in a replicat-
the incoming deoxynucleoside triphosphate. The separa-
ing cell. If a mammalian genome (3 × 109 bp) repli-
tion of the DNA double helix is promoted by SSBs, spe-
cated at the same rate as bacteria (ie, 3 × 105 bp/min)
cific protein molecules that stabilize the single-stranded
from but a single ori, replication would take over 150
structure as the replication fork progresses. These stabi-
Origin of replication
“Replication bubble”
3′
5′
5′
3′
Unwinding proteins
at replication forks
Directions
of replication
Figure 36-17. The generation of “replication bubbles” during the process of DNA synthesis. The bidirectional
replication and the proposed positions of unwinding proteins at the replication forks are depicted.
332
/
CHAPTER 36
lizing proteins bind cooperatively and stoichiometrically
without requiring energy input, because of the forma-
to the single strands without interfering with the abili-
tion of a high-energy covalent bond between the nicked
ties of the nucleotides to serve as templates
(Figure
phosphodiester backbone and the nicking-sealing en-
36-13). In addition to separating the two strands of the
zyme. The nicking-resealing enzymes are called DNA
double helix, there must be an unwinding of the mole-
topoisomerases. This process is depicted diagrammati-
cule (once every 10 nucleotide pairs) to allow strand sep-
cally in Figure
36-18 and there compared with the
aration. This must happen in segments, given the time
ATP-dependent resealing carried out by the DNA li-
during which DNA replication occurs. There are multi-
gases. Topoisomerases are also capable of unwinding su-
ple “swivels” interspersed in the DNA molecules of all
percoiled DNA. Supercoiled DNA is a higher-ordered
organisms. The swivel function is provided by specific
structure occurring in circular DNA molecules wrapped
enzymes that introduce “nicks” in one strand of the
around a core, as depicted in Figure 36-19.
unwinding double helix, thereby allowing the unwind-
There exists in one species of animal viruses (retro-
ing process to proceed. The nicks are quickly resealed
viruses) a class of enzymes capable of synthesizing a sin-
Step 1
DNA topoisomerase I = E
DNA ligase = E
E + ATP
E
P R A
(AMP-Enzyme)
5′
5′
P -E
P
Enzyme (E) -generated
Single-strand nick
single-strand nick
present
O 3′
O
3′
H
H
3′
5′
3′
5′
E P R A
Step 2
E
5′
5′
P
-E
P
P R A
Formation of high-
O
O
energy bond
H
H
Step 3
E
P R A
(AMP)
Nick repaired
Nick repaired
Figure 36-18. Comparison of two types of nick-sealing reactions on DNA. The
series of reactions at left is catalyzed by DNA topoisomerase I, that at right by
DNA ligase; P = phosphate, R = ribose, A = ademine. (Slightly modified and repro-
duced, with permission, from Lehninger AL: Biochemistry, 2nd ed. Worth, 1975.)
DNA ORGANIZATION, REPLICATION, & REPAIR
/
333
preexisting and newly assembled histone octamers are
randomly distributed to each arm of the replication
fork.
DNA Synthesis Occurs During
the S Phase of the Cell Cycle
In animal cells, including human cells, the replication
of the DNA genome occurs only at a specified time
during the life span of the cell. This period is referred to
as the synthetic or S phase. This is usually temporally
separated from the mitotic phase by nonsynthetic peri-
ods referred to as gap 1 (G1) and gap 2 (G2), occurring
before and after the S phase, respectively
(Figure
36-20). Among other things, the cell prepares for DNA
synthesis in G1 and for mitosis in G2. The cell regu-
lates its DNA synthesis grossly by allowing it to occur
only at specific times and mostly in cells preparing to
divide by a mitotic process.
It appears that all eukaryotic cells have gene prod-
ucts that govern the transition from one phase of the
cell cycle to another. The cyclins are a family of pro-
teins whose concentration increases and decreases
throughout the cell cycle—thus their name. The cyclins
Figure 36-19.
Supercoiling of DNA. A left-handed
turn on, at the appropriate time, different cyclin-
toroidal (solenoidal) supercoil, at left, will convert to a
dependent protein kinases (CDKs) that phosphory-
right-handed interwound supercoil, at right, when the
late substrates essential for progression through the cell
cylindric core is removed. Such a transition is analogous
cycle (Figure 36-21). For example, cyclin D levels rise
to that which occurs when nucleosomes are disrupted
in late G1 phase and allow progression beyond the start
by the high salt extraction of histones from chromatin.
(yeast) or restriction point (mammals), the point be-
yond which cells irrevocably proceed into the S or
DNA synthesis phase.
The D cyclins activate CDK4 and CDK6. These
gle-stranded and then a double-stranded DNA mole-
two kinases are also synthesized during G1 in cells un-
cule from a single-stranded RNA template. This poly-
dergoing active division. The D cyclins and CDK4 and
merase, RNA-dependent DNA polymerase, or “reverse
CDK6 are nuclear proteins that assemble as a complex
transcriptase,” first synthesizes a DNA-RNA hybrid
in late G1 phase. The complex is an active serine-
molecule utilizing the RNA genome as a template. A
threonine protein kinase. One substrate for this kinase
specific nuclease, RNase H, degrades the RNA strand,
is the retinoblastoma (Rb) protein. Rb is a cell cycle
and the remaining DNA strand in turn serves as a tem-
regulator because it binds to and inactivates a transcrip-
plate to form a double-stranded DNA molecule con-
tion factor (E2F) necessary for the transcription of cer-
taining the information originally present in the RNA
tain genes (histone genes, DNA replication proteins,
genome of the animal virus.
etc) needed for progression from G1 to S phase. The
phosphorylation of Rb by CDK4 or CDK6 results in
the release of E2F from Rb-mediated transcription re-
Reconstitution of Chromatin Structure
pression—thus, gene activation ensues and cell cycle
There is evidence that nuclear organization and chro-
progression takes place.
matin structure are involved in determining the regu-
Other cyclins and CDKs are involved in different
lation and initiation of DNA synthesis. As noted
aspects of cell cycle progression (Table 36-7). Cyclin E
above, the rate of polymerization in eukaryotic cells,
and CDK2 form a complex in late G1. Cyclin E is
which have chromatin and nucleosomes, is tenfold
rapidly degraded, and the released CDK2 then forms a
slower than that in prokaryotic cells, which have
complex with cyclin A. This sequence is necessary for
naked DNA. It is also clear that chromatin structure
the initiation of DNA synthesis in S phase. A complex
must be re-formed after replication. Newly replicated
between cyclin B and CDK1 is rate-limiting for the
DNA is rapidly assembled into nucleosomes, and the
G2/M transition in eukaryotic cells.
334
/
CHAPTER 36
Improper spindle
detected
Figure 36-20. Mammalian cell cycle and cell
M
cycle checkpoints. DNA, chromosome, and chro-
mosome segregation integrity is continuously
G2
monitored throughout the cell cycle. If DNA dam-
Damaged DNA
Gl
age is detected in either the G1 or the G2 phase of
detected
the cell cycle, if the genome is incompletely repli-
S
cated, or if normal chromosome segregation ma-
Damaged DNA
detected
chinery is incomplete (ie, a defective spindle), cells
will not progress through the phase of the cycle in
Incomplete
which defects are detected. In some cases, if the
replication
damage cannot be repaired, such cells undergo
detected
programmed cell death (apoptosis).
Many of the cancer-causing viruses (oncoviruses)
duced by several DNA viruses target the Rb transcrip-
and cancer-inducing genes (oncogenes) are capable of
tion repressor for inactivation, inducing cell division in-
alleviating or disrupting the apparent restriction that
appropriately.
normally controls the entry of mammalian cells from
During the S phase, mammalian cells contain
G1 into the S phase. From the foregoing, one might
greater quantities of DNA polymerase than during the
have surmised that excessive production of a cyclin—or
nonsynthetic phases of the cell cycle. Furthermore,
production at an inappropriate time—might result in
those enzymes responsible for formation of the sub-
abnormal or unrestrained cell division. In this context it
strates for DNA synthesis—ie, deoxyribonucleoside
is noteworthy that the bcl oncogene associated with B
triphosphates—are also increased in activity, and their
cell lymphoma appears to be the cyclin D1 gene. Simi-
activity will diminish following the synthetic phase
larly, the oncoproteins (or transforming proteins) pro-
until the reappearance of the signal for renewed DNA
Cdk1-cyclin B
Cdk1-cyclin A
G2
M
Cdk4-cyclin D
Cdk6-cyclin D
G1
S
Restriction
point
Cdk2-cyclin A
Figure 36-21. Schematic illustration of the
points during the mammalian cell cycle during
which the indicated cyclins and cyclin-dependent
kinases are activated. The thickness of the various
Cdk2-cyclin E
colored lines is indicative of the extent of activity.
DNA ORGANIZATION, REPLICATION, & REPAIR
/
335
Table 36-7. Cyclins and cyclin-dependent kinases
ring more frequently than once every 108-1010 base
involved in cell cycle progression.
pairs of DNA synthesized. The mechanisms responsible
for this monitoring mechanism in E coli include the 3′
to 5′ exonuclease activities of one of the subunits of the
Cyclin
Kinase
Function
pol III complex and of the pol I molecule. The analo-
D
CDK4, CDK6
Progression past restriction point at
gous mammalian enzymes (δ and α) do not seem to
G1/S boundary
possess such a nuclease proofreading function. Other
E, A
CDK2
Initiation of DNA synthesis in early S
enzymes provide this repair function.
phase
Replication errors, even with a very efficient repair
system, lead to the accumulation of mutations. A
B
CDK1
Transition from G2 to M
human has 1014 nucleated cells each with 3 × 109 base
pairs of DNA. If about 1016 cell divisions occur in a
lifetime and 10−10 mutations per base pair per cell gen-
eration escape repair, there may eventually be as many
synthesis. During the S phase, the nuclear DNA is
as one mutation per 106 bp in the genome. Fortunately,
completely replicated once and only once. It seems
most of these will probably occur in DNA that does not
that once chromatin has been replicated, it is marked so
encode proteins or will not affect the function of en-
as to prevent its further replication until it again passes
coded proteins and so are of no consequence. In addi-
through mitosis. The molecular mechanisms for this
tion, spontaneous and chemically induced damage to
phenomenon have yet to be elucidated.
DNA must be repaired.
In general, a given pair of chromosomes will repli-
Damage to DNA by environmental, physical, and
cate simultaneously and within a fixed portion of the S
chemical agents may be classified into four types
phase upon every replication. On a chromosome, clus-
(Table 36-8). Abnormal regions of DNA, either from
ters of replication units replicate coordinately. The na-
copying errors or DNA damage, are replaced by four
ture of the signals that regulate DNA synthesis at these
mechanisms: (1) mismatch repair, (2) base excision-
levels is unknown, but the regulation does appear to be
repair, (3) nucleotide excision-repair, and (4) double-
an intrinsic property of each individual chromosome.
strand break repair (Table 36-9). These mechanisms
exploit the redundancy of information inherent in the
Enzymes Repair Damaged DNA
double helical DNA structure. The defective region in
one strand can be returned to its original form by rely-
The maintenance of the integrity of the information in
ing on the complementary information stored in the
DNA molecules is of utmost importance to the survival
unaffected strand.
of a particular organism as well as to survival of the
species. Thus, it can be concluded that surviving species
have evolved mechanisms for repairing DNA damage
occurring as a result of either replication errors or envi-
ronmental insults.
Table 36-8. Types of damage to DNA.
As described in Chapter 35, the major responsibility
for the fidelity of replication resides in the specific pair-
I.
Single-base alteration
ing of nucleotide bases. Proper pairing is dependent
A. Depurination
upon the presence of the favored tautomers of the
B. Deamination of cytosine to uracil
purine and pyrimidine nucleotides, but the equilibrium
C. Deamination of adenine to hypoxanthine
whereby one tautomer is more stable than another is
D. Alkylation of base
only about 104 or 105 in favor of that with the greater
E. Insertion or deletion of nucleotide
stability. Although this is not favorable enough to en-
F. Base-analog incorporation
sure the high fidelity that is necessary, favoring of the
II.
Two-base alteration
preferred tautomers—and thus of the proper base pair-
A. UV light-induced thymine-thymine (pyrimidine) dimer
ing—could be ensured by monitoring the base pairing
B. Bifunctional alkylating agent cross-linkage
twice. Such double monitoring does appear to occur in
III. Chain breaks
both bacterial and mammalian systems: once at the
A. Ionizing radiation
time of insertion of the deoxyribonucleoside triphos-
B. Radioactive disintegration of backbone element
phates, and later by a follow-up energy-requiring mech-
C. Oxidative free radical formation
IV. Cross-linkage
anism that removes all improper bases which may occur
A. Between bases in same or opposite strands
in the newly formed strand. This “proofreading” pre-
B. Between DNA and protein molecules (eg, histones)
vents tautomer-induced misincorporation from occur-
336
/
CHAPTER 36
Table 36-9. Mechanism of DNA repair
CH3
CH3
3′
5′
Mechanism
Problem
Solution
5′
3′
Mismatch
Copying errors (single
Methyl-directed
SINGLE-SITE STRAND CUT
repair
base or two- to five-
strand cutting, exo-
BY GATC ENDONUCLEASE
base unpaired loops)
nuclease digestion,
CH3
CH3
and replacement
3′
5′
Base
Spontaneous, chem-
Base removal by N-
5′
3′
excision-
ical, or radiation dam-
glycosylase, abasic
repair
age to a single base
sugar removal, re-
DEFECT REMOVED
placement
BY EXONUCLEASE
Nucleotide
Spontaneous, chem-
Removal of an ap-
CH3
CH3
excision-
ical, or radiation dam-
proximately 30-
3′
5′
repair
age to a DNA segment
nucleotide oligomer
5′
3′
and replacement
DEFECT REPAIRED
Double-
Ionizing radiation,
Synapsis, unwind-
BY POLYMERASE
strand
chemotherapy,
ing, alignment,
break repair
oxidative free
ligation
CH3
CH3
radicals
3′
5′
RELIGATED
BY LIGASE
Mismatch Repair
CH3
CH3
Mismatch repair corrects errors made when DNA is
3′
5′
copied. For example, a C could be inserted opposite an
A, or the polymerase could slip or stutter and insert two
5′
3′
to five extra unpaired bases. Specific proteins scan the
Figure 36-22.
Mismatch repair of DNA. This mecha-
newly synthesized DNA, using adenine methylation
nism corrects a single mismatch base pair (eg, C to A
within a GATC sequence as the point of reference (Fig-
rather than T to A) or a short region of unpaired DNA.
ure 36-22). The template strand is methylated, and the
The defective region is recognized by an endonuclease
newly synthesized strand is not. This difference allows
that makes a single-strand cut at an adjacent methy-
the repair enzymes to identify the strand that contains
lated GATC sequence. The DNA strand is removed
the errant nucleotide which requires replacement. If a
mismatch or small loop is found, a GATC endonucle-
through the mutation, replaced, and religated.
ase cuts the strand bearing the mutation at a site corre-
sponding to the GATC. An exonuclease then digests
this strand from the GATC through the mutation, thus
E coli MutS protein that is involved in mismatch repair
removing the faulty DNA. This can occur from either
(see above). Mutations of hMSH2 account for 50-60%
end if the defect is bracketed by two GATC sites. This
of HNPCC cases. Another gene, hMLH1, is associated
defect is then filled in by normal cellular enzymes ac-
with most of the other cases. hMLH1 is the human ana-
cording to base pairing rules. In E coli, three proteins
log of the bacterial mismatch repair gene MutL. How
(Mut S, Mut C, and Mut H) are required for recogni-
does faulty mismatch repair result in colon cancer? The
tion of the mutation and nicking of the strand. Other
human genes were localized because microsatellite in-
cellular enzymes, including ligase, polymerase, and
stability was detected. That is, the cancer cells had a mi-
SSBs, remove and replace the strand. The process is
crosatellite of a length different from that found in the
somewhat more complicated in mammalian cells, as
normal cells of the individual. It appears that the af-
about six proteins are involved in the first steps.
fected cells, which harbor a mutated hMSH2
or
Faulty mismatch repair has been linked to heredi-
hMLH1 mismatch repair enzyme, are unable to remove
tary nonpolyposis colon cancer (HNPCC), one of the
small loops of unpaired DNA, and the microsatellite
most common inherited cancers. Genetic studies linked
thus increases in size. Ultimately, microsatellite DNA
HNPCC in some families to a region of chromosome
expansion must affect either the expression or the func-
2. The gene located, designated hMSH2, was sub-
tion of a protein critical in surveillance of the cell cycle
sequently shown to encode the human analog of the
in these colon cells.
DNA ORGANIZATION, REPLICATION, & REPAIR
/
337
Base Excision-Repair
3′
5′
A
T
C G G C T C A T C C G A T
The depurination of DNA, which happens sponta-
neously owing to the thermal lability of the purine N-
T
A G C C G A G T A G G C T A
5′
3′
glycosidic bond, occurs at a rate of 5000-10,000/cell/d
Heat energy
at 37 °C. Specific enzymes recognize a depurinated site
and replace the appropriate purine directly, without in-
A
T
C G G C T
U A T C C G A T
terruption of the phosphodiester backbone.
Cytosine, adenine, and guanine bases in DNA spon-
T
A G C C G A
G T A G G C T A
taneously form uracil, hypoxanthine, or xanthine, re-
spectively. Since none of these normally exist in DNA,
U
URACIL DNA GLYCOSYLASE
it is not surprising that specific N-glycosylases can rec-
ognize these abnormal bases and remove the base itself
A
T
C G G C T
A
T C C G A T
from the DNA. This removal marks the site of the de-
fect and allows an apurinic or apyrimidinic endonu-
T
A G C C G A G T A G G C T A
clease to excise the abasic sugar. The proper base is
NUCLEASES
then replaced by a repair DNA polymerase, and a ligase
returns the DNA to its original state (Figure 36-23).
A
T
C G G C
T C C G A T
This series of events is called base excision-repair. By a
similar series of steps involving initially the recognition
T
A G C C G A G T A G G C T A
of the defect, alkylated bases and base analogs can be re-
moved from DNA and the DNA returned to its origi-
DNA POLYMERASE + DNA LIGASE
nal informational content. This mechanism is suitable
for replacement of a single base but is not effective at
A
T
C G G C T C A T C C G A T
replacing regions of damaged DNA.
T
A G C C G A G T A G G C T A
Nucleotide Excision-Repair
Figure 36-23. Base excision-repair of DNA. The en-
This mechanism is used to replace regions of damaged
zyme uracil DNA glycosylase removes the uracil created
DNA up to 30 bases in length. Common examples of
by spontaneous deamination of cytosine in the DNA. An
DNA damage include ultraviolet (UV) light, which in-
endonuclease cuts the backbone near the defect; then,
duces the formation of cyclobutane pyrimidine-pyrimi-
after an endonuclease removes a few bases, the defect
dine dimers, and smoking, which causes formation of
is filled in by the action of a repair polymerase and the
benzo[a]pyrene-guanine adducts. Ionizing radiation,
strand is rejoined by a ligase. (Courtesy of B Alberts.)
cancer chemotherapeutic agents, and a variety of chemi-
cals found in the environment cause base modification,
strand breaks, cross-linkage between bases on opposite
strands or between DNA and protein, and numerous
marked sensitivity to sunlight (ultraviolet) with subse-
other defects. These are repaired by a process called nu-
quent formation of multiple skin cancers and prema-
cleotide excision-repair
(Figure 36-24). This complex
ture death. The risk of developing skin cancer is in-
process, which involves more gene products than the two
creased 1000- to 2000-fold. The inherited defect seems
other types of repair, essentially involves the hydrolysis of
to involve the repair of damaged DNA, particularly
two phosphodiester bonds on the strand containing the
thymine dimers. Cells cultured from patients with xero-
defect. A special excision nuclease (exinuclease), consist-
derma pigmentosum exhibit low activity for the nu-
ing of at least three subunits in E coli and 16 polypep-
cleotide excision-repair process. Seven complementa-
tides in humans, accomplishes this task. In eukaryotic
tion groups have been identified using hybrid cell
cells the enzymes cut between the third to fifth phospho-
analyses, so at least seven gene products (XPA-XPG)
diester bond 3′ from the lesion, and on the 5′ side the cut
are involved. Two of these (XPA and XPC) are in-
is somewhere between the twenty-first and twenty-fifth
volved in recognition and excision. XPB and XPD are
bonds. Thus, a fragment of DNA 27-29 nucleotides
helicases and, interestingly, are subunits of the tran-
long is excised. After the strand is removed it is replaced,
scription factor TFIIH (see Chapter 37).
again by exact base pairing, through the action of yet an-
other polymerase (δ/ε in humans), and the ends are
Double-Strand Break Repair
joined to the existing strands by DNA ligase.
Xeroderma pigmentosum (XP) is an autosomal re-
The repair of double-strand breaks is part of the physio-
cessive genetic disease. The clinical syndrome includes
logic process of immunoglobulin gene rearrangement. It
338
/
CHAPTER 36
3′
5′
ase; and the gaps are filled and closed by DNA ligase.
This repair mechanism is illustrated in Figure 36-25.
5′
3′
RECOGNITION AND UNWINDING
Some Repair Enzymes Are Multifunctional
Somewhat surprising is the recent observation that
3′
5′
DNA repair proteins can serve other purposes. For ex-
ample, some repair enzymes are also found as compo-
5′
3′
nents of the large TFIIH complex that plays a central
role in gene transcription (Chapter 37). Another com-
OLIGONUCLEOTIDE EXCISION
ponent of TFIIH is involved in cell cycle regulation.
BY CUTTING AT TWO SITES
Thus, three critical cellular processes may be linked
3′
5′
through use of common proteins. There is also good
evidence that some repair enzymes are involved in gene
5′
3′
rearrangements that occur normally.
In patients with ataxia-telangiectasia, an autosomal
DEGRADATION OF MUTATED DNA
recessive disease in humans resulting in the development
RESYNTHESIS AND RELIGATION
of cerebellar ataxia and lymphoreticular neoplasms,
3′
5′
there appears to exist an increased sensitivity to damage
by x-ray. Patients with Fanconi’s anemia, an autosomal
5′
3′
recessive anemia characterized also by an increased fre-
Figure 36-24. Nucleotide excision-repair. This
quency of cancer and by chromosomal instability, prob-
mechanism is employed to correct larger defects in
ably have defective repair of cross-linking damage.
DNA and generally involves more proteins than either
mismatch or base excision-repair. After defect recogni-
tion (indicated by XXXX) and unwinding of the DNA en-
compassing the defect, an excision nuclease (exinucle-
Ku and DNA-PK bind
ase) cuts the DNA upstream and downstream of the
defective region. This gap is then filled in by a poly-
merase (δ/ε in humans) and religated.
Approximation
is also an important mechanism for repairing damaged
P
DNA, such as occurs as a result of ionizing radiation or
P
oxidative free radical generation. Some chemotherapeu-
Unwinding
tic agents destroy cells by causing ds breaks or prevent-
ing their repair.
Two proteins are initially involved in the nonho-
mologous rejoining of a ds break. Ku, a heterodimer of
70 kDa and 86 kDa subunits, binds to free DNA ends
Alignment and base pairing
and has latent ATP-dependent helicase activity. The
DNA-bound Ku heterodimer recruits a unique protein
kinase, DNA-dependent protein kinase (DNA-PK).
DNA-PK has a binding site for DNA free ends and an-
other for dsDNA just inside these ends. It therefore al-
Ligation
lows for the approximation of the two separated ends.
The free end DNA-Ku-DNA-PK complex activates the
kinase activity in the latter. DNA-PK reciprocally phos-
Figure 36-25. Double-strand break repair of DNA.
phorylates Ku and the other DNA-PK molecule, on the
The proteins Ku and DNA-dependent protein kinase
opposing strand, in trans. DNA-PK then dissociates
combine to approximate the two strands and unwind
from the DNA and Ku, resulting in activation of the
them. The aligned fragments form base pairs; the extra
Ku helicase. This results in unwinding of the two ends.
ends are removed, probably by a DNA-PK-associated
The unwound, approximated DNA forms base pairs;
endo- or exonuclease, and the gaps are filled in; and
the extra nucleotide tails are removed by an exonucle-
continuity is restored by ligation.
DNA ORGANIZATION, REPLICATION, & REPAIR
/
339
All three of these clinical syndromes are associated
•
Much of the DNA is associated with histone proteins
with an increased frequency of cancer. It is likely that
to form a structure called the nucleosome. Nucleo-
other human diseases resulting from disordered DNA
somes are composed of an octamer of histones and
repair capabilities will be found in the future.
150 bp of DNA.
•
Nucleosomes and higher-order structures formed
DNA & Chromosome Integrity Is
from them serve to compact the DNA.
Monitored Throughout the Cell Cycle
•
As much as 90% of DNA may be transcriptionally
inactive as a result of being nuclease-resistant, highly
Given the importance of normal DNA and chromosome
compacted, and nucleosome-associated.
function to survival, it is not surprising that eukaryotic
•
DNA in transcriptionally active regions is sensitive to
cells have developed elaborate mechanisms to monitor
the integrity of the genetic material. As detailed above, a
nuclease attack; some regions are exceptionally sensi-
tive and are often found to contain transcription
number of complex multi-subunit enzyme systems have
evolved to repair damaged DNA at the nucleotide se-
control sites.
quence level. Similarly, DNA mishaps at the chromo-
•
Transcriptionally active DNA (the genes) is often
some level are also monitored and repaired. As shown in
clustered in regions of each chromosome. Within
Figure
36-20, DNA integrity and chromosomal in-
these regions, genes may be separated by inactive
tegrity are continuously monitored throughout the cell
DNA in nucleosomal structures. The transcription
cycle. The four specific steps at which this monitoring
unit—that portion of a gene that is copied by RNA
occurs have been termed checkpoint controls. If prob-
polymerase—consists of coding regions of DNA
lems are detected at any of these checkpoints, progression
(exons) interrupted by intervening sequences of non-
through the cycle is interrupted and transit through the
coding DNA (introns).
cell cycle is halted until the damage is repaired. The mol-
•
After transcription, during RNA processing, introns
ecular mechanisms underlying detection of DNA dam-
are removed and the exons are ligated together to
age during the G1 and G2 phases of the cycle are under-
form the mature mRNA that appears in the cyto-
stood better than those operative during S and M phases.
plasm.
The tumor suppressor p53, a protein of MW 53
•
DNA in each chromosome is exactly replicated ac-
kDa, plays a key role in both G1 and G2 checkpoint con-
cording to the rules of base pairing during the S
trol. Normally a very unstable protein, p53 is a DNA
phase of the cell cycle.
binding transcription factor, one of a family of related
•
Each strand of the double helix is replicated simulta-
proteins, that is somehow stabilized in response to DNA
neously but by somewhat different mechanisms. A
damage, perhaps by direct p53-DNA interactions. In-
complex of proteins, including DNA polymerase,
creased levels of p53 activate transcription of an ensemble
replicates the leading strand continuously in the 5′ to
of genes that collectively serve to delay transit through the
3′ direction. The lagging strand is replicated discon-
cycle. One of these induced proteins, p21CIP, is a potent
tinuously, in short pieces of 150-250 nucleotides, in
CDK-cyclin inhibitor (CKI) that is capable of efficiently
the 3′ to 5′ direction.
inhibiting the action of all CDKs. Clearly, inhibition of
•
DNA replication occurs at several sites—called repli-
CDKs will halt progression through the cell cycle (see
cation bubbles—in each chromosome. The entire
Figures 36-19 and 36-20). If DNA damage is too exten-
process takes about 9 hours in a typical cell.
sive to repair, the affected cells undergo apoptosis (pro-
grammed cell death) in a p53-dependent fashion. In this
•
A variety of mechanisms employing different en-
case, p53 induces the activation of a collection of genes
zymes repair damaged DNA, as after exposure to
that induce apoptosis. Cells lacking functional p53 fail to
chemical mutagens or ultraviolet radiation.
undergo apoptosis in response to high levels of radiation
or DNA-active chemotherapeutic agents. It may come as
REFERENCES
no surprise, then, that p53 is one of the most frequently
DePamphilis ML: Origins of DNA replication in metazoan chro-
mutated genes in human cancers. Additional research into
mosomes. J Biol Chem 1993;268:1.
the mechanisms of checkpoint control will prove invalu-
Hartwell LH, Kastan MB: Cell cycle control and cancer. Science
able for the development of effective anticancer therapeu-
1994;266:1821.
tic options.
Jenuwein T, Allis CD: Translating the histone code. Science 2001;
293:1074.
Lander ES et al: Initial sequencing and analysis of the human
SUMMARY
genome. Nature 2001;409:860.
• DNA in eukaryotic cells is associated with a variety
Luger L et al: Crystal structure of the nucleosome core particle at
of proteins, resulting in a structure called chromatin.
2.8 Å resolution. Nature 1997;398:251.
340
/
CHAPTER 36
Marians KJ: Prokaryotic DNA replication. Annu Rev Biochem
Sullivan et al: Determining centromere identity: cyclical stories and
1992;61:673.
forking paths. Nat Rev Genet 2001;2:584.
Michelson RJ, Weinart T: Closing the gaps among a web of DNA
van Holde K, Zlatanova J: Chromatin higher order: chasing a mi-
repair disorders. Bioessays J 2002;22:966.
rage? J Biol Chem 1995;270:8373.
Moll UM, Erster S, Zaika A: p53, p63 and p73—solos, alliances
Venter JC et al: The sequence of the human genome. Science
and feuds among family members. Biochim Biophys Acta
2002;291:1304.
2001;1552:47.
Wallace DC: Mitochondrial DNA in aging and disease. Sci Am
Mouse Genome Sequencing Consortium: Initial sequencing and
1997 Aug;277:40.
comparative analysis of the mouse genome. Nature 2002;
Wood RD: Nucleotide excision repair in mammalian cells. J Biol
420:520.
Chem 1997;272:23465.
Narlikar GJ et al: Cooperation between complexes that regulate
chromatin structure and transcription. Cell 2002;108:475.
RNA Synthesis, Processing,
37
& Modification
Daryl K. Granner, MD, & P. Anthony Weil, PhD
BIOMEDICAL IMPORTANCE
following: (1) ribonucleotides are used in RNA synthe-
sis rather than deoxyribonucleotides; (2) U replaces T
The synthesis of an RNA molecule from DNA is a
as the complementary base pair for A in RNA; (3) a
complex process involving one of the group of RNA
primer is not involved in RNA synthesis; (4) only a very
polymerase enzymes and a number of associated pro-
small portion of the genome is transcribed or copied
teins. The general steps required to synthesize the pri-
into RNA, whereas the entire genome must be copied
mary transcript are initiation, elongation, and termina-
during DNA replication; and (5) there is no proofread-
tion. Most is known about initiation. A number of
ing function during RNA transcription.
DNA regions (generally located upstream from the ini-
The process of synthesizing RNA from a DNA tem-
tiation site) and protein factors that bind to these se-
plate has been characterized best in prokaryotes. Al-
quences to regulate the initiation of transcription have
though in mammalian cells the regulation of RNA syn-
been identified. Certain RNAs—mRNAs in particu-
thesis and the processing of the RNA transcripts are
lar—have very different life spans in a cell. It is impor-
different from those in prokaryotes, the process of RNA
tant to understand the basic principles of messenger
synthesis per se is quite similar in these two classes of
RNA synthesis and metabolism, for modulation of this
organisms. Therefore, the description of RNA synthesis
process results in altered rates of protein synthesis and
in prokaryotes, where it is better understood, is applica-
thus a variety of metabolic changes. This is how all or-
ble to eukaryotes even though the enzymes involved
ganisms adapt to changes of environment. It is also how
and the regulatory signals are different.
differentiated cell structures and functions are estab-
lished and maintained. The RNA molecules synthe-
sized in mammalian cells are made as precursor mole-
cules that have to be processed into mature, active
The Template Strand of DNA
RNA. Errors or changes in synthesis, processing, and
Is Transcribed
splicing of mRNA transcripts are a cause of disease.
The sequence of ribonucleotides in an RNA molecule is
complementary to the sequence of deoxyribonu-
RNA EXISTS IN FOUR MAJOR CLASSES
cleotides in one strand of the double-stranded DNA
All eukaryotic cells have four major classes of RNA: ri-
molecule (Figure 35-8). The strand that is transcribed
bosomal RNA (rRNA), messenger RNA (mRNA), trans-
or copied into an RNA molecule is referred to as the
fer RNA (tRNA), and small nuclear RNA (snRNA).
template strand of the DNA. The other DNA strand is
The first three are involved in protein synthesis, and
frequently referred to as the coding strand of that gene.
snRNA is involved in mRNA splicing. As shown in
It is called this because, with the exception of T for U
Table 37-1, these various classes of RNA are different
changes, it corresponds exactly to the sequence of the
in their diversity, stability, and abundance in cells.
primary transcript, which encodes the protein product
of the gene. In the case of a double-stranded DNA mol-
ecule containing many genes, the template strand for
RNA IS SYNTHESIZED FROM A DNA
each gene will not necessarily be the same strand of the
TEMPLATE BY AN RNA POLYMERASE
DNA double helix (Figure 37-1). Thus, a given strand
The processes of DNA and RNA synthesis are similar
of a double-stranded DNA molecule will serve as the
in that they involve (1) the general steps of initiation,
template strand for some genes and the coding strand
elongation, and termination with 5′ to 3′ polarity; (2)
of other genes. Note that the nucleotide sequence of an
large, multicomponent initiation complexes; and (3)
RNA transcript will be the same (except for U replacing
adherence to Watson-Crick base-pairing rules. These
T) as that of the coding strand. The information in the
processes differ in several important ways, including the
template strand is read out in the 3′ to 5′ direction.
341
342
/
CHAPTER 37
Table 37-1. Classes of eukaryotic RNA.
RNA transcript
5′ P-P-P
β′
β
RNA
Types
Abundance
Stability
Ribosomal
28S, 18S, 5.8S, 5S
80% of total
Very stable
3′
3′OH
Transcription
5′
(rRNA)
Messenger
~105 different
2-5% of total
Unstable to
σ
(mRNA)
species
very
α
α
5′
stable
3′
Transfer
~60 different
~15% of total
Very stable
(tRNA)
species
RNAP complex
Small nuclear
~30 different
≤ 1% of total
Very stable
Figure 37-2. RNA polymerase (RNAP) catalyzes the
(snRNA)
species
polymerization of ribonucleotides into an RNA se-
quence that is complementary to the template strand
of the gene. The RNA transcript has the same polarity
DNA-Dependent RNA Polymerase
(5′ to 3′) as the coding strand but contains U rather
Initiates Transcription at a Distinct
than T. E coli RNAP consists of a core complex of two
Site, the Promoter
α subunits and two β subunits (β and β′). The holoen-
DNA-dependent RNA polymerase is the enzyme re-
zyme contains the σ subunit bound to the α2ββ′ core
sponsible for the polymerization of ribonucleotides into
assembly. The ω subunit is not shown. The transcription
a sequence complementary to the template strand of
“bubble” is an approximately 20-bp area of melted
the gene (see Figures 37-2 and 37-3). The enzyme at-
DNA, and the entire complex covers 30-75 bp, depend-
taches at a specific site—the promoter—on the tem-
ing on the conformation of RNAP.
plate strand. This is followed by initiation of RNA syn-
thesis at the starting point, and the process continues
until a termination sequence is reached (Figure 37-3).
(1) Template binding
p
A transcription unit is defined as that region of DNA
ATP + NTP
that includes the signals for transcription initiation,
elongation, and termination. The RNA product, which
RNAP
p
is synthesized in the 5′ to 3′ direction, is the primary
(2) Chain initiation
transcript. In prokaryotes, this can represent the prod-
pppApN
uct of several contiguous genes; in mammalian cells, it
pppApN
pppApN
usually represents the product of a single gene. The 5′
NTPs
terminals of the primary RNA transcript and the ma-
(5) Chain termination
ture cytoplasmic RNA are identical. Thus, the starting
and RNAP release
(3) Promoter
pppApN
clearance
point of transcription corresponds to the 5
nu-
NTPs
cleotide of the mRNA. This is designated position +1,
as is the corresponding nucleotide in the DNA. The
(4) Chain elongation
Figure 37-3. The transcription cycle in bacteria. Bac-
Gene A Gene B Gene C
Gene D
terial RNA transcription is described in four steps:
5′
3′
(1) Template binding: RNA polymerase (RNAP) binds
3′
5′
to DNA and locates a promoter (P) melts the two DNA
strands to form a preinitiation complex (PIC). (2) Chain
Template strands
initiation: RNAP holoenzyme (core + one of multiple
Figure 37-1. This figure illustrates that genes can be
sigma factors) catalyzes the coupling of the first base
transcribed off both strands of DNA. The arrowheads in-
(usually ATP or GTP) to a second ribonucleoside
dicate the direction of transcription (polarity). Note that
triphosphate to form a dinucleotide. (3) Chain elonga-
the template strand is always read in the 3′ to 5′ direc-
tion: Successive residues are added to the 3′-OH termi-
tion. The opposite strand is called the coding strand be-
nus of the nascent RNA molecule. (4) Chain termina-
cause it is identical (except for T for U changes) to the
tion and release: The completed RNA chain and RNAP
mRNA transcript (the primary transcript in eukaryotic
are released from the template. The RNAP holoenzyme
cells) that encodes the protein product of the gene.
re-forms, finds a promoter, and the cycle is repeated.
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
343
numbers increase as the sequence proceeds downstream.
Table 37-2. Nomenclature and properties of
This convention makes it easy to locate particular re-
mammalian nuclear DNA-dependent RNA
gions, such as intron and exon boundaries. The nu-
polymerases.
cleotide in the promoter adjacent to the transcription
initiation site is designated −1, and these negative num-
Form of RNA
Sensitivity to
bers increase as the sequence proceeds upstream, away
Polymerase
-Amanitin
Major Products
from the initiation site. This provides a conventional
way of defining the location of regulatory elements in
I (A)
Insensitive
rRNA
the promoter.
II (B)
High sensitivity
mRNA
The primary transcripts generated by RNA polym-
III (C)
Intermediate sensitivity
tRNA/5S rRNA
erase II—one of three distinct nuclear DNA-depen-
dent RNA polymerases in eukaryotes—are promptly
capped by 7-methylguanosine triphosphate caps (Fig-
ferent sets of genes. The sizes of the RNA polymerases
ure 35-10) that persist and eventually appear on the 5′
range from MW 500,000 to MW 600,000. These en-
end of mature cytoplasmic mRNA. These caps are nec-
zymes are much more complex than prokaryotic RNA
essary for the subsequent processing of the primary
polymerases. They all have two large subunits and a
transcript to mRNA, for the translation of the mRNA,
number of smaller subunits—as many as 14 in the case
and for protection of the mRNA against exonucleolytic
of RNA pol III. The eukaryotic RNA polymerases have
attack.
extensive amino acid homologies with prokaryotic
RNA polymerases. This homology has been shown re-
Bacterial DNA-Dependent RNA
cently to extend to the level of three-dimensional struc-
Polymerase Is a Multisubunit Enzyme
tures. The functions of each of the subunits are not yet
fully understood. Many could have regulatory func-
The DNA-dependent RNA polymerase (RNAP) of the
tions, such as serving to assist the polymerase in the
bacterium Escherichia coli exists as an approximately
recognition of specific sequences like promoters and
400 kDa core complex consisting of two identical α
termination signals.
subunits, similar but not identical β and β′ subunits,
One peptide toxin from the mushroom Amanita
and an ω subunit. Beta is thought to be the catalytic
phalloides, α-amanitin, is a specific differential inhibitor
subunit (Figure 37-2). RNAP, a metalloenzyme, also
of the eukaryotic nuclear DNA-dependent RNA polym-
contains two zinc molecules. The core RNA polymerase
erases and as such has proved to be a powerful research
associates with a specific protein factor (the sigma [σ]
tool (Table 37-2). α-Amanitin blocks the translocation
factor) that helps the core enzyme recognize and bind
of RNA polymerase during transcription.
to the specific deoxynucleotide sequence of the pro-
moter region (Figure 37-5) to form the preinitiation
complex (PIC). Sigma factors have a dual role in the
process of promoter recognition; σ association with
RNA SYNTHESIS IS A CYCLICAL PROCESS
core RNA polymerase decreases its affinity for nonpro-
& INVOLVES INITIATION, ELONGATION,
moter DNA while simultaneously increasing holoen-
& TERMINATION
zyme affinity for promoter DNA. Bacteria contain mul-
tiple σ factors, each of which acts as a regulatory
The process of RNA synthesis in bacteria—depicted in
protein that modifies the promoter recognition speci-
Figure 37-3—involves first the binding of the RNA
ficity of the RNA polymerase. The appearance of dif-
holopolymerase molecule to the template at the pro-
ferent σ factors can be correlated temporally with vari-
moter site to form a PIC. Binding is followed by a con-
ous programs of gene expression in prokaryotic systems
formational change of the RNAP, and the first nu-
such as bacteriophage development, sporulation, and
cleotide (almost always a purine) then associates with
the response to heat shock.
the initiation site on the β subunit of the enzyme. In
the presence of the appropriate nucleotide, the RNAP
catalyzes the formation of a phosphodiester bond, and
Mammalian Cells Possess Three
the nascent chain is now attached to the polymerization
Distinct Nuclear DNA-Dependent
site on the β subunit of RNAP. (The analogy to the A
RNA Polymerases
and P sites on the ribosome should be noted; see Figure
The properties of mammalian polymerases are de-
38-9.)
scribed in Table 37-2. Each of these DNA-dependent
Initiation of formation of the RNA molecule at its
RNA polymerases is responsible for transcription of dif-
5′ end then follows, while elongation of the RNA mole-
344
/
CHAPTER 37
cule from the 5′ to its 3′ end continues cyclically, an-
tiparallel to its template. The enzyme polymerizes the
ribonucleotides in a specific sequence dictated by the
template strand and interpreted by Watson-Crick base-
pairing rules. Pyrophosphate is released in the polymer-
ization reaction. This pyrophosphate (PPi) is rapidly
degraded to 2 mol of inorganic phosphate (Pi ) by ubiq-
uitous pyrophosphatases, thereby providing irreversibil-
ity on the overall synthetic reaction. In both prokary-
otes and eukaryotes, a purine ribonucleotide is usually
the first to be polymerized into the RNA molecule. As
with eukaryotes, 5′ triphosphate of this first nucleotide
is maintained in prokaryotic mRNA.
As the elongation complex containing the core
RNA polymerase progresses along the DNA molecule,
DNA unwinding must occur in order to provide access
for the appropriate base pairing to the nucleotides of
the coding strand. The extent of this transcription bub-
ble (ie, DNA unwinding) is constant throughout tran-
scription and has been estimated to be about 20 base
pairs per polymerase molecule. Thus, it appears that the
size of the unwound DNA region is dictated by the
polymerase and is independent of the DNA sequence in
the complex. This suggests that RNA polymerase has
associated with it an “unwindase” activity that opens
Figure 37-4.
Electron photomicrograph of multiple
the DNA helix. The fact that the DNA double helix
copies of amphibian ribosomal RNA genes in the
must unwind and the strands part at least transiently
process of being transcribed. The magnification is
for transcription implies some disruption of the nucleo-
about 6000 ×. Note that the length of the transcripts in-
some structure of eukaryotic cells. Topoisomerase both
creases as the RNA polymerase molecules progress
precedes and follows the progressing RNAP to prevent
along the individual ribosomal RNA genes; transcrip-
the formation of superhelical complexes.
Termination of the synthesis of the RNA molecule
tion start sites (filled circles) to transcription termina-
in bacteria is signaled by a sequence in the template
tion sites (open circles). RNA polymerase I (not visual-
strand of the DNA molecule—a signal that is recog-
ized here) is at the base of the nascent rRNA transcripts.
nized by a termination protein, the rho (ρ) factor. Rho
Thus, the proximal end of the transcribed gene has
is an ATP-dependent RNA-stimulated helicase that
short transcripts attached to it, while much longer tran-
disrupts the nascent RNA-DNA complex. After termi-
scripts are attached to the distal end of the gene. The
nation of synthesis of the RNA molecule, the enzyme
arrows indicate the direction (5′ to 3′) of transcription.
separates from the DNA template and probably disso-
(Reproduced with permission, from Miller OL Jr, Beatty BR:
ciates to free core enzyme and free σ factor. With the
Portrait of a gene. J Cell Physiol 1969;74[Suppl 1]:225.)
assistance of another σ factor, the core enzyme then
recognizes a promoter at which the synthesis of a new
RNA molecule commences. In eukaryotic cells, termi-
nation is less well defined. It appears to be somehow
THE FIDELITY & FREQUENCY OF
linked both to initiation and to addition of the 3′
TRANSCRIPTION IS CONTROLLED
polyA tail of mRNA and could involve destabilization
BY PROTEINS BOUND TO CERTAIN
of the RNA-DNA complex at a region of A-U base
DNA SEQUENCES
pairs. More than one RNA polymerase molecule may
transcribe the same template strand of a gene simulta-
The DNA sequence analysis of specific genes has al-
neously, but the process is phased and spaced in such a
lowed the recognition of a number of sequences impor-
way that at any one moment each is transcribing a dif-
tant in gene transcription. From the large number of
ferent portion of the DNA sequence. An electron mi-
bacterial genes studied it is possible to construct con-
crograph of extremely active RNA synthesis is shown
sensus models of transcription initiation and termina-
in Figure 37-4.
tion signals.
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
345
The question, “How does RNAP find the correct
tion start site there is a consensus sequence of eight nu-
site to initiate transcription?” is not trivial when the
cleotide pairs (5′-TGTTGACA-3′) to which the RNAP
complexity of the genome is considered. E coli has
binds to form the so-called closed complex. More
4 × 103 transcription initiation sites in 4 × 106 base
proximal to the transcription start site—about ten nu-
pairs (bp) of DNA. The situation is even more complex
cleotides upstream—is a six-nucleotide-pair A+T-rich
in humans, where perhaps 105 transcription initiation
sequence (5′-TATAAT-3′). These conserved sequence
sites are distributed throughout in 3 × 109 bp of DNA.
elements comprising the promoter are shown schemati-
RNAP can bind to many regions of DNA, but it scans
cally in Figure 37-5. The latter sequence has a low
the DNA sequence—at a rate of ≥ 103 bp/s—until it
melting temperature because of its deficiency of GC
recognizes certain specific regions of DNA to which it
nucleotide pairs. Thus, the TATA box is thought to
binds with higher affinity. This region is called the pro-
ease the dissociation between the two DNA strands so
moter, and it is the association of RNAP with the pro-
that RNA polymerase bound to the promoter region
moter that ensures accurate initiation of transcription.
can have access to the nucleotide sequence of its imme-
The promoter recognition-utilization process is the tar-
diately downstream template strand. Once this process
get for regulation in both bacteria and humans.
occurs, the combination of RNA polymerase plus pro-
moter is called the open complex. Other bacteria have
slightly different consensus sequences in their promot-
Bacterial Promoters Are Relatively Simple
ers, but all generally have two components to the pro-
Bacterial promoters are approximately 40 nucleotides
moter; these tend to be in the same position relative to
(40 bp or four turns of the DNA double helix) in
the transcription start site, and in all cases the sequences
length, a region small enough to be covered by an
between the boxes have no similarity but still provide
E coli RNA holopolymerase molecule. In this consensus
critical spacing functions facilitating recognition of −35
promoter region are two short, conserved sequence ele-
and −10 sequence by RNA polymerase holoenzyme.
ments. Approximately 35 bp upstream of the transcrip-
Within a bacterial cell, different sets of genes are often
TRANSCRIPTION UNIT
Promoter
Transcribed region
Transcription
start site
+1
Coding strand 5′
Termination
3′
Template strand 3′TGTTGACATATAAT
signals
5′DNA
−35
−10
region
region
OH
PPP
RNA
3′
5′
5′ Flanking
3′ Flanking
sequences
sequences
Figure 37-5. Bacterial promoters, such as that from E coli shown here,
share two regions of highly conserved nucleotide sequence. These regions
are located 35 and 10 bp upstream (in the 5′ direction of the coding strand)
from the start site of transcription, which is indicated as +1. By convention,
all nucleotides upstream of the transcription initiation site (at +1) are num-
bered in a negative sense and are referred to as 5′-flanking sequences. Also
by convention, the DNA regulatory sequence elements (TATA box, etc) are
described in the 5′ to 3′ direction and as being on the coding strand. These
elements function only in double-stranded DNA, however. Note that the
transcript produced from this transcription unit has the same polarity or
“sense” (ie, 5′ to 3′ orientation) as the coding strand. Termination cis-
elements reside at the end of the transcription unit (see Figure 37-6 for
more detail). By convention the sequences downstream of the site at which
transcription termination occurs are termed 3′-flanking sequences.
346
/
CHAPTER 37
coordinately regulated. One important way that this is
simplex virus, which utilizes transcription factors of its
accomplished is through the fact that these co-regulated
mammalian host for gene expression, there is a single
genes share unique −35 and −10 promoter sequences.
unique transcription start site, and accurate transcription
These unique promoters are recognized by different σ
from this start site depends upon a nucleotide sequence
factors bound to core RNA polymerase.
located 32 nucleotides upstream from the start site (ie, at
Rho-dependent transcription termination signals
−32) (Figure 37-7). This region has the sequence of
in E coli also appear to have a distinct consensus se-
TATAAAAG and bears remarkable similarity to the
quence, as shown in Figure 37-6. The conserved con-
functionally related TATA box that is located about 10
sensus sequence, which is about 40 nucleotide pairs in
bp upstream from the prokaryotic mRNA start site (Fig-
length, can be seen to contain a hyphenated or inter-
ure 37-5). Mutation or inactivation of the TATA box
rupted inverted repeat followed by a series of AT base
markedly reduces transcription of this and many other
pairs. As transcription proceeds through the hyphen-
genes that contain this consensus cis element (see Figures
ated, inverted repeat, the generated transcript can form
37-7, 37-8). Most mammalian genes have a TATA box
the intramolecular hairpin structure, also depicted in
that is usually located 25-30 bp upstream from the tran-
Figure 37-6.
scription start site. The consensus sequence for a TATA
Transcription continues into the AT region, and
box is TATAAA, though numerous variations have been
with the aid of the ρ termination protein the RNA
characterized. The TATA box is bound by 34 kDa
polymerase stops, dissociates from the DNA template,
TATA binding protein (TBP), which in turn binds sev-
and releases the nascent transcript.
eral other proteins called TBP-associated factors
(TAFs). This complex of TBP and TAFs is referred to as
TFIID. Binding of TFIID to the TATA box sequence is
Eukaryotic Promoters Are More Complex
thought to represent the first step in the formation of the
It is clear that the signals in DNA which control tran-
transcription complex on the promoter.
scription in eukaryotic cells are of several types. Two
A small number of genes lack a TATA box. In such
types of sequence elements are promoter-proximal. One
instances, two additional cis elements, an initiator se-
of these defines where transcription is to commence
quence (Inr) and the so-called downstream promoter
along the DNA, and the other contributes to the mecha-
element (DPE), direct RNA polymerase II to the pro-
nisms that control how frequently this event is to occur.
moter and in so doing provide basal transcription start-
For example, in the thymidine kinase gene of the herpes
ing from the correct site. The Inr element spans the start
Direction of transcription
Coding strand
5′
AGCCCGC
GCGGGCT
TTTTTTTT
3′
DNA
Template strand
3′
TCGGGCG
CGCCCGA
AAAAAAAA
5′
Coding strand
5′
3′
DNA
Template strand
3′
AAAAAAAA
5′
UUUUUUU-3′
U
A
U
G
C
C
G
C
G
C
G
G
C
C
G
RNA transcript
5′
Figure 37-6. The predominant bacterial transcription termination signal contains an inverted, hyphenated re-
peat (the two boxed areas) followed by a stretch of AT base pairs (top figure). The inverted repeat, when tran-
scribed into RNA, can generate the secondary structure in the RNA transcript shown at the bottom of the figure.
Formation of this RNA hairpin causes RNA polymerase to pause and subsequently the ρ termination factor inter-
acts with the paused polymerase and somehow induces chain termination.
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
347
Promoter proximal
Promoter
upstream elements
TFIID
Sp1
+1
GC
CAAT
GC
TATA box
tk coding region
CTF
−25
Sp1
Figure 37-7. Transcription elements and binding factors in the herpes simplex virus thymidine ki-
nase (tk) gene. DNA-dependent RNA polymerase II binds to the region of the TATA box (which is bound
by transcription factor TFIID) to form a multicomponent preinitiation complex capable of initiating
transcription at a single nucleotide (+1). The frequency of this event is increased by the presence of up-
stream cis-acting elements (the GC and CAAT boxes). These elements bind trans-acting transcription
factors, in this example Sp1 and CTF (also called C/EBP, NF1, NFY). These cis elements can function inde-
pendently of orientation (arrows).
Regulated expression
“Basal” expression
Distal
Promoter
regulatory
proximal
Promoter
elements
elements
+1
Other
Enhancer (+)
Promoter
regulatory
and
proximal
Inr
DPE
elements
repressor (−)
elements
TATA
Coding region
elements
(GC/CAAT, etc)
Figure 37-8. Schematic diagram showing the transcription control regions in a hypothetical class II
(mRNA-producing) eukaryotic gene. Such a gene can be divided into its coding and regulatory regions,
as defined by the transcription start site (arrow; +1). The coding region contains the DNA sequence that
is transcribed into mRNA, which is ultimately translated into protein. The regulatory region consists of
two classes of elements. One class is responsible for ensuring basal expression. These elements gener-
ally have two components. The proximal component, generally the TATA box, or Inr or DPE elements di-
rect RNA polymerase II to the correct site (fidelity). In TATA-less promoters, an initiator (Inr) element that
spans the initiation site (+1) may direct the polymerase to this site. Another component, the upstream
elements, specifies the frequency of initiation. Among the best studied of these is the CAAT box, but
several other elements (Sp1, NF1, AP1, etc) may be used in various genes. A second class of regulatory
cis-acting elements is responsible for regulated expression. This class consists of elements that enhance
or repress expression and of others that mediate the response to various signals, including hormones,
heat shock, heavy metals, and chemicals. Tissue-specific expression also involves specific sequences of
this sort. The orientation dependence of all the elements is indicated by the arrows within the boxes. For
example, the proximal element (the TATA box) must be in the 5′ to 3′ orientation. The upstream ele-
ments work best in the 5′ to 3′ orientation, but some of them can be reversed. The locations of some el-
ements are not fixed with respect to the transcription start site. Indeed, some elements responsible for
regulated expression can be located either interspersed with the upstream elements, or they can be lo-
cated downstream from the start site.
348
/
CHAPTER 37
site (from −3 to +5) and consists of the general consen-
below and Figures 37-9 and 37-10). The protein-
sus sequence TCA+1 G/T T T/C which is similar to the
DNA interaction at the TATA box involving RNA
initiation site sequence per se. (A+1 indicates the first
polymerase II and other components of the basal tran-
nucleotide transcribed.) The proteins that bind to Inr in
scription machinery ensures the fidelity of initiation.
order to direct pol II binding include TFIID. Promoters
Together, then, the promoter and promoter-proxi-
that have both a TATA box and an Inr may be stronger
mal cis-active upstream elements confer fidelity and fre-
than those that have just one of these elements. The
quency of initiation upon a gene. The TATA box has a
DPE has the consensus sequence A/GGA/T CGTG and
particularly rigid requirement for both position and ori-
is localized about 25 bp downstream of the +1 start site.
entation. Single-base changes in any of these cis ele-
Like the Inr, DPE sequences are also bound by the TAF
ments have dramatic effects on function by reducing
subunits of TFIID. In a survey of over 200 eukaryotic
the binding affinity of the cognate trans factors (either
genes, roughly 30% contained a TATA box and Inr,
TFIID/TBP or Sp1, CTF, and similar factors). The
25% contained Inr and DPE, 15% contained all three
spacing of these elements with respect to the transcrip-
elements, while ~30% contained just the Inr.
tion start site can also be critical. This is particularly
Sequences farther upstream from the start site deter-
true for the TATA box Inr and DPE.
mine how frequently the transcription event occurs.
A third class of sequence elements can either increase
Mutations in these regions reduce the frequency of
or decrease the rate of transcription initiation of eukary-
transcriptional starts tenfold to twentyfold. Typical of
otic genes. These elements are called either enhancers or
these DNA elements are the GC and CAAT boxes, so
repressors (or silencers), depending on which effect
named because of the DNA sequences involved. As il-
they have. They have been found in a variety of locations
lustrated in Figure 37-7, each of these boxes binds a
both upstream and downstream of the transcription start
protein, Sp1 in the case of the GC box and CTF (or
site and even within the transcribed portions of some
C/EPB,NF1,NFY) by the CAAT box; both bind
genes. In contrast to proximal and upstream promoter el-
through their distinct DNA binding domains (DBDs).
ements, enhancers and silencers can exert their effects
The frequency of transcription initiation is a conse-
when located hundreds or even thousands of bases away
quence of these protein-DNA interactions and complex
from transcription units located on the same chromo-
interactions between particular domains of the tran-
some. Surprisingly, enhancers and silencers can function
scription factors (distinct from the DBD domains—so-
in an orientation-independent fashion. Literally hun-
called activation domains; ADs) of these proteins and
dreds of these elements have been described. In some
the rest of the transcription machinery (RNA polym-
cases, the sequence requirements for binding are rigidly
erase II and the basal factors TFIIA, B, D, E, F). (See
constrained; in others, considerable sequence variation is
H
F
A
E
D
B
TATA
pol II
-50
-30
-10
+10
+30
+50
Figure 37-9. The eukaryotic basal transcription complex. Formation of the basal transcription complex begins
when TFIID binds to the TATA box. It directs the assembly of several other components by protein-DNA and
protein-protein interactions. The entire complex spans DNA from position −30 to +30 relative to the initiation site
(+1, marked by bent arrow). The atomic level, x-ray-derived structures of RNA polymerase II alone and of TBP
bound to TATA promoter DNA in the presence of either TFIIB or TFIIA have all been solved at 3 Å resolution. The
structure of TFIID complexes have been determined by electron microscopy at 30 Å resolution. Thus, the molecu-
lar structures of the transcription machinery are beginning to be elucidated. Much of this structural information is
consistent with the models presented here.
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
349
Rate of
Rate of
transcription
transcription
Basal
TBP
complex
TAF
TAF
CTF TAF
CCAAT
TBP
Basal complex
CAAT
TATA
nil
+
CTF
Basal
TBP
complex
TAF
CTF
CTF
TAF TAF
CAAT
TATA
nil
TAF
TBP
Basal
Basal complex
TBP
complex
TATA
A
B
Figure 37-10. Two models for assembly of the active transcription complex and for how activators and coacti-
vators might enhance transcription. Shown here as a small oval is TBP, which contains TFIID, a large oval that con-
tains all the components of the basal transcription complex illustrated in Figure 37-9 (ie, RNAP II and TFIIA, TFIIB,
TFIIE, TFIIF, and TFIIH). Panel A: The basal transcription complex is assembled on the promoter after the TBP sub-
unit of TFIID is bound to the TATA box. Several TAFs (coactivators) are associated with TBP. In this example, a tran-
scription activator, CTF, is shown bound to the CAAT box, forming a loop complex by interacting with a TAF
bound to TBP. Panel B: The recruitment model. The transcription activator CTF binds to the CAAT box and inter-
acts with a coactivator (TAF in this case). This allows for an interaction with the preformed TBP-basal transcription
complex. TBP can now bind to the TATA box, and the assembled complex is fully active.
allowed. Some sequences bind only a single protein, but
stood. However, it appears that the termination signals
the majority bind several different proteins. Similarly, a
exist far downstream of the coding sequence of eukary-
single protein can bind to more than one element.
otic genes. For example, the transcription termination
Hormone response elements (for steroids, T3, reti-
signal for mouse β-globin occurs at several positions
noic acid, peptides, etc) act as—or in conjunction with—
1000-2000 bases beyond the site at which the poly(A)
enhancers or silencers (Chapter 43). Other processes
tail will eventually be added. Little is known about the
that enhance or silence gene expression—such as the re-
termination process or whether specific termination
sponse to heat shock, heavy metals (Cd2+ and Zn2+),
factors similar to the bacterial ρ factor are involved.
and some toxic chemicals (eg, dioxin)—are mediated
However, it is known that the mRNA 3′ terminal is
through specific regulatory elements. Tissue-specific ex-
generated posttranscriptionally, is somehow coupled to
pression of genes (eg, the albumin gene in liver, the he-
events or structures formed at the time and site of initi-
moglobin gene in reticulocytes) is also mediated by spe-
ation, depends on a special structure in one of the sub-
cific DNA sequences.
units of RNA polymerase II (the CTD; see below), and
appears to involve at least two steps. After RNA polym-
erase II has traversed the region of the transcription
Specific Signals Regulate
unit encoding the 3′ end of the transcript, an RNA en-
Transcription Termination
donuclease cleaves the primary transcript at a position
The signals for the termination of transcription by
about 15 bases 3′ of the consensus sequence AAUAAA
eukaryotic RNA polymerase II are very poorly under-
that serves in eukaryotic transcripts as a cleavage signal.
350
/
CHAPTER 37
Finally, this newly formed 3′ terminal is polyadenylated
distinct, polymerase-specific sets of TAFs, is also an im-
in the nucleoplasm, as described below.
portant component of class I and class III initiation
complexes even if they do not contain TATA boxes.
The binding of TBP marks a specific promoter for
THE EUKARYOTIC
transcription and is the only step in the assembly process
TRANSCRIPTION COMPLEX
that is entirely dependent on specific, high-affinity pro-
A complex apparatus consisting of as many as
50
tein-DNA interaction. Of several subsequent in vitro
unique proteins provides accurate and regulatable tran-
steps, the first is the binding of TFIIB to the TFIID-
scription of eukaryotic genes. The RNA polymerase en-
promoter complex. This results in a stable ternary com-
zymes (pol I, pol II, and pol III for class I, II, and III
plex which is then more precisely located and more
genes, respectively) transcribe information contained in
tightly bound at the transcription initiation site. This
the template strand of DNA into RNA. These polym-
complex then attracts and tethers the pol II-TFIIF com-
erases must recognize a specific site in the promoter in
plex to the promoter. TFIIF is structurally and func-
order to initiate transcription at the proper nucleotide.
tionally similar to the bacterial σ factor and is required
In contrast to the situation in prokaryotes, eukaryotic
for the delivery of pol II to the promoter. TFIIA binds
RNA polymerases alone are not able to discriminate be-
to this assembly and may allow the complex to respond
tween promoter sequences and other regions of DNA;
to activators, perhaps by the displacement of repressors.
thus, other proteins known as general transcription fac-
Addition of TFIIE and TFIIH is the final step in the as-
tors or GTFs facilitate promoter-specific binding of
sembly of the PIC. TFIIE appears to join the complex
these enzymes and formation of the preinitiation com-
with pol II-TFIIF, and TFIIH is then recruited. Each of
plex (PIC). This combination of components can cat-
these binding events extends the size of the complex so
alyze basal or (non)-unregulated transcription in vitro.
that finally about 60 bp (from −30 to +30 relative to +1,
Another set of proteins—coactivators—help regulate
the nucleotide from which transcription commences)
the rate of transcription initiation by interacting with
are covered (Figure 37-9). The PIC is now complete
transcription activators that bind to upstream DNA el-
and capable of basal transcription initiated from the cor-
ements (see below).
rect nucleotide. In genes that lack a TATA box, the
same factors, including TBP, are required. In such cases,
Formation of the Basal
an Inr or the DPEs (see Figure 37-8) position the com-
plex for accurate initiation of transcription.
Transcription Complex
In bacteria, a σ factor-polymerase complex selectively
Phosphorylation Activates Pol II
binds to DNA in the promoter forming the PIC. The
situation is more complex in eukaryotic genes. Class II
Eukaryotic pol II consists of
12 subunits. The two
genes—those transcribed by pol II to make mRNA—
largest subunits, both about 200 kDa, are homologous
are described as an example. In class II genes, the func-
to the bacterial β and β′ subunits. In addition to the in-
tion of σ factors is assumed by a number of proteins.
creased number of subunits, eukaryotic pol II differs
Basal transcription requires, in addition to pol II, a
from its prokaryotic counterpart in that it has a series of
number of GTFs called TFIIA, TFIIB, TFIID,
heptad repeats with consensus sequence Tyr-Ser-Pro-
TFIIE, TFIIF, and TFIIH. These GTFs serve to pro-
Thr-Ser-Pro-Ser at the carboxyl terminal of the largest
mote RNA polymerase II transcription on essentially all
pol II subunit. This carboxyl terminal repeat domain
genes. Some of these GTFs are composed of multiple
(CTD) has 26 repeated units in brewers’ yeast and 52
subunits. TFIID, which binds to the TATA box pro-
units in mammalian cells. The CTD is both a substrate
moter element, is the only one of these factors capa-
for several kinases, including the kinase component of
ble of binding to specific sequences of DNA. As de-
TFIIH, and a binding site for a wide array of proteins.
scribed above, TFIID consists of TATA binding
The CTD has been shown to interact with RNA pro-
protein (TBP) and 14 TBP-associated factors (TAFs).
cessing enzymes; such binding may be involved with
TBP binds to the TATA box in the minor groove of
RNA polyadenylation. The association of the factors
DNA (most transcription factors bind in the major
with the CTD of RNA polymerase II (and other com-
groove) and causes an approximately 100-degree bend
ponents of the basal machinery) somehow serves to
or kink of the DNA helix. This bending is thought to
couple initiation with mRNA 3′ end formation. Pol II
facilitate the interaction of TBP-associated factors with
is activated when phosphorylated on the Ser and Thr
other components of the transcription initiation com-
residues and displays reduced activity when the CTD is
plex and possibly with factors bound to upstream ele-
dephosphorylated. Pol II lacking the CTD tail is inca-
ments. Although defined as a component of class II
pable of activating transcription, which underscores the
gene promoters, TBP, by virtue of its association with
importance of this domain.
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
351
Pol II associates with other proteins to form a
TAFs. It is conceivable that different combinations of
holoenzyme complex. In yeast, at least nine gene prod-
TAFs with TBP—or one of several recently discovered
ucts—called Srb
(for suppressor of RNA polymer-
TBP-like factors (TLFs)—may bind to different pro-
ase B)—bind to the CTD. The Srb proteins—or medi-
moters, and recent reports suggest that this may ac-
ators, as they are also called—are essential for pol II
count for selective activation noted in various promot-
transcription, though their exact role in this process has
ers and for the different strengths of certain promoters.
not been defined. Related proteins comprising even
TAFs, since they are required for the action of acti-
more complex forms of RNA polymerase II have been
vators, are often called coactivators. There are thus
described in human cells.
three classes of transcription factors involved in the reg-
ulation of class II genes: basal factors, coactivators, and
The Role of Transcription Activators
activator-repressors (Table 37-4). How these classes of
proteins interact to govern both the site and frequency
& Coactivators
of transcription is a question of central importance.
TFIID was originally considered to be a single protein.
However, several pieces of evidence led to the impor-
Two Models Explain the Assembly
tant discovery that TFIID is actually a complex consist-
of the Preinitiation Complex
ing of TBP and the 14 TAFs. The first evidence that
TFIID was more complex than just the TBP molecules
The formation of the PIC described above is based on
came from the observation that TBP binds to a 10-bp
the sequential addition of purified components in in
segment of DNA, immediately over the TATA box of
vitro experiments. An essential feature of this model is
the gene, whereas native holo-TFIID covers a 35 bp or
that the assembly takes place on the DNA template.
larger region (Figure 37-9). Second, TBP has a molec-
Accordingly, transcription activators, which have au-
ular mass of 20-40 kDa (depending on the species),
tonomous DNA binding and activation domains (see
whereas the TFIID complex has a mass of about 1000
Chapter 39), are thought to function by stimulating ei-
kDa. Finally, and perhaps most importantly, TBP sup-
ther PIC formation or PIC function. The TAF coacti-
ports basal transcription but not the augmented tran-
vators are viewed as bridging factors that communicate
scription provided by certain activators, eg, Sp1 bound
between the upstream activators, the proteins associated
to the GC box. TFIID, on the other hand, supports
with pol II, or the many other components of TFIID.
both basal and enhanced transcription by Sp1, Oct1,
This view, which assumes that there is stepwise assem-
AP1, CTF, ATF, etc. (Table 37-3). The TAFs are es-
bly of the PIC—promoted by various interactions be-
sential for this activator-enhanced transcription. It is
tween activators, coactivators, and PIC components—
not yet clear whether there are one or several forms of
is illustrated in panel A of Figure 37-10. This model
TFIID that might differ slightly in their complement of
was supported by observations that many of these pro-
teins could indeed bind to one another in vitro.
Recent evidence suggests that there is another possi-
ble mechanism of PIC formation and transcription reg-
Table 37-3. Some of the transcription control
ulation. First, large preassembled complexes of GTFs
elements, their consensus sequences, and the
and pol II are found in cell extracts, and this complex
factors that bind to them which are found in
can associate with a promoter in a single step. Second,
mammalian genes transcribed by RNA
the rate of transcription achieved when activators are
polymerase II. A complete list would include
added to limiting concentrations of pol II holoenzyme
dozens of examples. The asterisks mean that
can be matched by increasing the concentration of the
there are several members of this family.
pol II holoenzyme in the absence of activators. Thus,
Element
Consensus Sequence
Factor
TATA box
TATAAA
TBP
Table 37-4. Three classes of transcription factors
CAAT box
CCAATC
C/EBP*, NF-Y*
in class II genes.
GC box
GGGCGG
Sp1*
CAACTGAC
Myo D
General Mechanisms
Specific Components
T/CGGA/CN5GCCAA
NF1*
lg octamer
ATGCAAAT
Oct1, 2, 4, 6*
Basal components
TBP, TFIIA, B, E, F, and H
AP1
TGAG/CTC/AA
Jun, Fos, ATF*
Coactivators
TAFs (TBP + TAFs) = TFIID; Srbs
Serum response
GATGCCCATA
SRF
Heat shock
(NGAAN)3
HSF
Activators
SP1, ATF, CTF, AP1, etc
352
/
CHAPTER 37
activators are not in themselves absolutely essential for
precursor molecules is required for the generation of
PIC formation. These observations led to the “recruit-
the mature functional molecules.
ment” hypothesis, which has now been tested experi-
Nearly all eukaryotic RNA primary transcripts un-
mentally. Simply stated, the role of activators and
dergo extensive processing between the time they are
coactivators may be solely to recruit a preformed
synthesized and the time at which they serve their ulti-
holoenzyme-GTF complex to the promoter. The re-
mate function, whether it be as mRNA or as a com-
quirement for an activation domain is circumvented
ponent of the translation machinery such as rRNA,
when either a component of TFIID or the pol II
5S RNA, or tRNA or RNA processing machinery,
holoenzyme is artificially tethered, using recombinant
snRNAs. Processing occurs primarily within the nu-
DNA techniques, to the DNA binding domain (DBD)
cleus and includes nucleolytic cleavage to smaller mole-
of an activator. This anchoring, through the DBD
cules and coupled nucleolytic and ligation reactions
component of the activator molecule, leads to a tran-
(splicing of exons). In mammalian cells, 50-75% of
scriptionally competent structure, and there is no fur-
the nuclear RNA does not contribute to the cytoplas-
ther requirement for the activation domain of the acti-
mic mRNA. This nuclear RNA loss is significantly
vator. In this view, the role of activation domains and
greater than can be reasonably accounted for by the loss
TAFs is to form an assembly that directs the preformed
of intervening sequences alone (see below). Thus, the
holoenzyme-GTF complex to the promoter; they do
exact function of the seemingly excessive transcripts in
not assist in PIC assembly (see panel B, Figure 37-10).
the nucleus of a mammalian cell is not known.
The efficiency of this recruitment process determines
the rate of transcription at a given promoter.
Hormones—and other effectors that serve to trans-
The Coding Portions (Exons)
mit information related to the extracellular environ-
of Most Eukaryotic Genes
ment—modulate gene expression by influencing the as-
Are Interrupted by Introns
sembly and activity of the activator and coactivator
Interspersed within the amino acid-coding portions
complexes and the subsequent formation of the PIC at
(exons) of many genes are long sequences of DNA that
the promoter of target genes (see Chapter 43). The nu-
do not contribute to the genetic information ultimately
merous components involved provide for an abundance
translated into the amino acid sequence of a protein
of possible combinations and therefore a range of tran-
molecule (see Chapter 36). In fact, these sequences ac-
scriptional activity of a given gene. It is important to
tually interrupt the coding region of structural genes.
note that the two models are not mutually exclusive—
These intervening sequences (introns) exist within
stepwise versus holoenzyme-mediated PIC formation.
most but not all mRNA encoding genes of higher eu-
Indeed, one can envision various more complex models
karyotes. The primary transcripts of the structural genes
invoking elements of both models operating on a gene.
contain RNA complementary to the interspersed se-
quences. However, the intron RNA sequences are
cleaved out of the transcript, and the exons of the tran-
RNA MOLECULES ARE USUALLY
script are appropriately spliced together in the nucleus
PROCESSED BEFORE THEY
before the resulting mRNA molecule appears in the cy-
BECOME FUNCTIONAL
toplasm for translation
(Figures
37-11 and 37-12).
One speculation is that exons, which often encode an
In prokaryotic organisms, the primary transcripts of
activity domain of a protein, represent a convenient
mRNA-encoding genes begin to serve as translation
means of shuffling genetic information, permitting or-
templates even before their transcription has been com-
ganisms to quickly test the results of combining novel
pleted. This is because the site of transcription is not
protein functional domains.
compartmentalized into a nucleus as it is in eukaryotic
organisms. Thus, transcription and translation are cou-
pled in prokaryotic cells. Consequently, prokaryotic
Introns Are Removed & Exons
mRNAs are subjected to little processing prior to carry-
Are Spliced Together
ing out their intended function in protein synthesis. In-
deed, appropriate regulation of some genes (eg, the Trp
The mechanisms whereby introns are removed from
operon) relies upon this coupling of transcription and
the primary transcript in the nucleus, exons are ligated
translation. Prokaryotic rRNA and tRNA molecules are
to form the mRNA molecule, and the mRNA molecule
transcribed in units considerably longer than the ulti-
is transported to the cytoplasm are being elucidated.
mate molecule. In fact, many of the tRNA transcription
Four different splicing reaction mechanisms have been
units contain more than one molecule. Thus, in
described. The one most frequently used in eukaryotic
prokaryotes the processing of these rRNA and tRNA
cells is described below. Although the sequences of nu-
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
353
Exon 1
Exon 2
Intron
5′ Cap
G
G
A G
An
3′ Primary transcript
Cap
G G
A G
An
Nucleophilic attack
at 5′ end of intron
Cap
G OH
A G
n
A Lariat formation
Cap
G
OH
G
An
Cut at 3′ end of intron
Cap
G—G
An
and
Ligation of 3′ end of exon
1 to 5′ end of exon 2
Intron is digested
Figure 37-11. The processing of the primary transcript to mRNA. In this hy-
pothetical transcript, the 5′ (left) end of the intron is cut (↓) and a lariat forms
between the G at the 5′ end of the intron and an A near the 3′ end, in the con-
sensus sequence UACUAAC. This sequence is called the branch site, and it is the
3′ most A that forms the 5′-2’ bond with the G. The 3′ (right) end of the intron is
then cut (⇓). This releases the lariat, which is digested, and exon 1 is joined to
exon 2 at G residues.
cleotides in the introns of the various eukaryotic tran-
mary transcript, five small nuclear RNAs (U1, U2, U5,
scripts—and even those within a single transcript—are
U4, and U6) and more than 60 proteins. Collectively,
quite heterogeneous, there are reasonably conserved se-
these form a small nucleoprotein (snRNP) complex,
quences at each of the two exon-intron (splice) junc-
sometimes called a “snurp.” It is likely that this penta-
tions and at the branch site, which is located 20-40 nu-
snRNP spliceosome forms prior to interaction with
cleotides upstream from the 3′ splice site (see consensus
mRNA precursors. Snurps are thought to position the
sequences in Figure 37-12). A special structure, the
RNA segments for the necessary splicing reactions. The
spliceosome, is involved in converting the primary
splicing reaction starts with a cut at the junction of the
transcript into mRNA. Spliceosomes consist of the pri-
5′ exon (donor or left) and intron (Figure 37-11). This
Consensus sequences
A
5′
AG
G
UAAGU
UACUAAC 28-37 nucleotides C
AG
G
3′
C
5′ Exon
Intron
Exon 3′
Figure 37-12. Consensus sequences at splice junctions. The 5′ (donor or left) and 3′ (ac-
ceptor or right) sequences are shown. Also shown is the yeast consensus sequence
(UACUAAC) for the branch site. In mammalian cells, this consensus sequence is PyNPyPy-
PuAPy, where Py is a pyrimidine, Pu is a purine, and N is any nucleotide. The branch site is lo-
cated 20-40 nucleotides upstream from the 3′ site.
354
/
CHAPTER 37
is accomplished by a nucleophilic attack by an adenylyl
above, the sequence of exon-intron splicing events gen-
residue in the branch point sequence located just up-
erally follows a hierarchical order for a given gene. The
stream from the 3′ end of this intron. The free 5′ termi-
fact that very complex RNA structures are formed dur-
nal then forms a loop or lariat structure that is linked
ing splicing—and that a number of snRNAs and pro-
by an unusual 5′-2′ phosphodiester bond to the reac-
teins are involved—affords numerous possibilities for a
tive A in the PyNPyPyPuAPy branch site sequence
change of this order and for the generation of different
(Figure 37-12). This adenylyl residue is typically lo-
mRNAs. Similarly, the use of alternative termination-
cated 28-37 nucleotides upstream from the 3′ end of
cleavage-polyadenylation sites also results in mRNA
the intron being removed. The branch site identifies
heterogeneity. Some schematic examples of these
the 3′ splice site. A second cut is made at the junction
processes, all of which occur in nature, are shown in
of the intron with the 3′ exon (donor on right). In this
Figure 37-13.
second transesterification reaction, the 3′ hydroxyl of
Faulty splicing can cause disease. At least one
the upstream exon attacks the
5′ phosphate at the
form of β-thalassemia, a disease in which the β-globin
downstream exon-intron boundary, and the lariat
gene of hemoglobin is severely underexpressed, appears
structure containing the intron is released and hy-
to result from a nucleotide change at an exon-intron
drolyzed. The 5′ and 3′ exons are ligated to form a con-
junction, precluding removal of the intron and there-
tinuous sequence.
fore leading to diminished or absent synthesis of the
The snRNAs and associated proteins are required
β-chain protein. This is a consequence of the fact that
for formation of the various structures and intermedi-
the normal translation reading frame of the mRNA is
ates. U1 within the snRNP complex binds first by base
disrupted—a defect in this fundamental process (splic-
pairing to the 5′ exon-intron boundary. U2 within the
ing) that underscores the accuracy which the process of
snRNP complex then binds by base pairing to the
RNA-RNA splicing must achieve.
branch site, and this exposes the nucleophilic A residue.
U5/U4/U6 within the snRNP complex mediates an
Alternative Promoter Utilization
ATP-dependent protein-mediated unwinding that re-
Provides a Form of Regulation
sults in disruption of the base-paired U4-U6 complex
with the release of U4. U6 is then able to interact first
Tissue-specific regulation of gene expression can be
with U2, then with U1. These interactions serve to ap-
provided by control elements in the promoter or by the
proximate the 5′ splice site, the branch point with its
reactive A, and the 3′ splice site. This alignment is en-
hanced by U5. This process also results in the forma-
mRNA precursor
tion of the loop or lariat structure. The two ends are
1
2
3
AAUAA AAUAA
(A)n
cleaved, probably by the U2-U6 within the snRNP
complex. U6 is certainly essential, since yeasts deficient
in this snRNA are not viable. It is important to note
Selective splicing
that RNA serves as the catalytic agent. This sequence is
then repeated in genes containing multiple introns. In
1
2
3
AAUAA AAUAA
(A)n
such cases, a definite pattern is followed for each gene,
and the introns are not necessarily removed in se-
Alternative 5′ donor site
quence—1, then 2, then 3, etc.
The relationship between hnRNA and the corre-
1′
2
3
AAUAA AAUAA
(A)n
sponding mature mRNA in eukaryotic cells is now ap-
parent. The hnRNA molecules are the primary tran-
Alternative 3′ acceptor site
scripts plus their early processed products, which, after
the addition of caps and poly(A) tails and removal of
1
2′
3
AAUAA AAUAA
(A)n
the portion corresponding to the introns, are trans-
Alternative polyadenylation site
ported to the cytoplasm as mature mRNA molecules.
1
2
3
AAUAA
(A)n
Alternative Splicing Provides
for Different mRNAs
Figure 37-13. Mechanisms of alternative process-
The processing of hnRNA molecules is a site for reg-
ing of mRNA precursors. This form of RNA processing
ulation of gene expression. Alternative patterns of
involves the selective inclusion or exclusion of exons,
RNA splicing result from tissue-specific adaptive and
the use of alternative 5′ donor or 3′ acceptor sites, and
developmental control mechanisms. As mentioned
the use of different polyadenylation sites.
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
355
use of alternative promoters. The glucokinase
(GK)
mature tRNA. A small fraction of tRNAs even contain
gene consists of ten exons interrupted by nine introns.
introns.
The sequence of exons 2-10 is identical in liver and
pancreatic B cells, the primary tissues in which GK pro-
RNAS CAN BE EXTENSIVELY MODIFIED
tein is expressed. Expression of the GK gene is regulated
very differently—by two different promoters—in these
Essentially all RNAs are covalently modified after tran-
two tissues. The liver promoter and exon 1L are located
scription. It is clear that at least some of these modifica-
near exons 2-10; exon 1L is ligated directly to exon 2.
tions are regulatory.
In contrast, the pancreatic B cell promoter is located
about 30 kbp upstream. In this case, the 3′ boundary of
Messenger RNA (mRNA) Is Modified
exon 1B is ligated to the 5′ boundary of exon 2. The
at the 5 & 3 Ends
liver promoter and exon 1L are excluded and removed
As mentioned above, mammalian mRNA molecules
during the splicing reaction (see Figure 37-14). The ex-
contain a 7-methylguanosine cap structure at their 5′
istence of multiple distinct promoters allows for cell-
terminal, and most have a poly(A) tail at the 3′ termi-
and tissue-specific expression patterns of a particular
nal. The cap structure is added to the 5′ end of the
gene (mRNA).
newly transcribed mRNA precursor in the nucleus
prior to transport of the mRNA molecule to the cyto-
plasm. The 5
cap of the RNA transcript is required
Both Ribosomal RNAs & Most
both for efficient translation initiation and protection
Transfer RNAs Are Processed
of the 5′ end of mRNA from attack by 5′ → 3′ exonu-
From Larger Precursors
cleases. The secondary methylations of mRNA mole-
In mammalian cells, the three rRNA molecules are
cules, those on the 2′-hydroxy and the N6 of adenylyl
transcribed as part of a single large precursor molecule.
residues, occur after the mRNA molecule has appeared
The precursor is subsequently processed in the nu-
in the cytoplasm.
cleolus to provide the RNA component for the ribo-
Poly(A) tails are added to the 3′ end of mRNA mol-
some subunits found in the cytoplasm. The rRNA
ecules in a posttranscriptional processing step. The
genes are located in the nucleoli of mammalian cells.
mRNA is first cleaved about 20 nucleotides down-
Hundreds of copies of these genes are present in every
stream from an AAUAA recognition sequence. Another
cell. This large number of genes is required to synthe-
enzyme, poly(A) polymerase, adds a poly(A) tail which
size sufficient copies of each type of rRNA to form the
is subsequently extended to as many as 200 A residues.
107
ribosomes required for each cell replication.
The poly(A) tail appears to protect the
3′ end of
Whereas a single mRNA molecule may be copied into
mRNA from 3′ → 5′ exonuclease attack. The presence
105 protein molecules, providing a large amplification,
or absence of the poly(A) tail does not determine
the rRNAs are end products. This lack of amplification
whether a precursor molecule in the nucleus appears in
requires a large number of genes. Similarly, transfer
the cytoplasm, because all poly(A)-tailed hnRNA mole-
RNAs are often synthesized as precursors, with extra se-
cules do not contribute to cytoplasmic mRNA, nor do
quences both 5′ and 3′ of the sequences comprising the
all cytoplasmic mRNA molecules contain poly(A) tails
30 kb)
(˜
Liver
1B
1L
2A 2 3
4
56
7
8
9
10
30 kb)
(˜
B cell /pituitary
1B
1L
2A 2 3
4
56
7
8
9
10
Figure 37-14. Alternative promoter use in the liver and pancreatic B cell glucokinase
genes. Differential regulation of the glucokinase (GK) gene is accomplished by the use of
tissue-specific promoters. The B cell GK gene promoter and exon 1B are located about
30 kbp upstream from the liver promoter and exon 1L. Each promoter has a unique
structure and is regulated differently. Exons 2-10 are identical in the two genes, and the
GK proteins encoded by the liver and B cell mRNAs have identical kinetic properties.
356
/
CHAPTER 37
(the histones are most notable in this regard). Cytoplas-
mRNA into protein sequences. The tRNAs contain
mic enzymes in mammalian cells can both add and re-
many modifications of the standard bases A, U, G, and
move adenylyl residues from the poly(A) tails; this
C, including methylation, reduction, deamination, and
process has been associated with an alteration of mRNA
rearranged glycosidic bonds. Further modification of
stability and translatability.
the tRNA molecules includes nucleotide alkylations
The size of some cytoplasmic mRNA molecules,
and the attachment of the characteristic CpCpAOH ter-
even after the poly(A) tail is removed, is still consider-
minal at the 3′ end of the molecule by the enzyme nu-
ably greater than the size required to code for the spe-
cleotidyl transferase. The 3′ OH of the A ribose is the
cific protein for which it is a template, often by a factor
point of attachment for the specific amino acid that is
of 2 or 3. The extra nucleotides occur in untrans-
to enter into the polymerization reaction of protein
lated (non-protein coding) regions both 5′ and 3′ of
synthesis. The methylation of mammalian tRNA pre-
the coding region; the longest untranslated sequences
cursors probably occurs in the nucleus, whereas the
are usually at the 3′ end. The exact function of these se-
cleavage and attachment of CpCpAOH are cytoplasmic
quences is unknown, but they have been implicated in
functions, since the terminals turn over more rapidly
RNA processing, transport, degradation, and transla-
than do the tRNA molecules themselves. Enzymes
tion; each of these reactions potentially contributes ad-
within the cytoplasm of mammalian cells are required
ditional levels of control of gene expression.
for the attachment of amino acids to the CpCpAOH
residues. (See Chapter 38.)
RNA Editing Changes mRNA
After Transcription
RNA CAN ACT AS A CATALYST
The central dogma states that for a given gene and gene
In addition to the catalytic action served by the
product there is a linear relationship between the cod-
snRNAs in the formation of mRNA, several other
ing sequence in DNA, the mRNA sequence, and the
enzymatic functions have been attributed to RNA.
protein sequence (Figure 36-7). Changes in the DNA
Ribozymes are RNA molecules with catalytic activity.
sequence should be reflected in a change in the mRNA
These generally involve transesterification reactions,
sequence and, depending on codon usage, in protein se-
and most are concerned with RNA metabolism (splic-
quence. However, exceptions to this dogma have been
ing and endoribonuclease). Recently, a ribosomal RNA
recently documented. Coding information can be
component was noted to hydrolyze an aminoacyl ester
changed at the mRNA level by RNA editing. In such
and thus to play a central role in peptide bond function
cases, the coding sequence of the mRNA differs from
(peptidyl transferases; see Chapter 38). These observa-
that in the cognate DNA. An example is the apolipo-
tions, made in organelles from plants, yeast, viruses,
protein B (apoB) gene and mRNA. In liver, the single
and higher eukaryotic cells, show that RNA can act as
apoB gene is transcribed into an mRNA that directs the
an enzyme. This has revolutionized thinking about en-
synthesis of a 100-kDa protein, apoB100. In the intes-
zyme action and the origin of life itself.
tine, the same gene directs the synthesis of the primary
transcript; however, a cytidine deaminase converts a
CAA codon in the mRNA to UAA at a single specific
SUMMARY
site. Rather than encoding glutamine, this codon be-
• RNA is synthesized from a DNA template by the en-
comes a termination signal, and a
48-kDa protein
zyme RNA polymerase.
(apoB48) is the result. ApoB100 and apoB48 have dif-
• There are three distinct nuclear DNA-dependent
ferent functions in the two organs. A growing number
RNA polymerases in mammals: RNA polymerases I,
of other examples include a glutamine to arginine
II, and III. These enzymes control the transcriptional
change in the glutamate receptor and several changes
function—the transcription of rRNA, mRNA, and
in trypanosome mitochondrial mRNAs, generally in-
small RNA (tRNA/5S rRNA, snRNA) genes, respec-
volving the addition or deletion of uridine. The exact
tively.
extent of RNA editing is unknown, but current esti-
mates suggest that < 0.01% of mRNAs are edited in
• RNA polymerases interact with unique cis-active re-
this fashion.
gions of genes, termed promoters, in order to form
preinitiation complexes (PICs) capable of initiation.
In eukaryotes the process of PIC formation is facili-
Transfer RNA (tRNA) Is Extensively
tated by multiple general transcription factors
Processed & Modified
(GTFs), TFIIA, B, D, E, F, and H.
As described in Chapters 35 and 38, the tRNA mole-
• Eukaryotic PIC formation can occur either step-
cules serve as adapter molecules for the translation of
wise—by the sequential, ordered interactions of
RNA SYNTHESIS, PROCESSING, & MODIFICATION
/
357
GTFs and RNA polymerase with promoters—or in
REFERENCES
one step by the recognition of the promoter by a pre-
Busby S, Ebright RH: Promoter structure, promoter recognition,
formed GTF-RNA polymerase holoenzyme complex.
and transcription activation in prokaryotes. Cell
1994;79:
•
Transcription exhibits three phases: initiation, elon-
743.
gation, and termination. All are dependent upon dis-
Cramer P, Bushnell DA, Kornberg R: Structural basis of transcrip-
tinct DNA cis-elements and can be modulated by
tion: RNA polymerase II at 2.8 angstrom resolution. Science
distinct trans-acting protein factors.
2001;292:1863.
•
Most eukaryotic RNAs are synthesized as precursors
Fedor MJ: Ribozymes. Curr Biol 1998;8:R441.
that contain excess sequences which are removed
Gott JM, Emeson RB: Functions and mechanisms of RNA editing.
Ann Rev Genet 2000;34:499.
prior to the generation of mature, functional RNA.
Hirose Y, Manley JL: RNA polymerase II and the integration of
•
Eukaryotic mRNA synthesis results in a pre-mRNA
nuclear events. Genes Dev 2000;14:1415.
precursor that contains extensive amounts of excess
Keaveney M, Struhl K: Activator-mediated recruitment of the
RNA (introns) that must be precisely removed by
RNA polymerase machinery is the predominant mechanism
RNA splicing to generate functional, translatable
for transcriptional activation in yeast. Mol Cell 1998;1:917.
mRNA composed of exonic coding and noncoding
Lemon B, Tjian R: Orchestrated response: a symphony of tran-
sequences.
scription factors for gene control. Genes Dev 2000;14:2551.
•
All steps—from changes in DNA template, sequence,
Maniatis T, Reed R: An extensive network of coupling among gene
and accessibility in chromatin to RNA stability—are
expression machines. Nature 2002;416:499.
subject to modulation and hence are potential con-
Orphanides G, Reinberg D: A unified theory of gene expression.
Cell 2002;108:439.
trol sites for eukaryotic gene regulation.
Shatkin AJ, Manley JL: The ends of the affair: capping and poly-
adenylation. Nat Struct Biol 2000;7:838.
Stevens SW et al: Composition and functional characterization of
the yeast spliceosomal penta-snRNP. Mol Cell 2002;9:31.
Tucker M, Parker R: Mechanisms and control of mRNA decap-
ping in Saccharomyces cerevisiae. Ann Rev Biochem 2000;69:
571.
Woychik NA, Hampsey M: The RNA polymerase II machinery:
structure illuminates function. Cell 2002;108:453.
Protein Synthesis & the
38
Genetic Code
Daryl K. Granner, MD
BIOMEDICAL IMPORTANCE
The cell must possess the machinery necessary to
translate information accurately and efficiently from
The letters A, G, T, and C correspond to the nu-
the nucleotide sequence of an mRNA into the sequence
cleotides found in DNA. They are organized into three-
of amino acids of the corresponding specific protein.
letter code words called codons, and the collection of
Clarification of our understanding of this process,
these codons makes up the genetic code. It was impos-
which is termed translation, awaited deciphering of the
sible to understand protein synthesis—or to explain
genetic code. It was realized early that mRNA mole-
mutations—before the genetic code was elucidated.
cules themselves have no affinity for amino acids and,
The code provides a foundation for explaining the way
therefore, that the translation of the information in the
in which protein defects may cause genetic disease and
mRNA nucleotide sequence into the amino acid se-
for the diagnosis and perhaps the treatment of these
quence of a protein requires an intermediate adapter
disorders. In addition, the pathophysiology of many
molecule. This adapter molecule must recognize a spe-
viral infections is related to the ability of these agents to
cific nucleotide sequence on the one hand as well as a
disrupt host cell protein synthesis. Many antibacterial
specific amino acid on the other. With such an adapter
agents are effective because they selectively disrupt pro-
molecule, the cell can direct a specific amino acid into
tein synthesis in the invading bacterial cell but do not
the proper sequential position of a protein during its
affect protein synthesis in eukaryotic cells.
synthesis as dictated by the nucleotide sequence of the
specific mRNA. In fact, the functional groups of the
amino acids do not themselves actually come into con-
GENETIC INFORMATION FLOWS
tact with the mRNA template.
FROM DNA TO RNA TO PROTEIN
The genetic information within the nucleotide se-
THE NUCLEOTIDE SEQUENCE
quence of DNA is transcribed in the nucleus into the
specific nucleotide sequence of an RNA molecule. The
OF AN mRNA MOLECULE CONSISTS
sequence of nucleotides in the RNA transcript is com-
OF A SERIES OF CODONS THAT SPECIFY
plementary to the nucleotide sequence of the template
THE AMINO ACID SEQUENCE OF THE
strand of its gene in accordance with the base-pairing
ENCODED PROTEIN
rules. Several different classes of RNA combine to di-
rect the synthesis of proteins.
Twenty different amino acids are required for the syn-
In prokaryotes there is a linear correspondence be-
thesis of the cellular complement of proteins; thus,
tween the gene, the messenger RNA (mRNA) tran-
there must be at least 20 distinct codons that make up
scribed from the gene, and the polypeptide product.
the genetic code. Since there are only four different nu-
The situation is more complicated in higher eukaryotic
cleotides in mRNA, each codon must consist of more
cells, in which the primary transcript is much larger
than a single purine or pyrimidine nucleotide. Codons
than the mature mRNA. The large mRNA precursors
consisting of two nucleotides each could provide for
contain coding regions (exons) that will form the ma-
only 16 (42) specific codons, whereas codons of three
ture mRNA and long intervening sequences (introns)
nucleotides could provide 64 (43) specific codons.
that separate the exons. The hnRNA is processed
It is now known that each codon consists of a se-
within the nucleus, and the introns, which often make
quence of three nucleotides; ie, it is a triplet code
up much more of this RNA than the exons, are re-
(see Table 38-1). The deciphering of the genetic code
moved. Exons are spliced together to form mature
depended heavily on the chemical synthesis of nu-
mRNA, which is transported to the cytoplasm, where it
cleotide polymers, particularly triplets in repeated se-
is translated into protein.
quence.
358
PROTEIN SYNTHESIS & THE GENETIC CODE
/
359
Table 38-1. The genetic code (codon
the code. However, for any specific codon, only a single
assignments in mammalian messenger RNA).1
amino acid is indicated; with rare exceptions, the ge-
netic code is unambiguous—ie, given a specific codon,
only a single amino acid is indicated. The distinction
First
Second
Third
between ambiguity and degeneracy is an important
Nucleotide
Nucleotide
Nucleotide
concept.
U
C
A
G
The unambiguous but degenerate code can be ex-
plained in molecular terms. The recognition of specific
Phe
Ser
Tyr
Cys
U
Phe
Ser
Tyr
Cys
C
codons in the mRNA by the tRNA adapter molecules is
U
Leu
Ser
Term
Term2
A
dependent upon their anticodon region and specific
Leu
Ser
Term
Trp
G
base-pairing rules. Each tRNA molecule contains a spe-
cific sequence, complementary to a codon, which is
Leu
Pro
His
Arg
U
termed its anticodon. For a given codon in the mRNA,
Leu
Pro
His
Arg
C
only a single species of tRNA molecule possesses the
C
Leu
Pro
Gln
Arg
A
proper anticodon. Since each tRNA molecule can be
Leu
Pro
Gln
Arg
G
charged with only one specific amino acid, each codon
Ile
Thr
Asn
Ser
U
therefore specifies only one amino acid. However, some
Ile
Thr
Asn
Ser
C
tRNA molecules can utilize the anticodon to recognize
A
Ile2
Thr
Lys
Arg2
A
more than one codon. With few exceptions, given a
Met
Thr
Lys
Arg2
G
specific codon, only a specific amino acid will be in-
Val
Ala
Asp
Gly
U
corporated—although, given a specific amino acid,
Val
Ala
Asp
Gly
C
more than one codon may be used.
G
Val
Ala
Glu
Gly
A
As discussed below, the reading of the genetic code
Val
Ala
Glu
Gly
G
during the process of protein synthesis does not involve
any overlap of codons. Thus, the genetic code is
1The terms first, second, and third nucleotide refer to the indi-
nonoverlapping. Furthermore, once the reading is
vidual nucleotides of a triplet codon. U, uridine nucleotide;
C, cytosine nucleotide; A, adenine nucleotide; G, guanine nu-
commenced at a specific codon, there is no punctua-
cleotide; Term, chain terminator codon. AUG, which codes for
tion between codons, and the message is read in a con-
Met, serves as the initiator codon in mammalian cells and en-
tinuing sequence of nucleotide triplets until a transla-
codes for internal methionines in a protein. (Abbreviations of
tion stop codon is reached.
amino acids are explained in Chapter 3.)
Until recently, the genetic code was thought to be
2In mammalian mitochondria, AUA codes for Met and UGA for
universal. It has now been shown that the set of tRNA
Trp, and AGA and AGG serve as chain terminators.
molecules in mitochondria (which contain their own
separate and distinct set of translation machinery) from
lower and higher eukaryotes, including humans, reads
THE GENETIC CODE IS DEGENERATE,
four codons differently from the tRNA molecules in
the cytoplasm of even the same cells. As noted in Table
UNAMBIGUOUS, NONOVERLAPPING,
38-1, the codon AUA is read as Met, and UGA codes
WITHOUT PUNCTUATION, & UNIVERSAL
for Trp in mammalian mitochondria. In addition, in
Three of the 64 possible codons do not code for specific
mitochondria, the codons AGA and AGG are read as
amino acids; these have been termed nonsense codons.
stop or chain terminator codons rather than as Arg. As
These nonsense codons are utilized in the cell as termi-
a result, mitochondria require only 22 tRNA molecules
nation signals; they specify where the polymerization
to read their genetic code, whereas the cytoplasmic
of amino acids into a protein molecule is to stop. The
translation system possesses a full complement of 31
remaining 61 codons code for 20 amino acids (Table
tRNA species. These exceptions noted, the genetic
38-1). Thus, there must be “degeneracy” in the ge-
code is universal. The frequency of use of each amino
netic code—ie, multiple codons must decode the same
acid codon varies considerably between species and
amino acid. Some amino acids are encoded by several
among different tissues within a species. The specific
codons; for example, six different codons specify serine.
tRNA levels generally mirror these codon usage biases.
Other amino acids, such as methionine and trypto-
Thus, a particular abundantly used codon is decoded
phan, have a single codon. In general, the third nu-
by a similarly abundant specific tRNA which recognizes
cleotide in a codon is less important than the first two
that particular codon. Tables of codon usage are be-
in determining the specific amino acid to be incorpo-
coming more accurate as more genes are sequenced.
rated, and this accounts for most of the degeneracy of
This is of considerable importance because investigators
360
/
CHAPTER 38
Table 38-2. Features of the genetic code.
tRNA synthetases. They form an activated intermedi-
ate of aminoacyl-AMP-enzyme complex (Figure 38-1).
The specific aminoacyl-AMP-enzyme complex then
• Degenerate
recognizes a specific tRNA to which it attaches the
• Unambiguous
• Nonoverlapping
aminoacyl moiety at the 3′-hydroxyl adenosyl terminal.
• Not punctuated
The charging reactions have an error rate of less than
• Universal
10−4 and so are extremely accurate. The amino acid re-
mains attached to its specific tRNA in an ester linkage
until it is polymerized at a specific position in the fabri-
cation of a polypeptide precursor of a protein molecule.
often need to deduce mRNA structure from the pri-
The regions of the tRNA molecule referred to in
mary sequence of a portion of protein in order to syn-
Chapter 35 (and illustrated in Figure 35-11) now be-
thesize an oligonucleotide probe and initiate a recombi-
come important. The thymidine-pseudouridine-cyti-
nant DNA cloning project. The main features of the
dine (TΨC) arm is involved in binding of the amino-
genetic code are listed in Table 38-2.
acyl-tRNA to the ribosomal surface at the site of
protein synthesis. The D arm is one of the sites impor-
tant for the proper recognition of a given tRNA species
AT LEAST ONE SPECIES OF TRANSFER
by its proper aminoacyl-tRNA synthetase. The acceptor
arm, located at the 3′-hydroxyl adenosyl terminal, is the
RNA (tRNA) EXISTS FOR EACH OF THE
site of attachment of the specific amino acid.
20 AMINO ACIDS
The anticodon region consists of seven nucleotides,
tRNA molecules have extraordinarily similar functions
and it recognizes the three-letter codon in mRNA (Fig-
and three-dimensional structures. The adapter function
ure 38-2). The sequence read from the 3′ to 5′ direc-
of the tRNA molecules requires the charging of each
tion in that anticodon loop consists of a variable
specific tRNA with its specific amino acid. Since there
base-modified purine-XYZ-pyrimidine-pyrimidine-
is no affinity of nucleic acids for specific functional
5′. Note that this direction of reading the anticodon is
groups of amino acids, this recognition must be carried
3′ to 5′, whereas the genetic code in Table 38-1 is read
out by a protein molecule capable of recognizing both a
5′ to 3′, since the codon and the anticodon loop of the
specific tRNA molecule and a specific amino acid. At
mRNA and tRNA molecules, respectively, are antipar-
least 20 specific enzymes are required for these specific
allel in their complementarity just like all other inter-
recognition functions and for the proper attachment of
molecular interactions between nucleic acid strands.
the 20 amino acids to specific tRNA molecules. The
The degeneracy of the genetic code resides mostly in
process of recognition and attachment
(charging)
the last nucleotide of the codon triplet, suggesting that
proceeds in two steps by one enzyme for each of the 20
the base pairing between this last nucleotide and the
amino acids. These enzymes are termed aminoacyl-
corresponding nucleotide of the anticodon is not strictly
ATP
PPi
AMP + Enz
O
O
HOOC HC R
Enz
•
Adenosine
O P O
C
CH
R
H2N
OH
NH2
Enzyme (Enz)
Enz•AMP-aa
tRNA
tRNA-aa
(Activated amino acid)
AMINOACYL-
tRNA SYNTHETASE
Amino acid (aa)
Aminoacyl-AMP-enzyme
Aminoacyl-tRNA
complex
Figure 38-1. Formation of aminoacyl-tRNA. A two-step reaction, involving the enzyme
aminoacyl-tRNA synthetase, results in the formation of aminoacyl-tRNA. The first reaction in-
volves the formation of an AMP-amino acid-enzyme complex. This activated amino acid is next
transferred to the corresponding tRNA molecule. The AMP and enzyme are released, and the lat-
ter can be reutilized. The charging reactions have an error rate of less than 10-4 and so are ex-
tremely accurate.
PROTEIN SYNTHESIS & THE GENETIC CODE
/
361
mRNA
purine is changed to the other purine. Transversions are
Codon
5′
3′
changes from a purine to either of the two pyrimidines
U • U • U
Anticodon
or the change of a pyrimidine into either of the two
A • A • A
•
•
Py
purines, as shown in Figure 38-3.
•Pu*
•
N
Anticodon
Py
If the nucleotide sequence of the gene containing
arm
the mutation is transcribed into an RNA molecule,
then the RNA molecule will possess a complementary
D
base change at this corresponding locus.
TψC
arm
arm
Single-base changes in the mRNA molecules may
have one of several effects when translated into protein:
Phenylalanyl-
(1) There may be no detectable effect because of the
tRNA
degeneracy of the code. This would be more likely if
5′
the changed base in the mRNA molecule were to be at
C
•
the third nucleotide of a codon; such mutations are
C
Acceptor arm
often referred to as silent mutations. Because of wob-
•
A
ble, the translation of a codon is least sensitive to a
3′
change at the third position.
Phe
(2) A missense effect will occur when a different
amino acid is incorporated at the corresponding site in
Figure 38-2. Recognition of the codon by the anti-
the protein molecule. This mistaken amino acid—or
codon. One of the codons for phenylalanine is UUU.
missense, depending upon its location in the specific
tRNA charged with phenylalanine (Phe) has the com-
protein—might be acceptable, partially acceptable, or
plementary sequence AAA; hence, it forms a base-pair
unacceptable to the function of that protein molecule.
complex with the codon. The anticodon region typi-
From a careful examination of the genetic code, one
cally consists of a sequence of seven nucleotides: vari-
can conclude that most single-base changes would re-
able (N), modified purine ((Pu*), X, Y, Z, and two pyrim-
sult in the replacement of one amino acid by another
idines (Py) in the 3′ to 5′ direction.
with rather similar functional groups. This is an effec-
tive mechanism to avoid drastic change in the physical
properties of a protein molecule. If an acceptable mis-
sense effect occurs, the resulting protein molecule may
by the Watson-Crick rule. This is called wobble; the
pairing of the codon and anticodon can “wobble” at this
not be distinguishable from the normal one. A partially
acceptable missense will result in a protein molecule
specific nucleotide-to-nucleotide pairing site. For exam-
ple, the two codons for arginine, AGA and AGG, can
with partial but abnormal function. If an unacceptable
missense effect occurs, then the protein molecule will
bind to the same anticodon having a uracil at its 5′ end
(UCU). Similarly, three codons for glycine—GGU,
not be capable of functioning in its assigned role.
(3) A nonsense codon may appear that would then
GGC, and GGA—can form a base pair from one anti-
codon, CCI. I is an inosine nucleotide, another of the
result in the premature termination of amino acid in-
corporation into a peptide chain and the production of
peculiar bases appearing in tRNA molecules.
only a fragment of the intended protein molecule. The
probability is high that a prematurely terminated pro-
MUTATIONS RESULT WHEN CHANGES
tein molecule or peptide fragment will not function in
OCCUR IN THE NUCLEOTIDE SEQUENCE
its assigned role.
Although the initial change may not occur in the tem-
plate strand of the double-stranded DNA molecule for
that gene, after replication, daughter DNA molecules
T
C
T
A A
T
with mutations in the template strand will segregate
and appear in the population of organisms.
Some Mutations Occur
A
G
C
G G
C
by Base Substitution
Transitions
Transversions
Single-base changes (point mutations) may be transi-
tions or transversions. In the former, a given pyrimi-
Figure 38-3. Diagrammatic representation of transi-
dine is changed to the other pyrimidine or a given
tion mutations and transversion mutations.
362
/
CHAPTER 38
Hemoglobin Illustrates the Effects of
to possess at position 67 a codon GUA or GUG in
Single-Base Changes in Structural Genes
order that a single nucleotide change could provide for
the appearance of the glutamic acid codons GAA or
Some mutations have no apparent effect. The gene
GAG. Hemoglobin Sydney, which contains an alanine
system that encodes hemoglobin is one of the best-
at position 67, could have arisen by the change of a sin-
studied in humans. The lack of effect of a single-base
gle nucleotide in any of the four codons for valine
change is demonstrable only by sequencing the nu-
(GUU, GUC, GUA, or GUG) to the alanine codons
cleotides in the mRNA molecules or structural genes.
(GCU, GCC, GCA, or GCG, respectively).
The sequencing of a large number of hemoglobin
mRNAs and genes from many individuals has shown
that the codon for valine at position 67 of the β chain
Substitution of Amino Acids Causes
of hemoglobin is not identical in all persons who pos-
Missense Mutations
sess a normally functional β chain of hemoglobin. He-
A. ACCEPTABLE MISSENSE MUTATIONS
moglobin Milwaukee has at position 67 a glutamic
acid; hemoglobin Bristol contains aspartic acid at posi-
An example of an acceptable missense mutation (Figure
tion 67. In order to account for the amino acid change
38-4, top) in the structural gene for the β chain of he-
by the change of a single nucleotide residue in the
moglobin could be detected by the presence of an elec-
codon for amino acid 67, one must infer that the
trophoretically altered hemoglobin in the red cells of an
mRNA encoding hemoglobin Bristol possessed a GUU
apparently healthy individual. Hemoglobin Hikari has
or GUC codon prior to a later change to GAU or
been found in at least two families of Japanese people.
GAC, both codons for aspartic acid. However, the
This hemoglobin has asparagine substituted for lysine
mRNA encoding hemoglobin Milwaukee would have
at the 61 position in the β chain. The corresponding
Protein molecule
Amino acid
Codons
Hb A, β chain
61 Lysine
AAA
or
AAG
Acceptable
missense
Hb Hikari, β chain
Asparagine
AAU
or
AAC
Hb A, β chain
6 Glutamate
GAA
or
GAG
Partially
acceptable
missense
Hb S, β chain
Valine
GUA
or
GUG
Hb A, α chain
58 Histidine
CAU
or
CAC
Unacceptable
missense
Hb M (Boston), α chain
Tyrosine
UAU
or
UAC
Figure 38-4. Examples of three types of missense mutations resulting in abnormal hemoglo-
bin chains. The amino acid alterations and possible alterations in the respective codons are indi-
cated. The hemoglobin Hikari β-chain mutation has apparently normal physiologic properties
but is electrophoretically altered. Hemoglobin S has a β-chain mutation and partial function; he-
moglobin S binds oxygen but precipitates when deoxygenated. Hemoglobin M Boston, an
α-chain mutation, permits the oxidation of the heme ferrous iron to the ferric state and so will
not bind oxygen at all.
PROTEIN SYNTHESIS & THE GENETIC CODE
/
363
transversion might be either AAA or AAG changed to
mal termination codon (nonsense codon), the reading
either AAU or AAC. The replacement of the specific ly-
of the normal termination signal is disturbed. Such a
sine with asparagine apparently does not alter the nor-
deletion might result in reading through a termination
mal function of the β chain in these individuals.
signal until another nonsense codon is encountered (ex-
ample 1, Figure 38-5). Examples of this phenomenon
B. PARTIALLY ACCEPTABLE MISSENSE MUTATIONS
are described in discussions of hemoglobinopathies.
A partially acceptable missense mutation (Figure 38-4,
Insertions of one or two or nonmultiples of three nu-
center) is best exemplified by hemoglobin S, which is
cleotides into a gene result in an mRNA in which the
found in sickle cell anemia. Here glutamic acid, the
reading frame is distorted upon translation, and the same
normal amino acid in position 6 of the β chain, has
effects that occur with deletions are reflected in the
been replaced by valine. The corresponding single nu-
mRNA translation. This may result in garbled amino
cleotide change within the codon would be GAA or
acid sequences distal to the insertion and the generation
GAG of glutamic acid to GUA or GUG of valine.
of a nonsense codon at or distal to the insertion, or per-
Clearly, this missense mutation hinders normal func-
haps reading through the normal termination codon.
tion and results in sickle cell anemia when the mutant
Following a deletion in a gene, an insertion (or vice
gene is present in the homozygous state. The gluta-
versa) can reestablish the proper reading frame (exam-
mate-to-valine change may be considered to be partially
ple 4, Figure 38-5). The corresponding mRNA, when
acceptable because hemoglobin S does bind and release
translated, would contain a garbled amino acid sequence
oxygen, although abnormally.
between the insertion and deletion. Beyond the reestab-
lishment of the reading frame, the amino acid sequence
C. UNACCEPTABLE MISSENSE MUTATIONS
would be correct. One can imagine that different com-
An unacceptable missense mutation (Figure 38-4, bot-
binations of deletions, of insertions, or of both would
tom) in a hemoglobin gene generates a nonfunctioning
result in formation of a protein wherein a portion is ab-
hemoglobin molecule. For example, the hemoglobin M
normal, but this portion is surrounded by the normal
mutations generate molecules that allow the Fe2+ of the
amino acid sequences. Such phenomena have been
heme moiety to be oxidized to Fe3+, producing methe-
demonstrated convincingly in a number of diseases.
moglobin. Methemoglobin cannot transport oxygen
(see Chapter 6).
Suppressor Mutations Can Counteract
Some of the Effects of Missense,
Frameshift Mutations Result From
Nonsense, & Frameshift Mutations
Deletion or Insertion of Nucleotides in
DNA That Generates Altered mRNAs
The above discussion of the altered protein products of
gene mutations is based on the presence of normally
The deletion of a single nucleotide from the coding
functioning tRNA molecules. However, in prokaryotic
strand of a gene results in an altered reading frame in
and lower eukaryotic organisms, abnormally function-
the mRNA. The machinery translating the mRNA does
ing tRNA molecules have been discovered that are
not recognize that a base was missing, since there is no
themselves the results of mutations. Some of these ab-
punctuation in the reading of codons. Thus, a major al-
normal tRNA molecules are capable of binding to and
teration in the sequence of polymerized amino acids, as
decoding altered codons, thereby suppressing the effects
depicted in example 1, Figure 38-5, results. Altering
of mutations in distant structural genes. These sup-
the reading frame results in a garbled translation of the
pressor tRNA molecules, usually formed as the result
mRNA distal to the single nucleotide deletion. Not
of alterations in their anticodon regions, are capable of
only is the sequence of amino acids distal to this dele-
suppressing missense mutations, nonsense mutations,
tion garbled, but reading of the message can also result
and frameshift mutations. However, since the suppres-
in the appearance of a nonsense codon and thus the
sor tRNA molecules are not capable of distinguishing
production of a polypeptide both garbled and prema-
between a normal codon and one resulting from a gene
turely terminated (example 3, Figure 38-5).
mutation, their presence in a cell usually results in de-
If three nucleotides or a multiple of three are deleted
creased viability. For instance, the nonsense suppressor
from a coding region, the corresponding mRNA when
tRNA molecules can suppress the normal termination
translated will provide a protein from which is missing
signals to allow a read-through when it is not desirable.
the corresponding number of amino acids (example 2,
Frameshift suppressor tRNA molecules may read a nor-
Figure 38-5). Because the reading frame is a triplet, the
mal codon plus a component of a juxtaposed codon to
reading phase will not be disturbed for those codons
provide a frameshift, also when it is not desirable. Sup-
distal to the deletion. If, however, deletion of one or
pressor tRNA molecules may exist in mammalian cells,
two nucleotides occurs just prior to or within the nor-
since read-through transcription occurs.
364
/
CHAPTER 38
Normal
Wild type
mRNA
5'...
UAG UUUG AUG GCC UCU UGC AAA GGC UAU AGU AGU UAG...
3'
Polypeptide
Met
Ala
Ser
Cys
Lys
Gly
Tyr
Ser
Ser
STOP
Example 1
Deletion (-1)
-1 U
mRNA
5'...
UAG UUUG AUG GCC CUU GCA AAG GCU AUA GUA GUU AG...
3'
Polypeptide
Met
Ala
Leu
Ala
Lys
Ala
Thr
Val
Val
Ser
Garbled
Example 2
Deletion (- 3)
- 3 UGC
mRNA
5'...
UAG UUUG AUG GCC UCU AAA GGC UAU AGU AGU UAG...
3'
Polypeptide
Met
Ala
Ser
Lys
Gly
Try
Ser
Ser
STOP
Example 3
Insertion (+1)
+1 C
mRNA
5'...
UAG UUUG AUG GCC CUC UUG CAA AGG CUA UAG UAG UUAG...
3'
Polypeptide
Met
Ala
Leu
Leu
Gln
Arg
Leu
STOP
Garbled
Example 4
Insertion (+1)
Deletion (-1)
+1 U
-1 C
mRNA
5'...
UAG UUUG AUG GCC UCU UUG CAA AGG UAU AGU AGU UAG...
3'
Polypeptide
Met
Ala
Ser
Leu
Gln
Arg
Tyr
Ser
Ser
STOP
Garbled
Figure 38-5. Examples of the effects of deletions and insertions in a gene on the sequence of the mRNA
transcript and of the polypeptide chain translated therefrom. The arrows indicate the sites of deletions or inser-
tions, and the numbers in the ovals indicate the number of nucleotide residues deleted or inserted. Blue type
indicates amino acids in correct order.
LIKE TRANSCRIPTION, PROTEIN
The translation of the mRNA commences near its 5′
terminal with the formation of the corresponding
SYNTHESIS CAN BE DESCRIBED
amino terminal of the protein molecule. The message is
IN THREE PHASES: INITIATION,
read from 5′ to 3′, concluding with the formation of
ELONGATION, & TERMINATION
the carboxyl terminal of the protein. Again, the concept
of polarity is apparent. As described in Chapter 37, the
The general structural characteristics of ribosomes and
transcription of a gene into the corresponding mRNA
their self-assembly process are discussed in Chapter 37.
or its precursor first forms the 5′ terminal of the RNA
These particulate entities serve as the machinery on
molecule. In prokaryotes, this allows for the beginning
which the mRNA nucleotide sequence is translated into
of mRNA translation before the transcription of the
the sequence of amino acids of the specified protein.
gene is completed. In eukaryotic organisms, the process
PROTEIN SYNTHESIS & THE GENETIC CODE
/
365
of transcription is a nuclear one; mRNA translation oc-
particularly interesting in this regard. This kinase is ac-
curs in the cytoplasm. This precludes simultaneous
tivated by viruses and provides a host defense mecha-
transcription and translation in eukaryotic organisms
nism that decreases protein synthesis, thereby inhibit-
and makes possible the processing necessary to generate
ing viral replication. Phosphorylated eIF-2α binds
mature mRNA from the primary transcript—hnRNA.
tightly to and inactivates the GTP-GDP recycling pro-
tein eIF-2B. This prevents formation of the 43S preini-
tiation complex and blocks protein synthesis.
Initiation Involves Several Protein-RNA
Complexes (Figure 38-6)
OMPLEX
C. FORMATION OF THE 43S INITIATION C
Initiation of protein synthesis requires that an mRNA
The 5′ terminals of most mRNA molecules in eukary-
molecule be selected for translation by a ribosome.
otic cells are “capped,” as described in Chapter 37. This
Once the mRNA binds to the ribosome, the latter finds
methyl-guanosyl triphosphate cap facilitates the bind-
the correct reading frame on the mRNA, and transla-
ing of mRNA to the 43S preinitiation complex. A cap
tion begins. This process involves tRNA, rRNA,
binding protein complex, eIF-4F (4F), which consists
mRNA, and at least ten eukaryotic initiation factors
of eIF-4E and the eIF-4G (4G)-eIF4A (4A) complex,
(eIFs), some of which have multiple (three to eight)
binds to the cap through the 4E protein. Then eIF-4A
subunits. Also involved are GTP, ATP, and amino
(4A) and eIF-4B (4B) bind and reduce the complex sec-
acids. Initiation can be divided into four steps: (1) dis-
ondary structure of the 5′ end of the mRNA through
sociation of the ribosome into its 40S and 60S sub-
ATPase and ATP-dependent helicase activities. The as-
units; (2) binding of a ternary complex consisting of
sociation of mRNA with the 43S preinitiation complex
met-tRNAi, GTP, and eIF-2 to the 40S ribosome to
to form the 48S initiation complex requires ATP hy-
form a preinitiation complex; (3) binding of mRNA to
drolysis. eIF-3 is a key protein because it binds with
the 40S preinitiation complex to form a 43S initiation
high affinity to the 4G component of 4F, and it links
complex; and (4) combination of the 43S initiation
this complex to the 40S ribosomal subunit. Following
complex with the 60S ribosomal subunit to form the
association of the 43S preinitiation complex with the
80S initiation complex.
mRNA cap and reduction (“melting”) of the secondary
structure near the 5′ end of the mRNA, the complex
A. RIBOSOMAL DISSOCIATION
scans the mRNA for a suitable initiation codon. Gener-
Two initiation factors, eIF-3 and eIF-1A, bind to the
ally this is the 5′-most AUG, but the precise initiation
newly dissociated 40S ribosomal subunit. This delays
codon is determined by so-called Kozak consensus se-
its reassociation with the 60S subunit and allows other
quences that surround the AUG:
translation initiation factors to associate with the 40S
subunit.
−3
−1
+4
B. FORMATION OF THE 43S PREINITIATION COMPLEX
GCCA / GCCAUGG
The first step in this process involves the binding of
GTP by eIF-2. This binary complex then binds to met-
Most preferred is the presence of a purine at positions
tRNAi, a tRNA specifically involved in binding to the
−3 and +4 relative to the AUG.
initiation codon AUG. (There are two tRNAs for me-
thionine. One specifies methionine for the initiator
D. ROLE OF THE POLY(A) TAIL IN INITIATION
codon, the other for internal methionines. Each has a
unique nucleotide sequence.) This ternary complex
Biochemical and genetic experiments in yeast have re-
binds to the 40S ribosomal subunit to form the 43S
vealed that the 3′ poly(A) tail and its binding protein,
preinitiation complex, which is stabilized by association
Pab1p, are required for efficient initiation of protein
with eIF-3 and eIF-1A.
synthesis. Further studies showed that the poly(A) tail
eIF-2 is one of two control points for protein syn-
stimulates recruitment of the 40S ribosomal subunit to
thesis initiation in eukaryotic cells. eIF-2 consists of
the mRNA through a complex set of interactions.
α, β, and γ subunits. eIF-2α is phosphorylated (on
Pab1p, bound to the poly(A) tail, interacts with eIF-4G,
serine
51) by at least four different protein kinases
which in turn binds to eIF-4E that is bound to the cap
(HCR, PKR, PERK, and GCN2) that are activated
structure. It is possible that a circular structure is
when a cell is under stress and when the energy expen-
formed and that this helps direct the 40S ribosomal
diture required for protein synthesis would be deleteri-
subunit to the 5′ end of the mRNA. This helps explain
ous. Such conditions include amino acid and glucose
how the cap and poly(A) tail structures have a synergis-
starvation, virus infection, misfolded proteins, serum
tic effect on protein synthesis. It appears that a similar
deprivation, hyperosmolality, and heat shock. PKR is
mechanism is at work in mammalian cells.
Ternary complex
Formation of the 80S
Activation of mRNA
formation
initiation complex
80S
Ribosomal
dissociation
3
1A
60S
Met
40S
3
1A
eIF-2C
2
Cap
AUG
(A)n
Ternary
ATP
4F
=
4E
+
4G
4A
complex
Met
2
43S Preinitiation
4F
Cap
AUG
(A)
complex
n
3
ATP
4A 4B
2B
1A
2
i
ADP + P
Met
4A
4F
Cap
AUG
(A)
n
4B
2B
2
4F
Cap
AUG
(A)n
3
GDP
1A
2
Met
GTP
ATP
ADP + Pi
2B
2
Cap
AUG
(A)n
48S Initiation
3
complex
1A
2
2B
Met
eIF-5
2
+ P
i
+
1A
+
3
Cap
(A)n
P site
A site
Met
80S Initiation
complex
Elongation
Figure 38-6.
Diagrammatic representation of the initiation of protein synthesis on the mRNA template contain-
ing a 5′ cap (GmTP-5′) and 3′ poly(A) terminal [3′(A)n]. This process proceeds in three steps: (1) activation of mRNA;
(2) formation of the ternary complex consisting of tRNAmeti, initiation factor eIF-2, and GTP; and (3) formation of the
active 80S initiation complex. (See text for details.) GTP, •; GDP, . The various initiation factors appear in abbrevi-
ated form as circles or squares, eg, eIF-3 ( 3 ), eIF-4F ( 4F ). 4•F is a complex consisting of 4E and 4A bound to 4G (see
Figure 38-7). The constellation of protein factors and the 40S ribosomal subunit comprise the 43S preinitiation com-
plex. When bound to mRNA, this forms the 48S preinitiation complex.
PROTEIN SYNTHESIS AND THE GENETIC CODE
/
367
E. FORMATION OF THE 80S INITIATION COMPLEX
PO4
The binding of the 60S ribosomal subunit to the 48S
initiation complex involves hydrolysis of the GTP
PO4
bound to eIF-2 by eIF-5. This reaction results in release
of the initiation factors bound to the 48S initiation
eIF-4E
eIF-4E
complex (these factors then are recycled) and the rapid
Insulin
association of the 40S and 60S subunits to form the
(kinase
80S ribosome. At this point, the met-tRNAi is on the P
activation)
eIF-4G
site of the ribosome, ready for the elongation cycle to
commence.
eIF-4A
The Regulation of eIF-4E Controls
the Rate of Initiation
The 4F complex is particularly important in controlling
the rate of protein translation. As described above, 4F is
eIF-4G
eIF-4F
a complex consisting of 4E, which binds to the m7G
complex
eIF-4E
eIF-4A
cap structure at the 5′ end of the mRNA, and 4G,
which serves as a scaffolding protein. In addition to
PO4
binding 4E, 4G binds to eIF-3, which links the com-
plex to the 40S ribosomal subunit. It also binds 4A and
4B, the ATPase-helicase complex that helps unwind the
4F
Cap
AUG
(A)n
RNA (Figure 38-7).
4E is responsible for recognition of the mRNA cap
Figure 38-7. Activation of eIF-4E by insulin and for-
structure, which is a rate-limiting step in translation.
mation of the cap binding eIF-4F complex. The 4F-cap
This process is regulated at two levels. Insulin and mi-
mRNA complex is depicted as in Figure 38-6. The 4F
togenic growth factors result in the phosphorylation of
complex consists of eIF-4E (4E), eIF-4A, and eIF-4G. 4E is
4E on ser 209 (or thr 210). Phosphorylated 4E binds to
inactive when bound by one of a family of binding pro-
the cap much more avidly than does the nonphospho-
teins (4E-BPs). Insulin and mitogenic factors (eg, IGF-1,
rylated form, thus enhancing the rate of initiation. A
PDGF, interleukin-2, and angiotensin II) activate a serine
component of the MAP kinase pathway (see Figure
protein kinase in the mTOR pathway, and this results in
43-8) appears to be involved in this phosphorylation
the phosphorylation of 4E-BP. Phosphorylated 4E-BP
reaction.
dissociates from 4E, and the latter is then able to form
The activity of 4E is regulated in a second way, and
the 4F complex and bind to the mRNA cap. These
this also involves phosphorylation. A recently discov-
growth peptides also phosphorylate 4E itself by activat-
ered set of proteins bind to and inactivate 4E. These
ing a component of the MAP kinase pathway. Phos-
proteins include 4E-BP1 (BP1, also known as PHAS-1)
phorylated 4E binds much more avidly to the cap than
and the closely related proteins 4E-BP2 and 4E-BP3.
does nonphosphorylated 4E.
BP1 binds with high affinity to 4E. The [4E]•[BP1] as-
sociation prevents 4E from binding to 4G (to form 4F).
Since this interaction is essential for the binding of 4F
to the ribosomal 40S subunit and for correctly posi-
increase of protein synthesis in liver, adipose tissue, and
tioning this on the capped mRNA, BP-1 effectively in-
muscle.
hibits translation initiation.
Insulin and other growth factors result in the phos-
Elongation Also Is a Multistep Process
phorylation of BP-1 at five unique sites. Phosphoryla-
(Figure 38-8)
tion of BP-1 results in its dissociation from 4E, and it
cannot rebind until critical sites are dephosphorylated.
Elongation is a cyclic process on the ribosome in which
The protein kinase responsible has not been identified,
one amino acid at a time is added to the nascent peptide
but it appears to be different from the one that phos-
chain. The peptide sequence is determined by the order
phorylates 4E. A kinase in the mammalian target of
of the codons in the mRNA. Elongation involves several
rapamycin (mTOR) pathway, perhaps mTOR itself, is
steps catalyzed by proteins called elongation factors (EFs).
involved. These effects on the activation of 4E explain
These steps are (1) binding of aminoacyl-tRNA to the A
in part how insulin causes a marked posttranscriptional
site, (2) peptide bond formation, and (3) translocation.
368
/
CHAPTER 38
n
n+1
A. BINDING OF AMINOACYL-TRNA TO THE A SITE
Gm TP—5′
3′ (A)n
In the complete
80S ribosome formed during the
process of initiation, the A site (aminoacyl or acceptor
site) is free. The binding of the proper aminoacyl-
P site
A site
tRNA in the A site requires proper codon recognition.
Elongation factor EF1A forms a ternary complex with
n
GTP and the entering aminoacyl-tRNA (Figure 38-8).
n-1
Peptidyl-
n-2
This complex then allows the aminoacyl-tRNA to enter
tRNA
the A site with the release of EF1A•GDP and phos-
+
m et
phate. GTP hydrolysis is catalyzed by an active site on
the ribosome. As shown in Figure 38-8, EF1A-GDP
then recycles to EF1A-GTP with the aid of other solu-
GTP
+
GTP
ble protein factors and GTP.
EFIA
B. PEPTIDE BOND FORMATION
n+1
n+1
The α-amino group of the new aminoacyl-tRNA in the
A site carries out a nucleophilic attack on the esterified
GDP
carboxyl group of the peptidyl-tRNA occupying the P
site (peptidyl or polypeptide site). At initiation, this site
GTP
n
n+1
is occupied by aminoacyl-tRNA meti. This reaction is
5′
3′
catalyzed by a peptidyltransferase, a component of the
28S RNA of the 60S ribosomal subunit. This is another
example of ribozyme activity and indicates an impor-
Pi + GDP
+
tant—and previously unsuspected—direct role for
EFIA
RNA in protein synthesis (Table 38-3). Because the
n
n+1
amino acid on the aminoacyl-tRNA is already “acti-
E site
n-1
vated,” no further energy source is required for this re-
n-2
action. The reaction results in attachment of the grow-
ing peptide chain to the tRNA in the A site.
met
C. TRANSLOCATION
n
n+1
5′
3′
The now deacylated tRNA is attached by its anticodon
to the P site at one end and by the open CCA tail to an
exit
(E) site on the large ribosomal subunit (Figure
38-8). At this point, elongation factor 2 (EF2) binds
to and displaces the peptidyl tRNA from the A site to
n+1
the P site. In turn, the deacylated tRNA is on the E site,
GTP +
+
n
from which it leaves the ribosome. The EF2-GTP com-
n-1
plex is hydrolyzed to EF2-GDP, effectively moving the
EF2
n-2
mRNA forward by one codon and leaving the A site
open for occupancy by another ternary complex of
m et
amino acid tRNA-EF1A-GTP and another cycle of
elongation.
n
n+1
n+2
Codon
Gm TP—5′
3′ (A)n
Pi + GDP +
+
Figure 38-8. Diagrammatic representation of the peptide
EF2
elongation process of protein synthesis. The small circles la-
n+1
n
beled n − 1, n, n + 1, etc, represent the amino acid residues of
n-1
the newly formed protein molecule. EFIA and EF2 represent
n-2
elongation factors 1 and 2, respectively. The peptidyl-tRNA and
aminoacyl-tRNA sites on the ribosome are represented by P site
met
and A site, respectively.
PROTEIN SYNTHESIS & THE GENETIC CODE
/
369
Termination
(stop)
codon
Gm TP-5′
3′ (A)n
P site
A site
+
Releasing factor (RF1)
GTP
Releasing factor (RF3)
C
+
N
5′
3′
GTP
O
H2
C
N
Gm TP-5′
3′ (A)n
N
C
+
+
40S
+
60S
+
+
GDP
+
Pi
Peptide
RF1
RF3
tRNA
Figure 38-9. Diagrammatic representation of the termination process of protein synthesis. The peptidyl-tRNA and
aminoacyl-tRNA sites are indicated as P site and A site, respectively. The termination (stop) codon is indicated by the
three vertical bars. Releasing factor RF1 binds to the stop codon. Releasing factor RF3, with bound GTP, binds to RF1.
Hydrolysis of the peptidyl-tRNA complex is shown by the entry of H2O. N and C indicate the amino and carboxyl termi-
nal amino acids, respectively, and illustrate the polarity of protein synthesis.
370
/
CHAPTER 38
Table 38-3. Evidence that rRNA
proteins that hydrolyze the peptidyl-tRNA bond when
is peptidyltransferase.
a stop codon occupies the A site. The mRNA is then re-
leased from the ribosome, which dissociates into its
component 40S and 60S subunits, and another cycle
• Ribosomes can make peptide bonds even when proteins
can be repeated.
are removed or inactivated.
• Certain parts of the rRNA sequence are highly conserved in
all species.
Polysomes Are Assemblies of Ribosomes
• These conserved regions are on the surface of the RNA
Many ribosomes can translate the same mRNA mole-
molecule.
cule simultaneously. Because of their relatively large
• RNA can be catalytic.
size, the ribosome particles cannot attach to an mRNA
• Mutations that result in antibiotic resistance at the level of
protein synthesis are more often found in rRNA than in the
any closer than 35 nucleotides apart. Multiple ribo-
protein components of the ribosome.
somes on the same mRNA molecule form a polyribo-
some, or “polysome.” In an unrestricted system, the
number of ribosomes attached to an mRNA (and thus
the size of polyribosomes) correlates positively with the
The charging of the tRNA molecule with the
length of the mRNA molecule. The mass of the mRNA
aminoacyl moiety requires the hydrolysis of an ATP to
molecule is, of course, quite small compared with the
an AMP, equivalent to the hydrolysis of two ATPs to
mass of even a single ribosome.
two ADPs and phosphates. The entry of the aminoacyl-
A single mammalian ribosome is capable of synthe-
tRNA into the A site results in the hydrolysis of one
sizing about 400 peptide bonds each minute. Polyribo-
GTP to GDP. Translocation of the newly formed pep-
somes actively synthesizing proteins can exist as free
tidyl-tRNA in the A site into the P site by EF2 similarly
particles in the cellular cytoplasm or may be attached to
results in hydrolysis of GTP to GDP and phosphate.
sheets of membranous cytoplasmic material referred to
Thus, the energy requirements for the formation of one
as endoplasmic reticulum. Attachment of the particu-
peptide bond include the equivalent of the hydrolysis of
late polyribosomes to the endoplasmic reticulum is re-
two ATP molecules to ADP and of two GTP molecules
sponsible for its “rough” appearance as seen by electron
to GDP, or the hydrolysis of four high-energy phos-
microscopy. The proteins synthesized by the attached
phate bonds. A eukaryotic ribosome can incorporate as
polyribosomes are extruded into the cisternal space be-
many as six amino acids per second; prokaryotic ribo-
tween the sheets of rough endoplasmic reticulum and
somes incorporate as many as 18 per second. Thus, the
are exported from there. Some of the protein products
process of peptide synthesis occurs with great speed and
of the rough endoplasmic reticulum are packaged by
accuracy until a termination codon is reached.
the Golgi apparatus into zymogen particles for eventual
export (see Chapter 46). The polyribosomal particles
free in the cytosol are responsible for the synthesis of
Termination Occurs When a Stop
proteins required for intracellular functions.
Codon Is Recognized (Figure 38-9)
In comparison to initiation and elongation, termina-
The Machinery of Protein Synthesis Can
tion is a relatively simple process. After multiple cycles
Respond to Environmental Threats
of elongation culminating in polymerization of the spe-
cific amino acids into a protein molecule, the stop or
Ferritin, an iron-binding protein, prevents ionized iron
terminating codon of mRNA (UAA, UAG, UGA) ap-
(Fe2+) from reaching toxic levels within cells. Elemental
pears in the A site. Normally, there is no tRNA with an
iron stimulates ferritin synthesis by causing the release of
anticodon capable of recognizing such a termination
a cytoplasmic protein that binds to a specific region in
signal. Releasing factor RF1 recognizes that a stop
the 5′ nontranslated region of ferritin mRNA. Disrup-
codon resides in the A site (Figure 38-9). RF1 is bound
tion of this protein-mRNA interaction activates ferritin
by a complex consisting of releasing factor RF3 with
mRNA and results in its translation. This mechanism
bound GTP. This complex, with the peptidyl trans-
provides for rapid control of the synthesis of a protein
ferase, promotes hydrolysis of the bond between the
that sequesters Fe2+, a potentially toxic molecule.
peptide and the tRNA occupying the P site. Thus, a
water molecule rather than an amino acid is added.
Many Viruses Co-opt the Host Cell
This hydrolysis releases the protein and the tRNA from
Protein Synthesis Machinery
the P site. Upon hydrolysis and release, the 80S ribo-
some dissociates into its 40S and 60S subunits, which
The protein synthesis machinery can also be modified
are then recycled. Therefore, the releasing factors are
in deleterious ways. Viruses replicate by using host
PROTEIN SYNTHESIS & THE GENETIC CODE
/
371
cell processes, including those involved in protein syn-
4G
thesis. Some viral mRNAs are translated much more ef-
4E
ficiently than those of the host cell (eg, encephalomyo-
Cap
AUG
carditis virus). Others, such as reovirus and vesicular
4G
stomatitis virus, replicate abundantly, and their mRNAs
4E
have a competitive advantage over host cell mRNAs for
4G
4E
limited translation factors. Other viruses inhibit host
IRES
AUG
cell protein synthesis by preventing the association of
Poliovirus
mRNA with the 40S ribosome.
protease
Poliovirus and other picornaviruses gain a selective
advantage by disrupting the function of the 4F complex
Nil
to their advantage. The mRNAs of these viruses do not
4G
Cap
AUG
have a cap structure to direct the binding of the 40S ri-
4E
4G
bosomal subunit (see above). Instead, the 40S ribosomal
subunit contacts an internal ribosomal entry site
(IRES) in a reaction that requires 4G but not 4E. The
IRES
AUG
virus gains a selective advantage by having a protease that
Figure 38-10. Picornaviruses disrupt the 4F com-
attacks 4G and removes the amino terminal 4E binding
plex. The 4E-4G complex (4F) directs the 40S ribosomal
site. Now the 4E-4G complex (4F) cannot form, so the
subunit to the typical capped mRNA (see text). 4G
40S ribosomal subunit cannot be directed to capped
alone is sufficient for targeting the 40S subunit to the
mRNAs. Host cell translation is thus abolished. The 4G
internal ribosomal entry site (IRES) of viral mRNAs. To
fragment can direct binding of the 40S ribosomal sub-
gain selective advantage, certain viruses (eg, poliovirus)
unit to IRES-containing mRNAs, so viral mRNA trans-
lation is very efficient (Figure 38-10). These viruses also
have a protease that cleaves the 4E binding site from
promote the dephosphorylation of BP1
(PHAS-1),
the amino terminal end of 4G. This truncated 4G can di-
thereby decreasing cap (4E)-dependent translation.
rect the 40S ribosomal subunit to mRNAs that have an
IRES but not to those that have a cap. The widths of the
arrows indicate the rate of translation initiation from
POSTTRANSLATIONAL PROCESSING
the AUG codon in each example.
AFFECTS THE ACTIVITY OF
MANY PROTEINS
Some animal viruses, notably poliovirus and hepatitis A
lagen polypeptide molecules, frequently not identical in
virus, synthesize long polycistronic proteins from one
sequence, align themselves in a particular way that is
long mRNA molecule. These protein molecules are
dependent upon the existence of specific amino termi-
subsequently cleaved at specific sites to provide the sev-
nal peptides. Specific enzymes then carry out hydrox-
eral specific proteins required for viral function. In ani-
ylations and oxidations of specific amino acid residues
mal cells, many proteins are synthesized from the
within the procollagen molecules to provide cross-links
mRNA template as a precursor molecule, which then
for greater stability. Amino terminal peptides are
must be modified to achieve the active protein. The
cleaved off the molecule to form the final product—a
prototype is insulin, which is a low-molecular-weight
strong, insoluble collagen molecule. Many other post-
protein having two polypeptide chains with interchain
translational modifications of proteins occur. Covalent
and intrachain disulfide bridges. The molecule is syn-
modification by acetylation, phosphorylation, methyla-
thesized as a single chain precursor, or prohormone,
tion, ubiquitinylation, and glycosylation is common,
which folds to allow the disulfide bridges to form. A
for example.
specific protease then clips out the segment that con-
nects the two chains which form the functional insulin
MANY ANTIBIOTICS WORK BECAUSE
molecule (see Figure 42-12).
THEY SELECTIVELY INHIBIT PROTEIN
Many other peptides are synthesized as proproteins
SYNTHESIS IN BACTERIA
that require modifications before attaining biologic ac-
tivity. Many of the posttranslational modifications in-
Ribosomes in bacteria and in the mitochondria of
volve the removal of amino terminal amino acid
higher eukaryotic cells differ from the mammalian ribo-
residues by specific aminopeptidases. Collagen, an
some described in Chapter 35. The bacterial ribosome
abundant protein in the extracellular spaces of higher
is smaller (70S rather than 80S) and has a different,
eukaryotes, is synthesized as procollagen. Three procol-
somewhat simpler complement of RNA and protein
372
/
CHAPTER 38
molecules. This difference is exploited for clinical pur-
Other antibiotics inhibit protein synthesis on all ri-
poses because many effective antibiotics interact specifi-
bosomes (puromycin) or only on those of eukaryotic
cally with the proteins and RNAs of prokaryotic ribo-
cells (cycloheximide). Puromycin (Figure 38-11) is a
somes and thus inhibit protein synthesis. This results in
structural analog of tyrosinyl-tRNA. Puromycin is in-
growth arrest or death of the bacterium. The most use-
corporated via the A site on the ribosome into the car-
ful members of this class of antibiotics (eg, tetracy-
boxyl terminal position of a peptide but causes the pre-
clines, lincomycin, erythromycin, and chlorampheni-
mature release of the polypeptide. Puromycin, as a
col) do not interact with components of eukaryotic
tyrosinyl-tRNA analog, effectively inhibits protein syn-
ribosomal particles and thus are not toxic to eukaryotes.
thesis in both prokaryotes and eukaryotes. Cyclohex-
Tetracycline prevents the binding of aminoacyl-tRNAs
imide inhibits peptidyltransferase in the 60S ribosomal
to the A site. Chloramphenicol and the macrolide class
subunit in eukaryotes, presumably by binding to an
of antibiotics work by binding to 23S rRNA, which is
rRNA component.
interesting in view of the newly appreciated role of
Diphtheria toxin, an exotoxin of Corynebacterium
rRNA in peptide bond formation through its peptidyl-
diphtheriae infected with a specific lysogenic phage, cat-
transferase activity. It should be mentioned that the
alyzes the ADP-ribosylation of EF-2 on the unique
close similarity between prokaryotic and mitochondrial
amino acid diphthamide in mammalian cells. This
ribosomes can lead to complications in the use of some
modification inactivates EF-2 and thereby specifically
antibiotics.
inhibits mammalian protein synthesis. Many animals
(eg, mice) are resistant to diphtheria toxin. This resis-
tance is due to inability of diphtheria toxin to cross the
cell membrane rather than to insensitivity of mouse
N(CH3)2
EF-2 to diphtheria toxin-catalyzed ADP-ribosylation
N
by NAD.
N
Ricin, an extremely toxic molecule isolated from the
castor bean, inactivates eukaryotic 28S ribosomal RNA
N
N
by providing the N-glycolytic cleavage or removal of a
HOCH2
O
single adenine.
Many of these compounds—puromycin and cyclo-
H
H H
H
heximide in particular—are not clinically useful but
have been important in elucidating the role of protein
NH OH
synthesis in the regulation of metabolic processes, par-
ticularly enzyme induction by hormones.
O
C
CH
CH2
OCH3
NH2
SUMMARY
• The flow of genetic information follows the sequence
DNA → RNA → protein.
• The genetic information in the structural region of a
NH2
gene is transcribed into an RNA molecule such that
N
the sequence of the latter is complementary to that in
N
the DNA.
O-
• Several different types of RNA, including ribosomal
N
N
tRNA
O P O
CH2
O
RNA (rRNA), transfer RNA (tRNA), and messenger
RNA (mRNA), are involved in protein synthesis.
O
H
• The information in mRNA is in a tandem array of
H H
H
codons, each of which is three nucleotides long.
O
OH
• The mRNA is read continuously from a start codon
(AUG) to a termination codon (UAA, UAG, UGA).
O
C
CH
CH2
OH
• The open reading frame of the mRNA is the series of
NH2
codons, each specifying a certain amino acid, that de-
termines the precise amino acid sequence of the pro-
Figure 38-11. The comparative structures of the an-
tein.
tibiotic puromycin (top) and the 3′ terminal portion of
• Protein synthesis, like DNA and RNA synthesis, fol-
tyrosinyl-tRNA (bottom).
lows a 5′ to 3′ polarity and can be divided into three
PROTEIN SYNTHESIS & THE GENETIC CODE
/
373
processes: initiation, elongation, and termination.
Kozak M: Structural features in eukaryotic mRNAs that modulate
the initiation of translation. J Biol Chem 1991;266:1986.
Mutant proteins arise when single-base substitutions
Lawrence JC, Abraham RT: PHAS/4E-BPs as regulators of mRNA
result in codons that specify a different amino acid at
translation and cell proliferation. Trends Biochem Sci
a given position, when a stop codon results in a trun-
1997;22:345.
cated protein, or when base additions or deletions
Sachs AB, Buratowski S: Common themes in translational and
alter the reading frame, so different codons are read.
transcriptional regulation. Trends Biochem Sci 1997;22:189.
• A variety of compounds, including several antibi-
Sachs AB, Sarnow P, Hentze MW: Starting at the beginning, mid-
otics, inhibit protein synthesis by affecting one or
dle and end: translation initiation in eukaryotes. Cell 1997;
more of the steps involved in protein synthesis.
98:831.
Weatherall DJ et al: The hemoglobinopathies. In: The Metabolic
and Molecular Bases of Inherited Disease, 8th ed. Scriver CR et
REFERENCES
al (editors). McGraw-Hill, 2001.
Crick F et al: The genetic code. Nature 1961;192:1227.
Green R, Noller HF: Ribosomes and translation. Annu Rev
Biochem 1997;66:679.
Regulation of Gene Expression
39
Daryl K. Granner, MD, & P. Anthony Weil, PhD
BIOMEDICAL IMPORTANCE
suring that the organism can respond to complex envi-
ronmental challenges.
Organisms adapt to environmental changes by altering
In simple terms, there are only two types of gene
gene expression. The process of alteration of gene ex-
regulation: positive regulation and negative regula-
pression has been studied in detail and often involves
tion (Table 39-1). When the expression of genetic in-
modulation of gene transcription. Control of transcrip-
formation is quantitatively increased by the presence of
tion ultimately results from changes in the interaction
a specific regulatory element, regulation is said to be
of specific binding regulatory proteins with various re-
positive; when the expression of genetic information is
gions of DNA in the controlled gene. This can have a
diminished by the presence of a specific regulatory ele-
positive or negative effect on transcription. Transcrip-
ment, regulation is said to be negative. The element or
tion control can result in tissue-specific gene expres-
molecule mediating negative regulation is said to be a
sion, and gene regulation is influenced by hormones,
negative regulator or repressor; that mediating positive
heavy metals, and chemicals. In addition to transcrip-
regulation is a positive regulator or activator. However,
tion level controls, gene expression can also be modu-
a double negative has the effect of acting as a positive.
lated by gene amplification, gene rearrangement, post-
Thus, an effector that inhibits the function of a nega-
transcriptional modifications, and RNA stabilization.
tive regulator will appear to bring about a positive regu-
Many of the mechanisms that control gene expression
lation. Many regulated systems that appear to be in-
are used to respond to hormones and therapeutic
duced are in fact derepressed at the molecular level.
agents. Thus, a molecular understanding of these
(See Chapter 9 for explanation of these terms.)
processes will lead to development of agents that alter
pathophysiologic mechanisms or inhibit the function or
arrest the growth of pathogenic organisms.
BIOLOGIC SYSTEMS EXHIBIT THREE
TYPES OF TEMPORAL RESPONSES
REGULATED EXPRESSION OF GENES
TO A REGULATORY SIGNAL
IS REQUIRED FOR DEVELOPMENT,
Figure 39-1 depicts the extent or amount of gene ex-
DIFFERENTIATION, & ADAPTATION
pression in three types of temporal response to an in-
The genetic information present in each somatic cell of
ducing signal. A type A response is characterized by an
a metazoan organism is practically identical. The excep-
increased extent of gene expression that is dependent
tions are found in those few cells that have amplified or
upon the continued presence of the inducing signal.
rearranged genes in order to perform specialized cellular
When the inducing signal is removed, the amount of
functions. Expression of the genetic information must
gene expression diminishes to its basal level, but the
be regulated during ontogeny and differentiation of the
amount repeatedly increases in response to the reap-
organism and its cellular components. Furthermore, in
pearance of the specific signal. This type of response is
order for the organism to adapt to its environment and
commonly observed in prokaryotes in response to sud-
to conserve energy and nutrients, the expression of
den changes of the intracellular concentration of a nu-
genetic information must be cued to extrinsic signals
trient. It is also observed in many higher organisms
and respond only when necessary. As organisms have
after exposure to inducers such as hormones, nutrients,
evolved, more sophisticated regulatory mechanisms
or growth factors (Chapter 43).
have appeared which provide the organism and its cells
A type B response exhibits an increased amount of
with the responsiveness necessary for survival in a com-
gene expression that is transient even in the continued
plex environment. Mammalian cells possess about 1000
presence of the regulatory signal. After the regulatory
times more genetic information than does the bac-
signal has terminated and the cell has been allowed to
terium Escherichia coli. Much of this additional genetic
recover, a second transient response to a subsequent
information is probably involved in regulation of gene
regulatory signal may be observed. This phenomenon
expression during the differentiation of tissues and bio-
of response-desensitization-recovery characterizes the
logic processes in the multicellular organism and in en-
action of many pharmacologic agents, but it is also a
374
REGULATION OF GENE EXPRESSION
/
375
Table 39-1. Effects of positive and negative
feature of many naturally occurring processes. This type
regulation on gene expression.
of response commonly occurs during development of
an organism, when only the transient appearance of a
specific gene product is required although the signal
Rate of Gene Expression
persists.
Negative
Positive
The type C response pattern exhibits, in response
Regulation
Regulation
to the regulatory signal, an increased extent of gene ex-
pression that persists indefinitely even after termination
Regulator present
Decreased
Increased
of the signal. The signal acts as a trigger in this pattern.
Regulator absent
Increased
Decreased
Once expression of the gene is initiated in the cell, it
cannot be terminated even in the daughter cells; it is
therefore an irreversible and inherited alteration. This
type of response typically occurs during the develop-
ment of differentiated function in a tissue or organ.
Type A
Prokaryotes Provide Models for the Study
of Gene Expression in Mammalian Cells
Analysis of the regulation of gene expression in
prokaryotic cells helped establish the principle that in-
formation flows from the gene to a messenger RNA to a
specific protein molecule. These studies were aided by
the advanced genetic analyses that could be performed
in prokaryotic and lower eukaryotic organisms. In re-
Time
cent years, the principles established in these early stud-
Signal
ies, coupled with a variety of molecular biology tech-
Type B
niques, have led to remarkable progress in the analysis
of gene regulation in higher eukaryotic organisms, in-
cluding mammals. In this chapter, the initial discussion
will center on prokaryotic systems. The impressive ge-
netic studies will not be described, but the physiology
of gene expression will be discussed. However, nearly
all of the conclusions about this physiology have been
derived from genetic studies and confirmed by molecu-
lar genetic and biochemical studies.
Recovery
Time
Signal
Some Features of Prokaryotic Gene
Expression Are Unique
Type C
Before the physiology of gene expression can be ex-
plained, a few specialized genetic and regulatory terms
must be defined for prokaryotic systems. In prokary-
otes, the genes involved in a metabolic pathway are
often present in a linear array called an operon, eg, the
lac operon. An operon can be regulated by a single pro-
moter or regulatory region. The cistron is the smallest
unit of genetic expression. As described in Chapter 9,
some enzymes and other protein molecules are com-
posed of two or more nonidentical subunits. Thus, the
Time
Signal
“one gene, one enzyme” concept is not necessarily
valid. The cistron is the genetic unit coding for the
Figure 39-1.
Diagrammatic representations of the
structure of the subunit of a protein molecule, acting as
responses of the extent of expression of a gene to spe-
it does as the smallest unit of genetic expression. Thus,
cific regulatory signals such as a hormone.
the one gene, one enzyme idea might more accurately
376
/
CHAPTER 39
be regarded as a one cistron, one subunit concept. A
Promoter
Operator
single mRNA that encodes more than one separately
site
translated protein is referred to as a polycistronic
lacI
lacZ
lacY lacA
mRNA. For example, the polycistronic lac operon
mRNA is translated into three separate proteins (see
lac operon
below). Operons and polycistronic mRNAs are com-
mon in bacteria but not in eukaryotes.
Figure 39-2. The positional relationships of the
An inducible gene is one whose expression increases
structural and regulatory genes of the lac operon. lacZ
in response to an inducer or activator, a specific posi-
encodes β-galactosidase, lacY encodes a permease, and
tive regulatory signal. In general, inducible genes have
lacA encodes a thiogalactoside transacetylase. lacI en-
relatively low basal rates of transcription. By contrast,
codes the lac operon repressor protein.
genes with high basal rates of transcription are often
subject to down-regulation by repressors.
The expression of some genes is constitutive, mean-
encodes the primary transcript. Although there are
ing that they are expressed at a reasonably constant rate
many historical exceptions, a gene is generally italicized
and not known to be subject to regulation. These are
in lower case and the encoded protein, when abbrevi-
often referred to as housekeeping genes. As a result of
ated, is expressed in roman type with the first letter cap-
mutation, some inducible gene products become con-
italized. For example, the gene lacI encodes the repres-
stitutively expressed. A mutation resulting in constitu-
sor protein LacI. When E coli is presented with lactose
tive expression of what was formerly a regulated gene is
or some specific lactose analogs under appropriate non-
called a constitutive mutation.
repressing conditions (eg, high concentrations of lac-
tose, no or very low glucose in media; see below), the
expression of the activities of β-galactosidase, galacto-
Analysis of Lactose Metabolism in E coli
side permease, and thiogalactoside transacetylase is in-
Led to the Operon Hypothesis
creased 100-fold to 1000-fold. This is a type A re-
Jacob and Monod in 1961 described their operon
sponse, as depicted in Figure 39-1. The kinetics of
model in a classic paper. Their hypothesis was to a
induction can be quite rapid; lac-specific mRNAs are
large extent based on observations on the regulation of
fully induced within 5-6 minutes after addition of lac-
lactose metabolism by the intestinal bacterium E coli.
tose to a culture; β-galactosidase protein is maximal
The molecular mechanisms responsible for the regula-
within 10 minutes. Under fully induced conditions,
tion of the genes involved in the metabolism of lactose
there can be up to 5000 β-galactosidase molecules per
are now among the best-understood in any organism.
cell, an amount about 1000 times greater than the
β-Galactosidase hydrolyzes the β-galactoside lactose to
basal, uninduced level. Upon removal of the signal, ie,
galactose and glucose. The structural gene for β-galac-
the inducer, the synthesis of these three enzymes de-
tosidase (lacZ) is clustered with the genes responsible
clines.
for the permeation of galactose into the cell (lacY) and
When E coli is exposed to both lactose and glucose
for thiogalactoside transacetylase (lacA). The structural
as sources of carbon, the organisms first metabolize
genes for these three enzymes, along with the lac pro-
the glucose and then temporarily stop growing until the
moter and lac operator (a regulatory region), are physi-
genes of the lac operon become induced to provide the
cally associated to constitute the lac operon as depicted
ability to metabolize lactose as a usable energy source.
in Figure 39-2. This genetic arrangement of the struc-
Although lactose is present from the beginning of the
tural genes and their regulatory genes allows for coordi-
bacterial growth phase, the cell does not induce those
nate expression of the three enzymes concerned with
enzymes necessary for catabolism of lactose until the
lactose metabolism. Each of these linked genes is tran-
glucose has been exhausted. This phenomenon was first
scribed into one large mRNA molecule that contains
thought to be attributable to repression of the lac
multiple independent translation start (AUG) and stop
operon by some catabolite of glucose; hence, it was
(UAA) codons for each cistron. Thus, each protein is
termed catabolite repression. It is now known that
translated separately, and they are not processed from a
catabolite repression is in fact mediated by a catabolite
single large precursor protein. This type of mRNA mol-
gene activator protein
(CAP) in conjunction with
ecule is called a polycistronic mRNA. Polycistronic
cAMP (Figure 18-5). This protein is also referred to as
mRNAs are predominantly found in prokaryotic organ-
the cAMP regulatory protein (CRP). The expression of
isms.
many inducible enzyme systems or operons in E coli
It is now conventional to consider that a gene in-
and other prokaryotes is sensitive to catabolite repres-
cludes regulatory sequences as well as the region that
sion, as discussed below.
REGULATION OF GENE EXPRESSION
/
377
A
Operator
Promoter
lacI gene
lacZ gene
lacY gene
lacA gene
RNA polymerase
No inducer
cannot transcribe
↑ RNA
operator or distal
or
polymerase
genes (Z,Y, A)
Inducer
and
glucose
Repressor
subunits
Repressor
(tetramer)
B
CAP-cAMP
lacI
lacZ
lacY
lacA
With inducer
and
RNA polymerases transcribing genes
no glucose
Inactive
repressor
mRNA
β-Galacto-
Permease Transacetylase
Inducers
sidase
protein
protein
protein
Figure 39-3. The mechanism of repression and derepression of the lac operon. When either no inducer is
present or inducer is present with glucose (A), the lacI gene products that are synthesized constitutively form
a repressor tetramer molecule which binds at the operator locus to prevent the efficient initiation of transcrip-
tion by RNA polymerase at the promoter locus and thus to prevent the subsequent transcription of the lacZ,
lacY, and lacA structural genes. When inducer is present (B), the constitutively expressed lacI gene forms re-
pressor molecules that are conformationally altered by the inducer and cannot efficiently bind to the operator
locus (affinity of binding reduced > 1000-fold). In the presence of cAMP and its binding protein (CAP), the RNA
polymerase can transcribe the structural genes lacZ, lacY, and lacA, and the polycistronic mRNA molecule
formed can be translated into the corresponding protein molecules β-galactosidase, permease, and
transacetylase, allowing for the catabolism of lactose.
The physiology of induction of the lac operon is
an inverted palindrome (indicated by solid lines about
well understood at the molecular level (Figure 39-3).
the dotted axis) in a region that is 21 base pairs long, as
Expression of the normal lacI gene of the lac operon is
shown below:
constitutive; it is expressed at a constant rate, resulting
:
in formation of the subunits of the lac repressor. Four
identical subunits with molecular weights of 38,000 as-
5′ − AAT
TGTGAGC G GATAACAATT
semble into a lac repressor molecule. The LacI repressor
3′−
TTA ACACTCG C CTATTGTTAA
protein molecule, the product of lacI, has a high affinity
:
(Kd about 10−13 mol/L) for the operator locus. The op-
erator locus is a region of double-stranded DNA 27
The minimum effective size of an operator for LacI
base pairs long with a twofold rotational symmetry and
repressor binding is 17 base pairs (boldface letters in the
378
/
CHAPTER 39
above sequence). At any one time, only two subunits of
priate bioengineered constructs is commonly used to ex-
the repressors appear to bind to the operator, and within
press mammalian recombinant proteins in E coli.
the 17-base-pair region at least one base of each base
In order for the RNA polymerase to efficiently form
pair is involved in LacI recognition and binding. The
a PIC at the promoter site, there must also be present
binding occurs mostly in the major groove without in-
the catabolite gene activator protein (CAP) to which
terrupting the base-paired, double-helical nature of the
cAMP is bound. By an independent mechanism, the
operator DNA. The operator locus is between the pro-
bacterium accumulates cAMP only when it is starved
moter site, at which the DNA-dependent RNA polym-
for a source of carbon. In the presence of glucose—or
erase attaches to commence transcription, and the tran-
of glycerol in concentrations sufficient for growth—the
scription initiation site of the lacZ gene, the structural
bacteria will lack sufficient cAMP to bind to CAP be-
gene for β-galactosidase (Figure 39-2). When attached
cause the glucose inhibits adenylyl cyclase, the enzyme
to the operator locus, the LacI repressor molecule pre-
that converts ATP to cAMP (see Chapter 42). Thus, in
vents transcription of the operator locus as well as of the
the presence of glucose or glycerol, cAMP-saturated
distal structural genes, lacZ, lacY, and lacA. Thus, the
CAP is lacking, so that the DNA-dependent RNA
LacI repressor molecule is a negative regulator; in its
polymerase cannot initiate transcription of the lac
presence (and in the absence of inducer; see below), ex-
operon. In the presence of the CAP-cAMP complex,
pression from the lacZ, lacY, and lacA genes is pre-
which binds to DNA just upstream of the promoter
vented. There are normally 20-40 repressor tetramer
site, transcription then occurs (Figure 39-3). Studies
molecules in the cell, a concentration of tetramer suffi-
indicate that a region of CAP contacts the RNA polym-
cient to effect, at any given time, > 95% occupancy of
erase α subunit and facilitates binding of this enzyme to
the one lac operator element in a bacterium, thus ensur-
the promoter. Thus, the CAP-cAMP regulator is acting
ing low (but not zero) basal lac operon gene transcrip-
as a positive regulator because its presence is required
tion in the absence of inducing signals.
for gene expression. The lac operon is therefore con-
A lactose analog that is capable of inducing the lac
trolled by two distinct, ligand-modulated DNA bind-
operon while not itself serving as a substrate for β-galac-
ing trans factors; one that acts positively (cAMP-CRP
tosidase is an example of a gratuitous inducer. An ex-
complex) and one that acts negatively (LacI repressor).
ample is isopropylthiogalactoside (IPTG). The addition
Maximal activity of the lac operon occurs when glucose
of lactose or of a gratuitous inducer such as IPTG to
levels are low (high cAMP with CAP activation) and
bacteria growing on a poorly utilized carbon source
lactose is present (LacI is prevented from binding to the
(such as succinate) results in prompt induction of the
operator).
lac operon enzymes. Small amounts of the gratuitous in-
When the lacI gene has been mutated so that its
ducer or of lactose are able to enter the cell even in the
product, LacI, is not capable of binding to operator
absence of permease. The LacI repressor molecules—
DNA, the organism will exhibit constitutive expres-
both those attached to the operator loci and those free in
sion of the lac operon. In a contrary manner, an organ-
the cytosol—have a high affinity for the inducer. Bind-
ism with a lacI gene mutation that produces a LacI pro-
ing of the inducer to a repressor molecule attached to
tein which prevents the binding of an inducer to the
the operator locus induces a conformational change in
repressor will remain repressed even in the presence of
the structure of the repressor and causes it to dissociate
the inducer molecule, because the inducer cannot bind
from the DNA because its affinity for the operator is
to the repressor on the operator locus in order to dere-
now 103 times lower (Kd about 10−9 mol/L) than that of
press the operon. Similarly, bacteria harboring muta-
LacI in the absence of IPTG. If DNA-dependent RNA
tions in their lac operator locus such that the operator
polymerase has already attached to the coding strand at
sequence will not bind a normal repressor molecule
the promoter site, transcription will begin. The polym-
constitutively express the lac operon genes. Mechanisms
erase generates a polycistronic mRNA whose 5′ terminal
of positive and negative regulation comparable to those
is complementary to the template strand of the operator.
described here for the lac system have been observed in
In such a manner, an inducer derepresses the lac
eukaryotic cells (see below).
operon and allows transcription of the structural genes
for β-galactosidase, galactoside permease, and thiogalac-
The Genetic Switch of Bacteriophage
toside transacetylase. Translation of the polycistronic
Lambda ( ) Provides a Paradigm
mRNA can occur even before transcription is com-
for Protein-DNA Interactions
pleted. Derepression of the lac operon allows the cell to
in Eukaryotic Cells
synthesize the enzymes necessary to catabolize lactose as
an energy source. Based on the physiology just de-
Like some eukaryotic viruses (eg, herpes simplex, HIV),
scribed, IPTG-induced expression of transfected plas-
some bacterial viruses can either reside in a dormant
mids bearing the lac operator-promoter ligated to appro-
state within the host chromosomes or can replicate
REGULATION OF GENE EXPRESSION
/
379
within the bacterium and eventually lead to lysis and
1
killing of the bacterial host. Some E coli harbor such a
“temperate” virus, bacteriophage lambda
(λ). When
lambda infects an organism of that species it injects its
45,000-bp, double-stranded, linear DNA genome into
the cell (Figure 39-4). Depending upon the nutritional
2
state of the cell, the lambda DNA will either integrate
into the host genome (lysogenic pathway) and remain
dormant until activated (see below), or it will com-
mence replicating until it has made about 100 copies
of complete, protein-packaged virus, at which point it
causes lysis of its host (lytic pathway). The newly gen-
erated virus particles can then infect other susceptible
3
hosts.
When integrated into the host genome in its dor-
mant state, lambda will remain in that state until acti-
Lysogenic
Lytic
vated by exposure of its lysogenic bacterial host to
pathway
pathway
DNA-damaging agents. In response to such a noxious
4
6
stimulus, the dormant bacteriophage becomes
“in-
duced” and begins to transcribe and subsequently trans-
late those genes of its own genome which are necessary
for its excision from the host chromosome, its DNA
replication, and the synthesis of its protein coat and
lysis enzymes. This event acts like a trigger or type C
5
10
7
(Figure 39-1) response; ie, once lambda has committed
itself to induction, there is no turning back until the
cell is lysed and the replicated bacteriophage released.
Ultraviolet
This switch from a dormant or prophage state to a
radiation
Induction
lytic infection is well understood at the genetic and
9
8
molecular levels and will be described in detail here.
The switching event in lambda is centered around
an 80-bp region in its double-stranded DNA genome
referred to as the “right operator” (OR) (Figure 39-5A).
The right operator is flanked on its left side by the
Figure 39-4. Infection of the bacterium E coli by
structural gene for the lambda repressor protein, the cI
phage lambda begins when a virus particle attaches it-
protein, and on its right side by the structural gene en-
self to the bacterial cell (1) and injects its DNA (shaded
coding another regulatory protein called Cro. When
line) into the cell (2, 3). Infection can take either of two
lambda is in its prophage state—ie, integrated into the
courses depending on which of two sets of viral genes
host genome—the cI repressor gene is the only lambda
gene cI protein that is expressed. When the bacterio-
is turned on. In the lysogenic pathway, the viral DNA
phage is undergoing lytic growth, the cI repressor gene
becomes integrated into the bacterial chromosome (4,
is not expressed, but the cro gene—as well as many
5), where it replicates passively as the bacterial cell di-
other genes in lambda—is expressed. That is, when the
vides. The dormant virus is called a prophage, and the
repressor gene is on, the cro gene is off, and when
cell that harbors it is called a lysogen. In the alternative
the cro gene is on, the repressor gene is off. As we
lytic mode of infection, the viral DNA replicates itself (6)
shall see, these two genes regulate each other’s expres-
and directs the synthesis of viral proteins (7). About 100
sion and thus, ultimately, the decision between lytic
new virus particles are formed. The proliferating viruses
and lysogenic growth of lambda. This decision be-
lyse, or burst, the cell (8). A prophage can be “induced”
tween repressor gene transcription and cro gene
by a DNA damaging agent such as ultraviolet radiation
transcription is a paradigmatic example of a molecu-
(9). The inducing agent throws a switch, so that a differ-
lar switch.
ent set of genes is turned on. Viral DNA loops out of the
The 80-bp λ right operator, OR, can be subdivided
chromosome (10) and replicates; the virus proceeds
into three discrete, evenly spaced,
17-bp cis-active
along the lytic pathway. (Reproduced, with permission,
DNA elements that represent the binding sites for ei-
from Ptashne M, Johnson AD, Pabo CO: A genetic switch
ther of two bacteriophage λ regulatory proteins. Impor-
in a bacterial virus. Sci Am [Nov] 1982;247:128.)
380
/
CHAPTER 39
Gene for repressor (cl)
Gene for Cro
A
OR
Repressor RNA
OR3
OR2
OR1
B
Repressor promoter
cro Promoter
cro RNA
T A C C T C T G G C G G T G A T A
C
A T G G A G A C C G C C A C T A T
Figure 39-5. Right operator (OR) is shown in increasing detail in this series of drawings.
The operator is a region of the viral DNA some 80 base pairs long (A). To its left lies the
gene encoding lambda repressor (cI), to its right the gene (cro) encoding the regulator pro-
tein Cro. When the operator region is enlarged (B), it is seen to include three subregions,
OR1, OR2, and OR3, each 17 base pairs long. They are recognition sites to which both repres-
sor and Cro can bind. The recognition sites overlap two promoters—sequences of bases to
which RNA polymerase binds in order to transcribe these genes into mRNA (wavy lines),
that are translated into protein. Site OR1 is enlarged (C) to show its base sequence. Note
that in this region of the λ chromosome, both strands of DNA act as a template for tran-
scription (Chapter 39). (Reproduced, with permission, from Ptashne M, Johnson AD, Pabo CO:
A genetic switch in a bacterial virus. Sci Am [Nov] 1982;247:128.)
tantly, the nucleotide sequences of these three tandemly
39-6D). The Cro protein’s single domain
mediates
arranged sites are similar but not identical
(Figure
both operator binding and dimerization.
39-5B). The three related cis elements, termed opera-
In a lysogenic bacterium—ie, a bacterium containing
tors OR1, OR2, and OR3, can be bound by either cI or
a lambda prophage—the lambda repressor dimer binds
Cro proteins. However, the relative affinities of cI and
preferentially to OR1 but in so doing, by a cooperative
Cro for each of the sites varies, and this differential
interaction, enhances the binding (by a factor of 10) of
binding affinity is central to the appropriate operation
another repressor dimer to OR2 (Figure 39-7). The
of the λ phage lytic or lysogenic “molecular switch.”
affinity of repressor for OR3 is the least of the three oper-
The DNA region between the cro and repressor genes
ator subregions. The binding of repressor to OR1 has two
also contains two promoter sequences that direct the
major effects. The occupation of OR1 by repressor
binding of RNA polymerase in a specified orientation,
blocks the binding of RNA polymerase to the right-
where it commences transcribing adjacent genes. One
ward promoter and in that way prevents expression of
promoter directs RNA polymerase to transcribe in the
cro. Second, as mentioned above, repressor dimer bound
rightward direction and, thus, to transcribe cro and
to OR1 enhances the binding of repressor dimer to OR2.
other distal genes, while the other promoter directs the
The binding of repressor to OR2 has the important
transcription of the repressor gene in the leftward di-
added effect of enhancing the binding of RNA polym-
rection (Figure 39-5B).
erase to the leftward promoter that overlaps OR2 and
The product of the repressor gene, the 236-amino-
thereby enhances transcription and subsequent expres-
acid,
27 kDa repressor protein, exists as a two-
sion of the repressor gene. This enhancement of tran-
domain molecule in which the amino terminal domain
scription is apparently mediated through direct protein-
binds to operator DNA and the carboxyl terminal
protein interactions between promoter-bound RNA
domain promotes the association of one repressor
polymerase and OR2-bound repressor. Thus, the lambda
protein with another to form a dimer. A dimer of re-
repressor is both a negative regulator, by preventing
pressor molecules binds to operator DNA much more
transcription of cro, and a positive regulator, by enhanc-
tightly than does the monomeric form (Figure 39-6A
ing transcription of its own gene, the repressor gene.
to 39-6C).
This dual effect of repressor is responsible for the stable
The product of the cro gene, the 66-amino-acid,
state of the dormant lambda bacteriophage; not only
9 kDa Cro protein, has a single domain but also binds
does the repressor prevent expression of the genes neces-
the operator DNA more tightly as a dimer (Figure
sary for lysis, but it also promotes expression of itself to
REGULATION OF GENE EXPRESSION
/
381
A
B
C
D
Amino acids
COOH
COOH
COOH
COOH COOH
132 - 236
Cro
NH2
Amino acids
NH2
NH2
NH2
NH2
1 - 92
OR1
OR3
Figure 39-6. Schematic molecular structures of cI (lambda repressor, shown in A, B, and C)
and Cro (D). Lambda repressor protein is a polypeptide chain 236 amino acids long. The chain
folds itself into a dumbbell shape with two substructures: an amino terminal (NH2) domain and
a carboxyl terminal (COOH) domain. The two domains are linked by a region of the chain that is
susceptible to cleavage by proteases (indicated by the two arrows in A). Single repressor mole-
cules (monomers) tend to associate to form dimers (B); a dimer can dissociate to form
monomers again. A dimer is held together mainly by contact between the carboxyl terminal
domains (hatching). Repressor dimers bind to (and can dissociate from) the recognition sites in
the operator region; they display the greatest affinity for site OR1 (C). It is the amino terminal
domain of the repressor molecule that makes contact with the DNA (hatching). Cro (D) has a
single domain with sites that promote dimerization and other sites that promote binding of
dimers to operator, preferentially to OR3. (Reproduced, with permission, from Ptashne M, Johnson
AD, Pabo CO: A genetic switch in a bacterial virus. Sci Am [Nov] 1982;247:128.)
stabilize this state of differentiation. In the event that in-
The resulting newly synthesized Cro protein also
tracellular repressor protein concentration becomes very
binds to the operator region as a dimer, but its order of
high, this excess repressor will bind to OR3 and by so
preference is opposite to that of repressor
(Figure
doing diminish transcription of the repressor gene from
39-7). That is, Cro binds most tightly to OR3, but
the leftward promoter until the repressor concentration
there is no cooperative effect of Cro at OR3 on the
drops and repressor dissociates itself from OR3.
binding of Cro to OR2. At increasingly higher concen-
With such a stable, repressive, cI-mediated, lyso-
trations of Cro, the protein will bind to OR2 and even-
genic state, one might wonder how the lytic cycle could
tually to OR1.
ever be entered. However, this process does occur quite
Occupancy of OR3 by Cro immediately turns off
efficiently. When a DNA-damaging signal, such as ul-
transcription from the leftward promoter and in that
traviolet light, strikes the lysogenic host bacterium,
way prevents any further expression of the repressor
fragments of single-stranded DNA are generated that
gene. The molecular switch is thus completely
activate a specific protease coded by a bacterial gene
“thrown” in the lytic direction. The cro gene is now ex-
and referred to as recA (Figure 39-7). The activated
pressed, and the repressor gene is fully turned off. This
recA protease hydrolyzes the portion of the repressor
event is irreversible, and the expression of other lambda
protein that connects the amino terminal and carboxyl
genes begins as part of the lytic cycle. When Cro repres-
terminal domains of that molecule (see Figure 39-6A).
sor concentration becomes quite high, it will eventually
Such cleavage of the repressor domains causes the re-
occupy OR1 and in so doing reduce the expression of its
pressor dimers to dissociate, which in turn causes dis-
own gene, a process that is necessary in order to effect
sociation of the repressor molecules from OR2 and
the final stages of the lytic cycle.
eventually from OR1. The effects of removal of repres-
The three-dimensional structures of Cro and of the
sor from OR1 and OR2 are predictable. RNA polym-
lambda repressor protein have been determined by
erase immediately has access to the rightward promoter
x-ray crystallography, and models for their binding and ef-
and commences transcribing the cro gene, and the en-
fecting the above-described molecular and genetic events
hancement effect of the repressor at OR2 on leftward
have been proposed and tested. Both bind to DNA using
transcription is lost (Figure 39-7).
helix-turn-helix DNA binding domain motifs (see below).
Prophage
OR3
OR2
OR1
RNA polymerase
Repressor promoter
cro promoter
OR3
OR2
OR1
Induction (1)
RNA polymerase
recA
Repressor promoter
cro promoter
Ultraviolet radiation
OR3
OR2
OR1
RNA polymerase
Induction (2)
Repressor promoter
cro promoter
Early lytic growth
OR3
OR2
OR1
RNA polymerase
Repressor promoter
cro promoter
Figure 39-7. Configuration of the switch is shown at four stages of lambda’s life cycle. The lysogenic pathway (in
which the virus remains dormant as a prophage) is selected when a repressor dimer binds to OR1, thereby making it
likely that OR2 will be filled immediately by another dimer. In the prophage (top), the repressor dimers bound at OR1
and OR2 prevent RNA polymerase from binding to the rightward promoter and so block the synthesis of Cro (nega-
tive control). The repressors also enhance the binding of polymerase to the leftward promoter (positive control),
with the result that the repressor gene is transcribed into RNA (wavy line) and more repressor is synthesized, main-
taining the lysogenic state. The prophage is induced when ultraviolet radiation activates the protease recA, which
cleaves repressor monomers. The equilibrium of free monomers, free dimers, and bound dimers is thereby shifted,
and dimers leave the operator sites. RNA polymerase is no longer encouraged to bind to the leftward promoter, so
that repressor is no longer synthesized. As induction proceeds, all the operator sites become vacant, and so polym-
erase can bind to the rightward promoter and Cro is synthesized. During early lytic growth, a single Cro dimer binds
to OR3 shaded circles, the site for which it has the highest affinity. Consequently, RNA polymerase cannot bind to the
leftward promoter, but the rightward promoter remains accessible. Polymerase continues to bind there, transcribing
cro and other early lytic genes. Lytic growth ensues. (Reproduced, with permission, from Ptashne M, Johnson AD, Pabo
CO: A genetic switch in a bacterial virus. Sci Am [Nov] 1982;247:128.)
382
REGULATION OF GENE EXPRESSION
/
383
To date, this system provides the best understanding of
tors with specific DNA regions. The dynamics of the for-
the molecular events involved in gene regulation.
mation and disruption of nucleosome structure are there-
Detailed analysis of the lambda repressor led to the
fore an important part of eukaryotic gene regulation.
important concept that transcription regulatory proteins
Histone acetylation and deacetylation is an im-
have several functional domains. For example, lambda
portant determinant of gene activity. The surprising
repressor binds to DNA with high affinity. Repressor
discovery that histone acetylase activity is associated
monomers form dimers, dimers interact with each
with TAFs and the coactivators involved in hormonal
other, and repressor interacts with RNA polymerase.
regulation of gene transcription (see Chapter 43) has
The protein-DNA interface and the three protein-
provided a new concept of gene regulation. Acetylation
protein interfaces all involve separate and distinct do-
is known to occur on lysine residues in the amino ter-
mains of the repressor molecule. As will be noted below
minal tails of histone molecules. This modification re-
(see Figure 39-17), this is a characteristic shared by
duces the positive charge of these tails and decreases the
most (perhaps all) molecules that regulate transcription.
binding affinity of histone for the negatively charged
DNA. Accordingly, the acetylation of histone could re-
sult in disruption of nucleosomal structure and allow
SPECIAL FEATURES ARE INVOLVED
readier access of transcription factors to cognate regula-
IN REGULATION OF EUKARYOTIC
tory DNA elements. As discussed previously, this
GENE TRANSCRIPTION
would enhance binding of the basal transcription ma-
chinery to the promoter. Histone deacetylation would
Most of the DNA in prokaryotic cells is organized into
have the opposite effect. Different proteins with specific
genes, and the templates can always be transcribed. A
acetylase and deacetylase activities are associated with
very different situation exists in mammalian cells, in
various components of the transcription apparatus. The
which relatively little of the total DNA is organized
specificity of these processes is under investigation, as
into genes and their associated regulatory regions. The
are a variety of mechanisms of action. Some specific ex-
function of the extra DNA is unknown. In addition, as
amples are illustrated in Chapter 43.
described in Chapter 36, the DNA in eukaryotic cells is
There is evidence that the methylation of deoxycy-
extensively folded and packed into the protein-DNA
tidine residues (in the sequence 5′-mCpG-3′) in DNA
complex called chromatin. Histones are an important
may effect gross changes in chromatin so as to preclude
part of this complex since they both form the structures
its active transcription, as described in Chapter 36. For
known as nucleosomes (see Chapter 36) and also factor
example, in mouse liver, only the unmethylated riboso-
significantly into gene regulatory mechanisms as out-
mal genes can be expressed, and there is evidence that
lined below.
many animal viruses are not transcribed when their
DNA is methylated. Acute demethylation of deoxycyti-
dine residues in a specific region of the tyrosine amino-
Chromatin Remodeling Is an Important
transferase gene—in response to glucocorticoid hor-
Aspect of Eukaryotic Gene Expression
mones—has been associated with an increased rate of
Chromatin structure provides an additional level of
transcription of the gene. However, it is not possible to
control of gene transcription. As discussed in Chapter
generalize that methylated DNA is transcriptionally in-
36, large regions of chromatin are transcriptionally inac-
active, that all inactive chromatin is methylated, or that
tive while others are either active or potentially active.
active DNA is not methylated.
With few exceptions, each cell contains the same com-
Finally, the binding of specific transcription factors
plement of genes (antibody-producing cells are a notable
to cognate DNA elements may result in disruption of
exception). The development of specialized organs, tis-
nucleosomal structure. Many eukaryotic genes have
sues, and cells and their function in the intact organism
multiple protein-binding DNA elements. The serial
depend upon the differential expression of genes.
binding of transcription factors to these elements—in a
Some of this differential expression is achieved by
combinatorial fashion—may either directly disrupt the
having different regions of chromatin available for tran-
structure of the nucleosome or prevent its re-formation
scription in cells from various tissues. For example, the
or recruit, via protein-protein interactions, multipro-
DNA containing the β-globin gene cluster is in “active”
tein coactivator complexes that have the ability to cova-
chromatin in the reticulocyte but in “inactive” chro-
lently modify or remodel nucleosomes. These reactions
matin in muscle cells. All the factors involved in the de-
result in chromatin-level structural changes that in the
termination of active chromatin have not been eluci-
end increase DNA accessibility to other factors and the
dated. The presence of nucleosomes and of complexes of
transcription machinery.
histones and DNA (see Chapter 36) certainly provides a
Eukaryotic DNA that is in an “active” region of
barrier against the ready association of transcription fac-
chromatin can be transcribed. As in prokaryotic cells, a
384
/
CHAPTER 39
promoter dictates where the RNA polymerase will ini-
(Enhancer
Promoter
Structural gene
response element)
tiate transcription, but this promoter cannot be neatly
defined as containing a −35 and −10 box, particularly
in mammalian cells (Chapter 37). In addition, the
A
SV40
β globin
β globin
trans-acting factors generally come from other chromo-
somes (and so act in trans), whereas this consideration
is moot in the case of the single chromosome-contain-
ing prokaryotic cells. Additional complexity is added by
B SV40
β globin
β globin
elements or factors that enhance or repress transcrip-
tion, define tissue-specific expression, and modulate the
actions of many effector molecules.
C
mt
tk
hGH
Certain DNA Elements Enhance or Repress
Transcription of Eukaryotic Genes
In addition to gross changes in chromatin affecting
D
GRE
PEPCK
CAT
transcriptional activity, certain DNA elements facilitate
Figure 39-8. A schematic explanation of the action
or enhance initiation at the promoter. For example, in
of enhancers and other cis-acting regulatory elements.
simian virus 40 (SV40) there exists about 200 bp up-
stream from the promoter of the early genes a region of
These model chimeric genes consist of a reporter
two identical, tandem 72-bp lengths that can greatly in-
(structural) gene that encodes a protein which can be
crease the expression of genes in vivo. Each of these
readily assayed, a promoter that ensures accurate initia-
72-bp elements can be subdivided into a series of
tion of transcription, and the putative regulatory ele-
smaller elements; therefore, some enhancers have a very
ments. In all cases, high-level transcription from the in-
complex structure. Enhancer elements differ from the
dicated chimeras depends upon the presence of
promoter in two remarkable ways. They can exert their
enhancers, which stimulate transcription ≥ 100-fold
positive influence on transcription even when separated
over basal transcriptional levels (ie, transcription of the
by thousands of base pairs from a promoter; they work
same chimeric genes containing just promoters fused
when oriented in either direction; and they can work
to the structural genes). Examples A and B illustrate the
upstream (5′) or downstream (3′) from the promoter.
fact that enhancers (eg, SV40) work in either orientation
Enhancers are promiscuous; they can stimulate any
and upon a heterologous promoter. Example C illus-
promoter in the vicinity and may act on more than one
trates that the metallothionein (mt) regulatory element
promoter. The SV40 enhancer element can exert an in-
(which under the influence of cadmium or zinc induces
fluence on, for example, the transcription of β-globin
transcription of the endogenous mt gene and hence
by increasing its transcription 200-fold in cells contain-
the metal-binding mt protein) will work through the
ing both the enhancer and the β-globin gene on the
thymidine kinase (tk) promoter to enhance transcrip-
same plasmid (see below and Figure 39-8). The en-
tion of the human growth hormone (hGH) gene. The
hancer element does not produce a product that in turn
engineered genetic constructions were introduced into
acts on the promoter, since it is active only when it ex-
the male pronuclei of single-cell mouse embryos and
ists within the same DNA molecule as (ie, cis to) the
the embryos placed into the uterus of a surrogate
promoter. Enhancer binding proteins are responsible
for this effect. The exact mechanisms by which these
mother to develop as transgenic animals. Offspring
transcription activators work are subject to much de-
have been generated under these conditions, and in
bate. Certainly, enhancer binding trans factors have
some the addition of zinc ions to their drinking water
been shown to interact with a plethora of other tran-
effects an increase in liver growth hormone. In this
scription proteins. These interactions include chro-
case, these transgenic animals have responded to the
matin-modifying coactivators as well as the individual
high levels of growth hormone by becoming twice as
components of the basal RNA polymerase II transcrip-
large as their normal litter mates. Example D illustrates
tion machinery. Ultimately, trans-factor-enhancer DNA
that a glucocorticoid response element (GRE) will work
binding events result in an increase in the binding of
through homologous (PEPCK gene) or heterologous
the basal transcription machinery to the promoter. En-
promoters (not shown; ie, tk promoter, SV40 promoter,
hancer elements and associated binding proteins often
β-globin promoter, etc).
convey nuclease hypersensitivity to those regions where
they reside (Chapter 36). A summary of the properties
of enhancers is presented in Table 39-2. One of the
REGULATION OF GENE EXPRESSION
/
385
Table 39-2. Summary of the properties
duces β-interferon gene transcription—rather, it is the
of enhancers.
formation of the enhanceosome proper that provides
appropriate surfaces for the recruitment of coactivators
that results in the enhanced formation of the PIC on
• Work when located long distances from the promoter
the cis-linked promoter and thus transcription activa-
• Work when upstream or downstream from the promoter
tion.
• Work when oriented in either direction
The cis-acting elements that decrease or repress the
• Work through heterologous promoters
expression of specific genes have also been identified.
• Work by binding one or more proteins
• Work by facilitating binding of the basal transcription com-
Because fewer of these elements have been studied, it is
plex to the promoter
not possible to formulate generalizations about their
mechanism of action—though again, as for gene activa-
tion, chromatin level covalent modifications of histones
and other proteins by (repressor)-recruited multisub-
best-understood mammalian enhancer systems is that
unit corepressors have been implicated.
of the β-interferon gene. This gene is induced upon
viral infection of mammalian cells. One goal of the cell,
Tissue-Specific Expression May
once virally infected, is to attempt to mount an antivi-
Result From the Action
ral response—if not to save the infected cell, then to
of Enhancers or Repressors
help to save the entire organism from viral infection.
Interferon production is one mechanism by which this
Many genes are now recognized to harbor enhancer or
is accomplished. This family of proteins is secreted by
activator elements in various locations relative to their
virally infected cells. They interact with neighboring
coding regions. In addition to being able to enhance
cells to cause an inhibition of viral replication by a vari-
gene transcription, some of these enhancer elements
ety of mechanisms, thereby limiting the extent of viral
clearly possess the ability to do so in a tissue-specific
infection. The enhancer element controlling induction
manner. Thus, the enhancer element associated with
of this gene, located between nucleotides −110 and −45
the immunoglobulin genes between the J and C regions
of the β-interferon gene, is well characterized. This en-
enhances the expression of those genes preferentially in
hancer is composed of four distinct clustered cis ele-
lymphoid cells. Similarly to the SV40 enhancer, which
ments, each of which is bound by distinct trans factors.
is capable of promiscuously activating a variety of cis-
One cis element is bound by the trans-acting factor
linked genes, enhancer elements associated with the
NF-κB, one by a member of the IRF (interferon regula-
genes for pancreatic enzymes are capable of enhancing
tory factor) family of trans factors, and a third by the
even unrelated but physically linked genes preferentially
heterodimeric leucine zipper factor ATF-2/c-Jun. The
in the pancreatic cells of mice into which the specifi-
fourth factor is the ubiquitous, architectural transcrip-
cally engineered gene constructions were introduced
tion factor known as HMG I(Y). Upon binding to its
microsurgically at the single-cell embryo stage. This
degenerate, A+T-rich binding sites, HMG I(Y) induces
transgenic animal approach has proved useful in
a significant bend in the DNA. There are four such
studying tissue-specific gene expression. For example,
HMG I(Y) binding sites interspersed throughout the
DNA containing a pancreatic B cell tissue-specific en-
enhancer. These sites play a critical role in forming the
hancer (from the insulin gene), when ligated in a vector
enhanceosome, along with the aforementioned three
to polyoma large-T antigen, an oncogene, produced
trans factors, by inducing a series of critically spaced
B cell tumors in transgenic mice. Tumors did not de-
DNA bends. Consequently, HMG I(Y) induces the co-
velop in any other tissue. Tissue-specific gene expres-
operative formation of a unique, stereospecific, three
sion may therefore be mediated by enhancers or en-
dimensional structure within which all four factors are
hancer-like elements.
active when viral infection signals are sensed by the cell.
The structure formed by the cooperative assembly of
Reporter Genes Are Used to Define
these four factors is termed the β-interferon enhanceo-
Enhancers & Other Regulatory Elements
some (see Figure 39-9), so named because of its obvi-
ous structural similarity to the nucleosome, also a
By ligating regions of DNA suspected of harboring reg-
unique three-dimensional protein DNA structure that
ulatory sequences to various reporter genes
(the re-
wraps DNA about an assembly of proteins (see Figures
porter or chimeric gene approach) (Figures 39-10
36-1 and 36-2). The enhanceosome, once formed, in-
and 39-11), one can determine which regions in the
duces a large increase in β-interferon gene transcription
vicinity of structural genes have an influence on their
upon virus infection. It is not simply the protein occu-
expression. Pieces of DNA thought to harbor regula-
pancy of the linearly apposed cis element sites that in-
tory elements are ligated to a suitable reporter gene and
386
/
CHAPTER 39
HMG PRDIV HMG PRDI-III PRDII HMG NRDI HMG
HMGI-Y
ATF-2
cJun
NF-κB
IRF(IRF3/7)
HMGI
cJun
ATF-2
HMGI
NF-κB
HMGI
IRF3
IRF7
HMGI
Figure 39-9. Formation and putative structure of the enhanceosome formed on the human β-interferon gene
enhancer. Diagramatically represented at the top is the distribution of the multiple cis-elements (HMG, PRDIV,
PRDI-III, PRDII, NRDI) composing the β-interferon gene enhancer. The intact enhancer mediates transcriptional in-
duction of the β-interferon gene (over 100-fold) upon virus infection of human cells. The cis-elements of this modu-
lar enhancer represent the binding sites for the trans-factors HMG I(Y), cJun-ATF-2, IRF3, IRF7, and NF-κB, respec-
tively. The factors interact with these DNA elements in an obligatory, ordered, and highly cooperative fashion as
indicated by the arrow. Initial binding of four HMG I(Y) proteins induces sharp DNA bends in the enhancer, causing
the entire 70-80 bp region to assume a high level of curvature. This curvature is integral to the subsequent highly
cooperative binding of the other trans-factors since this enables the DNA-bound factors to make important, direct
protein-protein interactions that both contribute to the formation and stability of the enhanceosome and generate
a unique three-dimensional surface that serves to recruit chromatin-modifying activities (eg, Swi/Snf and P/CAF) as
well as the general transcription machinery (RNA polymerase II and GTFs). Although four of the five cis-elements
(PRDIV, PRDI-III, PRDII, NRDI) independently can modestly stimulate (~tenfold) transcription of a reporter gene in
transfected cells (see Figures 39-10 and 39-12), all five cis-elements, in appropriate order, are required to form an
enhancer that can appropriately stimulate mRNA gene transcription (ie, ≥ 100-fold) in response to viral infection of a
human cell. This distinction indicates the strict requirement for appropriate enhanceosome architecture for efficient
trans-activation. Similar enhanceosomes, involving distinct cis- and trans-factors, are proposed to form on many
other mammalian genes.
introduced into a host cell (Figure 39-10). Basal ex-
This strategy, using transfected cells in culture
pression of the reporter gene will be increased if the
and transgenic animals, has led to the identification
DNA contains an enhancer. Addition of a hormone or
of dozens of enhancers, repressors, tissue-specific ele-
heavy metal to the culture medium will increase expres-
ments, and hormone, heavy metal, and drug-response
sion of the reporter gene if the DNA contains a hor-
elements. The activity of a gene at any moment re-
mone or metal response element (Figure 39-11). The
flects the interaction of these numerous cis-acting
location of the element can be pinpointed by using pro-
DNA elements with their respective trans-acting fac-
gressively shorter pieces of DNA, deletions, or point
tors. The challenge now is to figure out how this oc-
mutations (Figure 39-11).
curs.
REGULATION OF GENE EXPRESSION
/
387
Test promoter
Reporter gene
ner, but the process in most genes, especially in mam-
5′
3′
5′
3′
mals, is much more complicated. Signals representing a
GENE
CAT
number of complex environmental stimuli may con-
ENHANCER-PROMOTER
REPORTER GENE:
verge on a single gene. The response of the gene to
TEST ENHANCER-PROMOTER
DRIVING TRANSCRIPTION
these signals can have several physiologic characteristics.
CAT GENE
First, the response may extend over a considerable
range. This is accomplished by having additive and syn-
CAT
ergistic positive responses counterbalanced by negative
or repressing effects. In some cases, either the positive
or the negative response can be dominant. Also re-
quired is a mechanism whereby an effector such as a
TRANSFECT CELLS USING CaPO4
PRECIPITATED DNA
hormone can activate some genes in a cell while repress-
ing others and leaving still others unaffected. When all
of these processes are coupled with tissue-specific ele-
Divide and re-plate
ment factors, considerable flexibility is afforded. These
physiologic variables obviously require an arrangement
much more complicated than an on-off switch. The
Cells
array of DNA elements in a promoter specifies—with
associated factors—how a given gene will respond.
Control
Hormones
Some simple examples are illustrated in Figure 39-12.
HARVEST 24 HOURS LATER
ASSAY FOR CAT ACTIVITY
Transcription Domains Can Be Defined by
Identification of
control elements
Locus Control Regions & Insulators
Figure 39-10. The use of reporter genes to define
The large number of genes in eukaryotic cells and the
DNA regulatory elements. A DNA fragment from the
complex arrays of transcription regulatory factors pre-
gene in question—in this example, approximately 2 kb
sents an organizational problem. Why are some genes
of 5′-flanking DNA and cognate promoter—is ligated
available for transcription in a given cell whereas others
are not? If enhancers can regulate several genes and are
into a plasmid vector that contains a suitable reporter
not position- and orientation-dependent, how are they
gene—in this case, the bacterial enzyme chlorampheni-
prevented from triggering transcription randomly? Part
col transferase (CAT). The enzyme luciferase (abbrevi-
of the solution to these problems is arrived at by having
ated LUC) is another popular reporter gene. Neither
the chromatin arranged in functional units that restrict
LUC nor CAT is present in mammalian cells; hence, de-
patterns of gene expression. This may be achieved by
tection of these activities in a cell extract means that
having the chromatin form a structure with the nuclear
the cell was successfully transfected by the plasmid. An
matrix or other physical entity, or compartments
increase of CAT activity over the basal level, eg, after
within the nucleus. Alternatively, some regions are con-
addition of one or more hormones, means that the re-
trolled by complex DNA elements called locus control
gion of DNA inserted into the reporter gene plasmid
regions (LCRs). An LCR—with associated bound pro-
contains functional hormone response elements (HRE).
teins—controls the expression of a cluster of genes. The
Progressively shorter pieces of DNA, regions with inter-
best-defined LCR regulates expression of the globin
nal deletions, or regions with point mutations can be
gene family over a large region of DNA. Another mech-
constructed and inserted to pinpoint the response ele-
anism is provided by insulators. These DNA elements,
ment (see Figure 39-11 for deletion mapping of the rel-
also in association with one or more proteins, prevent
evant HREs).
an enhancer from acting on a promoter on the other
side of an insulator in another transcription domain.
Combinations of DNA Elements
SEVERAL MOTIFS MEDIATE THE BINDING
& Associated Proteins Provide
OF REGULATORY PROTEINS TO DNA
Diversity in Responses
The specificity involved in the control of transcription
Prokaryotic genes are often regulated in an on-off man-
requires that regulatory proteins bind with high affinity
ner in response to simple environmental cues. Some eu-
to the correct region of DNA. Three unique motifs—
karyotic genes are regulated in the simple on-off man-
the helix-turn-helix, the zinc finger, and the leucine
388
/
CHAPTER 39
REPORTER GENE CONSTRUCTS
HORMONE-DEPENDENT
WITH VARIABLE AMOUNTS
TRANSCRIPTION
OF 5'-FLANKING DNA
INDUCTION
A
B C
+
+
+
+
+
+
+
+
+
Figure 39-11.
Location of hormone response ele-
CAT
ments (HREs) A, B, and C using the reporter
5′
gene-transfection approach. A family of reporter
2000
1000
+1
genes, constructed as described in Figure 39-10, can
Nucleotide position
be transfected individually into a recipient cell. By an-
alyzing when certain hormone responses are lost in
HRE
HRE
HRE
comparison to the 5′ deletion, specific hormone-
A
B
C
responsive elements can be located.
zipper—account for many of these specific protein-
DNA interactions. Examples of proteins containing
3
1
2
these motifs are given in Table 39-3.
Gene
A
Comparison of the binding activities of the proteins
that contain these motifs leads to several important
4
generalizations.
3
1
(1) Binding must be of high affinity to the specific
Gene
B
site and of low affinity to other DNA.
(2) Small regions of the protein make direct contact
2
with DNA; the rest of the protein, in addition to pro-
3
1
5
Gene C
Table 39-3. Examples of transcription regulatory
Figure 39-12. Combinations of DNA elements and
proteins that contain the various binding motifs.
proteins provide diversity in the response of a gene.
Gene A is activated (the width of the arrow indicates
Binding Motif
Organism
Regulatory Protein
the extent) by the combination of activators 1, 2, and 3
Helix-turn-helix
E coli
lac repressor
(probably with coactivators, as shown in Figure 37-10).
CAP
Gene B is activated, in this case more effectively, by the
Phage
λcI, cro, and tryptophan and
combination of 1, 3, and 4; note that 4 does not contact
434 repressors
DNA directly in this example. The activators could form
Mammals
homeo box proteins
a linear bridge that links the basal machinery to the
Pit-1, Oct1, Oct2
promoter, or this could be accomplished by looping
Zinc finger
E coli
Gene 32 protein
out of the DNA. In either case, the purpose is to direct
Yeast
GaI4
the basal transcription machinery to the promoter.
Drosophila
Serendipity, Hunchback
Gene C is inactivated by the combination of 1, 5, and 3;
Xenopus
TFIIIA
in this case, factor 5 is shown to preclude the essential
Mammals
steroid receptor family, Sp1
binding of factor 2 to DNA, as occurs in example A. If
Leucine zipper
Yeast
GCN4
activator 1 helps repressor 5 bind and if activator 1
Mammals
C/EBP, fos, Jun, Fra-1,
binding requires a ligand (solid dot), it can be seen how
CRE binding protein,
the ligand could activate one gene in a cell (gene A)
c-myc, n-myc, I-myc
and repress another (gene C).
REGULATION OF GENE EXPRESSION
/
389
viding the trans-activation domains, may be involved in
The Helix-Turn-Helix Motif
the dimerization of monomers of the binding protein,
may provide a contact surface for the formation of het-
The first motif described—and the one studied most
erodimers, may provide one or more ligand-binding
extensively—is the helix-turn-helix. Analysis of the
sites, or may provide surfaces for interaction with coac-
three-dimensional structure of the λ Cro transcription
tivators or corepressors.
regulator has revealed that each monomer consists of
(3) The protein-DNA interactions are maintained
three antiparallel β sheets and three α helices (Figure
by hydrogen bonds and van der Waals forces.
39-13). The dimer forms by association of the antipar-
(4) The motifs found in these proteins are unique;
allel β3 sheets. The α3 helices form the DNA recogni-
their presence in a protein of unknown function sug-
tion surface, and the rest of the molecule appears to be
gests that the protein may bind to DNA.
involved in stabilizing these structures. The average di-
(5) Proteins with the helix-turn-helix or leucine zip-
ameter of an α helix is 1.2 nm, which is the approxi-
per motifs form symmetric dimers, and their respective
mate width of the major groove in the B form of DNA.
DNA binding sites are symmetric palindromes. In pro-
The DNA recognition domain of each Cro monomer
teins with the zinc finger motif, the binding site is re-
interacts with 5 bp and the dimer binding sites span
peated two to nine times. These features allow for co-
3.4 nm, allowing fit into successive half turns of the
operative interactions between binding sites and
major groove on the same surface (Figure 39-13). X-ray
enhance the degree and affinity of binding.
analyses of the λ cI repressor, CAP (the cAMP receptor
α2
α3
α1
N C
α2
β1
34 Å
β2
β3
α3
Twofold
axis of
β3
symmetry
β2
β1
α3
α
2
C N
α3
α1
34 Å
α2
Figure 39-13. A schematic representation of the three-dimensional structure of Cro protein and its binding to
DNA by its helix-turn-helix motif. The Cro monomer consists of three antiparallel β sheets (β1-β3) and three
α-helices (α1-α3). The helix-turn-helix motif is formed because the α3 and α2 helices are held at about 90 degrees
to each other by a turn of four amino acids. The α3 helix of Cro is the DNA recognition surface (shaded). Two
monomers associate through the antiparallel β3 sheets to form a dimer that has a twofold axis of symmetry
(right). A Cro dimer binds to DNA through its α3 helices, each of which contacts about 5 bp on the same surface of
the major groove. The distance between comparable points on the two DNA α-helices is 34 Å, which is the dis-
tance required for one complete turn of the double helix. (Courtesy of B Mathews.)
390
/
CHAPTER 39
protein of E coli), tryptophan repressor, and phage 434
The Leucine Zipper Motif
repressor all also display this dimeric helix-turn-helix
Careful analysis of a 30-amino-acid sequence in the car-
structure that is present in eukaryotic DNA proteins as
boxyl terminal region of the enhancer binding protein
well (see Table 39-3).
C/EBP revealed a novel structure. As illustrated in Fig-
ure 39-15, this region of the protein forms an α helix
The Zinc Finger Motif
in which there is a periodic repeat of leucine residues at
every seventh position. This occurs for eight helical
The zinc finger was the second DNA binding motif
turns and four leucine repeats. Similar structures have
whose atomic structure was elucidated. It was known
been found in a number of other proteins associated
that the protein TFIIIA, a positive regulator of 5S RNA
with the regulation of transcription in mammalian and
transcription, required zinc for activity. Structural and
yeast cells. It is thought that this structure allows two
biophysical analyses revealed that each TFIIIA molecule
identical monomers or heterodimers (eg, Fos-Jun or
contains nine zinc ions in a repeating coordination
Jun-Jun) to “zip together” in a coiled coil and form a
complex formed by closely spaced cysteine-cysteine
tight dimeric complex (Figure 39-15). This protein-
residues followed 12-13 amino acids later by a histi-
protein interaction may serve to enhance the associa-
dine-histidine pair (Figure 39-14). In some instances—
tion of the separate DNA binding domains with their
notably the steroid-thyroid receptor family—the His-
target (Figure 39-15).
His doublet is replaced by a second Cys-Cys pair. The
protein containing zinc fingers appears to lie on one
THE DNA BINDING & TRANS-ACTIVATION
face of the DNA helix, with successive fingers alterna-
DOMAINS OF MOST REGULATORY
tively positioned in one turn in the major groove. As is
the case with the recognition domain in the helix-turn-
PROTEINS ARE SEPARATE
helix protein, each TFIIIA zinc finger contacts about
& NONINTERACTIVE
5 bp of DNA. The importance of this motif in the ac-
DNA binding could result in a general conformational
tion of steroid hormones is underscored by an “experi-
change that allows the bound protein to activate tran-
ment of nature.” A single amino acid mutation in either
scription, or these two functions could be served by
of the two zinc fingers of the 1,25(OH)2-D3 receptor
separate and independent domains. Domain swap ex-
protein results in resistance to the action of this hor-
periments suggest that the latter is the case.
mone and the clinical syndrome of rickets.
The GAL1 gene product is involved in galactose me-
tabolism in yeast. Transcription of this gene is positively
regulated by the GAL4 protein, which binds to an up-
stream activator sequence (UAS), or enhancer, through
an amino terminal domain. The amino terminal 73-
amino-acid DNA-binding domain (DBD) of GAL4 was
removed and replaced with the DBD of LexA, an E coli
DNA-binding protein. This domain swap resulted in a
molecule that did not bind to the GAL1 UAS and, of
course, did not activate the GAL1 gene (Figure 39-16).
C
C
C
H
If, however, the lexA operator—the DNA sequence nor-
Zn
Zn
mally bound by the lexA DBD—was inserted into the
C
C
C
H
promoter region of the GAL gene, the hybrid protein
bound to this promoter (at the lexA operator) and it ac-
Cys-Cys zinc finger
Cys-His zinc finger
tivated transcription of GAL1. This experiment, which
has been repeated a number of times, affords solid evi-
Figure 39-14. Zinc fingers are a series of repeated
dence that the carboxyl terminal region of GAL4 causes
domains (two to nine) in which each is centered on a
transcriptional activation. These data also demonstrate
tetrahedral coordination with zinc. In the case of TFIIIA,
that the DNA-binding DBD and trans-activation do-
the coordination is provided by a pair of cysteine
mains (ADs) are independent and noninteractive. The
residues (C) separated by 12-13 amino acids from a
hierarchy involved in assembling gene transcription acti-
pair of histidine (H) residues. In other zinc finger pro-
vating complexes includes proteins that bind DNA and
teins, the second pair also consists of C residues. Zinc
trans-activate; others that form protein-protein com-
fingers bind in the major groove, with adjacent fingers
plexes which bridge DNA-binding proteins to trans-
making contact with 5 bp along the same face of the
activating proteins; and others that form protein-protein
helix.
complexes with components of the basal transcription
REGULATION OF GENE EXPRESSION
/
391
A
B
L
L
22
L
15
NH2
L
8
F
1
L
I
V
E
R
N
D
4
5
COOH
COOH
2
T
R
7
R
Q
T
SR
Q
NH2
3
6
S
D
K
E
R
G
DR
Figure 39-15. The leucine zipper motif. A shows a helical wheel analysis of a carboxyl terminal portion of the
DNA binding protein C/EBP. The amino acid sequence is displayed end-to-end down the axis of a schematic
α-helix. The helical wheel consists of seven spokes that correspond to the seven amino acids that comprise every
two turns of the α-helix. Note that leucine residues (L) occur at every seventh position. Other proteins with
“leucine zippers” have a similar helical wheel pattern. B is a schematic model of the DNA binding domain of C/EBP.
Two identical C/EBP polypeptide chains are held in dimer formation by the leucine zipper domain of each
polypeptide (denoted by the rectangles and attached ovals). This association is apparently required to hold the
DNA binding domains of each polypeptide (the shaded rectangles) in the proper conformation for DNA binding.
(Courtesy of S McKnight.)
apparatus. A given protein may thus have several sur-
prokaryotes. These RNA processing steps in eukaryotes,
faces or domains that serve different functions (see Fig-
described in detail in Chapter 37, include capping of
ure 39-17). As described in Chapter 37, the primary
the 5′ ends of the primary transcripts, addition of a
purpose of these complex assemblies is to facilitate the
polyadenylate tail to the 3′ ends of transcripts, and exci-
assembly of the basal transcription apparatus on the cis-
sion of intron regions to generate spliced exons in the
linked promoter.
mature mRNA molecule. To date, analyses of eukary-
otic gene expression provide evidence that regulation
occurs at the level of transcription, nuclear RNA pro-
GENE REGULATION IN PROKARYOTES
cessing, and mRNA stability. In addition, gene ampli-
fication and rearrangement influence gene expression.
& EUKARYOTES DIFFERS IN
Owing to the advent of recombinant DNA technol-
IMPORTANT RESPECTS
ogy, much progress has been made in recent years in
In addition to transcription, eukaryotic cells employ a
the understanding of eukaryotic gene expression. How-
variety of mechanisms to regulate gene expression
ever, because most eukaryotic organisms contain so
(Table 39-4). The nuclear membrane of eukaryotic
much more genetic information than do prokaryotes
cells physically segregates gene transcription from trans-
and because manipulation of their genes is so much
lation, since ribosomes exist only in the cytoplasm.
more limited, molecular aspects of eukaryotic gene reg-
Many more steps, especially in RNA processing, are in-
ulation are less well understood than the examples
volved in the expression of eukaryotic genes than of
discussed earlier in this chapter. This section briefly de-
prokaryotic genes, and these steps provide additional
scribes a few different types of eukaryotic gene regula-
sites for regulatory influences that cannot exist in
tion.
392
/
CHAPTER 39
GAL4
+1
Active
A
UAS
GAL1 gene
LexA-GAL4
+1
Inactive
B
UAS
GAL1 gene
+1
LexA-GAL4
Active
lexA
GAL1 gene
C
operator
Figure 39-16. Domain-swap experiments demonstrate the independent nature of DNA binding and transcrip-
tion activation domains. The GAL1 gene promoter contains an upstream activating sequence (UAS) or enhancer that
binds the regulatory protein GAL4 (A). This interaction results in a stimulation of GAL1 gene transcription. A chimeric
protein, in which the amino terminal DNA binding domain of GAL4 is removed and replaced with the DNA binding
region of the E coli protein LexA, fails to stimulate GAL1 transcription because the LexA domain cannot bind to the
UAS (B). The LexA-GAL4 fusion protein does increase GAL1 transcription when the lexA operator (its natural target)
is inserted into the GAL1 promoter region (C).
Eukaryotic Genes Can Be Amplified
or Rearranged During Development
or in Response to Drugs
During early development of metazoans, there is an
abrupt increase in the need for specific molecules such
2
as ribosomal RNA and messenger RNA molecules for
Activation
proteins that make up such organs as the eggshell. One
domains
1- 4
way to increase the rate at which such molecules can be
3
formed is to increase the number of genes available for
Ligand-binding domain
1
transcription of these specific molecules. Among the
repetitive DNA sequences are hundreds of copies of ri-
4
bosomal RNA genes and tRNA genes. These genes pre-
DNA-binding domain
exist repetitively in the genomic material of the gametes
Table 39-4. Gene expression is regulated by
transcription and in numerous other ways in
Figure 39-17. Proteins that regulate transcription
eukaryotic cells.
have several domains. This hypothetical transcription
factor has a DNA-binding domain (DBD) that is distinct
from a ligand-binding domain (LBD) and several activa-
• Gene amplification
tion domains (ADs) (1-4). Other proteins may lack the
• Gene rearrangement
• RNA processing
DBD or LBD and all may have variable numbers of
• Alternate mRNA splicing
domains that contact other proteins, including
• Transport of mRNA from nucleus to cytoplasm
co-regulators and those of the basal transcription
• Regulation of mRNA stability
complex (see also Chapters 42 and 43).
REGULATION OF GENE EXPRESSION
/
393
and thus are transmitted in high copy numbers from
both immunologic flexibility and specificity. However,
generation to generation. In some specific organisms
a given functional IgG light chain transcription unit—
such as the fruit fly (drosophila), there occurs during
like all other
“normal” mammalian transcription
oogenesis an amplification of a few preexisting genes
units—contains only the coding sequences for a single
such as those for the chorion (eggshell) proteins. Subse-
protein. Thus, before a particular IgG light chain can
quently, these amplified genes, presumably generated
be expressed, single VL, JL, and CL coding sequences
by a process of repeated initiations during DNA syn-
must be recombined to generate a single, contiguous
thesis, provide multiple sites for gene transcription
transcription unit excluding the multiple nonutilized
(Figures 36-16 and 39-18).
segments (ie, the other approximately 300 unused VL
As noted in Chapter 37, the coding sequences re-
segments, the other four unused JL segments, and the
sponsible for the generation of specific protein mole-
other nine unused CL segments). This deletion of un-
cules are frequently not contiguous in the mammalian
used genetic information is accomplished by selective
genome. In the case of antibody encoding genes, this is
DNA recombination that removes the unwanted cod-
particularly true. As described in detail in Chapter 50,
ing DNA while retaining the required coding se-
immunoglobulins are composed of two polypeptides,
quences: one VL, one JL, and one CL sequence. (VL se-
the so-called heavy (about 50 kDa) and light (about 25
quences are subjected to additional point mutagenesis
kDa) chains. The mRNAs encoding these two protein
to generate even more variability—hence the name.)
subunits are encoded by gene sequences that are sub-
The newly recombined sequences thus form a single
jected to extensive DNA sequence-coding changes.
transcription unit that is competent for RNA polym-
These DNA coding changes are integral to generating
erase II-mediated transcription. Although the IgG
the requisite recognition diversity central to appropriate
genes represent one of the best-studied instances of di-
immune function.
rected DNA rearrangement modulating gene expres-
IgG heavy and light chain mRNAs are encoded by
sion, other cases of gene regulatory DNA rearrange-
several different segments that are tandemly repeated in
ment have been described in the literature. Indeed, as
the germline. Thus, for example, the IgG light chain is
detailed below, drug-induced gene amplification is an
composed of variable (VL ), joining (JL ), and constant
important complication of cancer chemotherapy.
(CL ) domains or segments. For particular subsets of
In recent years, it has been possible to promote the
IgG light chains, there are roughly 300 tandemly re-
amplification of specific genetic regions in cultured
peated VL gene coding segments, five tandemly
mammalian cells. In some cases, a several thousand-fold
arranged JL coding sequences, and roughly ten CL gene
increase in the copy number of specific genes can be
coding segments. All of these multiple, distinct coding
achieved over a period of time involving increasing doses
regions are located in the same region of the same chro-
of selective drugs. In fact, it has been demonstrated in
mosome, and each type of coding segment (VL, JL, and
patients receiving methotrexate for cancer that malig-
CL ) is tandemly repeated in head-to-tail fashion within
nant cells can develop drug resistance by increasing the
the segment repeat region. By having multiple VL, JL,
number of genes for dihydrofolate reductase, the target
and CL segments to choose from, an immune cell has a
of methotrexate. Gene amplification events such as these
greater repertoire of sequences to work with to develop
occur spontaneously in vivo—ie, in the absence of ex-
ogenously supplied selective agents—and these unsched-
uled extra rounds of replication can become “frozen” in
the genome under appropriate selective pressures.
Unamplified
s36
s38
Alternative RNA Processing
s36
s38
Is Another Control Mechanism
In addition to affecting the efficiency of promoter uti-
lization, eukaryotic cells employ alternative RNA pro-
Amplified
cessing to control gene expression. This can result when
alternative promoters, intron-exon splice sites, or
polyadenylation sites are used. Occasionally, hetero-
geneity within a cell results, but more commonly the
same primary transcript is processed differently in dif-
Figure 39-18. Schematic representation of the am-
ferent tissues. A few examples of each of these types of
plification of chorion protein genes s36 and s38. (Repro-
regulation are presented below.
duced, with permission, from Chisholm R: Gene amplifica-
The use of alternative transcription start sites re-
tion during development. Trends Biochem Sci 1982;7:161.)
sults in a different 5′ exon on mRNAs corresponding to
394
/
CHAPTER 39
mouse amylase and myosin light chain, rat glucokinase,
cap structure in eukaryotic mRNA prevents attack by 5′
and drosophila alcohol dehydrogenase and actin. Alter-
exonucleases, and the poly(A) tail prohibits the action
native polyadenylation sites in the µ immunoglobulin
of
3′ exonucleases. In mRNA molecules with those
heavy chain primary transcript result in mRNAs that
structures, it is presumed that a single endonucleolytic
are either 2700 bases long (µm) or 2400 bases long (µs).
cut allows exonucleases to attack and digest the entire
This results in a different carboxyl terminal region of
molecule. Other structures (sequences) in the 5′ non-
the encoded proteins such that the µm protein remains
coding sequence (5′ NCS), the coding region, and the
attached to the membrane of the B lymphocyte and the
3′ NCS are thought to promote or prevent this initial
µs immunoglobulin is secreted. Alternative splicing
endonucleolytic action (Figure 39-19). A few illustra-
and processing results in the formation of seven
tive examples will be cited.
unique α-tropomyosin mRNAs in seven different tis-
Deletion of the 5′ NCS results in a threefold to five-
sues. It is not clear how these processing-splicing deci-
fold prolongation of the half-life of c-myc mRNA. Short-
sions are made or whether these steps can be regulated.
ening the coding region of histone mRNA results in a
prolonged half-life. A form of autoregulation of mRNA
stability indirectly involves the coding region. Free tubu-
Regulation of Messenger RNA Stability
lin binds to the first four amino acids of a nascent chain
Provides Another Control Mechanism
of tubulin as it emerges from the ribosome. This appears
Although most mRNAs in mammalian cells are very
to activate an RNase associated with the ribosome (RNP)
stable (half-lives measured in hours), some turn over
which then digests the tubulin mRNA.
very rapidly (half-lives of 10-30 minutes). In certain in-
Structures at the 3′ end, including the poly(A) tail,
stances, mRNA stability is subject to regulation. This
enhance or diminish the stability of specific mRNAs.
has important implications since there is usually a di-
The absence of a poly(A) tail is associated with rapid
rect relationship between mRNA amount and the
degradation of mRNA, and the removal of poly(A)
translation of that mRNA into its cognate protein.
from some RNAs results in their destabilization. His-
Changes in the stability of a specific mRNA can there-
tone mRNAs lack a poly(A) tail but have a sequence
fore have major effects on biologic processes.
near the 3′ terminal that can form a stem-loop struc-
Messenger RNAs exist in the cytoplasm as ribonu-
ture, and this appears to provide resistance to exonucle-
cleoprotein particles (RNPs). Some of these proteins
olytic attack. Histone H4 mRNA, for example, is de-
protect the mRNA from digestion by nucleases, while
graded in the 3′ to 5′ direction but only after a single
others may under certain conditions promote nuclease
endonucleolytic cut occurs about nine nucleotides from
attack. It is thought that mRNAs are stabilized or desta-
the 3′ end in the region of the putative stem-loop struc-
bilized by the interaction of proteins with these various
ture. Stem-loop structures in the
3′ noncoding se-
structures or sequences. Certain effectors, such as hor-
quence are also critical for the regulation, by iron, of
mones, may regulate mRNA stability by increasing or
the mRNA encoding the transferrin receptor. Stem-
decreasing the amount of these proteins.
loop structures are also associated with mRNA stability
It appears that the ends of mRNA molecules are
in bacteria, suggesting that this mechanism may be
involved in mRNA stability (Figure 39-19). The 5′
commonly employed.
Cap
5′ NCS
Coding
3′ NCS
A-A-A-A-An
AUUUA
Figure 39-19. Structure of a typical eukaryotic mRNA showing
elements that are involved in regulating mRNA stability. The typical
eukaryotic mRNA has a 5′ noncoding sequence (5′ NCS), a coding
region, and a 3′ NCS. All are capped at the 5′ end, and most have a
polyadenylate sequence at the 3′ end. The 5′ cap and 3′ poly(A) tail
protect the mRNA against exonuclease attack. Stem-loop structures
in the 5′ and 3′ NCS, features in the coding sequence, and the AU-
rich region in the 3′ NCS are thought to play roles in mRNA stability.
REGULATION OF GENE EXPRESSION
/
395
Other sequences in the 3′ ends of certain eukaryotic
REFERENCES
mRNAs appear to be involved in the destabilization of
Albright SR, Tjian R: TAFs revisited: more data reveal new twists
these molecules. Of particular interest are AU-rich re-
and confirm old ideas. Gene 2000;242:1.
gions, many of which contain the sequence AUUUA.
Bird AP, Wolffe AP: Methylation-induced repression—belts, braces
This sequence appears in mRNAs that have a very short
and chromatin. Cell 1999;99:451.
half-life, including some encoding oncogene proteins
Berger SL, Felsenfeld G: Chromatin goes global. Mol Cell 2001;
and cytokines. The importance of this region is under-
8:263.
scored by an experiment in which a sequence corre-
Busby S, Ebright RH: Promoter structure, promoter recognition,
sponding to the 3′ noncoding region of the short-half-
and transcription activation in prokaryotes. Cell
1994;79:
life colony-stimulating factor
(CSF) mRNA, which
743.
contains the AUUUA motif, was added to the 3′ end of
Busby S, Ebright RH: Transcription activation by catabolite activa-
the β-globin mRNA. Instead of becoming very stable,
tor protein (CAP). J Mol Biol 1999;293:199.
this hybrid β-globin mRNA now had the short-half-life
Cowell IG: Repression versus activation in the control of gene tran-
characteristic of CSF mRNA.
scription. Trends Biochem Sci 1994;1:38.
From the few examples cited, it is clear that a num-
Ebright RH: RNA polymerase: structural similarities between bac-
ber of mechanisms are used to regulate mRNA stabil-
terial RNA polymerase and eukaryotic RNA polymerase II.
J Mol Biol 2000;304:687.
ity—just as several mechanisms are used to regulate the
Fugman SD: RAG1 and RAG2 in V(D)J recombination and trans-
synthesis of mRNA. Coordinate regulation of these two
position. Immunol Res 2001;23:23.
processes confers on the cell remarkable adaptability.
Jacob F, Monod J: Genetic regulatory mechanisms in protein syn-
thesis. J Mol Biol 1961;3:318.
SUMMARY
Lemon B, Tjian R: Orchestrated response: a symphony of tran-
scription factors for gene control. Genes Dev 2000;14:2551.
• The genetic constitutions of nearly all metazoan so-
Letchman DS: Transcription factor mutations and disease. N Engl
matic cells are identical.
J Med 1996;334:28.
• Phenotype (tissue or cell specificity) is dictated by
Merika M, Thanos D: Enhanceosomes. Curr Opin Genet Dev
differences in gene expression of this complement of
2001;11:205.
genes.
Naar AM, Lemon BD, Tjian R: Transcriptional coactivator com-
• Alterations in gene expression allow a cell to adapt to
plexes. Annu Rev Biochem 2001;70:475.
environmental changes.
Narlikar GJ, Fan HY, Kingston RE: Cooperation between com-
plexes that regulate chromatin structure and transcription.
• Gene expression can be controlled at multiple levels
Cell 2002;108:475.
by changes in transcription, RNA processing, local-
Oltz EM: Regulation of antigen receptor gene assembly in lympho-
ization, and stability or utilization. Gene amplifica-
cytes. Immunol Res 2001;23:121.
tion and rearrangements also influence gene expres-
Ptashne M: Control of gene transcription: an outline. Nat Med
sion.
1997;3:1069.
• Transcription controls operate at the level of protein-
Ptashne M: A Genetic Switch, 2nd ed. Cell Press and Blackwell Sci-
DNA and protein-protein interactions. These inter-
entific Publications, 1992.
actions display protein domain modularity and high
Sterner DE, Berger SL: Acetylation of histones and transcription-
specificity.
related factors. Microbiol Mol Biol Rev 2000;64:435.
Wu R, Bahl CP, Narang SA: Lactose operator-repressor interac-
• Several different classes of DNA-binding domains
tion. Curr Top Cell Regul 1978;13:137.
have been identified in transcription factors.
• Chromatin modifications are important in eukary-
otic transcription control.
Molecular Genetics, Recombinant
40
DNA, & Genomic Technology
Daryl K. Granner, MD, & P. Anthony Weil, PhD
BIOMEDICAL IMPORTANCE*
ELUCIDATION OF THE BASIC FEATURES
OF DNA LED TO RECOMBINANT
The development of recombinant DNA, high-density,
high-throughput screening, and other molecular ge-
DNA TECHNOLOGY
netic methodologies has revolutionized biology and is
DNA Is a Complex Biopolymer
having an increasing impact on clinical medicine.
Organized as a Double Helix
Much has been learned about human genetic disease
from pedigree analysis and study of affected proteins,
The fundamental organizational element is the se-
but in many cases where the specific genetic defect is
quence of purine (adenine [A] or guanine [G]) and
unknown, these approaches cannot be used. The new
pyrimidine (cytosine [C] or thymine [T]) bases. These
technologies circumvent these limitations by going di-
bases are attached to the C-1′ position of the sugar de-
rectly to the DNA molecule for information. Manipu-
oxyribose, and the bases are linked together through
lation of a DNA sequence and the construction of
joining of the sugar moieties at their 3′ and 5′ positions
chimeric molecules—so-called genetic engineering—
via a phosphodiester bond (Figure 35-1). The alternat-
provides a means of studying how a specific segment of
ing deoxyribose and phosphate groups form the back-
DNA works. Novel molecular genetic tools allow inves-
bone of the double helix (Figure 35-2). These 3′-5′
tigators to query and manipulate genomic sequences as
linkages also define the orientation of a given strand of
well as to examine both cellular mRNA and protein
the DNA molecule, and, since the two strands run in
profiles at the molecular level.
opposite directions, they are said to be antiparallel.
Understanding this technology is important for sev-
eral reasons: (1) It offers a rational approach to under-
standing the molecular basis of a number of diseases
Base Pairing Is a Fundamental Concept
(eg, familial hypercholesterolemia, sickle cell disease,
of DNA Structure & Function
the thalassemias, cystic fibrosis, muscular dystrophy).
(2) Human proteins can be produced in abundance for
Adenine and thymine always pair, by hydrogen bonding,
therapy (eg, insulin, growth hormone, tissue plasmino-
as do guanine and cytosine (Figure 35-3). These base
gen activator). (3) Proteins for vaccines (eg, hepatitis B)
pairs are said to be complementary, and the guanine
and for diagnostic testing (eg, AIDS tests) can be ob-
content of a fragment of double-stranded DNA will al-
tained. (4) This technology is used to diagnose existing
ways equal its cytosine content; likewise, the thymine
diseases and predict the risk of developing a given dis-
and adenine contents are equal. Base pairing and hy-
ease. (5) Special techniques have led to remarkable ad-
drophobic base-stacking interactions hold the two DNA
vances in forensic medicine. (6) Gene therapy for sickle
strands together. These interactions can be reduced by
cell disease, the thalassemias, adenosine deaminase defi-
heating the DNA to denature it. The laws of base pairing
ciency, and other diseases may be devised.
predict that two complementary DNA strands will rean-
neal exactly in register upon renaturation, as happens
when the temperature of the solution is slowly reduced to
normal. Indeed, the degree of base-pair matching (or
* See glossary of terms at the end of this chapter.
mismatching) can be estimated from the temperature re-
396
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
397
quired for denaturation-renaturation. Segments of DNA
exons. Regulatory regions for specific eukaryotic genes
with high degrees of base-pair matching require more en-
are usually located in the DNA that flanks the tran-
ergy input (heat) to accomplish denaturation—or, to put
scription initiation site at its
5′ end
(5′ flanking-
it another way, a closely matched segment will withstand
sequence DNA). Occasionally, such sequences are
more heat before the strands separate. This reaction is
found within the gene itself or in the region that flanks
used to determine whether there are significant differ-
the 3′ end of the gene. In mammalian cells, each gene
ences between two DNA sequences, and it underlies the
has its own regulatory region. Many eukaryotic genes
concept of hybridization, which is fundamental to the
(and some viruses that replicate in mammalian cells)
processes described below.
have special regions, called enhancers, that increase the
There are about 3
109 base pairs (bp) in each
rate of transcription. Some genes also have DNA se-
human haploid genome. If an average gene length is
quences, known as silencers, that repress transcription.
3 × 103 bp (3 kilobases [kb]), the genome could consist
Mammalian genes are obviously complicated, multi-
of 106 genes, assuming that there is no overlap and that
component structures.
transcription proceeds in only one direction. It is
thought that there are < 105 genes in the human and
Genes Are Transcribed Into RNA
that only 1-2% of the DNA codes for proteins. The
Information generally flows from DNA to mRNA to
exact function of the remaining ~98% of the human
protein, as illustrated in Figure 40-1 and discussed in
genome has not yet been defined.
more detail in Chapter 39. This is a rigidly controlled
The double-helical DNA is packaged into a more
process involving a number of complex steps, each of
compact structure by a number of proteins, most
which no doubt is regulated by one or more enzymes or
notably the basic proteins called histones. This con-
factors; faulty function at any of these steps can cause
densation may serve a regulatory role and certainly has
disease.
a practical purpose. The DNA present within the nu-
cleus of a cell, if simply extended, would be about
RECOMBINANT DNA TECHNOLOGY
1 meter long. The chromosomal proteins compact this
long strand of DNA so that it can be packaged into a
INVOLVES ISOLATION & MANIPULATION
nucleus with a volume of a few cubic micrometers.
OF DNA TO MAKE CHIMERIC MOLECULES
Isolation and manipulation of DNA, including end-to-
DNA Is Organized Into Genes
end joining of sequences from very different sources to
make chimeric molecules
(eg, molecules containing
In general, prokaryotic genes consist of a small regula-
both human and bacterial DNA sequences in a se-
tory region (100-500 bp) and a large protein-coding
quence-independent fashion), is the essence of recom-
segment (500-10,000 bp). Several genes are often con-
binant DNA research. This involves several unique
trolled by a single regulatory unit. Most mammalian
techniques and reagents.
genes are more complicated in that the coding regions
are interrupted by noncoding regions that are elimi-
Restriction Enzymes Cut DNA
nated when the primary RNA transcript is processed
Chains at Specific Locations
into mature messenger RNA (mRNA). The coding re-
gions (those regions that appear in the mature RNA
Certain endonucleases—enzymes that cut DNA at spe-
species) are called exons, and the noncoding regions,
cific DNA sequences within the molecule (as opposed
which interpose or intervene between the exons, are
to exonucleases, which digest from the ends of DNA
called introns
(Figure
40-1). Introns are always re-
molecules)—are a key tool in recombinant DNA re-
moved from precursor RNA before transport into the
search. These enzymes were called restriction enzymes
cytoplasm occurs. The process by which introns are re-
because their presence in a given bacterium restricted
moved from precursor RNA and by which exons are
the growth of certain bacterial viruses called bacterio-
ligated together is called RNA splicing. Incorrect pro-
phages. Restriction enzymes cut DNA of any source
cessing of the primary transcript into the mature
into short pieces in a sequence-specific manner—in
mRNA can result in disease in humans (see below); this
contrast to most other enzymatic, chemical, or physical
underscores the importance of these posttranscriptional
methods, which break DNA randomly. These defensive
processing steps. The variation in size and complexity
enzymes (hundreds have been discovered) protect the
of some human genes is illustrated in Table 40-1. Al-
host bacterial DNA from DNA from foreign organisms
though there is a 300-fold difference in the sizes of the
(primarily infective phages). However, they are present
genes illustrated, the mRNA sizes vary only about 20-
only in cells that also have a companion enzyme which
fold. This is because most of the DNA in genes is pres-
methylates the host DNA, rendering it an unsuitable
ent as introns, and introns tend to be much larger than
substrate for digestion by the restriction enzyme. Thus,
398
/
CHAPTER 40
Regulatory
Basal
Transcription
Poly(A)
region
promoter
start site
addition
region
site
Exon
Exon
DNA
5′
CAAT
TATA
AATAAA
3′
5′
Intron
3′
Noncoding
Noncoding
region
region
Transcription
NUCLEUS
Primary RNA transcript
PPP
Modification of
5′ and 3′ ends
Modified transcript
AAA---A
Poly(A) tail
Cap
Removal of introns
and splicing of exons
Processed nuclear mRNA
AAA---A
Transmembrane
CYTOPLASM
transport
AAA---A
mRNA
Translation
Protein
NH2
COOH
Figure 40-1. Organization of a eukaryotic transcription unit and the pathway of eukaryotic gene expres-
sion. Eukaryotic genes have structural and regulatory regions. The structural region consists of the coding
DNA and 5′ and 3′ noncoding DNA sequences. The coding regions are divided into two parts: (1) exons, which
eventually are ligated together to become mature RNA, and (2) introns, which are processed out of the pri-
mary transcript. The structural region is bounded at its 5′ end by the transcription initiation site and at its
3′ end by the polyadenylate addition or termination site. The promoter region, which contains specific DNA
sequences that interact with various protein factors to regulate transcription, is discussed in detail in Chap-
ters 37 and 39. The primary transcript has a special structure, a cap, at the 5′ end and a stretch of As at the 3′
end. This transcript is processed to remove the introns; and the mature mRNA is then transported to the cyto-
plasm, where it is translated into protein.
site-specific DNA methylases and restriction enzymes
HpaI) or overlapping (sticky) ends (eg, BamHI) (Figure
always exist in pairs in a bacterium.
40-2), depending on the mechanism used by the en-
Restriction enzymes are named after the bac-
zyme. Sticky ends are particularly useful in constructing
terium from which they are isolated. For example,
hybrid or chimeric DNA molecules (see below). If the
EcoRI is from Escherichia coli, and BamHI is from Bacil-
four nucleotides are distributed randomly in a given
lus amyloliquefaciens (Table 40-2). The first three letters
DNA molecule, one can calculate how frequently a
in the restriction enzyme name consist of the first letter
given enzyme will cut a length of DNA. For each posi-
of the genus (E) and the first two letters of the species
tion in the DNA molecule, there are four possibilities
(co). These may be followed by a strain designation (R)
(A, C, G, and T); therefore, a restriction enzyme that
and a roman numeral (I) to indicate the order of discov-
recognizes a 4-bp sequence cuts, on average, once every
ery (eg, EcoRI, EcoRII). Each enzyme recognizes and
256 bp (44), whereas another enzyme that recognizes a
cleaves a specific double-stranded DNA sequence that is
6-bp sequence cuts once every 4096 bp (46). A given
4-7 bp long. These DNA cuts result in blunt ends (eg,
piece of DNA has a characteristic linear array of sites for
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
399
Table 40-1. Variations in the size and complexity
Table 40-2. Selected restriction endonucleases
of some human genes and mRNAs.1
and their sequence specificities.1
mRNA
Sequence Recognized
Bacterial
Gene Size
Number
Size
Endonuclease
Cleavage Sites Shown
Source
Gene
(kb)
of Introns
(kb)
↓
β-Globin
1.5
2
0.6
BamHI
GGATCC
Bacillus amylo-
Insulin
1.7
2
0.4
CCTAGG
liquefaciens H
β-Adrenergic receptor
3
0
2.2
↑
Albumin
25
14
2.1
↓
LDL receptor
45
17
5.5
BgIII
AGATCT
Bacillus glolbigii
Factor VIII
186
25
9.0
TCTAGA
Thyroglobulin
300
36
8.7
↑
1The sizes are given in kilobases (kb). The sizes of the genes in-
↓
clude some proximal promoter and regulatory region sequences;
EcoRI
GAATTC
Escherichia coli
these are generally about the same size for all genes. Genes vary
CTTAAG
RY13
in size from about 1500 base pairs (bp) to over 2 × 106 bp. There is
↑
also great variation in the number of introns and exons. The
↓
β-adrenergic receptor gene is intronless, and the thyroglobulin
EcoRII
CCTGG
Escherichia coli
gene has 36 introns. As noted by the smaller difference in mRNA
GGACC
R245
sizes, introns comprise most of the gene sequence.
↑
↓
HindIII
AAGCTT
Haemophilus
the various enzymes dictated by the linear sequence of
TTCGAA
influenzae Rd
its bases; hence, a restriction map can be constructed.
↑
When DNA is digested with a given enzyme, the ends
↓
of all the fragments have the same DNA sequence. The
Hhal
GCGC
Haemophilus
fragments produced can be isolated by electrophoresis
CGCG
haemolyticus
on agarose or polyacrylamide gels (see the discussion of
↑
blot transfer, below); this is an essential step in cloning
and a major use of these enzymes.
↓
Hpal
GTTAAC
Haemophilus
A number of other enzymes that act on DNA and
CAATTG
parainfluenzae
RNA are an important part of recombinant DNA tech-
↑
nology. Many of these are referred to in this and subse-
quent chapters (Table 40-3).
↓
MstII
CCTNAGG
Microcoleus
GGANTCC
strain
Restriction Enzymes & DNA Ligase Are
↑
Used to Prepare Chimeric DNA Molecules
↓
Sticky-end ligation is technically easy, but some special
PstI
CTGCAG
Providencia
techniques are often required to overcome problems in-
GACGTC
stuartii 164
herent in this approach. Sticky ends of a vector may re-
↑
connect with themselves, with no net gain of DNA.
↓
Sticky ends of fragments can also anneal, so that tandem
Taql
TCGA
Thermus
heterogeneous inserts form. Also, sticky-end sites may
AGCT
aquaticus YTI
not be available or in a convenient position. To circum-
↑
vent these problems, an enzyme that generates blunt
1A, adenine; C, cytosine; G, guanine, T, thymine. Arrows show the site
ends is used, and new ends are added using the enzyme
of cleavage; depending on the site, sticky ends (BamHI) or blunt ends
terminal transferase. If poly d(G) is added to the 3′ ends
(Hpal) may result. The length of the recognition sequence can be 4 bp
of the vector and poly d(C) is added to the 3′ ends of
(Taql), 5 bp (EcoRII), 6 bp (EcoRI), or 7 bp (MstII) or longer. By conven-
tion, these are written in the 5′ or 3′ direction for the upper strand of
the foreign DNA, the two molecules can only anneal to
each recognition sequence, and the lower strand is shown with the
each other, thus circumventing the problems listed
opposite (ie, 3′ or 5′) polarity. Note that most recognition sequences
above. This procedure is called homopolymer tailing.
are palindromes (ie, the sequence reads the same in opposite direc-
Sometimes, synthetic blunt-ended duplex oligonu-
tions on the two strands). A residue designated N means that any nu-
cleotide linkers with a convenient restriction enzyme se-
cleotide is permitted.
400
/
CHAPTER 40
A. Sticky or staggered ends
5’
G
G
A
T
C
C
3’
G
G
A
T
C
C
BamHI
Figure 40-2. Results of restriction en-
+
donuclease digestion. Digestion with a re-
3’
C
C
T
A
G
G
5’
C
C
T
A G
G
striction endonuclease can result in the for-
B. Blunt ends
mation of DNA fragments with sticky, or
5’
G
T
T
A
A
C
3’
G
T
T
A
A
C
cohesive, ends (A) or blunt ends (B). This is
HpaI
+
an important consideration in devising
3’
C A A T T G
5’
C A A
T T G
cloning strategies.
quence are ligated to the blunt-ended DNA. Direct
terized or used for other purposes. This technique is
blunt-end ligation is accomplished using the enzyme
based on the fact that chimeric or hybrid DNA molecules
bacteriophage T4 DNA ligase. This technique, though
can be constructed in cloning vectors—typically bacter-
less efficient than sticky-end ligation, has the advantage
ial plasmids, phages, or cosmids—which then continue
of joining together any pairs of ends. The disadvantages
to replicate in a host cell under their own control systems.
are that there is no control over the orientation of inser-
In this way, the chimeric DNA is amplified. The general
tion or the number of molecules annealed together, and
procedure is illustrated in Figure 40-3.
there is no easy way to retrieve the insert.
Bacterial plasmids are small, circular, duplex DNA
molecules whose natural function is to confer antibiotic
resistance to the host cell. Plasmids have several proper-
Cloning Amplifies DNA
ties that make them extremely useful as cloning vectors.
A clone is a large population of identical molecules, bac-
They exist as single or multiple copies within the bac-
teria, or cells that arise from a common ancestor. Molec-
terium and replicate independently from the bacterial
ular cloning allows for the production of a large number
DNA. The complete DNA sequence of many plasmids is
of identical DNA molecules, which can then be charac-
known; hence, the precise location of restriction enzyme
Table 40-3. Some of the enzymes used in recombinant DNA research.1
Enzyme
Reaction
Primary Use
Alkaline phosphatase
Dephosphorylates 5′ ends of RNA and DNA.
Removal of 5′-PO4 groups prior to kinase labeling to prevent
self-ligation.
BAL 31 nuclease
Degrades both the 3′ and 5′ ends of DNA.
Progressive shortening of DNA molecules.
DNA ligase
Catalyzes bonds between DNA molecules.
Joining of DNA molecules.
DNA polymerase I
Synthesizes double-stranded DNA from
Synthesis of double-stranded cDNA; nick translation; gener-
single-stranded DNA.
ation of blunt ends from sticky ends.
DNase I
Under appropriate conditions, produces
Nick translation; mapping of hypersensitive sites; mapping
single-stranded nicks in DNA.
protein-DNA interactions.
Exonuclease III
Removes nucleotides from 3′ ends of DNA.
DNA sequencing; mapping of DNA-protein interactions.
λ exonuclease
Removes nucleotides from 5′ ends of DNA.
DNA sequencing.
Polynucleotide kinase
Transfers terminal phosphate (γ position)
32P labeling of DNA or RNA.
from ATP to 5′-OH groups of DNA or RNA.
Reverse transcriptase
Synthesizes DNA from RNA template.
Synthesis of cDNA from mRNA; RNA (5′ end) mapping
studies.
S1 nuclease
Degrades single-stranded DNA.
Removal of “hairpin” in synthesis of cDNA; RNA mapping
studies (both 5′ and 3′ ends).
Terminal transferase
Adds nucleotides to the 3′ ends of DNA.
Homopolymer tailing.
1Adapted and reproduced, with permission, from Emery AEH: Page 41 in: An Introduction to Recombinant DNA. Wiley, 1984.
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
401
EcoRI
T
restriction
T
A
endonuclease
A
A
A
T
Human DNA
T
Circular plasmid DNA
Linear plasmid DNA
with sticky ends
EcoRI restriction
endonuclease
AA T T
A
A
A
A
T
T
T T
AA
T
T
T
T
TA
Piece of human DNA cut with
A
T AA
DNA
same restriction nuclease and
ligase
containing same sticky ends
T
T
AT
A
AT
A
A
A
A
A
T
T
T
T
Plasmid DNA molecule with human DNA insert
(recombinant DNA molecule)
Figure 40-3. Use of restriction nucleases to make new recombinant or chimeric DNA molecules. When in-
serted back into a bacterial cell (by the process called transformation), typically only a single plasmid is taken up
by a single cell, and the plasmid DNA replicates not only itself but also the physically linked new DNA insert. Since
recombining the sticky ends, as indicated, regenerates the same DNA sequence recognized by the original restric-
tion enzyme, the cloned DNA insert can be cleanly cut back out of the recombinant plasmid circle with this en-
donuclease. If a mixture of all of the DNA pieces created by treatment of total human DNA with a single restriction
nuclease is used as the source of human DNA, a million or so different types of recombinant DNA molecules can
be obtained, each pure in its own bacterial clone. (Modified and reproduced, with permission, from Cohen SN: The
manipulation of genes. Sci Am [July] 1975;233:34.)
cleavage sites for inserting the foreign DNA is available.
Larger fragments of DNA can be cloned in cosmids,
Plasmids are smaller than the host chromosome and are
which combine the best features of plasmids and
therefore easily separated from the latter, and the desired
phages. Cosmids are plasmids that contain the DNA se-
plasmid-inserted DNA is readily removed by cutting the
quences, so-called cos sites, required for packaging
plasmid with the enzyme specific for the restriction site
lambda DNA into the phage particle. These vectors
into which the original piece of DNA was inserted.
grow in the plasmid form in bacteria, but since much of
Phages usually have linear DNA molecules into
the unnecessary lambda DNA has been removed, more
which foreign DNA can be inserted at several restric-
chimeric DNA can be packaged into the particle head.
tion enzyme sites. The chimeric DNA is collected after
It is not unusual for cosmids to carry inserts of chimeric
the phage proceeds through its lytic cycle and produces
DNA that are 35-50 kb long. Even larger pieces of
mature, infective phage particles. A major advantage of
DNA can be incorporated into bacterial artificial chro-
phage vectors is that while plasmids accept DNA pieces
mosome (BAC), yeast artificial chromosome (YAC), or
about 6-10 kb long, phages can accept DNA fragments
E. coli bacteriophage P1-based (PAC) vectors. These
10-20 kb long, a limitation imposed by the amount of
vectors will accept and propagate DNA inserts of sev-
DNA that can be packed into the phage head.
eral hundred kilobases or more and have largely re-
402
/
CHAPTER 40
Table 40-4. Cloning capacities of common
ferent recombinant clones is called a library. A genomic
cloning vectors.
library is prepared from the total DNA of a cell line or
tissue. A cDNA library comprises complementary
DNA copies of the population of mRNAs in a tissue.
Vector
DNA Insert Size
Genomic DNA libraries are often prepared by perform-
Plasmid pBR322
0.01-10 kb
ing partial digestion of total DNA with a restriction en-
Lambda charon 4A
10-20 kb
zyme that cuts DNA frequently (eg, a four base cutter
Cosmids
35-50 kb
such as TaqI ). The idea is to generate rather large frag-
BAC, P1
50-250 kb
ments so that most genes will be left intact. The BAC,
YAC
500-3000 kb
YAC, and P1 vectors are preferred since they can accept
very large fragments of DNA and thus offer a better
placed the plasmid, phage, and cosmid vectors for some
chance of isolating an intact gene on a single DNA
cloning and gene mapping applications. A comparison
fragment.
of these vectors is shown in Table 40-4.
A vector in which the protein coded by the gene in-
Because insertion of DNA into a functional region
troduced by recombinant DNA technology is actually
of the vector will interfere with the action of this re-
synthesized is known as an expression vector. Such
gion, care must be taken not to interrupt an essential
vectors are now commonly used to detect specific
function of the vector. This concept can be exploited,
cDNA molecules in libraries and to produce proteins
however, to provide a selection technique. For example,
by genetic engineering techniques. These vectors are
the common plasmid vector pBR322 has both tetracy-
specially constructed to contain very active inducible
cline (tet) and ampicillin (amp) resistance genes. A
promoters, proper in-phase translation initiation
single PstI restriction enzyme site within the amp resis-
codons, both transcription and translation termination
tance gene is commonly used as the insertion site for a
signals, and appropriate protein processing signals, if
piece of foreign DNA. In addition to having sticky ends
needed. Some expression vectors even contain genes
(Table 40-2 and Figure 40-2), the DNA inserted at
that code for protease inhibitors, so that the final yield
this site disrupts the amp resistance gene and makes the
of product is enhanced.
bacterium carrying this plasmid amp-sensitive (Figure
40-4). Thus, the parental plasmid, which provides re-
Probes Search Libraries for Specific
sistance to both antibiotics, can be readily separated
Genes or cDNA Molecules
from the chimeric plasmid, which is resistant only to
A variety of molecules can be used to “probe” libraries in
tetracycline. YACs contain replication and segregation
functions that work in both bacteria and yeast cells and
search of a specific gene or cDNA molecule or to define
and quantitate DNA or RNA separated by electrophore-
therefore can be propagated in either organism.
In addition to the vectors described in Table 40-4
sis through various gels. Probes are generally pieces of
DNA or RNA labeled with a 32P-containing nu-
that are designed primarily for propagation in bacterial
cells, vectors for mammalian cell propagation and insert
cleotide—or fluorescently labeled nucleotides
(more
commonly now). Importantly, neither modification (32P
gene (cDNA)/protein expression have also been devel-
oped. These vectors are all based upon various eukary-
or fluorescent-label) affects the hybridization properties
of the resulting labeled nucleic acid probes. The probe
otic viruses that are composed of RNA or DNA
genomes. Notable examples of such viral vectors are
must recognize a complementary sequence to be effec-
tive. A cDNA synthesized from a specific mRNA can be
those utilizing adenoviral (DNA-based) and retroviral
(RNA-based) genomes. Though somewhat limited in
used to screen either a cDNA library for a longer cDNA
or a genomic library for a complementary sequence in
the size of DNA sequences that can be inserted, such
mammalian viral cloning vectors make up for this
the coding region of a gene. A popular technique for
finding specific genes entails taking a short amino acid
shortcoming because they will efficiently infect a wide
range of different cell types. For this reason, various
sequence and, employing the codon usage for that
species
(see Chapter 38), making an oligonucleotide
mammalian viral vectors are being investigated for use
in gene therapy experiments.
probe that will detect the corresponding DNA fragment
in a genomic library. If the sequences match exactly,
probes 15-20 nucleotides long will hybridize. cDNA
A Library Is a Collection
probes are used to detect DNA fragments on Southern
of Recombinant Clones
blot transfers and to detect and quantitate RNA on
The combination of restriction enzymes and various
Northern blot transfers. Specific antibodies can also be
cloning vectors allows the entire genome of an organ-
used as probes provided that the vector used synthesizes
ism to be packed into a vector. A collection of these dif-
protein molecules that are recognized by them.
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
403
Ampicillin
Tetracycline
resistance gene
resistance gene
EcoRI
EcoRI
Tetracycline
HindIII
resistance gene
HindIII
PstI
BamHI
BamHI
Cut open with
PstI
SalI
PstI
PstI
SalI
Then insert
PstI-cut DNA
Ampr
Amps
Tetr
Tetr
Host pBR322
Chimeric pBR322
Figure 40-4. A method of screening recombinants for inserted DNA fragments. Using the plasmid pBR322, a
piece of DNA is inserted into the unique PstI site. This insertion disrupts the gene coding for a protein that pro-
vides ampicillin resistance to the host bacterium. Hence, the chimeric plasmid will no longer survive when plated
on a substrate medium that contains this antibiotic. The differential sensitivity to tetracycline and ampicillin can
therefore be used to distinguish clones of plasmid that contain an insert. A similar scheme relying upon produc-
tion of an in-frame fusion of a newly inserted DNA producing a peptide fragment capable of complementing an
inactive, deleted form of the enzyme β-galactosidase allows for blue-white colony formation on agar plates con-
taining a dye hydrolyzable by β-galactoside. β-Galactosidase-positive colonies are blue.
Blotting & Hybridization Techniques Allow
renatured, and analyzed for an interaction by hybridiza-
Visualization of Specific Fragments
tion with a specific labeled DNA probe.
Colony or plaque hybridization is the method by
Visualization of a specific DNA or RNA fragment
which specific clones are identified and purified. Bacte-
among the many thousands of “contaminating” mole-
ria are grown on colonies on an agar plate and overlaid
cules requires the convergence of a number of tech-
with nitrocellulose filter paper. Cells from each colony
niques, collectively termed blot transfer. Figure 40-5
stick to the filter and are permanently fixed thereto by
illustrates the Southern (DNA), Northern (RNA), and
heat, which with NaOH treatment also lyses the cells
Western (protein) blot transfer procedures. (The first is
and denatures the DNA so that it will hybridize with
named for the person who devised the technique, and
the probe. A radioactive probe is added to the filter,
the other names began as laboratory jargon but are now
and (after washing) the hybrid complex is localized by
accepted terms.) These procedures are useful in deter-
exposing the filter to x-ray film. By matching the spot
mining how many copies of a gene are in a given tissue
on the autoradiograph to a colony, the latter can be
or whether there are any gross alterations in a gene
picked from the plate. A similar strategy is used to iden-
(deletions, insertions, or rearrangements). Occasionally,
tify fragments in phage libraries. Successive rounds of
if a specific base is changed and a restriction site is al-
this procedure result in a clonal isolate
(bacterial
tered, these procedures can detect a point mutation.
colony) or individual phage plaque.
The Northern and Western blot transfer techniques are
All of the hybridization procedures discussed in this
used to size and quantitate specific RNA and protein
section depend on the specific base-pairing properties
molecules, respectively. A fourth hybridization tech-
of complementary nucleic acid strands described above.
nique, the Southwestern blot, examines protein•DNA
Perfect matches hybridize readily and withstand high
interactions. Proteins are separated by electrophoresis,
temperatures in the hybridization and washing reac-
404
/
CHAPTER 40
Southern
Northern
Western
tions. Specific complexes also form in the presence of
low salt concentrations. Less than perfect matches do
DNA
RNA
Protein
not tolerate these stringent conditions (ie, elevated
temperatures and low salt concentrations); thus, hy-
bridization either never occurs or is disrupted during
Gel
the washing step. Gene families, in which there is some
electrophoresis
degree of homology, can be detected by varying the
stringency of the hybridization and washing steps.
Cross-species comparisons of a given gene can also be
made using this approach. Hybridization conditions ca-
pable of detecting just a single base pair mismatch be-
tween probe and target have been devised.
Transfer to paper
Manual & Automatic Techniques
Are Available to Determine
cDNA*
cDNA*
Antibody* Add probe
the Sequence of DNA
The segments of specific DNA molecules obtained by
recombinant DNA technology can be analyzed to de-
Autoradiograph
termine their nucleotide sequence. This method de-
pends upon having a large number of identical DNA
molecules. This requirement can be satisfied by cloning
Figure 40-5. The blot transfer procedure. In a
the fragment of interest, using the techniques described
Southern, or DNA, blot transfer, DNA isolated from a
above. The manual enzymatic method (Sanger) em-
cell line or tissue is digested with one or more restric-
ploys specific dideoxynucleotides that terminate DNA
tion enzymes. This mixture is pipetted into a well in an
strand synthesis at specific nucleotides as the strand is
agarose or polyacrylamide gel and exposed to a direct
synthesized on purified template nucleic acid. The reac-
electrical current. DNA, being negatively charged, mi-
tions are adjusted so that a population of DNA frag-
grates toward the anode; the smaller fragments move
ments representing termination at every nucleotide is
the most rapidly. After a suitable time, the DNA is dena-
obtained. By having a radioactive label incorporated at
tured by exposure to mild alkali and transferred to ni-
the end opposite the termination site, one can separate
trocellulose or nylon paper, in an exact replica of the
the fragments according to size using polyacrylamide
pattern on the gel, by the blotting technique devised
gel electrophoresis. An autoradiograph is made, and
by Southern. The DNA is bound to the paper by expo-
each of the fragments produces an image (band) on an
sure to heat, and the paper is then exposed to the
x-ray film. These are read in order to give the DNA se-
labeled cDNA probe, which hybridizes to complemen-
quence (Figure 40-6). Another manual method, that of
tary fragments on the filter. After thorough washing,
Maxam and Gilbert, employs chemical methods to
the paper is exposed to x-ray film, which is developed
cleave the DNA molecules where they contain the spe-
to reveal several specific bands corresponding to the
cific nucleotides. Techniques that do not require the
DNA fragment that recognized the sequences in the
use of radioisotopes are commonly employed in auto-
mated DNA sequencing. Most commonly employed is
cDNA probe. The RNA, or Northern, blot is conceptually
an automated procedure in which four different fluo-
similar. RNA is subjected to electrophoresis before blot
rescent labels—one representing each nucleotide—are
transfer. This requires some different steps from those
used. Each emits a specific signal upon excitation by a
of DNA transfer, primarily to ensure that the RNA re-
laser beam, and this can be recorded by a computer.
mains intact, and is generally somewhat more difficult.
In the protein, or Western, blot, proteins are elec-
trophoresed and transferred to nitrocellulose and then
Oligonucleotide Synthesis Is Now Routine
probed with a specific antibody or other probe mole-
The automated chemical synthesis of moderately long
cule. (Asterisks signify labeling, either radioactive or
oligonucleotides (about 100 nucleotides) of precise se-
fluorescent.)
quence is now a routine laboratory procedure. Each
synthetic cycle takes but a few minutes, so an entire
molecule can be made by synthesizing relatively short
segments that can then be ligated to one another.
Oligonucleotides are now indispensable for DNA se-
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
405
Reaction containing radiolabel:
Sequence of original strand:
ddGTP ddATP ddTTP ddCTP
- A - G - T -C - T - T - G - G- A - G - C - T
- 3′
G
A
T
C
A G T C T T G G A G C T
Bases terminated
Figure 40-6. Sequencing of DNA by the method devised by Sanger. The ladder-like arrays represent from bot-
tom to top all of the successively longer fragments of the original DNA strand. Knowing which specific dideoxynu-
cleotide reaction was conducted to produce each mixture of fragments, one can determine the sequence of nu-
cleotides from the labeled end (asterisk) toward the unlabeled end by reading up the gel. Automated sequencing
involves the reading of chemically modified deoxynucleotides. The base-pairing rules of Watson and Crick (A-T,
G-C) dictate the sequence of the other (complementary) strand. (Asterisks signify radiolabeling.)
quencing, library screening, protein-DNA binding,
quences, and extension of the annealed primers with
DNA mobility shift assays, the polymerase chain reac-
DNA polymerase result in the exponential amplifica-
tion (see below), site-directed mutagenesis, and numer-
tion of DNA segments of defined length. Early PCR re-
ous other applications.
actions used an E coli DNA polymerase that was de-
stroyed by each heat denaturation cycle. Substitution of
a heat-stable DNA polymerase from Thermus aquaticus
The Polymerase Chain Reaction
(or the corresponding DNA polymerase from other
(PCR) Amplifies DNA Sequences
thermophilic bacteria), an organism that lives and repli-
The polymerase chain reaction (PCR) is a method of
cates at 70-80 °C, obviates this problem and has made
amplifying a target sequence of DNA. PCR provides a
possible automation of the reaction, since the polym-
sensitive, selective, and extremely rapid means of ampli-
erase reactions can be run at 70 °C. This has also im-
fying a desired sequence of DNA. Specificity is based
proved the specificity and the yield of DNA.
on the use of two oligonucleotide primers that hy-
DNA sequences as short as 50-100 bp and as long
bridize to complementary sequences on opposite
as 10 kb can be amplified. Twenty cycles provide an
strands of DNA and flank the target sequence (Figure
amplification of 106 and 30 cycles of 109. The PCR al-
40-7). The DNA sample is first heated to separate the
lows the DNA in a single cell, hair follicle, or spermato-
two strands; the primers are allowed to bind to the
zoon to be amplified and analyzed. Thus, the applica-
DNA; and each strand is copied by a DNA polymerase,
tions of PCR to forensic medicine are obvious. The
starting at the primer site. The two DNA strands each
PCR is also used (1) to detect infectious agents, espe-
serve as a template for the synthesis of new DNA from
cially latent viruses; (2) to make prenatal genetic diag-
the two primers. Repeated cycles of heat denaturation,
noses; (3) to detect allelic polymorphisms; (4) to estab-
annealing of the primers to their complementary se-
lish precise tissue types for transplants; and (5) to study
406
/
CHAPTER 40
evolution, using DNA from archeological samples after
Targeted sequence
RNA copying and mRNA quantitation by the so-called
RT-PCR method (cDNA copies of mRNA generated
START
by a retroviral reverse transcriptase). There are an equal
number of applications of PCR to problems in basic
science, and new uses are developed every year.
CYCLE 1
PRACTICAL APPLICATIONS
OF RECOMBINANT DNA TECHNOLOGY
ARE NUMEROUS
The isolation of a specific gene from an entire genome
CYCLE 2
requires a technique that will discriminate one part in a
million. The identification of a regulatory region that
may be only 10 bp in length requires a sensitivity of
one part in 3 × 108; a disease such as sickle cell anemia
is caused by a single base change, or one part in 3 × 109.
Recombinant DNA technology is powerful enough to
accomplish all these things.
Gene Mapping Localizes Specific
Genes to Distinct Chromosomes
Gene localizing thus can define a map of the human
genome. This is already yielding useful information in
CYCLE 3
the definition of human disease. Somatic cell hybridiza-
tion and in situ hybridization are two techniques used
to accomplish this. In in situ hybridization, the sim-
pler and more direct procedure, a radioactive probe is
added to a metaphase spread of chromosomes on a glass
slide. The exact area of hybridization is localized by lay-
ering photographic emulsion over the slide and, after
exposure, lining up the grains with some histologic
identification of the chromosome. Fluorescence in situ
hybridization (FISH) is a very sensitive technique that
is also used for this purpose. This often places the gene
at a location on a given band or region on the chromo-
some. Some of the human genes localized using these
techniques are listed in Table 40-5. This table repre-
sents only a sampling, since thousands of genes have
been mapped as a result of the recent sequencing of the
Figure 40-7. The polymerase chain reaction is used to
amplify specific gene sequences. Double-stranded DNA is
heated to separate it into individual strands. These bind two
distinct primers that are directed at specific sequences on
opposite strands and that define the segment to be ampli-
fied. DNA polymerase extends the primers in each direction
and synthesizes two strands complementary to the original
two. This cycle is repeated several times, giving an amplified
product of defined length and sequence. Note that the two
CYCLES 4-n
primers are present in excess.
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
407
Table 40-5. Localization of human genes.1
Gene
Chromosome
Disease
Insulin
11p15
Prolactin
6p23-q12
Growth hormone
17q21-qter
Growth hormone deficiency
α-Globin
16p12-pter
α-Thalassemia
β-Globin
11p12
β-Thalassemia, sickle cell
Adenosine deaminase
20q13-qter
Adenosine deaminase deficiency
Phenylalanine hydroxylase
12q24
Phenylketonuria
Hypoxanthine-guanine
Xq26-q27
Lesch-Nyhan syndrome
phosphoribosyltransferase
DNA segment G8
4p
Huntington’s chorea
1This table indicates the chromosomal location of several genes and the diseases asso-
ciated with deficient or abnormal production of the gene products. The chromosome
involved is indicated by the first number or letter. The other numbers and letters refer
to precise localizations, as defined in McKusick VA: Mendelian Inheritance in Man, 6th
ed. John Hopkins Univ Press, 1983.
human genome. Once the defect is localized to a region
Recombinant DNA Technology Is Used
of DNA that has the characteristic structure of a gene
in the Molecular Analysis of Disease
(Figure 40-1), a synthetic gene can be constructed and
A. NORMAL GENE VARIATIONS
expressed in an appropriate vector and its function can
be assessed—or the putative peptide, deduced from the
There is a normal variation of DNA sequence just as is
open reading frame in the coding region, can be synthe-
true of more obvious aspects of human structure. Varia-
sized. Antibodies directed against this peptide can be
tions of DNA sequence, polymorphisms, occur ap-
used to assess whether this peptide is expressed in nor-
proximately once in every 500 nucleotides, or about
mal persons and whether it is absent in those with the
107 times per genome. There are without doubt dele-
genetic syndrome.
tions and insertions of DNA as well as single-base sub-
stitutions. In healthy people, these alterations obviously
occur in noncoding regions of DNA or at sites that
Proteins Can Be Produced
cause no change in function of the encoded protein.
This heritable polymorphism of DNA structure can be
for Research & Diagnosis
associated with certain diseases within a large kindred
A practical goal of recombinant DNA research is the
and can be used to search for the specific gene involved,
production of materials for biomedical applications.
as is illustrated below. It can also be used in a variety of
This technology has two distinct merits: (1) It can sup-
applications in forensic medicine.
ply large amounts of material that could not be ob-
tained by conventional purification methods (eg, inter-
B. GENE VARIATIONS CAUSING DISEASE
feron, tissue plasminogen activating factor). (2) It can
provide human material (eg, insulin, growth hormone).
Classic genetics taught that most genetic diseases were
The advantages in both cases are obvious. Although the
due to point mutations which resulted in an impaired
primary aim is to supply products—generally pro-
protein. This may still be true, but if on reading the
teins—for treatment
(insulin) and diagnosis
(AIDS
initial sections of this chapter one predicted that ge-
testing) of human and other animal diseases and for
netic disease could result from derangement of any of
disease prevention (hepatitis B vaccine), there are other
the steps illustrated in Figure 40-1, one would have
potential commercial applications, especially in agricul-
made a proper assessment. This point is nicely illus-
ture. An example of the latter is the attempt to engineer
trated by examination of the β-globin gene. This gene
plants that are more resistant to drought or temperature
is located in a cluster on chromosome
11 (Figure
extremes, more efficient at fixing nitrogen, or that pro-
40-8), and an expanded version of the gene is illus-
duce seeds containing the complete complement of es-
trated in Figure 40-9. Defective production of β-glo-
sential amino acids (rice, wheat, corn, etc).
bin results in a variety of diseases and is due to many
408
/
CHAPTER 40
Gγ
Aγ
Ψβ
δ
β
5′
LCR
3′
10 kb
Hemoglobinopathy
β0-Thalassemia
β0-Thalassemia
Hemoglobin Lepore
Inverted
(Aγδβ)0-Thalassemia
Figure 40-8. Schematic representation of the β-globin gene cluster and of the lesions in some ge-
netic disorders. The β-globin gene is located on chromosome 11 in close association with the two γ-glo-
bin genes and the δ-globin gene. The β-gene family is arranged in the order 5′-ε-Gγ-Aγ-ψβ-δ-β-3′. The
ε locus is expressed in early embryonic life (as a2ε2). The γ genes are expressed in fetal life, making fetal
hemoglobin (HbF, α2γ2). Adult hemoglobin consists of HbA (α2β2) or HbA2(α2δ2). The Ψβ is a pseudo-
gene that has sequence homology with β but contains mutations that prevent its expression. A locus
control region (LCR) located upstream (5′) from the ε gene controls the rate of transcription of the en-
tire β-globin gene cluster. Deletions (solid bar) of the β locus cause β-thalassemia (deficiency or ab-
sence [β0] of β-globin). A deletion of δ and β causes hemoglobin Lepore (only hemoglobin α is present).
An inversion (Aγδβ)0 in this region (colored bar) disrupts gene function and also results in thalassemia
(type III). Each type of thalassemia tends to be found in a certain group of people, eg, the (Aγδβ)0 dele-
tion inversion occurs in persons from India. Many more deletions in this region have been mapped, and
each causes some type of thalassemia.
different lesions in and around the β-globin gene
turn results in an A-to-U change in the mRNA corre-
(Table 40-6).
sponding to the sixth codon of the β-globin gene. The
altered codon specifies a different amino acid (valine
C. POINT MUTATIONS
rather than glutamic acid), and this causes a structural
The classic example is sickle cell disease, which is
abnormality of the β-globin molecule. Other point mu-
caused by mutation of a single base out of the 3 × 109
tations in and around the β-globin gene result in de-
in the genome, a T-to-A DNA substitution, which in
creased production or, in some instances, no produc-
5′
I
1
I
2
3′
Figure 40-9. Mutations in the β-globin gene causing β-thalassemia. The β-globin gene is shown in the 5′
to 3′ orientation. The cross-hatched areas indicate the 5′ and 3′ nontranslated regions. Reading from the 5′ to
3′ direction, the shaded areas are exons 1-3 and the clear spaces are introns 1 (I1) and 2 (I2). Mutations that af-
fect transcription control (•) are located in the 5′ flanking-region DNA. Examples of nonsense mutations (
),
mutations in RNA processing (
), and RNA cleavage mutations ( ) have been identified and are indicated. In
some regions, many mutations have been found. These are indicated by the brackets.
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
409
Table 40-6. Structural alterations of the β-globin
E. PEDIGREE ANALYSIS
gene.
Sickle cell disease again provides an excellent example
of how recombinant DNA technology can be applied
Alteration
Function Affected
Disease
to the study of human disease. The substitution of T
for A in the template strand of DNA in the β-globin
Point mutations
Protein folding
Sickle cell disease
gene changes the sequence in the region that corre-
Transcriptional control
β-Thalassemia
sponds to the sixth codon from
Frameshift and non-
β-Thalassemia
sense mutations
↓
RNA processing
β-Thalassemia
CCTGAGG
Coding strand
Deletion
mRNA production
β0-Thalassemia
GGAC T CC
Template strand
Hemoglobin
↑
Lepore
to
Rearrangement
mRNA production
β-Thalassemia
type III
CCTGTGG
Coding strand
GGAC A CC
Template strand
and destroys a recognition site for the restriction en-
tion of β-globin; β-thalassemia is the result of these
zyme MstII (CCTNAGG; denoted by the small vertical
mutations. (The thalassemias are characterized by de-
arrows; Table 40-2). Other MstII sites 5′ and 3′ from
fects in the synthesis of hemoglobin subunits, and so
this site (Figure 40-10) are not affected and so will be
β-thalassemia results when there is insufficient produc-
cut. Therefore, incubation of DNA from normal (AA),
tion of β-globin.) Figure 40-9 illustrates that point
heterozygous (AS), and homozygous (SS) individuals
mutations affecting each of the many processes in-
results in three different patterns on Southern blot
volved in generating a normal mRNA (and therefore a
transfer (Figure 40-10). This illustrates how a DNA
normal protein) have been implicated as a cause of
pedigree can be established using the principles dis-
β-thalassemia.
cussed in this chapter. Pedigree analysis has been ap-
D. DELETIONS, INSERTIONS, &
plied to a number of genetic diseases and is most useful
in those caused by deletions and insertions or the rarer
REARRANGEMENTS OF DNA
instances in which a restriction endonuclease cleavage
Studies of bacteria, viruses, yeasts, and fruit flies show
site is affected, as in the example cited in this para-
that pieces of DNA can move from one place to an-
graph. The analysis is facilitated by the PCR reaction,
other within a genome. The deletion of a critical piece
which can provide sufficient DNA for analysis from
of DNA, the rearrangement of DNA within a gene, or
just a few nucleated red blood cells.
the insertion of a piece of DNA within a coding or reg-
ulatory region can all cause changes in gene expression
F. PRENATAL DIAGNOSIS
resulting in disease. Again, a molecular analysis of
If the genetic lesion is understood and a specific probe
β-thalassemia produces numerous examples of these
is available, prenatal diagnosis is possible. DNA from
processes—particularly deletions—as causes of disease
cells collected from as little as 10 mL of amniotic fluid
(Figure 40-8). The globin gene clusters seem particu-
(or by chorionic villus biopsy) can be analyzed by
larly prone to this lesion. Deletions in the α-globin
Southern blot transfer. A fetus with the restriction pat-
cluster, located on chromosome
16, cause α-thal-
tern AA in Figure 40-10 does not have sickle cell dis-
assemia. There is a strong ethnic association for many
ease, nor is it a carrier. A fetus with the SS pattern will
of these deletions, so that northern Europeans, Fil-
develop the disease. Probes are now available for this
ipinos, blacks, and Mediterranean peoples have differ-
type of analysis of many genetic diseases.
ent lesions all resulting in the absence of hemoglobin A
and α-thalassemia.
G. RESTRICTION FRAGMENT LENGTH
A similar analysis could be made for a number of
POLYMORPHISM (RFLP)
other diseases. Point mutations are usually defined by
sequencing the gene in question, though occasionally, if
The differences in DNA sequence cited above can re-
the mutation destroys or creates a restriction enzyme
sult in variations of restriction sites and thus in the
site, the technique of restriction fragment analysis can
length of restriction fragments. An inherited difference
be used to pinpoint the lesion. Deletions or insertions
in the pattern of restriction (eg, a DNA variation occur-
of DNA larger than 50 bp can often be detected by the
ring in more than 1% of the general population) is
Southern blotting procedure.
known as a restriction fragment length polymorphism,
410
/
CHAPTER 40
A. MstII restriction sites around and in the β-globin gene
Normal (A)
5′
3′
1.15 kb
0.2
kb
Sickle (S)
5′
3′
1.35 kb
B. Pedigree analysis
Fragment
size
1.35 kb
1.15 kb
AS
AS
SS
AA
AS
AS
Phenotype
Figure 40-10. Pedigree analysis of sickle cell disease. The top part of the fig-
ure (A) shows the first part of the β-globin gene and the MstII restriction en-
zyme sites in the normal (A) and sickle cell (S) β-globin genes. Digestion with
the restriction enzyme MstII results in DNA fragments 1.15 kb and 0.2 kb long in
normal individuals. The T-to-A change in individuals with sickle cell disease
abolishes one of the three MstII sites around the β-globin gene; hence, a single
restriction fragment 1.35 kb in length is generated in response to MstII. This size
difference is easily detected on a Southern blot. (The 0.2-kb fragment would
run off the gel in this illustration.) (B) Pedigree analysis shows three possibili-
ties: AA = normal (open circle); AS = heterozygous (half-solid circles, half-solid
square); SS = homozygous (solid square). This approach allows for prenatal di-
agnosis of sickle cell disease (dash-sided square).
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
411
or RFLP. An extensive RFLP map of the human
H. MICROSATELLITE DNA POLYMORPHISMS
genome has been constructed. This is proving useful in
Short (2-6 bp), inherited, tandem repeat units of DNA
the human genome sequencing project and is an impor-
occur about
50,000-100,000 times in the human
tant component of the effort to understand various sin-
genome (Chapter 36). Because they occur more fre-
gle-gene and multigenic diseases. RFLPs result from
quently—and in view of the routine application of sen-
single-base changes (eg, sickle cell disease) or from dele-
sitive PCR methods—they are replacing RFLPs as the
tions or insertions of DNA into a restriction fragment
marker loci for various genome searches.
(eg, the thalassemias) and have proved to be useful di-
agnostic tools. They have been found at known gene
I. RFLPS & VNTRS IN FORENSIC MEDICINE
loci and in sequences that have no known function;
Variable numbers of tandemly repeated (VNTR) units
thus, RFLPs may disrupt the function of the gene or
are one common type of “insertion” that results in an
may have no biologic consequences.
RFLP. The VNTRs can be inherited, in which case
RFLPs are inherited, and they segregate in a
they are useful in establishing genetic association with a
mendelian fashion. A major use of RFLPs (thousands
disease in a family or kindred; or they can be unique to
are now known) is in the definition of inherited dis-
an individual and thus serve as a molecular fingerprint
eases in which the functional deficit is unknown.
of that person.
RFLPs can be used to establish linkage groups, which
in turn, by the process of chromosome walking, will
J. GENE THERAPY
eventually define the disease locus. In chromosome
Diseases caused by deficiency of a gene product (Table
walking (Figure 40-11), a fragment representing one
40-5) are amenable to replacement therapy. The strat-
end of a long piece of DNA is used to isolate another
egy is to clone a gene (eg, the gene that codes for
that overlaps but extends the first. The direction of ex-
adenosine deaminase) into a vector that will readily be
tension is determined by restriction mapping, and the
taken up and incorporated into the genome of a host
procedure is repeated sequentially until the desired se-
cell. Bone marrow precursor cells are being investigated
quence is obtained. The X chromosome-linked disor-
for this purpose because they presumably will resettle in
ders are particularly amenable to this approach, since
the marrow and replicate there. The introduced gene
only a single allele is expressed. Hence, 20% of the de-
would begin to direct the expression of its protein prod-
fined RFLPs are on the X chromosome, and a reason-
uct, and this would correct the deficiency in the host
ably complete linkage map of this chromosome exists.
cell.
The gene for the X-linked disorder, Duchenne-type
K. TRANSGENIC ANIMALS
muscular dystrophy, was found using RFLPs. Likewise,
the defect in Huntington’s disease was localized to the
The somatic cell gene replacement described above
terminal region of the short arm of chromosome 4, and
would obviously not be passed on to offspring. Other
the defect that causes polycystic kidney disease is linked
strategies to alter germ cell lines have been devised but
to the α-globin locus on chromosome 16.
have been tested only in experimental animals. A certain
Intact DNA
5′
Gene X
3′
Fragments
1
2
3
4
5
Initial
probe
Figure 40-11. The technique of chromosome walking. Gene X is to be isolated from a large piece
of DNA. The exact location of this gene is not known, but a probe (*——) directed against a frag-
ment of DNA (shown at the 5′ end in this representation) is available, as is a library containing a se-
ries of overlapping DNA fragments. For the sake of simplicity, only five of these are shown. The initial
probe will hybridize only with clones containing fragment 1, which can then be isolated and used as
a probe to detect fragment 2. This procedure is repeated until fragment 4 hybridizes with fragment
5, which contains the entire sequence of gene X.
412
/
CHAPTER 40
percentage of genes injected into a fertilized mouse ovum
square centimeters. By coupling such DNA microarrays
will be incorporated into the genome and found in both
with highly sensitive detection of hybridized fluores-
somatic and germ cells. Hundreds of transgenic animals
cently labeled nucleic acid probes derived from mRNA,
have been established, and these are useful for analysis of
investigators can rapidly and accurately generate profiles
tissue-specific effects on gene expression and effects of
of gene expression (eg, specific cellular mRNA content)
overproduction of gene products (eg, those from the
from cell and tissue samples as small as 1 gram or less.
growth hormone gene or oncogenes) and in discovering
Thus entire transcriptome information (the entire col-
genes involved in development—a process that hereto-
lection of cellular mRNAs) for such cell or tissue sources
fore has been difficult to study. The transgenic approach
can readily be obtained in only a few days. Transcrip-
has recently been used to correct a genetic deficiency in
tome information allows one to predict the collection of
mice. Fertilized ova obtained from mice with genetic hy-
proteins that might be expressed in a particular cell, tis-
pogonadism were injected with DNA containing the
sue, or organ in normal and disease states based upon the
coding sequence for the gonadotropin-releasing hormone
mRNAs present in those cells. Complementing this high-
(GnRH) precursor protein. This gene was expressed and
throughput, transcript-profiling method is the recent de-
regulated normally in the hypothalamus of a certain
velopment of high-sensitivity, high-throughput mass
number of the resultant mice, and these animals were in
spectrometry of complex protein samples. Newer mass
all respects normal. Their offspring also showed no evi-
spectrometry methods allow one to identify hundreds to
dence of GnRH deficiency. This is, therefore, evidence
thousands of proteins in proteins extracted from very
of somatic cell expression of the transgene and of its
small numbers of cells (< 1 g). This critical information
maintenance in germ cells.
tells investigators which of the many mRNAs detected in
transcript microarray mapping studies are actually trans-
lated into protein, generally the ultimate dictator of phe-
Targeted Gene Disruption or Knockout
notype. Microarray techniques and mass spectrometric
In transgenic animals, one is adding one or more copies
protein identification experiments both lead to the gen-
of a gene to the genome, and there is no way to control
eration of huge amounts of data. Appropriate data man-
where that gene eventually resides. A complementary—
agement and interpretation of the deluge of information
and much more difficult—approach involves the selec-
forthcoming from such studies has relied upon statistical
tive removal of a gene from the genome. Gene knock-
methods; and this new technology, coupled with the
out animals
(usually mice) are made by creating a
flood of DNA sequence information, has led to the de-
mutation that totally disrupts the function of a gene.
velopment of the field of bioinformatics, a new disci-
This is then used to replace one of the two genes in an
pline whose goal is to help manage, analyze, and inte-
embryonic stem cell that can be used to create a het-
grate this flood of biologically important information.
erozygous transgenic animal. The mating of two such
Future work at the intersection of bioinformatics and
animals will, by mendelian genetics, result in a ho-
transcript-protein profiling will revolutionize our under-
mozygous mutation in 25% of offspring. Several hun-
standing of biology and medicine.
dred strains of mice with knockouts of specific genes
have been developed.
SUMMARY
RNA Transcript & Protein Profiling
• A variety of very sensitive techniques can now be ap-
plied to the isolation and characterization of genes
The “-omic” revolution of the last several years has cul-
and to the quantitation of gene products.
minated in the determination of the nucleotide se-
• In DNA cloning, a particular segment of DNA is re-
quences of entire genomes, including those of budding
moved from its normal environment using one of
and fission yeasts, various bacteria, the fruit fly, the worm
many restriction endonucleases. This is then ligated
Caenorhabditis elegans, the mouse and, most notably, hu-
into one of several vectors in which the DNA seg-
mans. Additional genomes are being sequenced at an ac-
ment can be amplified and produced in abundance.
celerating pace. The availability of all of this DNA se-
quence information, coupled with engineering advances,
• The cloned DNA can be used as a probe in one of
has lead to the development of several revolutionary
several types of hybridization reactions to detect
methodologies, most of which are based upon high-den-
other related or adjacent pieces of DNA, or it can be
sity microarray technology. We now have the ability to
used to quantitate gene products such as mRNA.
deposit thousands of specific, known, definable DNA se-
• Manipulation of the DNA to change its structure, so-
quences (more typically now synthetic oligonucleotides)
called genetic engineering, is a key element in cloning
on a glass microscope-style slide in the space of a few
(eg, the construction of chimeric molecules) and can
MOLECULAR GENETICS, RECOMBINANT DNA, & GENOMIC TECHNOLOGY
/
413
also be used to study the function of a certain frag-
quences of a single strand of DNA or RNA.
ment of DNA and to analyze how genes are regulated.
Hybridization: The specific reassociation of com-
• Chimeric DNA molecules are introduced into cells
plementary strands of nucleic acids (DNA with
to make transfected cells or into the fertilized oocyte
DNA, DNA with RNA, or RNA with RNA).
to make transgenic animals.
Insert: An additional length of base pairs in DNA,
generally introduced by the techniques of recom-
• Techniques involving cloned DNA are used to locate
binant DNA technology.
genes to specific regions of chromosomes, to identify
Intron: The sequence of a gene that is transcribed
the genes responsible for diseases, to study how faulty
but excised before translation.
gene regulation causes disease, to diagnose genetic
Library: A collection of cloned fragments that rep-
diseases, and increasingly to treat genetic diseases.
resents the entire genome. Libraries may be either
genomic DNA (in which both introns and exons
GLOSSARY
are represented) or cDNA (in which only exons
are represented).
ARS: Autonomously replicating sequence; the ori-
Ligation: The enzyme-catalyzed joining in phos-
gin of replication in yeast.
phodiester linkage of two stretches of DNA or
Autoradiography: The detection of radioactive
RNA into one; the respective enzymes are DNA
molecules (eg, DNA, RNA, protein) by visualiza-
and RNA ligases.
tion of their effects on photographic film.
Lines: Long interspersed repeat sequences.
Bacteriophage: A virus that infects a bacterium.
Microsatellite polymorphism: Heterozygosity of a
Blunt-ended DNA: Two strands of a DNA duplex
certain microsatellite repeat in an individual.
having ends that are flush with each other.
Microsatellite repeat sequences: Dispersed or
cDNA: A single-stranded DNA molecule that is
group repeat sequences of 2-5 bp repeated up to
complementary to an mRNA molecule and is syn-
50 times. May occur at 50-100 thousand loca-
thesized from it by the action of reverse transcrip-
tions in the genome.
tase.
Nick translation: A technique for labeling DNA
Chimeric molecule: A molecule (eg, DNA, RNA,
based on the ability of the DNA polymerase from
protein) containing sequences derived from two
E coli to degrade a strand of DNA that has been
different species.
nicked and then to resynthesize the strand; if a ra-
Clone: A large number of organisms, cells or mole-
dioactive nucleoside triphosphate is employed, the
cules that are identical with a single parental or-
rebuilt strand becomes labeled and can be used as
ganism cell or molecule.
a radioactive probe.
Cosmid: A plasmid into which the DNA sequences
Northern blot: A method for transferring RNA
from bacteriophage lambda that are necessary for
from an agarose gel to a nitrocellulose filter, on
the packaging of DNA (cos sites) have been in-
which the RNA can be detected by a suitable
serted; this permits the plasmid DNA to be pack-
probe.
aged in vitro.
Oligonucleotide: A short, defined sequence of nu-
Endonuclease: An enzyme that cleaves internal
cleotides joined together in the typical phosphodi-
bonds in DNA or RNA.
ester linkage.
Excinuclease: The excision nuclease involved in nu-
Ori: The origin of DNA replication.
cleotide exchange repair of DNA.
PAC: A high capacity (70-95 kb) cloning vector
Exon: The sequence of a gene that is represented
based upon the lytic E. coli bacteriophage P1 that
(expressed) as mRNA.
replicates in bacteria as an extrachromosomal ele-
Exonuclease: An enzyme that cleaves nucleotides
ment.
from either the 3′ or 5′ ends of DNA or RNA.
Palindrome: A sequence of duplex DNA that is the
Fingerprinting: The use of RFLPs or repeat se-
same when the two strands are read in opposite di-
quence DNA to establish a unique pattern of
rections.
DNA fragments for an individual.
Plasmid: A small, extrachromosomal, circular mole-
Footprinting: DNA with protein bound is resistant
cule of DNA that replicates independently of the
to digestion by DNase enzymes. When a sequenc-
host DNA.
ing reaction is performed using such DNA, a pro-
Polymerase chain reaction (PCR): An enzymatic
tected area, representing the “footprint” of the
method for the repeated copying (and thus ampli-
bound protein, will be detected.
fication) of the two strands of DNA that make up
Hairpin: A double-helical stretch formed by base
a particular gene sequence.
pairing between neighboring complementary se-
414
/
CHAPTER 40
Primosome: The mobile complex of helicase and
Spliceosome: The macromolecular complex respon-
primase that is involved in DNA replication.
sible for precursor mRNA splicing. The spliceo-
Probe: A molecule used to detect the presence of a
some consists of at least five small nuclear RNAs
specific fragment of DNA or RNA in, for in-
(snRNA; U1, U2, U4, U5, and U6) and many
stance, a bacterial colony that is formed from a ge-
proteins.
netic library or during analysis by blot transfer
Splicing: The removal of introns from RNA ac-
techniques; common probes are cDNA molecules,
companied by the joining of its exons.
synthetic oligodeoxynucleotides of defined se-
Sticky-ended DNA: Complementary single strands
quence, or antibodies to specific proteins.
of DNA that protrude from opposite ends of a
Proteome: The entire collection of expressed pro-
DNA duplex or from the ends of different duplex
teins in an organism.
molecules (see also Blunt-ended DNA, above).
Pseudogene: An inactive segment of DNA arising
Tandem: Used to describe multiple copies of the
by mutation of a parental active gene.
same sequence (eg, DNA) that lie adjacent to one
Recombinant DNA: The altered DNA that results
another.
from the insertion of a sequence of deoxynu-
Terminal transferase: An enzyme that adds nu-
cleotides not previously present into an existing
cleotides of one type (eg, deoxyadenonucleotidyl
molecule of DNA by enzymatic or chemical
residues) to the 3′ end of DNA strands.
means.
Transcription: Template DNA-directed synthesis
Restriction enzyme: An endodeoxynuclease that
of nucleic acids; typically DNA-directed synthesis
causes cleavage of both strands of DNA at highly
of RNA.
specific sites dictated by the base sequence.
Transcriptome: The entire collection of expressed
Reverse transcription: RNA-directed synthesis of
mRNAs in an organism.
DNA, catalyzed by reverse transcriptase.
Transgenic: Describing the introduction of new
RT-PCR: A method used to quantitate mRNA lev-
DNA into germ cells by its injection into the nu-
els that relies upon a first step of cDNA copying of
cleus of the ovum.
mRNAs prior to PCR amplification and quantita-
Translation: Synthesis of protein using mRNA as
tion.
template.
Signal: The end product observed when a specific
Vector: A plasmid or bacteriophage into which for-
sequence of DNA or RNA is detected by autoradi-
eign DNA can be introduced for the purposes of
ography or some other method. Hybridization
cloning.
with a complementary radioactive polynucleotide
Western blot: A method for transferring protein to
(eg, by Southern or Northern blotting) is com-
a nitrocellulose filter, on which the protein can be
monly used to generate the signal.
detected by a suitable probe (eg, an antibody).
Sines: Short interspersed repeat sequences.
SNP: Single nucleotide polymorphism. Refers to
the fact that single nucleotide genetic variation in
REFERENCES
genome sequence exists at discrete loci throughout
Lewin B: Genes VII. Oxford Univ Press, 1999.
the chromosomes. Measurement of allelic SNP
Martin JB, Gusella JF: Huntington’s disease: pathogenesis and
differences is useful for gene mapping studies.
management. N Engl J Med 1986:315:1267.
snRNA: Small nuclear RNA. This family of RNAs
Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning: A Labora-
is best known for its role in mRNA processing.
tory Manual. Cold Spring Harbor Laboratory Press, 1989.
Southern blot: A method for transferring DNA
Spector DL, Goldman RD, Leinwand LA: Cells: A Laboratory
from an agarose gel to nitrocellulose filter, on
Manual. Cold Spring Harbor Laboratory Press, 1998.
which the DNA can be detected by a suitable
Watson JD et al: Recombinant DNA, 2nd ed. Scientific American
probe (eg, complementary DNA or RNA).
Books. Freeman, 1992.
Southwestern blot: A method for detecting pro-
Weatherall DJ: The New Genetics and Clinical Practice, 3rd ed. Ox-
tein-DNA interactions by applying a labeled DNA
ford Univ Press, 1991.
probe to a transfer membrane that contains a rena-
tured protein.
SECTION V
Biochemistry of Extracellular
& Intracellular Communication
Membranes: Structure & Function
41
Robert K. Murray, MD, PhD, & Daryl K. Granner, MD
BIOMEDICAL IMPORTANCE
MAINTENANCE OF A NORMAL INTRA-
& EXTRACELLULAR ENVIRONMENT
Membranes are highly viscous, plastic structures.
Plasma membranes form closed compartments around
IS FUNDAMENTAL TO LIFE
cellular protoplasm to separate one cell from another
Life originated in an aqueous environment; enzyme re-
and thus permit cellular individuality. The plasma
actions, cellular and subcellular processes, and so forth
membrane has selective permeabilities and acts as a
have therefore evolved to work in this milieu. Since
barrier, thereby maintaining differences in composition
mammals live in a gaseous environment, how is the
between the inside and outside of the cell. The selective
aqueous state maintained? Membranes accomplish this
permeabilities are provided mainly by channels and
by internalizing and compartmentalizing body water.
pumps for ions and substrates. The plasma membrane
also exchanges material with the extracellular environ-
The Body’s Internal Water
ment by exocytosis and endocytosis, and there are spe-
Is Compartmentalized
cial areas of membrane structure—the gap junctions—
through which adjacent cells exchange material. In
Water makes up about 60% of the lean body mass of
addition, the plasma membrane plays key roles in cell-
the human body and is distributed in two large com-
cell interactions and in transmembrane signaling.
partments.
Membranes also form specialized compartments
within the cell. Such intracellular membranes help
A. INTRACELLULAR FLUID (ICF)
shape many of the morphologically distinguishable
This compartment constitutes two-thirds of total body
structures (organelles), eg, mitochondria, ER, sarcoplas-
water and provides the environment for the cell (1) to
mic reticulum, Golgi complexes, secretory granules,
make, store, and utilize energy;
(2) to repair itself;
lysosomes, and the nuclear membrane. Membranes lo-
(3) to replicate; and (4) to perform special functions.
calize enzymes, function as integral elements in excita-
B. EXTRACELLULAR FLUID (ECF)
tion-response coupling, and provide sites of energy
transduction, such as in photosynthesis and oxidative
This compartment contains about one-third of total
phosphorylation.
body water and is distributed between the plasma and
Changes in membrane structure (eg caused by is-
interstitial compartments. The extracellular fluid is a
chemia) can affect water balance and ion flux and there-
delivery system. It brings to the cells nutrients (eg, glu-
fore every process within the cell. Specific deficiencies
cose, fatty acids, amino acids), oxygen, various ions and
or alterations of certain membrane components lead to
trace minerals, and a variety of regulatory molecules
a variety of diseases (see Table 41-5). In short, normal
(hormones) that coordinate the functions of widely sep-
cellular function depends on normal membranes.
arated cells. Extracellular fluid removes CO2, waste
415
416
/
CHAPTER 41
products, and toxic or detoxified materials from the im-
mediate cellular environment.
Myelin
0.23
The Ionic Compositions of Intracellular
Mouse
0.85
& Extracellular Fluids Differ Greatly
liver cells
As illustrated in Table 41-1, the internal environment
Retinal rods
is rich in K+ and Mg2+, and phosphate is its major
(bovine)
1.0
anion. Extracellular fluid is characterized by high Na+
and Ca2+ content, and Cl− is the major anion. Note
Human
1.1
erythrocyte
also that the concentration of glucose is higher in extra-
cellular fluid than in the cell, whereas the opposite is
true for proteins. Why is there such a difference? It is
Ameba
1.3
thought that the primordial sea in which life originated
was rich in K+ and Mg2+. It therefore follows that en-
HeLa cells
1.5
zyme reactions and other biologic processes evolved to
function best in that environment—hence the high
concentration of these ions within cells. Cells were
Mitochondrial
1.1
outer membrane
faced with strong selection pressure as the sea gradually
changed to a composition rich in Na+ and Ca2+. Vast
Sarcoplasmic
changes would have been required for evolution of a
2.0
reticulum
completely new set of biochemical and physiologic ma-
chinery; instead, as it happened, cells developed barri-
Mitochondrial
ers—membranes with associated “pumps”—to main-
3.2
inner membrane
tain the internal microenvironment.
0
1
2
3
4
MEMBRANES ARE COMPLEX
Ratio of protein to lipid
STRUCTURES COMPOSED OF LIPIDS,
Figure 41-1. Ratio of protein to lipid in different
PROTEINS, & CARBOHYDRATES
membranes. Proteins equal or exceed the quantity of
We shall mainly discuss the membranes present in eu-
lipid in nearly all membranes. The outstanding excep-
karyotic cells, although many of the principles de-
tion is myelin, an electrical insulator found on many
scribed also apply to the membranes of prokaryotes.
nerve fibers.
The various cellular membranes have different compo-
sitions, as reflected in the ratio of protein to lipid (Fig-
ure 41-1). This is not surprising, given their divergent
These sheet-like structures are noncovalent assemblies
functions. Membranes are asymmetric sheet-like en-
that are thermodynamically stable and metabolically ac-
closed structures with distinct inner and outer surfaces.
tive. Numerous proteins are located in membranes,
where they carry out specific functions of the organelle,
the cell, or the organism.
Table 41-1. Comparison of the mean
concentrations of various molecules outside and
The Major Lipids in Mammalian
inside a mammalian cell.
Membranes Are Phospholipids,
Glycosphingolipids, & Cholesterol
Substance
Extracellular Fluid
Intracellular Fluid
A. PHOSPHOLIPIDS
Na+
140 mmol/L
10 mmol/L
Of the two major phospholipid classes present in mem-
K+
4 mmol/L
140 mmol/L
branes, phosphoglycerides are the more common and
Ca2+ (free)
2.5 mmol/L
0.1 µmol/L
consist of a glycerol backbone to which are attached
Mg2+
1.5 mmol/L
30 mmol/L
two fatty acids in ester linkage and a phosphorylated al-
CI−
100 mmol/L
4 mmol/L
cohol (Figure 41-2). The fatty acid constituents are
−
HCO3
27 mmol/L
10 mmol/L
usually even-numbered carbon molecules, most com-
PO43−
2 mmol/L
60 mmol/L
monly containing 16 or 18 carbons. They are un-
Glucose
5.5 mmol/L
0-1 mmol/L
branched and can be saturated or unsaturated. The sim-
Protein
2 g/dL
16 g/dL
plest phosphoglyceride is phosphatidic acid, which is
MEMBRANES: STRUCTURE & FUNCTION
/
417
Fatty acids
Each eukaryotic cell membrane has a somewhat dif-
ferent lipid composition, though phospholipids are the
O
major class in all.
R1
C O
1CH2
Membrane Lipids Are Amphipathic
R
C O
2
CH
2
O-
All major lipids in membranes contain both hydropho-
O
3CH2
O
P
O
R3
bic and hydrophilic regions and are therefore termed
O
“amphipathic.” Membranes themselves are thus am-
phipathic. If the hydrophobic regions were separated
Glycerol
Alcohol
from the rest of the molecule, it would be insoluble in
water but soluble in oil. Conversely, if the hydrophilic
Figure 41-2. A phosphoglyceride showing the fatty
region were separated from the rest of the molecule, it
acids (R1 and R2), glycerol, and phosphorylated alcohol
would be insoluble in oil but soluble in water. The am-
components. In phosphatidic acid, R3 is hydrogen.
phipathic nature of a phospholipid is represented in
Figure 41-3. Thus, the polar head groups of the phos-
pholipids and the hydroxyl group of cholesterol inter-
1,2-diacylglycerol 3-phosphate, a key intermediate in
face with the aqueous environment; a similar situation
the formation of all other phosphoglycerides (Chapter
applies to the sugar moieties of the GSLs (see below).
24). In other phosphoglycerides, the 3-phosphate is es-
Saturated fatty acids have straight tails, whereas
terified to an alcohol such as ethanolamine, choline,
unsaturated fatty acids, which generally exist in the cis
serine, glycerol, or inositol (Chapter 14).
form in membranes, make kinked tails (Figure 41-3).
The second major class of phospholipids is com-
As more kinks are inserted in the tails, the membrane
posed of sphingomyelin, which contains a sphingosine
becomes less tightly packed and therefore more fluid.
backbone rather than glycerol. A fatty acid is attached
Detergents are amphipathic molecules that are impor-
by an amide linkage to the amino group of sphingosine,
tant in biochemistry as well as in the household. The
forming ceramide. The primary hydroxyl group of
molecular structure of a detergent is not unlike that of a
sphingosine is esterified to phosphorylcholine. Sphin-
phospholipid. Certain detergents are widely used to sol-
gomyelin, as the name implies, is prominent in myelin
ubilize membrane proteins as a first step in their purifi-
sheaths.
cation. The hydrophobic end of the detergent binds to
The amounts and fatty acid compositions of the var-
ious phospholipids vary among the different cellular
membranes.
B. GLYCOSPHINGOLIPIDS
Polar head group
The glycosphingolipids
(GSLs) are sugar-containing
lipids built on a backbone of ceramide; they include
galactosyl- and glucosylceramide (cerebrosides) and the
gangliosides. Their structures are described in Chapter
14. They are mainly located in the plasma membranes
of cells.
Apolar, hydrocarbon tails
C. STEROLS
The most common sterol in membranes is cholesterol
(Chapter 14), which resides mainly in the plasma mem-
branes of mammalian cells but can also be found in
S
U S S
lesser quantities in mitochondria, Golgi complexes, and
Figure 41-3. Diagrammatic representation of a
nuclear membranes. Cholesterol intercalates among the
phospholipid or other membrane lipid. The polar head
phospholipids of the membrane, with its hydroxyl
group is hydrophilic, and the hydrocarbon tails are hy-
group at the aqueous interface and the remainder of the
drophobic or lipophilic. The fatty acids in the tails are
molecule within the leaflet. Its effect on the fluidity of
membranes is discussed subsequently.
saturated (S) or unsaturated (U); the former are usually
All of the above lipids can be separated from one an-
attached to carbon 1 of glycerol and the latter to car-
other by techniques such as column, thin layer, and
bon 2. Note the kink in the tail of the unsaturated fatty
gas-liquid chromatography and their structures estab-
acid (U), which is important in conferring increased
lished by mass spectrometry.
membrane fluidity.
418
/
CHAPTER 41
Aqueous
hydrophobic regions of the proteins, displacing most of
their bound lipids. The polar end of the detergent is
Hydro-
philic
free, bringing the proteins into solution as detergent-
protein complexes, usually also containing some resid-
ual lipids.
Hydro-
phobic
Membrane Lipids Form Bilayers
The amphipathic character of phospholipids suggests
Hydro-
that the two regions of the molecule have incompatible
philic
solubilities; however, in a solvent such as water, phos-
Aqueous
pholipids organize themselves into a form that thermo-
dynamically serves the solubility requirements of both
Figure 41-5. Diagram of a section of a bilayer mem-
regions. A micelle
(Figure 41-4) is such a structure;
brane formed from phospholipid molecules. The unsat-
the hydrophobic regions are shielded from water, while
urated fatty acid tails are kinked and lead to more spac-
the hydrophilic polar groups are immersed in the aque-
ing between the polar head groups, hence to more
ous environment. However, micelles are usually rela-
room for movement. This in turn results in increased
tively small in size
(eg, approximately 200 nm) and
membrane fluidity. (Slightly modified and reproduced,
thus are limited in their potential to form membranes.
with permission, from Stryer L: Biochemistry, 2nd ed. Free-
As was recognized in 1925 by Gorter and Grendel, a
man, 1981.)
bimolecular layer, or lipid bilayer, can also satisfy the
thermodynamic requirements of amphipathic mole-
cules in an aqueous environment. Bilayers, not mi-
favorable environment, but even these exposed edges
celles, are indeed the key structures in biologic mem-
can be eliminated by folding the sheet back upon itself
branes. A bilayer exists as a sheet in which the
to form an enclosed vesicle with no edges. A bilayer can
hydrophobic regions of the phospholipids are protected
extend over relatively large distances (eg, 1 mm). The
from the aqueous environment, while the hydrophilic
closed bilayer provides one of the most essential proper-
regions are immersed in water (Figure 41-5). Only the
ties of membranes. It is impermeable to most water-
ends or edges of the bilayer sheet are exposed to an un-
soluble molecules, since they would be insoluble in the
hydrophobic core of the bilayer.
Lipid bilayers are formed by self-assembly, driven
by the hydrophobic effect. When lipid molecules
come together in a bilayer, the entropy of the surround-
ing solvent molecules increases.
Two questions arise from consideration of the
above. First, how many biologic materials are lipid-
soluble and can therefore readily enter the cell? Gases
such as oxygen, CO2, and nitrogen—small molecules
with little interaction with solvents—readily diffuse
through the hydrophobic regions of the membrane.
The permeability coefficients of several ions and of a
number of other molecules in a lipid bilayer are shown
in Figure 41-6. The three electrolytes shown (Na+, K+,
and Cl−) cross the bilayer much more slowly than
water. In general, the permeability coefficients of small
molecules in a lipid bilayer correlate with their solubili-
ties in nonpolar solvents. For instance, steroids more
readily traverse the lipid bilayer compared with elec-
trolytes. The high permeability coefficient of water it-
Figure 41-4. Diagrammatic cross-section of a mi-
self is surprising but is partly explained by its small size
celle. The polar head groups are bathed in water,
and relative lack of charge.
whereas the hydrophobic hydrocarbon tails are sur-
The second question concerns molecules that are
rounded by other hydrocarbons and thereby pro-
not lipid-soluble: How are the transmembrane concen-
tected from water. Micelles are relatively small (com-
tration gradients for non-lipid-soluble molecules main-
pared with lipid bilayers) spherical structures.
tained? The answer is that membranes contain proteins,
MEMBRANES: STRUCTURE & FUNCTION
/
419
ues for their transfer from the interior of a membrane
K+
Tryptophan
Indole
to water. Hydrophobic amino acids have positive val-
Na+
Cl-
Glucose
Urea,
H2O
glycerol
ues; polar amino acids have negative values. The total
free energy values for transferring successive sequences
of 20 amino acids in the protein are plotted, yielding a
so-called hydropathy plot. Values of over 20 kcal⋅mol−1
10-14
10-12
10-10
10-8
10-6
10-4
10-2
are consistent with—but do not prove—a transmem-
Permeability coefficient (cm/s)
brane location.
Low
High
Another aspect of the interaction of lipids and pro-
Permeability
teins is that some proteins are anchored to one leaflet or
another of the bilayer by covalent linkages to certain
Figure 41-6. Permeability coefficients of water,
lipids. Palmitate and myristate are fatty acids involved
some ions, and other small molecules in lipid bilayer
in such linkages to specific proteins. A number of other
membranes. Molecules that move rapidly through a
proteins
(see Chapter
47) are linked to glycophos-
given membrane are said to have a high permeability
phatidylinositol (GPI) structures.
coefficient. (Slightly modified and reproduced, with per-
mission, from Stryer L: Biochemistry, 2nd ed. Freeman,
Different Membranes Have Different
1981.)
Protein Compositions
The number of different proteins in a membrane
and proteins are also amphipathic molecules that can be
varies from less than a dozen in the sarcoplasmic reticu-
inserted into the correspondingly amphipathic lipid bi-
lum to over 100 in the plasma membrane. Most mem-
layer. Proteins form channels for the movement of ions
brane proteins can be separated from one another using
and small molecules and serve as transporters for larger
sodium dodecyl sulfate polyacrylamide gel electro-
molecules that otherwise could not pass the bilayer.
phoresis (SDS-PAGE), a technique that has revolution-
These processes are described below.
ized their study. In the absence of SDS, few membrane
proteins would remain soluble during electrophoresis.
Proteins are the major functional molecules of mem-
Membrane Proteins Are Associated
branes and consist of enzymes, pumps and channels,
With the Lipid Bilayer
structural components, antigens (eg, for histocompati-
Membrane phospholipids act as a solvent for mem-
bility), and receptors for various molecules. Because
brane proteins, creating an environment in which the
every membrane possesses a different complement of
latter can function. Of the 20 amino acids contributing
proteins, there is no such thing as a typical membrane
to the primary structure of proteins, the functional
structure. The enzymatic properties of several different
groups attached to the α carbon are strongly hydropho-
membranes are shown in Table 41-2.
bic in six, weakly hydrophobic in a few, and hy-
drophilic in the remainder. As described in Chapter 5,
Membranes Are Dynamic Structures
the α-helical structure of proteins minimizes the hy-
drophilic character of the peptide bonds themselves.
Membranes and their components are dynamic struc-
Thus, proteins can be amphipathic and form an inte-
tures. The lipids and proteins in membranes undergo
gral part of the membrane by having hydrophilic re-
turnover there just as they do in other compartments of
gions protruding at the inside and outside faces of the
the cell. Different lipids have different turnover rates,
membrane but connected by a hydrophobic region tra-
and the turnover rates of individual species of mem-
versing the hydrophobic core of the bilayer. In fact,
brane proteins may vary widely. The membrane itself
those portions of membrane proteins that traverse
can turn over even more rapidly than any of its con-
membranes do contain substantial numbers of hy-
stituents. This is discussed in more detail in the section
drophobic amino acids and almost invariably have ei-
on endocytosis.
ther a high α-helical or β-pleated sheet content. For
many membranes, a stretch of approximately 20 amino
Membranes Are Asymmetric Structures
acids in an α helix will span the bilayer.
It is possible to calculate whether a particular se-
This asymmetry can be partially attributed to the irreg-
quence of amino acids present in a protein is consistent
ular distribution of proteins within the membranes. An
with a transmembrane location. This can be done by
inside-outside asymmetry is also provided by the ex-
consulting a table that lists the hydrophobicities of each
ternal location of the carbohydrates attached to mem-
of the 20 common amino acids and the free energy val-
brane proteins. In addition, specific enzymes are lo-
420
/
CHAPTER 41
present in the two leaflets, contributing to the asym-
Table 41-2. Enzymatic markers of different
metric distribution of these lipid molecules. In addi-
membranes.1
tion, phospholipid exchange proteins recognize specific
phospholipids and transfer them from one membrane
Membrane
Enzyme
(eg, the endoplasmic reticulum [ER]) to others (eg, mi-
tochondrial and peroxisomal). There is further asym-
Plasma
5’-Nucleotidase
metry with regard to GSLs and also glycoproteins; the
Adenylyl cyclase
sugar moieties of these molecules all protrude outward
Na+-K+ ATPase
from the plasma membrane and are absent from its
Endoplasmic reticulum
Glucose-6-phosphatase
inner face.
Golgi apparatus
Cis
GlcNAc transferase I
Membranes Contain Integral
Medial
Golgi mannosidase II
& Peripheral Proteins
Trans
Galactosyl transferase
(Figure 41-7)
TGN
Sialyl transferase
It is useful to classify membrane proteins into two
Inner mitochondrial membrane
ATP synthase
types: integral and peripheral. Most membrane pro-
1Membranes contain many proteins, some of which have enzy-
teins fall into the integral class, meaning that they inter-
matic activity. Some of these enzymes are located only in certain
act extensively with the phospholipids and require the
membranes and can therefore be used as markers to follow the
use of detergents for their solubilization. Also, they gen-
purification of these membranes.
erally span the bilayer. Integral proteins are usually
TGN, trans golgi network.
globular and are themselves amphipathic. They consist
of two hydrophilic ends separated by an intervening hy-
cated exclusively on the outside or inside of mem-
drophobic region that traverses the hydrophobic core of
branes, as in the mitochondrial and plasma membranes.
the bilayer. As the structures of integral membrane pro-
There are regional asymmetries in membranes.
teins were being elucidated, it became apparent that
Some, such as occur at the villous borders of mucosal
certain ones (eg, transporter molecules, various recep-
cells, are almost macroscopically visible. Others, such as
tors, and G proteins) span the bilayer many times (see
those at gap junctions, tight junctions, and synapses,
Figure 46-5). Integral proteins are also asymmetrically
occupy much smaller regions of the membrane and
distributed across the membrane bilayer. This asym-
generate correspondingly smaller local asymmetries.
metric orientation is conferred at the time of their in-
There is also inside-outside (transverse) asymmetry
sertion in the lipid bilayer. The hydrophilic external re-
of the phospholipids. The choline-containing phos-
gion of an amphipathic protein, which is synthesized
pholipids
(phosphatidylcholine and sphingomyelin)
on polyribosomes, must traverse the hydrophobic core
are located mainly in the outer molecular layer; the
of its target membrane and eventually be found on the
aminophospholipids
(phosphatidylserine and phos-
outside of that membrane. The molecular mechanisms
phatidylethanolamine) are preferentially located in the
involved in insertion of proteins into membranes and
inner leaflet. Obviously, if this asymmetry is to exist at
the topic of membrane assembly are discussed in Chap-
all, there must be limited transverse mobility (flip-flop)
ter 46.
of the membrane phospholipids. In fact, phospholipids
Peripheral proteins do not interact directly with
in synthetic bilayers exhibit an extraordinarily slow
the phospholipids in the bilayer and thus do not require
rate of flip-flop; the half-life of the asymmetry can be
use of detergents for their release. They are weakly
measured in several weeks. However, when certain
bound to the hydrophilic regions of specific integral
membrane proteins such as the erythrocyte protein gly-
proteins and can be released from them by treatment
cophorin are inserted artificially into synthetic bilayers,
with salt solutions of high ionic strength. For example,
the frequency of phospholipid flip-flop may increase as
ankyrin, a peripheral protein, is bound to the integral
much as 100-fold.
protein “band 3” of erythrocyte membrane. Spectrin, a
The mechanisms involved in the establishment of
cytoskeletal structure within the erythrocyte, is in turn
lipid asymmetry are not well understood. The enzymes
bound to ankyrin and thereby plays an important role
involved in the synthesis of phospholipids are located
in maintenance of the biconcave shape of the erythro-
on the cytoplasmic side of microsomal membrane vesi-
cyte. Many hormone receptor molecules are integral
cles. Translocases (flippases) exist that transfer certain
proteins, and the specific polypeptide hormones that
phospholipids (eg, phosphatidylcholine) from the inner
bind to these receptor molecules may therefore be con-
to the outer leaflet. Specific proteins that preferen-
sidered peripheral proteins. Peripheral proteins, such as
tially bind individual phospholipids also appear to be
polypeptide hormones, may help organize the distribu-
MEMBRANES: STRUCTURE & FUNCTION
/
421
Figure 41-7. The fluid mosaic model of membrane structure. The membrane consists of a bimolecu-
lar lipid layer with proteins inserted in it or bound to either surface. Integral membrane proteins are
firmly embedded in the lipid layers. Some of these proteins completely span the bilayer and are called
transmembrane proteins, while others are embedded in either the outer or inner leaflet of the lipid bi-
layer. Loosely bound to the outer or inner surface of the membrane are the peripheral proteins. Many of
the proteins and lipids have externally exposed oligosaccharide chains. (Reproduced, with permission,
from Junqueira LC, Carneiro J: Basic Histology: Text & Atlas, 10th ed. McGraw-Hill, 2003.)
tion of integral proteins, such as their receptors, within
composition to permit systematic examination of the
the plane of the bilayer (see below).
effects of fatty acid composition on certain membrane
functions (eg, transport).
(2) Purified membrane proteins or enzymes can be
ARTIFICIAL MEMBRANES MODEL
incorporated into these vesicles in order to assess what
MEMBRANE FUNCTION
factors (eg, specific lipids or ancillary proteins) the pro-
teins require to reconstitute their function. Investiga-
Artificial membrane systems can be prepared by appro-
tions of purified proteins, eg, the Ca2+ ATPase of the
priate techniques. These systems generally consist of
sarcoplasmic reticulum, have in certain cases suggested
mixtures of one or more phospholipids of natural or
that only a single protein and a single lipid are required
synthetic origin that can be treated (eg, by using mild
to reconstitute an ion pump.
sonication) to form spherical vesicles in which the lipids
(3) The environment of these systems can be rigidly
form a bilayer. Such vesicles, surrounded by a lipid bi-
controlled and systematically varied (eg, ion concentra-
layer, are termed liposomes.
tions). The systems can also be exposed to known lig-
Some of the advantages and uses of artificial mem-
ands if, for example, the liposomes contain specific re-
brane systems in the study of membrane function can
ceptor proteins.
be briefly explained.
(4) When liposomes are formed, they can be made
(1) The lipid content of the membranes can be var-
to entrap certain compounds inside themselves, eg,
ied, allowing systematic examination of the effects of
drugs and isolated genes. There is interest in using lipo-
varying lipid composition on certain functions. For in-
somes to distribute drugs to certain tissues, and if com-
stance, vesicles can be made that are composed solely of
ponents (eg, antibodies to certain cell surface mole-
phosphatidylcholine or, alternatively, of known mix-
cules) could be incorporated into liposomes so that they
tures of different phospholipids, glycolipids, and cho-
would be targeted to specific tissues or tumors, the
lesterol. The fatty acid moieties of the lipids used can
therapeutic impact would be considerable. DNA en-
also be varied by employing synthetic lipids of known
trapped inside liposomes appears to be less sensitive to
422
/
CHAPTER 41
attack by nucleases; this approach may prove useful in
high cholesterol:phospholipid ratios, transition temper-
attempts at gene therapy.
atures are altogether indistinguishable.
The fluidity of a membrane significantly affects its
functions. As membrane fluidity increases, so does its
THE FLUID MOSAIC MODEL
permeability to water and other small hydrophilic mol-
OF MEMBRANE STRUCTURE
ecules. The lateral mobility of integral proteins in-
IS WIDELY ACCEPTED
creases as the fluidity of the membrane increases. If the
active site of an integral protein involved in a given
The fluid mosaic model of membrane structure pro-
function is exclusively in its hydrophilic regions, chang-
posed in 1972 by Singer and Nicolson (Figure 41-7) is
ing lipid fluidity will probably have little effect on the
now widely accepted. The model is often likened to ice-
activity of the protein; however, if the protein is in-
bergs (membrane proteins) floating in a sea of predomi-
volved in a transport function in which transport com-
nantly phospholipid molecules. Early evidence for the
ponents span the membrane, lipid phase effects may
model was the finding that certain species-specific inte-
significantly alter the transport rate. The insulin recep-
gral proteins
(detected by fluorescent labeling tech-
tor is an excellent example of altered function with
niques) rapidly and randomly redistributed in the
changes in fluidity. As the concentration of unsaturated
plasma membrane of an interspecies hybrid cell formed
fatty acids in the membrane is increased (by growing
by the artificially induced fusion of two different parent
cultured cells in a medium rich in such molecules), flu-
cells. It has subsequently been demonstrated that phos-
idity increases. This alters the receptor so that it binds
pholipids also undergo rapid redistribution in the plane
more insulin.
of the membrane. This diffusion within the plane of
A state of fluidity and thus of translational mobility
the membrane, termed translational diffusion, can be
in a membrane may be confined to certain regions of
quite rapid for a phospholipid; in fact, within the plane
membranes under certain conditions. For example,
of the membrane, one molecule of phospholipid can
protein-protein interactions may take place within the
move several micrometers per second.
plane of the membrane, such that the integral proteins
The phase changes—and thus the fluidity of mem-
form a rigid matrix—in contrast to the more usual situ-
branes—are largely dependent upon the lipid composi-
ation, where the lipid acts as the matrix. Such regions
tion of the membrane. In a lipid bilayer, the hydropho-
of rigid protein matrix can exist side by side in the same
bic chains of the fatty acids can be highly aligned or
membrane with the usual lipid matrix. Gap junctions
ordered to provide a rather stiff structure. As the tem-
and tight junctions are clear examples of such side-by-
perature increases, the hydrophobic side chains undergo
side coexistence of different matrices.
a transition from the ordered state (more gel-like or
crystalline phase) to a disordered one, taking on a more
Lipid Rafts & Caveolae Are Special
liquid-like or fluid arrangement. The temperature at
Features of Some Membranes
which the structure undergoes the transition from or-
dered to disordered (ie, melts) is called the “transition
While the fluid mosaic model of membrane structure
temperature” (Tm). The longer and more saturated
has stood up well to detailed scrutiny, additional fea-
fatty acid chains interact more strongly with each other
tures of membrane structure and function are con-
via their longer hydrocarbon chains and thus cause
stantly emerging. Two structures of particular current
higher values of Tm—ie, higher temperatures are re-
interest, located in surface membranes, are lipid rafts
quired to increase the fluidity of the bilayer. On the
and caveolae. The former are dynamic areas of the exo-
other hand, unsaturated bonds that exist in the cis con-
plasmic leaflet of the lipid bilayer enriched in choles-
figuration tend to increase the fluidity of a bilayer by de-
terol and sphingolipids; they are involved in signal
creasing the compactness of the side chain packing with-
transduction and possibly other processes. Caveolae
out diminishing hydrophobicity
(Figure
41-3). The
may derive from lipid rafts. Many if not all of them
phospholipids of cellular membranes generally contain
contain the protein caveolin-1, which may be involved
at least one unsaturated fatty acid with at least one cis
in their formation from rafts. Caveolae are observable
double bond.
by electron microscopy as flask-shaped indentations of
Cholesterol modifies the fluidity of membranes. At
the cell membrane. Proteins detected in caveolae in-
temperatures below the Tm, it interferes with the inter-
clude various components of the signal-transduction
action of the hydrocarbon tails of fatty acids and thus
system (eg, the insulin receptor and some G proteins),
increases fluidity. At temperatures above the Tm, it lim-
the folate receptor, and endothelial nitric oxide syn-
its disorder because it is more rigid than the hydrocar-
thase (eNOS). Caveolae and lipid rafts are active areas
bon tails of the fatty acids and cannot move in the
of research, and ideas concerning them and their possi-
membrane to the same extent, thus limiting fluidity. At
ble roles in various diseases are rapidly evolving.
MEMBRANES: STRUCTURE & FUNCTION
/
423
MEMBRANE SELECTIVITY ALLOWS
inversely proportionate to the number of hydrogen
bonds that must be broken in order for a solute in the
SPECIALIZED FUNCTIONS
external aqueous phase to become incorporated in the
If the plasma membrane is relatively impermeable, how
hydrophobic bilayer. Electrolytes, poorly soluble in
do most molecules enter a cell? How is selectivity of
lipid, do not form hydrogen bonds with water, but they
this movement established? Answers to such questions
do acquire a shell of water from hydration by electrosta-
are important in understanding how cells adjust to a
tic interaction. The size of the shell is directly propor-
constantly changing extracellular environment. Meta-
tionate to the charge density of the electrolyte. Elec-
zoan organisms also must have means of communicat-
trolytes with a large charge density have a larger shell of
ing between adjacent and distant cells, so that complex
hydration and thus a slower diffusion rate. Na+, for ex-
biologic processes can be coordinated. These signals
ample, has a higher charge density than K+. Hydrated
must arrive at and be transmitted by the membrane, or
Na+ is therefore larger than hydrated K+; hence, the lat-
they must be generated as a consequence of some inter-
ter tends to move more easily through the membrane.
action with the membrane. Some of the major mecha-
The following factors affect net diffusion of a sub-
nisms used to accomplish these different objectives are
stance: (1) Its concentration gradient across the mem-
listed in Table 41-3.
brane. Solutes move from high to low concentration.
(2) The electrical potential across the membrane.
Passive Mechanisms Move Some Small
Solutes move toward the solution that has the opposite
Molecules Across Membranes
charge. The inside of the cell usually has a negative
charge.
(3) The permeability coefficient of the sub-
Molecules can passively traverse the bilayer down elec-
stance for the membrane. (4) The hydrostatic pressure
trochemical gradients by simple diffusion or by facili-
gradient across the membrane. Increased pressure will
tated diffusion. This spontaneous movement toward
increase the rate and force of the collision between the
equilibrium contrasts with active transport, which re-
molecules and the membrane. (5) Temperature. In-
quires energy because it constitutes movement against
creased temperature will increase particle motion and
an electrochemical gradient. Figure 41-8 provides a
thus increase the frequency of collisions between exter-
schematic representation of these mechanisms.
nal particles and the membrane. In addition, a multi-
As described above, some solutes such as gases can
tude of channels exist in membranes that route the
enter the cell by diffusing down an electrochemical gra-
entry of ions into cells.
dient across the membrane and do not require meta-
bolic energy. The simple passive diffusion of a solute
Ion Channels Are Transmembrane
across the membrane is limited by the thermal agitation
of that specific molecule, by the concentration gradient
Proteins That Allow the Selective
across the membrane, and by the solubility of that
Entry of Various Ions
solute (the permeability coefficient, Figure 41-6) in the
In natural membranes, as opposed to synthetic mem-
hydrophobic core of the membrane bilayer. Solubility is
brane bilayers, there are transmembrane channels, pore-
like structures composed of proteins that constitute se-
lective ion channels. Cation-conductive channels have
Table 41-3. Transfer of material and information
an average diameter of about 5-8 nm and are negatively
across membranes.
charged within the channel. The permeability of a chan-
nel depends upon the size, the extent of hydration, and
Cross-membrane movement of small molecules
the extent of charge density on the ion. Specific chan-
Diffusion (passive and facilitated)
nels for Na+, K+, Ca2+, and Cl− have been identified; two
Active transport
such channels are illustrated in Figure 41-9. Both are
Cross-membrane movement of large molecules
seen to consist of four subunits. Each subunit consists of
Endocytosis
six α-helical transmembrane domains. The amino and
Exocytosis
carboxyl terminals of both ion channels are located in
Signal transmission across membranes
the cytoplasm, with both extracellular and intracellular
Cell surface receptors
loops being present. The actual pores in the channels
1. Signal transduction (eg, glucagon → cAMP)
through which the ions pass are not shown in the figure.
2. Signal internalization (coupled with endocytosis, eg,
They form the center (diameter about 5-8 nm) of a
the LDL receptor)
structure formed by apposition of the subunits. The
Movement to intracellular receptors (steroid hormones; a
channels are very selective, in most cases permitting the
form of diffusion)
passage of only one type of ion (Na+, Ca2+, etc). Many
Intercellular contact and communication
variations on the above structural themes are found, but
424
/
CHAPTER 41
Transported
molecule
Carrier
Channel
protein
protein
Electrochemical
Lipid
gradient
bilayer
Simple
Facilitated
diffusion
diffusion
Passive transport
Active transport
Figure 41-8. Many small uncharged molecules pass freely through the lipid
bilayer. Charged molecules, larger uncharged molecules, and some small un-
charged molecules are transferred through channels or pores or by specific carrier
proteins. Passive transport is always down an electrochemical gradient, toward
equilibrium. Active transport is against an electrochemical gradient and requires
an input of energy, whereas passive transport does not. (Redrawn and reproduced,
with permission, from Alberts B et al: Molecular Biology of the Cell. Garland, 1983.)
all ion channels are basically made up of transmembrane
Table 41-4; other aspects of ion channels are discussed
subunits that come together to form a central pore
briefly in Chapter 49.
through which ions pass selectively. The combination of
x-ray crystallography (where possible) and site-directed
Ionophores Are Molecules That Act as
mutagenesis affords a powerful approach to delineating
Membrane Shuttles for Various Ions
the structure-function relationships of ion channels.
The membranes of nerve cells contain well-studied
Certain microbes synthesize small organic molecules,
ion channels that are responsible for the action poten-
ionophores, that function as shuttles for the movement
tials generated across the membrane. The activity of
of ions across membranes. These ionophores contain hy-
some of these channels is controlled by neurotransmit-
drophilic centers that bind specific ions and are sur-
ters; hence, channel activity can be regulated. One ion
rounded by peripheral hydrophobic regions; this arrange-
can regulate the activity of the channel of another ion.
ment allows the molecules to dissolve effectively in the
For example, a decrease of Ca2+ concentration in the
membrane and diffuse transversely therein. Others, like
extracellular fluid increases membrane permeability and
the well-studied polypeptide gramicidin, form channels.
increases the diffusion of Na+. This depolarizes the
Microbial toxins such as diphtheria toxin and acti-
membrane and triggers nerve discharge, which may ex-
vated serum complement components can produce large
plain the numbness, tingling, and muscle cramps symp-
pores in cellular membranes and thereby provide macro-
tomatic of a low level of plasma Ca2+.
molecules with direct access to the internal milieu.
Channels are open transiently and thus are “gated.”
Gates can be controlled by opening or closing. In lig-
Aquaporins Are Proteins That Form Water
and-gated channels, a specific molecule binds to a re-
Channels in Certain Membranes
ceptor and opens the channel.Voltage-gated channels
open (or close) in response to changes in membrane po-
In certain cells (eg, red cells, cells of the collecting duc-
tential. Some properties of ion channels are listed in
tules of the kidney), the movement of water by simple
MEMBRANES: STRUCTURE & FUNCTION
/
425
Rat brain
Na+ channel
I
II
III
IV
Outside
1
2
3
4
5
6
1
2
3
4
5
6
1
2
3
4
5
6
1
2
3
4
5
6
Inside
C
N
Rabbit skeletal muscle
Ca2+ channel
I
II
III
IV
1
2
3
4
5
6
1
2
3
4
5
6
1
2
3
4
5
6
1
2
3
4
5
6
Figure 41-9. Diagrammatic representation of the structures of two ion channels. The Roman numer-
als indicate the four subunits of each channel and the Arabic numerals the α-helical transmembrane do-
mains of each subunit. The actual pores through which the ions pass are not shown but are formed by
apposition of the various subunits. The specific areas of the subunits involved in the opening and clos-
ing of the channels are also not indicated. (After WK Catterall. Modified and reproduced from Hall ZW: An
Introduction to Molecular Neurobiology. Sinauer, 1992.)
426
/
CHAPTER 41
Table 41-4. Some properties of ion channels.
• They are composed of transmembrane protein subunits.
Lipid
• Most are highly selective for one ion; a few are nonselec-
bilayer
tive.
• They allow impermeable ions to cross membranes at rates
approaching diffusion limits.
• They can permit ion fluxes of 106-107/s.
• Their activities are regulated.
Uniport
Symport
Antiport
• The two main types are voltage-gated and ligand-gated.
• They are usually highly conserved across species.
Cotransport
• Most cells have a variety of Na+, K+, Ca2+, and CI− channels.
• Mutations in genes encoding them can cause specific
Figure 41-10. Schematic representation of types of
diseases.1
transport systems. Transporters can be classified with
• Their activities are affected by certain drugs.
regard to the direction of movement and whether one
1Some diseases caused by mutations of ion channels are briefly
or more unique molecules are moved. (Redrawn and re-
discussed in Chapter 49.
produced, with permission, from Alberts B et al: Molecular
Biology of the Cell. Garland, 1983.)
diffusion is augmented by movement through water
Mutations in bacteria and mammalian cells (including
channels. These channels are composed of tetrameric
some that result in human disease) have supported
transmembrane proteins named aquaporins. At least
these conclusions. Facilitated diffusion and active trans-
five distinct aquaporins (AP-1 to AP-5) have been iden-
port resemble a substrate-enzyme reaction except
tified. Mutations in the gene encoding AP-2 have been
that no covalent interaction occurs. These points of re-
shown to be the cause of one type of nephrogenic dia-
semblance are as follows: (1) There is a specific binding
betes insipidus.
site for the solute. (2) The carrier is saturable, so it has a
maximum rate of transport
(Vmax; Figure
41-11).
(3) There is a binding constant (Km) for the solute, and
PLASMA MEMBRANES ARE INVOLVED
IN FACILITATED DIFFUSION, ACTIVE
TRANSPORT, & OTHER PROCESSES
Passive
Transport systems can be described in a functional
diffusion
Vmax
sense according to the number of molecules moved and
the direction of movement (Figure 41-10) or according
to whether movement is toward or away from equilib-
rium. A uniport system moves one type of molecule
Carrier-mediated
diffusion
bidirectionally. In cotransport systems, the transfer of
one solute depends upon the stoichiometric simultane-
ous or sequential transfer of another solute. A symport
moves these solutes in the same direction. Examples are
the proton-sugar transporter in bacteria and the Na+ -
sugar transporters (for glucose and certain other sugars)
and Na+-amino acid transporters in mammalian cells.
Km
Antiport systems move two molecules in opposite di-
Solute concentration
rections (eg, Na+ in and Ca2+ out).
Molecules that cannot pass freely through the lipid
Figure 41-11. A comparison of the kinetics of car-
bilayer membrane by themselves do so in association
rier-mediated (facilitated) diffusion with passive diffu-
with carrier proteins. This involves two processes—
sion. The rate of movement in the latter is directly pro-
facilitated diffusion and active transport—and highly
portionate to solute concentration, whereas the
specific transport systems.
process is saturable when carriers are involved. The
Facilitated diffusion and active transport share many
concentration at half-maximal velocity is equal to the
features. Both appear to involve carrier proteins, and
binding constant (Km) of the carrier for the solute. (Vmax,
both show specificity for ions, sugars, and amino acids.
maximal rate.)
MEMBRANES: STRUCTURE & FUNCTION
/
427
so the whole system has a Km (Figure 41-11). (4) Struc-
termined by the following factors: (1) The concentra-
turally similar competitive inhibitors block transport.
tion gradient across the membrane. (2) The amount of
Major differences are the following: (1) Facilitated
carrier available (this is a key control step). (3) The ra-
diffusion can operate bidirectionally, whereas active
pidity of the solute-carrier interaction. (4) The rapidity
transport is usually unidirectional. (2) Active transport
of the conformational change for both the loaded and
always occurs against an electrical or chemical gradient,
the unloaded carrier.
and so it requires energy.
Hormones regulate facilitated diffusion by changing
the number of transporters available. Insulin increases
Facilitated Diffusion
glucose transport in fat and muscle by recruiting trans-
porters from an intracellular reservoir. Insulin also en-
Some specific solutes diffuse down electrochemical gra-
hances amino acid transport in liver and other tissues.
dients across membranes more rapidly than might be
One of the coordinated actions of glucocorticoid hor-
expected from their size, charge, or partition coeffi-
mones is to enhance transport of amino acids into liver,
cients. This facilitated diffusion exhibits properties
where the amino acids then serve as a substrate for glu-
distinct from those of simple diffusion. The rate of fa-
coneogenesis. Growth hormone increases amino acid
cilitated diffusion, a uniport system, can be saturated;
transport in all cells, and estrogens do this in the uterus.
ie, the number of sites involved in diffusion of the spe-
There are at least five different carrier systems for
cific solutes appears finite. Many facilitated diffusion
amino acids in animal cells. Each is specific for a group
systems are stereospecific but, like simple diffusion, re-
of closely related amino acids, and most operate as Na+-
quire no metabolic energy.
symport systems (Figure 41-10).
As described earlier, the inside-outside asymmetry of
membrane proteins is stable, and mobility of proteins
Active Transport
across (rather than in) the membrane is rare; therefore,
transverse mobility of specific carrier proteins is not
The process of active transport differs from diffusion in
likely to account for facilitated diffusion processes ex-
that molecules are transported away from thermody-
cept in a few unusual cases.
namic equilibrium; hence, energy is required. This en-
A “Ping-Pong” mechanism (Figure 41-12) ex-
ergy can come from the hydrolysis of ATP, from elec-
plains facilitated diffusion. In this model, the carrier
tron movement, or from light. The maintenance of
protein exists in two principal conformations. In the
electrochemical gradients in biologic systems is so im-
“pong” state, it is exposed to high concentrations of
portant that it consumes perhaps 30-40% of the total
solute, and molecules of the solute bind to specific sites
energy expenditure in a cell.
on the carrier protein. Transport occurs when a confor-
In general, cells maintain a low intracellular Na+
mational change exposes the carrier to a lower concen-
concentration and a high intracellular K+ concentration
tration of solute (“ping” state). This process is com-
(Table 41-1), along with a net negative electrical po-
pletely reversible, and net flux across the membrane
tential inside. The pump that maintains these gradients
depends upon the concentration gradient. The rate at
is an ATPase that is activated by Na+ and K+ (Na+-K+
which solutes enter a cell by facilitated diffusion is de-
ATPase; see Figure 41-13). The ATPase is an integral
Pong
Ping
Figure 41-12. The “Ping-Pong” model of facilitated diffusion. A protein carrier (gray structure) in the lipid bi-
layer associates with a solute in high concentration on one side of the membrane. A conformational change en-
sues (“pong” to “ping”), and the solute is discharged on the side favoring the new equilibrium. The empty carrier
then reverts to the original conformation (“ping” to “pong”) to complete the cycle.
428
/
CHAPTER 41
only where the membrane is free of the insulation. The
INSIDE
OUTSIDE
myelin membrane is composed of phospholipids, cho-
Membrane
lesterol, proteins, and GSLs. Relatively few proteins are
3 Na+
found in the myelin membrane; those present appear to
ATP
3 Na+
hold together multiple membrane bilayers to form the
hydrophobic insulating structure that is impermeable
to ions and water. Certain diseases, eg, multiple sclero-
Mg2+
sis and the Guillain-Barré syndrome, are characterized
+
2 K
by demyelination and impaired nerve conduction.
ADP
+
Pi
2 K+
Glucose Transport Involves
Several Mechanisms
Figure 41-13. Stoichiometry of the Na+-K+ ATPase
A discussion of the transport of glucose summarizes
pump. This pump moves three Na+ ions from inside the
many of the points made in this chapter. Glucose must
cell to the outside and brings two K+ ions from the out-
enter cells as the first step in energy utilization. In
side to the inside for every molecule of ATP hydrolyzed
adipocytes and muscle, glucose enters by a specific
to ADP by the membrane-associated ATPase. Ouabain
transport system that is enhanced by insulin. Changes
and other cardiac glycosides inhibit this pump by act-
in transport are primarily due to alterations of Vmax
ing on the extracellular surface of the membrane.
(presumably from more or fewer active transporters),
(Courtesy of R Post.)
but changes in Km may also be involved. Glucose trans-
port involves different aspects of the principles of trans-
port discussed above. Glucose and Na+ bind to different
membrane protein and requires phospholipids for ac-
sites on the glucose transporter. Na+ moves into the cell
tivity. The ATPase has catalytic centers for both ATP
down its electrochemical gradient and “drags” glucose
and Na+ on the cytoplasmic side of the membrane, but
with it (Figure 41-14). Therefore, the greater the Na+
the K+ binding site is located on the extracellular side of
gradient, the more glucose enters; and if Na+ in extra-
the membrane. Ouabain or digitalis inhibits this ATP-
cellular fluid is low, glucose transport stops. To main-
ase by binding to the extracellular domain. Inhibition
tain a steep Na+ gradient, this Na+-glucose symport is
of the ATPase by ouabain can be antagonized by extra-
dependent on gradients generated by an Na+-K+ pump
cellular K+.
that maintains a low intracellular Na+ concentration.
Similar mechanisms are used to transport other sugars
as well as amino acids.
Nerve Impulses Are Transmitted
The transcellular movement of sugars involves one
Up & Down Membranes
additional component: a uniport that allows the glucose
The membrane forming the surface of neuronal cells
accumulated within the cell to move across a different
maintains an asymmetry of inside-outside voltage (elec-
surface toward a new equilibrium; this occurs in intesti-
trical potential) and is electrically excitable. When ap-
nal and renal cells, for example.
propriately stimulated by a chemical signal mediated by
a specific synaptic membrane receptor (see discussion of
Cells Transport Certain Macromolecules
the transmission of biochemical signals, below), gates in
Across the Plasma Membrane
the membrane are opened to allow the rapid influx of
Na+ or Ca2+ (with or without the efflux of K+), so that
The process by which cells take up large molecules is
the voltage difference rapidly collapses and that seg-
called
“endocytosis.” Some of these molecules (eg,
ment of the membrane is depolarized. However, as a re-
polysaccharides, proteins, and polynucleotides), when
sult of the action of the ion pumps in the membrane,
hydrolyzed inside the cell, yield nutrients. Endocytosis
the gradient is quickly restored.
provides a mechanism for regulating the content of cer-
When large areas of the membrane are depolarized
tain membrane components, hormone receptors being
in this manner, the electrochemical disturbance propa-
a case in point. Endocytosis can be used to learn more
gates in wave-like form down the membrane, generat-
about how cells function. DNA from one cell type can
ing a nerve impulse. Myelin sheets, formed by
be used to transfect a different cell and alter the latter’s
Schwann cells, wrap around nerve fibers and provide an
function or phenotype. A specific gene is often em-
electrical insulator that surrounds most of the nerve and
ployed in these experiments, and this provides a unique
greatly speeds up the propagation of the wave (signal)
way to study and analyze the regulation of that gene.
by allowing ions to flow in and out of the membrane
DNA transfection depends upon endocytosis; endocy-
MEMBRANES: STRUCTURE & FUNCTION
/
429
LUMEN
A
B
Glucose
Na+
(Symport)
CYTOSOL
Glucose
Na+
K+
CP
Na+
K+
Glucose
EXTRACELLULAR FLUID
Figure 41-14. The transcellular movement of glu-
V
cose in an intestinal cell. Glucose follows Na+ across the
luminal epithelial membrane. The Na+ gradient that
CV
drives this symport is established by Na+ -K+ exchange,
which occurs at the basal membrane facing the extra-
Figure 41-15. Two types of endocytosis. An endo-
cellular fluid compartment. Glucose at high concentra-
cytotic vesicle (V) forms as a result of invagination of a
tion within the cell moves “downhill” into the extracel-
portion of the plasma membrane. Fluid-phase endocy-
lular fluid by facilitated diffusion (a uniport mechanism).
tosis (A) is random and nondirected. Receptor-medi-
ated endocytosis (B) is selective and occurs in coated
pits (CP) lined with the protein clathrin (the fuzzy mate-
rial). Targeting is provided by receptors (black symbols)
tosis is responsible for the entry of DNA into the cell.
specific for a variety of molecules. This results in the for-
Such experiments commonly use calcium phosphate,
mation of a coated vesicle (CV).
since Ca2+ stimulates endocytosis and precipitates
DNA, which makes the DNA a better object for endo-
cytosis. Cells also release macromolecules by exocyto-
sis. Endocytosis and exocytosis both involve vesicle for-
reused in the cytoplasm. Endocytosis requires (1) en-
mation with or from the plasma membrane.
ergy, usually from the hydrolysis of ATP; (2) Ca2+ in
extracellular fluid; and (3) contractile elements in the
A. ENDOCYTOSIS
cell (probably the microfilament system) (Chapter 49).
All eukaryotic cells are continuously ingesting parts of
There are two general types of endocytosis. Phago-
their plasma membranes. Endocytotic vesicles are gen-
cytosis occurs only in specialized cells such as
erated when segments of the plasma membrane invagi-
macrophages and granulocytes. Phagocytosis involves
nate, enclosing a minute volume of extracellular fluid
the ingestion of large particles such as viruses, bacteria,
and its contents. The vesicle then pinches off as the fu-
cells, or debris. Macrophages are extremely active in
sion of plasma membranes seals the neck of the vesicle
this regard and may ingest 25% of their volume per
at the original site of invagination (Figure 41-15). This
hour. In so doing, a macrophage may internalize 3% of
vesicle fuses with other membrane structures and thus
its plasma membrane each minute or the entire mem-
achieves the transport of its contents to other cellular
brane every 30 minutes.
compartments or even back to the cell exterior. Most
Pinocytosis is a property of all cells and leads to the
endocytotic vesicles fuse with primary lysosomes to
cellular uptake of fluid and fluid contents. There are
form secondary lysosomes, which contain hydrolytic
two types. Fluid-phase pinocytosis is a nonselective
enzymes and are therefore specialized organelles for in-
process in which the uptake of a solute by formation of
tracellular disposal. The macromolecular contents are
small vesicles is simply proportionate to its concentra-
digested to yield amino acids, simple sugars, or nu-
tion in the surrounding extracellular fluid. The forma-
cleotides, and they diffuse out of the vesicles to be
tion of these vesicles is an extremely active process. Fi-
430
/
CHAPTER 41
broblasts, for example, internalize their plasma mem-
terol—released during the degradation of LDL. Disor-
brane at about one-third the rate of macrophages. This
ders of the LDL receptor and its internalization are
process occurs more rapidly than membranes are made.
medically important and are discussed in Chapter 25.
The surface area and volume of a cell do not change
Absorptive pinocytosis of extracellular glycopro-
much, so membranes must be replaced by exocytosis or
teins requires that the glycoproteins carry specific car-
by being recycled as fast as they are removed by endocy-
bohydrate recognition signals. These recognition signals
tosis.
are bound by membrane receptor molecules, which
The other type of pinocytosis, absorptive pinocyto-
play a role analogous to that of the LDL receptor. A
sis, is a receptor-mediated selective process primarily re-
galactosyl receptor on the surface of hepatocytes is in-
sponsible for the uptake of macromolecules for which
strumental in the absorptive pinocytosis of asialoglyco-
there are a finite number of binding sites on the plasma
proteins from the circulation (Chapter 47). Acid hydro-
membrane. These high-affinity receptors permit the se-
lases taken up by absorptive pinocytosis in fibroblasts
lective concentration of ligands from the medium, min-
are recognized by their mannose 6-phosphate moieties.
imize the uptake of fluid or soluble unbound macro-
Interestingly, the mannose
6-phosphate moiety also
molecules, and markedly increase the rate at which
seems to play an important role in the intracellular tar-
specific molecules enter the cell. The vesicles formed
geting of the acid hydrolases to the lysosomes of the
during absorptive pinocytosis are derived from invagi-
cells in which they are synthesized (Chapter 47).
nations (pits) that are coated on the cytoplasmic side
There is a dark side to receptor-mediated endocyto-
with a filamentous material. In many systems, the pro-
sis in that viruses which cause such diseases as hepatitis
tein clathrin is the filamentous material. It has a three-
(affecting liver cells), poliomyelitis
(affecting motor
limbed structure (called a triskelion), with each limb
neurons), and AIDS (affecting T cells) initiate their
being made up of one light and one heavy chain of
damage by this mechanism. Iron toxicity also begins
clathrin. The polymerization of clathrin into a vesicle is
with excessive uptake due to endocytosis.
directed by assembly particles, composed of four
B. EXOCYTOSIS
adapter proteins. These interact with certain amino
acid sequences in the receptors that become cargo, en-
Most cells release macromolecules to the exterior by ex-
suring selectivity of uptake. The lipid PIP2 also plays an
ocytosis. This process is also involved in membrane re-
important role in vesicle assembly. In addition, the pro-
modeling, when the components synthesized in the
tein dynamin, which both binds and hydrolyzes GTP,
Golgi apparatus are carried in vesicles to the plasma
is necessary for the pinching off of clathrin-coated vesi-
membrane. The signal for exocytosis is often a hor-
cles from the cell surface. Coated pits may constitute as
mone which, when it binds to a cell-surface receptor,
much as 2% of the surface of some cells.
induces a local and transient change in Ca2+ concentra-
As an example, the low-density lipoprotein (LDL)
tion. Ca2+ triggers exocytosis. Figure 41-16 provides a
molecule and its receptor (Chapter 25) are internalized
comparison of the mechanisms of exocytosis and endo-
by means of coated pits containing the LDL receptor.
cytosis.
These endocytotic vesicles containing LDL and its re-
Molecules released by exocytosis fall into three cate-
ceptor fuse to lysosomes in the cell. The receptor is re-
gories: (1) They can attach to the cell surface and be-
leased and recycled back to the cell surface membrane,
come peripheral proteins, eg, antigens. (2) They can be-
but the apoprotein of LDL is degraded and the choles-
come part of the extracellular matrix, eg, collagen and
teryl esters metabolized. Synthesis of the LDL receptor
glycosaminoglycans. (3) They can enter extracellular
is regulated by secondary or tertiary consequences of
fluid and signal other cells. Insulin, parathyroid hor-
pinocytosis, eg, by metabolic products—such as choles-
mone, and the catecholamines are all packaged in gran-
Exocytosis
Endocytosis
Figure 41-16. A comparison of the mechanisms of endocytosis and exocyto-
sis. Exocytosis involves the contact of two inside surface (cytoplasmic side) mono-
layers, whereas endocytosis results from the contact of two outer surface mono-
layers.
MEMBRANES: STRUCTURE & FUNCTION
/
431
ules and processed within cells, to be released upon ap-
amino acids, sugars, lipids, urate, anions, cations, water,
propriate stimulation.
and vitamins across the plasma membrane. Mutations
in genes encoding proteins in other membranes can
also have harmful consequences. For example, muta-
Some Signals Are Transmitted
tions in genes encoding mitochondrial membrane
Across Membranes
proteins involved in oxidative phosphorylation can
Specific biochemical signals such as neurotransmitters,
cause neurologic and other problems (eg, Leber’s hered-
hormones, and immunoglobulins bind to specific re-
itary optic neuropathy; LHON). Membrane proteins
ceptors (integral proteins) exposed to the outside of
can also be affected by conditions other than muta-
cellular membranes and transmit information through
tions. Formation of autoantibodies to the acetyl-
these membranes to the cytoplasm. This process, called
choline receptor in skeletal muscle causes myasthenia
transmembrane signaling, involves the generation of a
gravis. Ischemia can quickly affect the integrity of vari-
number of signals, including cyclic nucleotides, cal-
ous ion channels in membranes. Abnormalities of
cium, phosphoinositides, and diacylglycerol. It is dis-
membrane constituents other than proteins can also be
cussed in detail in Chapter 43.
harmful. With regard to lipids, excess of cholesterol
(eg, in familial hypercholesterolemia), of lysophospho-
lipid (eg, after bites by certain snakes, whose venom
Information Can Be Communicated
contains phospholipases), or of glycosphingolipids (eg,
by Intercellular Contact
in a sphingolipidosis) can all affect membrane function.
There are many areas of intercellular contact in a meta-
zoan organism. This necessitates contact between the
Cystic Fibrosis Is Due to Mutations in the
plasma membranes of the individual cells. Cells have
Gene Encoding a Chloride Channel
developed specialized regions on their membranes for
intercellular communication in close proximity. Gap
Cystic fibrosis (CF) is a recessive genetic disorder preva-
junctions mediate and regulate the passage of ions and
lent among whites in North America and certain parts
small molecules (up to 1000-2000 MW) through a
of northern Europe. It is characterized by chronic bac-
narrow hydrophilic core connecting the cytosol of adja-
terial infections of the airways and sinuses, fat maldiges-
cent cells. These structures are primarily composed of
tion due to pancreatic exocrine insufficiency, infertility
the protein connexin, which contains four membrane-
in males due to abnormal development of the vas defer-
spanning α helices. About a dozen genes encoding dif-
ens, and elevated levels of chloride in sweat
(>
60
ferent connexins have been cloned. An assembly of 12
mmol/L).
connexin molecules forms a structure
(a connexon)
After a Herculean landmark endeavor, the gene for
with a central channel that forms bridges between adja-
CF was identified in 1989 on chromosome 7. It was
cent cells. Ions and small molecules pass from the cy-
found to encode a protein of 1480 amino acids, named
tosol of one cell to that of another through the chan-
cystic fibrosis transmembrane regulator
(CFTR), a
nels, which open and close in a regulated fashion.
cyclic AMP-regulated Cl− channel (see Figure 41-17).
An abnormality of membrane Cl− permeability is be-
lieved to result in the increased viscosity of many bodily
MUTATIONS AFFECTING MEMBRANE
secretions, though the precise mechanisms are still
PROTEINS CAUSE DISEASES
under investigation. The commonest mutation (~70%
In view of the fact that membranes are located in so
in certain Caucasian populations) is deletion of three
many organelles and are involved in so many processes,
bases, resulting in loss of residue 508, a phenylalanine
it is not surprising that mutations affecting their pro-
(∆F508). However, more than 900 other mutations have
tein constituents should result in many diseases or dis-
been identified. These mutations affect CFTR in at
orders. Proteins in membranes can be classified as re-
least four ways: (1) its amount is reduced; (2) depend-
ceptors, transporters, ion channels, enzymes, and
ing upon the particular mutation, it may be susceptible
structural components. Members of all of these classes
to misfolding and retention within the ER or Golgi ap-
are often glycosylated, so that mutations affecting this
paratus; (3) mutations in the nucleotide-binding do-
process may alter their function. Examples of diseases
mains may affect the ability of the Cl− channel to open,
or disorders due to abnormalities in membrane proteins
an event affected by ATP; (4) the mutations may also
are listed in Table 41-5; these mainly reflect mutations
reduce the rate of ion flow through a channel, generat-
in proteins of the plasma membrane, with one affect-
ing less of a Cl− current.
ing lysosomal function (I-cell disease). Over 30 genetic
The most serious and life-threatening complication
diseases or disorders have been ascribed to mutations
is recurrent pulmonary infections due to overgrowth of
affecting various proteins involved in the transport of
various pathogens in the viscous secretions of the respi-
432
/
CHAPTER 41
Table 41-5. Some diseases or pathologic states resulting from or attributed to abnormalities
of membranes.1
Disease
Abnormality
Achondroplasia
Mutations in the gene encoding the fibroblast growth factor receptor 3
(MIM 100800)
Familial hypercholester-
Mutations in the gene encoding the LDL receptor
olemia (MIM 143890)
Cystic fibrosis
Mutations in the gene encoding the CFTR protein, a Cl− transporter
(MIM 219700)
Congenital long QT syn-
Mutations in genes encoding ion channels in the heart
drome (MIM 192500)
Wilson disease
Mutations in the gene encoding a copper-dependent ATPase
(MIM 277900)
I-cell disease
Mutations in the gene encoding GIcNAc phosphotransferase, resulting in absence of the Man 6-P
(MIM 252500)
signal for lysosomal localization of certain hydrolases
Hereditary spherocytosis
Mutations in the genes encoding spectrin or other structural proteins in the red cell membrane
(MIM 182900)
Metastasis
Abnormalities in the oligosaccharide chains of membrane glycoproteins and glycolipids are thought
to be of importance
Paroxysmal nocturnal
Mutation resulting in deficient attachment of the GPI anchor to certain proteins of the red cell
hemoglobinuria
membrane
(MIM 311770)
1The disorders listed are discussed further in other chapters. The table lists examples of mutations affecting receptors, a transporter, an
ion channel, an enzyme, and a structural protein. Examples of altered or defective glycosylation of glycoproteins are also presented. Most
of the conditions listed affect the plasma membrane.
ratory tract. Poor nutrition as a result of pancreatic in-
sufficiency worsens the situation. The treatment of CF
thus requires a comprehensive effort to maintain nutri-
tional status, to prevent and combat pulmonary infec-
tions, and to maintain physical and psychologic health.
Advances in molecular genetics mean that mutation
Amino
analysis can be performed for prenatal diagnosis and for
terminal
NBF1
carrier testing in families in which one child already has
R domain
NBF2
the condition. Efforts are in progress to use gene ther-
apy to restore the activity of CFTR. An aerosolized
Carboxyl
preparation of human DNase that digests the DNA of
terminal
microorganisms in the respiratory tract has proved
helpful in therapy.
Figure 41-17. Diagram of the structure of the CFTR
protein (not to scale). The protein contains twelve
SUMMARY
transmembrane segments (probably helical), two nu-
cleotide-binding folds or domains (NBF1 and NBF2),
• Membranes are complex structures composed of
and one regulatory (R) domain. NBF1 and NBF2 proba-
lipids, carbohydrates, and proteins.
bly bind ATP and couple its hydrolysis to transport of
• The basic structure of all membranes is the lipid bi-
Cl−. Phe 508, the major locus of mutations in cystic fi-
layer. This bilayer is formed by two sheets of phos-
brosis, is located in NBF1.
pholipids in which the hydrophilic polar head groups
MEMBRANES: STRUCTURE & FUNCTION
/
433
are directed away from each other and are exposed to
• Receptors may be integral components of mem-
the aqueous environment on the outer and inner sur-
branes (particularly the plasma membrane). The in-
faces of the membrane. The hydrophobic nonpolar
teraction of a ligand with its receptor may not in-
tails of these molecules are oriented toward each
volve the movement of either into the cell, but the
other, in the direction of the center of the mem-
interaction results in the generation of a signal that
brane.
influences intracellular processes
(transmembrane
•
Membrane proteins are classified as integral if they
signaling).
are firmly embedded in the bilayer and as peripheral
• Mutations that affect the structure of membrane pro-
if they are loosely attached to the outer or inner sur-
teins (receptors, transporters, ion channels, enzymes,
face.
and structural proteins) may cause diseases; examples
•
The 20 or so different membranes in a mammalian
include cystic fibrosis and familial hypercholes-
cell have intrinsic functions (eg, enzymatic activity),
terolemia.
and they define compartments, or specialized envi-
ronments, within the cell that have specific functions
(eg, lysosomes).
REFERENCES
•
Certain molecules freely diffuse across membranes,
but the movement of others is restricted because of
Doyle DA et al: The structure of the potassium channel: molecular
basis of K+ conductance and selectivity. Science 1998;280:
size, charge, or solubility.
69.
•
Various passive and active mechanisms are employed
Felix R: Channelopathies: ion channel defects linked to heritable
to maintain gradients of such molecules across differ-
clinical disorders. J Med Genet 2000;37:729.
ent membranes.
Garavito RM, Ferguson-Miller S: Detergents as tools in membrane
•
Certain solutes, eg, glucose, enter cells by facilitated
biochemistry. J Biol Chem 2001;276:32403.
diffusion, along a downhill gradient from high to low
Gillooly DJ, Stenmark H: A lipid oils the endocytosis machine. Sci-
concentration. Specific carrier molecules, or trans-
ence 2001;291;993.
porters, are involved in such processes.
Knowles MR, Durie PR: What is cystic fibrosis? N Engl J Med
2002;347:439.
•
Ligand- or voltage-gated ion channels are often em-
Longo N: Inherited defects of membrane transport. In: Harrison’s
ployed to move charged molecules (Na+, K+, Ca2+,
Principles of Internal Medicine, 15th ed. Braunwald E et al
etc) across membranes.
(editors). McGraw-Hill, 2001.
•
Large molecules can enter or leave cells through
Marx J: Caveolae: a once-elusive structure gets some respect. Sci-
mechanisms such as endocytosis or exocytosis. These
ence 2001;294;1862.
processes often require binding of the molecule to a
White SH et al: How membranes shape protein structure. J Biol
receptor, which affords specificity to the process.
Chem 2001:276:32395.
The Diversity of the
42
Endocrine System
Daryl K. Granner, MD
ACTH Adrenocorticotropic hormone
GH Growth hormone
ANF Atrial natriuretic factor
IGF-I Insulin-like growth factor-I
cAMP Cyclic adenosine monophosphate
LH Luteotropic hormone
CBG Corticosteroid-binding globulin
LPH Lipotropin
CG Chorionic gonadotropin
MIT Monoiodotyrosine
cGMP Cyclic guanosine monophosphate
MSH Melanocyte-stimulating hormone
CLIP Corticotropin-like intermediate lobe
OHSD Hydroxysteroid dehydrogenase
peptide
PNMT Phenylethanolamine-N-methyltransferase
DBH Dopamine β-hydroxylase
POMC Pro-opiomelanocortin
DHEA Dehydroepiandrosterone
SHBG Sex hormone-binding globulin
DHT Dihydrotestosterone
StAR Steroidogenic acute regulatory (protein)
DIT Diiodotyrosine
TBG Thyroxine-binding globulin
DOC Deoxycorticosterone
TEBG Testosterone-estrogen-binding globulin
EGF Epidermal growth factor
TRH Thyrotropin-releasing hormone
FSH Follicle-stimulating hormone
TSH Thyrotropin-stimulating hormone
BIOMEDICAL IMPORTANCE
tive because hormones can act on adjacent cells
(paracrine action) and on the cell in which they were
The survival of multicellular organisms depends on their
synthesized (autocrine action) without entering the sys-
ability to adapt to a constantly changing environment.
temic circulation. A diverse array of hormones—each
Intercellular communication mechanisms are necessary
with distinctive mechanisms of action and properties of
requirements for this adaptation. The nervous system
biosynthesis, storage, secretion, transport, and metabo-
and the endocrine system provide this intercellular, or-
lism—has evolved to provide homeostatic responses.
ganism-wide communication. The nervous system was
This biochemical diversity is the topic of this chapter.
originally viewed as providing a fixed communication
system, whereas the endocrine system supplied hor-
THE TARGET CELL CONCEPT
mones, which are mobile messages. In fact, there is a re-
markable convergence of these regulatory systems. For
There are about 200 types of differentiated cells in hu-
example, neural regulation of the endocrine system is
mans. Only a few produce hormones, but virtually all of
important in the production and secretion of some hor-
the 75 trillion cells in a human are targets of one or
mones; many neurotransmitters resemble hormones in
more of the over 50 known hormones. The concept of
their synthesis, transport, and mechanism of action; and
the target cell is a useful way of looking at hormone ac-
many hormones are synthesized in the nervous system.
tion. It was thought that hormones affected a single cell
The word “hormone” is derived from a Greek term that
type—or only a few kinds of cells—and that a hormone
means to arouse to activity. As classically defined, a hor-
elicited a unique biochemical or physiologic action. We
mone is a substance that is synthesized in one organ and
now know that a given hormone can affect several dif-
transported by the circulatory system to act on another
ferent cell types; that more than one hormone can affect
tissue. However, this original description is too restric-
a given cell type; and that hormones can exert many dif-
434
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
435
ferent effects in one cell or in different cells. With the
Table 42-2. Determinants of the target
discovery of specific cell-surface and intracellular hor-
cell response.
mone receptors, the definition of a target has been ex-
panded to include any cell in which the hormone (lig-
The number, relative activity, and state of occupancy of the
and) binds to its receptor, whether or not a biochemical
specific receptors on the plasma membrane or in the
or physiologic response has yet been determined.
cytoplasm or nucleus.
Several factors determine the response of a target cell
The metabolism (activation or inactivation) of the hormone in
to a hormone. These can be thought of in two general
the target cell.
ways: (1) as factors that affect the concentration of the
The presence of other factors within the cell that are neces-
hormone at the target cell (see Table 42-1) and (2) as
sary for the hormone response.
factors that affect the actual response of the target cell
Up- or down-regulation of the receptor consequent to the
to the hormone (see Table 42-2).
interaction with the ligand.
Postreceptor desensitzation of the cell, including down-
regulation of the receptor.
HORMONE RECEPTORS ARE
OF CENTRAL IMPORTANCE
Receptors Discriminate Precisely
logically relevant: (1) binding should be specific, ie, dis-
placeable by agonist or antagonist; (2) binding should
One of the major challenges faced in making the hor-
be saturable; and (3) binding should occur within the
mone-based communication system work is illustrated
concentration range of the expected biologic response.
in Figure 42-1. Hormones are present at very low con-
centrations in the extracellular fluid, generally in the
range of 10-15 to 10-9 mol/L. This concentration is
Both Recognition & Coupling
much lower than that of the many structurally similar
Domains Occur on Receptors
molecules (sterols, amino acids, peptides, proteins) and
All receptors have at least two functional domains. A
other molecules that circulate at concentrations in the
recognition domain binds the hormone ligand and a
10-5 to 10-3 mol/L range. Target cells, therefore, must
second region generates a signal that couples hormone
distinguish not only between different hormones pre-
recognition to some intracellular function. Coupling
sent in small amounts but also between a given hor-
(signal transduction) occurs in two general ways.
mone and the 106- to 109-fold excess of other similar
Polypeptide and protein hormones and the cate-
molecules. This high degree of discrimination is pro-
cholamines bind to receptors located in the plasma
vided by cell-associated recognition molecules called re-
membrane and thereby generate a signal that regulates
ceptors. Hormones initiate their biologic effects by
various intracellular functions, often by changing the
binding to specific receptors, and since any effective
activity of an enzyme. In contrast, steroid, retinoid, and
control system also must provide a means of stopping a
thyroid hormones interact with intracellular receptors,
response, hormone-induced actions generally terminate
and it is this ligand-receptor complex that directly pro-
when the effector dissociates from the receptor.
vides the signal, generally to specific genes whose rate of
A target cell is defined by its ability to selectively
transcription is thereby affected.
bind a given hormone to its cognate receptor. Several
The domains responsible for hormone recognition
biochemical features of this interaction are important in
and signal generation have been identified in the pro-
order for hormone-receptor interactions to be physio-
tein polypeptide and catecholamine hormone receptors.
Steroid, thyroid, and retinoid hormone receptors have
several functional domains: one site binds the hormone;
Table 42-1. Determinants of the concentration
another binds to specific DNA regions; a third is in-
of a hormone at the target cell.
volved in the interaction with other coregulator pro-
teins that result in the activation (or repression) of gene
The rate of synthesis and secretion of the hormones.
transcription; and a fourth may specify binding to one
The proximity of the target cell to the hormone source (dilu-
or more other proteins that influence the intracellular
tion effect).
trafficking of the receptor.
The dissociation constants of the hormone with specific
The dual functions of binding and coupling ulti-
plasma transport proteins (if any).
mately define a receptor, and it is the coupling of hor-
The conversion of inactive or suboptimally active forms of the
mone binding to signal transduction—so-called recep-
hormone into the fully active form.
tor-effector coupling—that provides the first step in
The rate of clearance from plasma by other tissues or by
amplification of the hormonal response. This dual pur-
digestion, metabolism, or excretion.
pose also distinguishes the target cell receptor from the
436
/
CHAPTER 42
◆
❁
❁
✴
❖
✪
❁
◗
❉
Figure 42-1. Specificity and selectivity of
✧
◆
hormone receptors. Many different molecules
❙
✪
✴
❙
ECF
circulate in the extracellular fluid (ECF), but
✧
❃
❃
content
❍
only a few are recognized by hormone recep-
✧
❍
◗
tors. Receptors must select these molecules
❉
❖
✪
❁
from among high concentrations of the other
❉
❁
molecules. This simplified drawing shows that
Hormone
Receptor
a cell may have no hormone receptors (1),
have one receptor (2+5+6), have receptors for
1
2
3
4
5
6
Cell types several hormones (3), or have a receptor but
no hormone in the vicinity (4).
plasma carrier proteins that bind hormone but do not
HORMONES CAN BE CLASSIFIED
generate a signal (see Table 42-6).
IN SEVERAL WAYS
Hormones can be classified according to chemical com-
Receptors Are Proteins
position, solubility properties, location of receptors,
Several classes of peptide hormone receptors have been
and the nature of the signal used to mediate hormonal
defined. For example, the insulin receptor is a het-
action within the cell. A classification based on the last
erotetramer (α2β2) linked by multiple disulfide bonds
two properties is illustrated in Table 42-3, and general
in which the extracellular α subunit binds insulin and
features of each group are illustrated in Table 42-4.
the membrane-spanning β subunit transduces the sig-
The hormones in group I are lipophilic. After secre-
nal through the tyrosine protein kinase domain located
tion, these hormones associate with plasma transport or
in the cytoplasmic portion of this polypeptide. The re-
carrier proteins, a process that circumvents the problem
ceptors for insulin-like growth factor I (IGF-I) and
of solubility while prolonging the plasma half-life of the
epidermal growth factor (EGF) are generally similar in
hormone. The relative percentages of bound and free
structure to the insulin receptor. The growth hormone
hormone are determined by the binding affinity and
and prolactin receptors also span the plasma mem-
binding capacity of the transport protein. The free hor-
brane of target cells but do not contain intrinsic pro-
mone, which is the biologically active form, readily tra-
tein kinase activity. Ligand binding to these receptors,
verses the lipophilic plasma membrane of all cells and
however, results in the association and activation of a
encounters receptors in either the cytosol or nucleus of
completely different protein kinase pathway, the Jak-
target cells. The ligand-receptor complex is assumed to
Stat pathway. Polypeptide hormone and catecho-
be the intracellular messenger in this group.
lamine receptors, which transduce signals by altering
The second major group consists of water-soluble
the rate of production of cAMP through G-proteins,
hormones that bind to the plasma membrane of the tar-
are characterized by the presence of seven domains that
get cell. Hormones that bind to the surfaces of cells
span the plasma membrane. Protein kinase activation
communicate with intracellular metabolic processes
and the generation of cyclic AMP,
(cAMP, 3′5′-
through intermediary molecules called second messen-
adenylic acid; see Figure 20-5) is a downstream action
gers (the hormone itself is the first messenger), which
of this class of receptor (see Chapter 43 for further de-
are generated as a consequence of the ligand-receptor
tails).
interaction. The second messenger concept arose from
A comparison of several different steroid receptors
an observation that epinephrine binds to the plasma
with thyroid hormone receptors revealed a remarkable
membrane of certain cells and increases intracellular
conservation of the amino acid sequence in certain re-
cAMP. This was followed by a series of experiments in
gions, particularly in the DNA-binding domains. This
which cAMP was found to mediate the effects of many
led to the realization that receptors of the steroid or
hormones. Hormones that clearly employ this mecha-
thyroid type are members of a large superfamily of nu-
nism are shown in group II.A of Table 42-3. To date,
clear receptors. Many related members of this family
only one hormone, atrial natriuretic factor (ANF), uses
have no known ligand at present and thus are called or-
cGMP as its second messenger, but other hormones
phan receptors. The nuclear receptor superfamily plays
will probably be added to group II.B. Several hor-
a critical role in the regulation of gene transcription by
mones, many of which were previously thought to af-
hormones, as described in Chapter 43.
fect cAMP, appear to use ionic calcium
(Ca2+) or
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
437
Table 42-3. Classification of hormones by
Table 42-4. General features of hormone classes.
mechanism of action.
Group I
Group II
I. Hormones that bind to intracellular receptors
Types
Steroids, iodothyro-
Polypeptides, proteins,
Androgens
nines, calcitriol,
glycoproteins, cate-
Calcitriol (1,25[OH]2-D3)
retinoids
cholamines
Estrogens
Glucocorticoids
Solubility
Lipophilic
Hydrophilic
Mineralocorticoids
Transport
Yes
No
Progestins
proteins
Retinoic acid
Thyroid hormones (T3 and T4)
Plasma half-
Long (hours to
Short (minutes)
II. Hormones that bind to cell surface receptors
life
days)
A. The second messenger is cAMP:
Receptor
Intracellular
Plasma membrane
α2-Adrenergic catecholamines
β-Adrenergic catecholamines
Mediator
Receptor-hormone
cAMP, cGMP, Ca2+,
Adrenocorticotropic hormone
complex
metabolites of complex
Antidiuretic hormone
phosphoinositols,
Calcitonin
kinase cascades
Chorionic gonadotropin, human
Corticotropin-releasing hormone
Follicle-stimulating hormone
metabolites of complex phosphoinositides (or both) as
Glucagon
the intracellular signal. These are shown in group II.C
Lipotropin
of the table. The intracellular messenger for group II.D
Luteinizing hormone
is a protein kinase-phosphatase cascade. Several of these
Melanocyte-stimulating hormone
Parathyroid hormone
have been identified, and a given hormone may use
Somatostatin
more than one kinase cascade. A few hormones fit into
Thyroid-stimulating hormone
more than one category, and assignments change as
B.
The second messenger is cGMP:
new information is brought forward.
Atrial natriuretic factor
Nitric oxide
C.
The second messenger is calcium or phosphatidyl-
DIVERSITY OF THE ENDOCRINE SYSTEM
inositols (or both):
Acetylcholine (muscarinic)
Hormones Are Synthesized in a
α1-Adrenergic catecholamines
Variety of Cellular Arrangements
Angiotensin II
Antidiuretic hormone (vasopressin)
Hormones are synthesized in discrete organs designed
Cholecystokinin
solely for this specific purpose, such as the thyroid (tri-
Gastrin
iodothyronine), adrenal (glucocorticoids and mineralo-
Gonadotropin-releasing hormone
corticoids), and the pituitary (TSH, FSH, LH, growth
Oxytocin
hormone, prolactin, ACTH). Some organs are designed
Platelet-derived growth factor
to perform two distinct but closely related functions.
Substance P
For example, the ovaries produce mature oocytes and
Thyrotropin-releasing hormone
the reproductive hormones estradiol and progesterone.
D.
The second messenger is a kinase or phosphatase
The testes produce mature spermatozoa and testos-
cascade:
terone. Hormones are also produced in specialized cells
Chorionic somatomammotropin
within other organs such as the small intestine
Epidermal growth factor
(glucagon-like peptide), thyroid (calcitonin), and kid-
Erythropoietin
ney (angiotensin II). Finally, the synthesis of some hor-
Fibroblast growth factor
mones requires the parenchymal cells of more than one
Growth hormone
organ—eg, the skin, liver, and kidney are required for
Insulin
the production of 1,25(OH)2-D3 (calcitriol). Examples
Insulin-like growth factors I and II
of this diversity in the approach to hormone synthesis,
Nerve growth factor
each of which has evolved to fulfill a specific purpose,
Platelet-derived growth factor
Prolactin
are discussed below.
438
/
CHAPTER 42
Hormones Are Chemically Diverse
MANY HORMONES ARE MADE
FROM CHOLESTEROL
Hormones are synthesized from a wide variety of chem-
ical building blocks. A large series is derived from cho-
Adrenal Steroidogenesis
lesterol. These include the glucocorticoids, mineralo-
corticoids, estrogens, progestins, and
1,25(OH)2-D3
The adrenal steroid hormones are synthesized from
(see Figure 42-2). In some cases, a steroid hormone is
cholesterol. Cholesterol is mostly derived from the
the precursor molecule for another hormone. For ex-
plasma, but a small portion is synthesized in situ from
ample, progesterone is a hormone in its own right but
acetyl-CoA via mevalonate and squalene. Much of the
is also a precursor in the formation of glucocorticoids,
cholesterol in the adrenal is esterified and stored in cy-
mineralocorticoids, testosterone, and estrogens. Testos-
toplasmic lipid droplets. Upon stimulation of the
terone is an obligatory intermediate in the biosynthesis
adrenal by ACTH, an esterase is activated, and the free
of estradiol and in the formation of dihydrotestosterone
cholesterol formed is transported into the mitochon-
(DHT). In these examples, described in detail below,
drion, where a cytochrome P450 side chain cleav-
the final product is determined by the cell type and the
age enzyme (P450scc) converts cholesterol to preg-
associated set of enzymes in which the precursor exists.
nenolone. Cleavage of the side chain involves sequential
The amino acid tyrosine is the starting point in the
hydroxylations, first at C22 and then at C20, followed by
synthesis of the catecholamines and of the thyroid hor-
side chain cleavage (removal of the six-carbon fragment
mones tetraiodothyronine (thyroxine; T4) and triiodo-
isocaproaldehyde) to give the 21-carbon steroid (Figure
thyronine (T3) (Figure 42-2). T3 and T4 are unique in
42-3, top). An ACTH-dependent steroidogenic acute
that they require the addition of iodine (as I−) for bioac-
regulatory (StAR) protein is essential for the transport
tivity. Because dietary iodine is very scarce in many
of cholesterol to P450scc in the inner mitochondrial
parts of the world, an intricate mechanism for accumu-
membrane.
lating and retaining I− has evolved.
All mammalian steroid hormones are formed from
Many hormones are polypeptides or glycoproteins.
cholesterol via pregnenolone through a series of reac-
These range in size from thyrotropin-releasing hor-
tions that occur in either the mitochondria or endoplas-
mone (TRH), a tripeptide, to single-chain polypeptides
mic reticulum of the adrenal cell. Hydroxylases that re-
like adrenocorticotropic hormone (ACTH; 39 amino
quire molecular oxygen and NADPH are essential, and
acids), parathyroid hormone (PTH; 84 amino acids),
dehydrogenases, an isomerase, and a lyase reaction are
and growth hormone (GH; 191 amino acids) (Figure
also necessary for certain steps. There is cellular speci-
42-2). Insulin is an AB chain heterodimer of 21 and 30
ficity in adrenal steroidogenesis. For instance,
18-
amino acids, respectively. Follicle-stimulating hormone
hydroxylase and
19-hydroxysteroid dehydrogenase,
(FSH), luteinizing hormone (LH), thyroid-stimulating
which are required for aldosterone synthesis, are found
hormone (TSH), and chorionic gonadotropin (CG) are
only in the zona glomerulosa cells (the outer region of
glycoprotein hormones of αβ heterodimeric structure.
the adrenal cortex), so that the biosynthesis of this min-
The α chain is identical in all of these hormones, and
eralocorticoid is confined to this region. A schematic
distinct β chains impart hormone uniqueness. These
representation of the pathways involved in the synthesis
hormones have a molecular mass in the range of 25-30
of the three major classes of adrenal steroids is pre-
kDa depending on the degree of glycosylation and the
sented in Figure 42-4. The enzymes are shown in the
length of the β chain.
rectangular boxes, and the modifications at each step
are shaded.
Hormones Are Synthesized & Modified
A. MINERALOCORTICOID SYNTHESIS
For Full Activity in a Variety of Ways
Synthesis of aldosterone follows the mineralocorticoid
pathway and occurs in the zona glomerulosa. Preg-
Some hormones are synthesized in final form and se-
nenolone is converted to progesterone by the action of
creted immediately. Included in this class are the hor-
two smooth endoplasmic reticulum enzymes,
3 -
mones derived from cholesterol. Others such as the cat-
hydroxysteroid dehydrogenase (3
-OHSD) and
5,4-
echolamines are synthesized in final form and stored in
isomerase. Progesterone is hydroxylated at the C21 posi-
the producing cells. Others are synthesized from pre-
tion to form 11-deoxycorticosterone (DOC), which is an
cursor molecules in the producing cell, then are
active (Na+-retaining) mineralocorticoid. The next hy-
processed and secreted upon a physiologic cue (insulin).
droxylation, at C11, produces corticosterone, which has
Finally, still others are converted to active forms from
glucocorticoid activity and is a weak mineralocorticoid (it
precursor molecules in the periphery (T3 and DHT).
has less than 5% of the potency of aldosterone). In some
All of these examples are discussed in more detail
species (eg, rodents), it is the most potent glucocorticoid.
below.
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
439
A. CHOLESTEROL DERIVATIVES
CH2OH
CH3
OH
OH
C O
C
O
OH
OH
CH2
HO
OH
HO
O
O
HO
17ß-Estradiol
Testosterone
Cortisol
Progesterone
1,25(OH)2-D3
B. TYROSINE DERIVATIVES
HO
H
I
I
O
H
OH
O
CH2CH
COOH
HO
C
C
NH2
I
NH2
H H
T3
Norepinephrine
HO
H
I
I
O
H CH3
OH
O
CH2CH
COOH
HO
C
C
NH
I
I
NH2
H H
T4
Epinephrine
C. PEPTIDES OF VARIOUS SIZES
1
2
3
4
5
6
7
8
9
10
11
12
Ser
Tyr
Ser
Mert
G
lu
H
ls
Phe
Arg
T
p
G
ly
Lys
P
ro
13
1
2
3
Conserved region; required for full biologic activity
Val
Glu Hls Pro
24
23
22
21
20
19
18
17
16
G
ly
(pyro)
NH2
14
25
Pro
Tyr
Val
Lys Val
Pro
Arg
Arg
Lys
Lys
Asp
26
Ala
27
Gly
Variable region; not required for biologic activity
TRH
Glu
Asp
G
ln
Ser A
la G
lu A
la
Phe Pro
Leu Glu
Phe
28
29
30
31
32
33
34
35
36
37
38
39
Structure of human ACTH.
D. GLYCOPROTEINS (TSH, FSH, LH)
ACTH
common
α
subunits
unique
β
subunits
Figure 42-2. Chemical diversity of hormones. A. Cholesterol derivatives. B. Tyrosine derivatives.
C. Peptides of various sizes D. Glycoproteins (TSH, FSH, LH) with common α subunits and unique β
subunits.
440
/
CHAPTER 42
Cholesterol side chain cleavage
21
CH3
C
C
C
C
C
C
C
20
C
O
C
18
H
12
17
C
ACTH
11
13
16
C
D
+
C C C C
(cAMP)
19
14
15
1
9
P450scc
2
10
8
O
C
A
B
3
5
7
HO
HO
4
6
Cholesterol
Pregnenolone + isocaproaldehyde
Basic steroid hormone structures
CH2OH
CH3
OH
OH
C
O
C
O
HO
OH
HO
O
O
O
17β—Estradiol
Testosterone
Cortisol
Progesterone
Estrane group (C18)
Androstane group (C19)
Pregnane group (C21)
Figure 42-3. Cholesterol side-chain cleavage and basic steroid hormone structures. The basic sterol rings are iden-
tified by the letters A-D. The carbon atoms are numbered 1-21 starting with the A ring. Note that the estrane group
has 18 carbons (C18), etc.
C21 hydroxylation is necessary for both mineralocorticoid
costerone or aldosterone, depending on the cell type).
and glucocorticoid activity, but most steroids with a C17
17α-Hydroxylase is a smooth endoplasmic reticulum
hydroxyl group have more glucocorticoid and less miner-
enzyme that acts upon either progesterone or, more
alocorticoid action. In the zona glomerulosa, which does
commonly, pregnenolone. 17α-Hydroxyprogesterone is
not have the smooth endoplasmic reticulum enzyme
hydroxylated at C21 to form 11-deoxycortisol, which is
17α-hydroxylase, a mitochondrial 18-hydroxylase is pres-
then hydroxylated at C11 to form cortisol, the most po-
ent. The 18-hydroxylase (aldosterone synthase) acts on
tent natural glucocorticoid hormone in humans. 21-Hy-
corticosterone to form 18-hydroxycorticosterone, which
droxylase is a smooth endoplasmic reticulum enzyme,
is changed to aldosterone by conversion of the 18-alcohol
whereas 11β-hydroxylase is a mitochondrial enzyme.
to an aldehyde. This unique distribution of enzymes and
Steroidogenesis thus involves the repeated shuttling of
the special regulation of the zona glomerulosa by K+ and
substrates into and out of the mitochondria.
angiotensin II have led some investigators to suggest that,
in addition to the adrenal being two glands, the adrenal
C. ANDROGEN SYNTHESIS
cortex is actually two separate organs.
The major androgen or androgen precursor produced by
B. GLUCOCORTICOID SYNTHESIS
the adrenal cortex is dehydroepiandrosterone (DHEA).
Most 17-hydroxypregnenolone follows the glucocorticoid
Cortisol synthesis requires three hydroxylases located in
pathway, but a small fraction is subjected to oxidative fis-
the fasciculata and reticularis zones of the adrenal cortex
sion and removal of the two-carbon side chain through
that act sequentially on the C17, C21, and C11 positions.
the action of 17,20-lyase. The lyase activity is actually
The first two reactions are rapid, while C11 hydroxyla-
part of the same enzyme (P450c17) that catalyzes 17α-
tion is relatively slow. If the C11 position is hydroxylated
hydroxylation. This is therefore a dual function protein.
first, the action of 17
-hydroxylase is impeded and the
The lyase activity is important in both the adrenals and
mineralocorticoid pathway is followed (forming corti-
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
441
Cholesterol
SCC
CH3
CH3
C O
C O
O
— OH
HO
HO
HO
Pregnenolone
17-Hydroxypregnenolone
Dehydroepiandrosterone
3β-HYDROXYSTEROID DEHYDROGENASE: ∆5,4 ISOMERASE
CH3
CH3
C O
C O
O
— OH
O
O
O
Progesterone
17-Hydroxyprogesterone
∆4 ANDROSTENE-3,17-DION
21-HYDROXYLASE
CH2OH
CH2OH
C O
C O
— OH
O
O
11-Deoxycorticosterone
11-Deoxycortisol
11β-HYDROXYLASE
CH2OH
CH2OH
C O
C O
HO
HO
— OH
O
O
Corticosterone
CORTISOL
18-HYDROXYLASE
18-HYDROXYDEHYDROGENASE
CH2OH
O
C O
H
C
HO
O
ALDOSTERONE
Figure 42-4. Pathways involved in the synthesis of the three major classes of
adrenal steroids (mineralocorticoids, glucocorticoids, and androgens). Enzymes
are shown in the rectangular boxes, and the modifications at each step are
shaded. Note that the 17α-hydroxylase and 17,20-lyase activities are both part of
one enzyme, designated P450c17. (Slightly modified and reproduced, with permis-
sion, from Harding BW: In: Endocrinology, vol 2. DeGroot LJ [editor]. Grune & Stratton,
1979.)
442
/
CHAPTER 42
the gonads and acts exclusively on 17α-hydroxy-contain-
are generally inactive or less active than the parent com-
ing molecules. Adrenal androgen production increases
pound. Metabolism by the second pathway, which is less
markedly if glucocorticoid biosynthesis is impeded by the
efficient, occurs primarily in target tissues and produces
lack of one of the hydroxylases
(adrenogenital syn-
the potent metabolite dihydrotestosterone (DHT).
drome). DHEA is really a prohormone, since the actions
The most significant metabolic product of testos-
of 3β-OHSD and ∆5,4-isomerase convert the weak andro-
terone is DHT, since in many tissues, including
gen DHEA into the more potent androstenedione.
prostate, external genitalia, and some areas of the skin,
Small amounts of androstenedione are also formed in the
this is the active form of the hormone. The plasma con-
adrenal by the action of the lyase on 17α-hydroxyproges-
tent of DHT in the adult male is about one-tenth that
terone. Reduction of androstenedione at the C17 position
of testosterone, and approximately 400 µg of DHT is
results in the formation of testosterone, the most potent
produced daily as compared with about 5 mg of testos-
adrenal androgen. Small amounts of testosterone are pro-
terone. About 50-100 µg of DHT are secreted by the
duced in the adrenal by this mechanism, but most of this
testes. The rest is produced peripherally from testos-
conversion occurs in the testes.
terone in a reaction catalyzed by the NADPH-depen-
dent 5
-reductase
(Figure
42-6). Testosterone can
thus be considered a prohormone, since it is converted
Testicular Steroidogenesis
into a much more potent compound (dihydrotestos-
Testicular androgens are synthesized in the interstitial
terone) and since most of this conversion occurs outside
tissue by the Leydig cells. The immediate precursor of
the testes. Some estradiol is formed from the peripheral
the gonadal steroids, as for the adrenal steroids, is cho-
aromatization of testosterone, particularly in males.
lesterol. The rate-limiting step, as in the adrenal, is de-
livery of cholesterol to the inner membrane of the mito-
Ovarian Steroidogenesis
chondria by the transport protein StAR. Once in the
proper location, cholesterol is acted upon by the side
The estrogens are a family of hormones synthesized in a
chain cleavage enzyme P450scc. The conversion of cho-
variety of tissues. 17β-Estradiol is the primary estrogen
lesterol to pregnenolone is identical in adrenal, ovary,
of ovarian origin. In some species, estrone, synthesized
and testis. In the latter two tissues, however, the reac-
in numerous tissues, is more abundant. In pregnancy,
tion is promoted by LH rather than ACTH.
relatively more estriol is produced, and this comes from
The conversion of pregnenolone to testosterone re-
the placenta. The general pathway and the subcellular
quires the action of five enzyme activities contained in
localization of the enzymes involved in the early steps
three proteins: (1) 3β-hydroxysteroid dehydrogenase (3β-
of estradiol synthesis are the same as those involved in
OHSD) and ∆5,4-isomerase; (2) 17α-hydroxylase and
androgen biosynthesis. Features unique to the ovary are
17,20-lyase; and (3) 17β-hydroxysteroid dehydrogenase
illustrated in Figure 42-7.
(17β-OHSD). This sequence, referred to as the proges-
Estrogens are formed by the aromatization of andro-
terone (or
4) pathway, is shown on the right side of Fig-
gens in a complex process that involves three hydroxyla-
ure 42-5. Pregnenolone can also be converted to testos-
tion steps, each of which requires O2 and NADPH. The
terone by the dehydroepiandrosterone (or
5) pathway,
aromatase enzyme complex is thought to include a
which is illustrated on the left side of Figure 42-5. The ∆5
P450 monooxygenase. Estradiol is formed if the sub-
route appears to be most used in human testes.
strate of this enzyme complex is testosterone, whereas es-
The five enzyme activities are localized in the micro-
trone results from the aromatization of androstenedione.
somal fraction in rat testes, and there is a close func-
The cellular source of the various ovarian steroids has
tional association between the activities of 3β-OHSD
been difficult to unravel, but a transfer of substrates be-
and ∆5,4-isomerase and between those of a 17α-hydrox-
tween two cell types is involved. Theca cells are the source
ylase and 17,20-lyase. These enzyme pairs, both con-
of androstenedione and testosterone. These are converted
tained in a single protein, are shown in the general reac-
by the aromatase enzyme in granulosa cells to estrone and
tion sequence in Figure 42-5.
estradiol, respectively. Progesterone, a precursor for all
steroid hormones, is produced and secreted by the corpus
luteum as an end-product hormone because these cells do
Dihydrotestosterone Is Formed From
not contain the enzymes necessary to convert proges-
Testosterone in Peripheral Tissues
terone to other steroid hormones (Figure 42-8).
Testosterone is metabolized by two pathways. One in-
Significant amounts of estrogens are produced by
volves oxidation at the 17 position, and the other in-
the peripheral aromatization of androgens. In human
volves reduction of the A ring double bond and the 3-ke-
males, the peripheral aromatization of testosterone to
tone. Metabolism by the first pathway occurs in many
estradiol (E2) accounts for 80% of the production of
tissues, including liver, and produces 17-ketosteroids that
the latter. In females, adrenal androgens are important
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
443
CH3
CH3
C O
C O
HO
HO
Pregnenolone
Progesterone
17α-HYDROXYLASE*
17α-HYDROXYLASE*
CH3
CH3
C O
C O
OH
OH
HO
O
17α-Hydroxypregnenolone
17α-Hydroxyprogesterone
17,20-LYASE*
17,20-LYASE*
O
O
HO
O
Dehydroepiandrosterone
Androstenedione
17β-HYDROXYSTEROID
17β-HYDROXYSTEROID
DEHYDROGENASE
DEHYDROGENASE
Figure 42-5. Pathways of testos-
terone biosynthesis. The pathway on
the left side of the figure is called the ∆5
OH
OH
or dehydroepiandrosterone pathway;
the pathway on the right side is called
the ∆4 or progesterone pathway. The as-
terisk indicates that the 17α-hydroxy-
lase and 17,20-lyase activities reside in a
HO
O
single protein, P450c17.
∆5-Androstenediol
TESTOSTERONE
444
/
CHAPTER 42
OH
OH
5α-REDUCTASE
NADPH
O
O
H
Testosterone
DIHYDROTESTOSTERONE (DHT)
Figure 42-6. Dihydrotestosterone is formed from testosterone through action of the
enzyme 5α-reductase.
Cholesterol
Pregnenolone
17α-Hydroxypregnenolone
Dehydroepiandrosterone
Progesterone
17α-Hydroxyprogesterone
Androstenedione
Testosterone
AROMATASE
AROMATASE
O
OH
Other
metabolites
HO
HO
ESTRONE (E1)
17β-ESTRADIOL (E2)
16α-Hydroxylase
Other metabolites
OH
OH
HO
Estriol
Figure 42-7. Biosynthesis of estrogens. (Slightly modified and reproduced, with permission, from Ganong
WF: Review of Medical Physiology, 20th ed. McGraw-Hill, 2001.)
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
445
Acetate
most of the precursor for 1,25(OH)2-D3 synthesis is
produced in the malpighian layer of the epidermis from
Cholesterol
7-dehydrocholesterol in an ultraviolet light-mediated,
CH3
nonenzymatic photolysis reaction. The extent of this
conversion is related directly to the intensity of the ex-
C O
posure and inversely to the extent of pigmentation in
the skin. There is an age-related loss of 7-dehydrocho-
lesterol in the epidermis that may be related to the neg-
ative calcium balance associated with old age.
B. LIVER
HO
A specific transport protein called the vitamin D-bind-
Pregnenolone
ing protein binds vitamin D3 and its metabolites and
moves vitamin D3 from the skin or intestine to the
CH3
liver, where it undergoes
25-hydroxylation, the first
obligatory reaction in the production of 1,25(OH)2-
C O
D3. 25-Hydroxylation occurs in the endoplasmic retic-
ulum in a reaction that requires magnesium, NADPH,
molecular oxygen, and an uncharacterized cytoplasmic
factor. Two enzymes are involved: an NADPH-depen-
dent cytochrome P450 reductase and a cytochrome
P450. This reaction is not regulated, and it also occurs
with low efficiency in kidney and intestine. The
O
25(OH)2-D3 enters the circulation, where it is the
Progesterone
major form of vitamin D found in plasma, and is trans-
ported to the kidney by the vitamin D-binding protein.
Figure 42-8.
Biosynthesis of progesterone in the
corpus luteum.
C. KIDNEY
25(OH)2-D3 is a weak agonist and must be modified
by hydroxylation at position C1 for full biologic activ-
substrates, since as much as 50% of the E2 produced
ity. This is accomplished in mitochondria of the renal
during pregnancy comes from the aromatization of an-
proximal convoluted tubule by a three-component
drogens. Finally, conversion of androstenedione to
monooxygenase reaction that requires NADPH, Mg2+,
estrone is the major source of estrogens in post-
molecular oxygen, and at least three enzymes: (1) a
menopausal women. Aromatase activity is present in
flavoprotein, renal ferredoxin reductase; (2) an iron sul-
adipose cells and also in liver, skin, and other tissues.
fur protein, renal ferredoxin; and (3) cytochrome P450.
Increased activity of this enzyme may contribute to the
This system produces 1,25(OH)2-D3, which is the most
“estrogenization” that characterizes such diseases as cir-
potent naturally occurring metabolite of vitamin D.
rhosis of the liver, hyperthyroidism, aging, and obesity.
CATECHOLAMINES & THYROID
1,25(OH)2-D3 (Calcitriol) Is Synthesized
HORMONES ARE MADE FROM TYROSINE
From a Cholesterol Derivative
Catecholamines Are Synthesized in Final
1,25(OH)2-D3 is produced by a complex series of enzy-
Form & Stored in Secretion Granules
matic reactions that involve the plasma transport of pre-
cursor molecules to a number of different tissues (Figure
Three amines—dopamine, norepinephrine, and epi-
42-9). One of these precursors is vitamin D—really not
nephrine—are synthesized from tyrosine in the chro-
a vitamin, but this common name persists. The active
maffin cells of the adrenal medulla. The major product
molecule, 1,25(OH)2-D3, is transported to other organs
of the adrenal medulla is epinephrine. This compound
where it activates biologic processes in a manner similar
constitutes about 80% of the catecholamines in the
to that employed by the steroid hormones.
medulla, and it is not made in extramedullary tissue. In
contrast, most of the norepinephrine present in organs
A. SKIN
innervated by sympathetic nerves is made in situ (about
Small amounts of the precursor for 1,25(OH)2-D3 syn-
80% of the total), and most of the rest is made in other
thesis are present in food (fish liver oil, egg yolk), but
nerve endings and reaches the target sites via the circu-
446
/
CHAPTER 42
Sunlight
7-Dehydrocholesterol
Previtamin D3
Vitamin D3
25-Hydroxylase
SKIN
LIVER
Other
25-Hydroxycholecalciferol (25[OH]-D3)
metabolites
24-Hydroxylase
1 α-Hydroxylase
24,25(OH)2-D3
KIDNEY
1,25(OH)2-D3
1,24,25(OH)3-D3
24
27
25
OH
26
CH2
CH2
HO
HO
HO
OH
7-Dehydrocholesterol
Vitamin D3
1,25(OH)2-D3
Figure 42-9. Formation and hydroxylation of vitamin D3. 25-Hydroxylation takes place in the liver, and the
other hydroxylations occur in the kidneys. 25,26(OH)2-D3 and 1,25,26(OH)3-D3 are probably formed as well. The
formulas of 7-dehydrocholesterol, vitamin D3, and 1,25(OH)2-D3 are also shown. (Modified and reproduced, with
permission, from Ganong WF: Review of Medical Physiology, 20th ed. McGraw-Hill, 2001.)
lation. Epinephrine and norepinephrine may be pro-
As the rate-limiting enzyme, tyrosine hydroxylase is regu-
duced and stored in different cells in the adrenal
lated in a variety of ways. The most important mecha-
medulla and other chromaffin tissues.
nism involves feedback inhibition by the catecholamines,
The conversion of tyrosine to epinephrine requires
which compete with the enzyme for the pteridine cofac-
four sequential steps: (1) ring hydroxylation; (2) decar-
tor. Catecholamines cannot cross the blood-brain barrier;
boxylation; (3) side chain hydroxylation to form norepi-
hence, in the brain they must be synthesized locally. In
nephrine; and (4) N-methylation to form epinephrine.
certain central nervous system diseases (eg, Parkinson’s
The biosynthetic pathway and the enzymes involved are
disease), there is a local deficiency of dopamine synthesis.
illustrated in Figure 42-10.
L-Dopa, the precursor of dopamine, readily crosses the
blood-brain barrier and so is an important agent in the
A. TYROSINE HYDROXYLASE IS RATE-LIMITING
treatment of Parkinson’s disease.
FOR CATECHOLAMINE BIOSYNTHESIS
B. DOPA DECARBOXYLASE IS PRESENT IN ALL TISSUES
Tyrosine is the immediate precursor of catecholamines,
and tyrosine hydroxylase is the rate-limiting enzyme in
This soluble enzyme requires pyridoxal phosphate for
catecholamine biosynthesis. Tyrosine hydroxylase is
the conversion of L-dopa to 3,4-dihydroxyphenylethyl-
found in both soluble and particle-bound forms only in
amine (dopamine). Compounds that resemble L-dopa,
tissues that synthesize catecholamines; it functions as an
such as α-methyldopa, are competitive inhibitors of
oxidoreductase, with tetrahydropteridine as a cofactor, to
this reaction. α-Methyldopa is effective in treating
convert L-tyrosine to L-dihydroxyphenylalanine (L-dopa).
some kinds of hypertension.
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
447
O
the conversion of dopamine to norepinephrine occurs
in this organelle.
H
C
OH
D. PHENYLETHANOLAMINE-N-METHYLTRANSFERASE
HO
C C
NH2
(PNMT) CATALYZES THE PRODUCTION
H H
OF EPINEPHRINE
Tyrosine
PNMT catalyzes the N-methylation of norepinephrine
TYROSINE
HYDROXYLASE
to form epinephrine in the epinephrine-forming cells
of the adrenal medulla. Since PNMT is soluble, it is as-
O
sumed that norepinephrine-to-epinephrine conversion
HO
H
C
OH
occurs in the cytoplasm. The synthesis of PNMT is in-
duced by glucocorticoid hormones that reach the
HO
C C
NH2
medulla via the intra-adrenal portal system. This special
H H
system provides for a 100-fold steroid concentration
Dopa
gradient over systemic arterial blood, and this high
DOPA
intra-adrenal concentration appears to be necessary for
DECARBOXYLASE
the induction of PNMT.
HO
T3 & T4 Illustrate the Diversity
H
H
in Hormone Synthesis
HO
C C
NH2
The formation of triiodothyronine (T3) and tetra-
H H
iodothyronine (thyroxine; T4) (see Figure 42-2) illus-
DOPAMINE
trates many of the principles of diversity discussed in
DOPAMINE
this chapter. These hormones require a rare element
β-HYDROXYLASE
(iodine) for bioactivity; they are synthesized as part of a
very large precursor molecule (thyroglobulin); they are
stored in an intracellular reservoir (colloid); and there is
HO
H
peripheral conversion of T4 to T3, which is a much
O
H
more active hormone.
HO
C C
NH2
The thyroid hormones T3 and T4 are unique in that
iodine (as iodide) is an essential component of both. In
H H
NOREPINEPHRINE
most parts of the world, iodine is a scarce component of
soil, and for that reason there is little in food. A com-
PNMT
plex mechanism has evolved to acquire and retain this
crucial element and to convert it into a form suitable
for incorporation into organic compounds. At the same
time, the thyroid must synthesize thyronine from tyro-
HO
H
O
H
CH3
sine, and this synthesis takes place in thyroglobulin
(Figure 42-11).
HO
C C
NH
Thyroglobulin is the precursor of T4 and T3. It is a
H H
large iodinated, glycosylated protein with a molecular
EPINEPHRINE
mass of 660 kDa. Carbohydrate accounts for 8-10% of
the weight of thyroglobulin and iodide for about
Figure 42-10. Biosynthesis of catecholamines.
0.2-1%, depending upon the iodine content in the
(PNMT, phenylethanolamine-N-methyltransferase.)
diet. Thyroglobulin is composed of two large subunits.
It contains 115 tyrosine residues, each of which is a po-
tential site of iodination. About
70% of the iodide
in thyroglobulin exists in the inactive precursors,
C. DOPAMINE
-HYDROXYLASE (DBH) CATALYZES
monoiodotyrosine (MIT) and diiodotyrosine (DIT),
THE CONVERSION OF DOPAMINE TO NOREPINEPHRINE
while 30% is in the iodothyronyl residues, T4 and T3.
DBH is a monooxygenase and uses ascorbate as an elec-
When iodine supplies are sufficient, the T4:T3 ratio is
tron donor, copper at the active site, and fumarate as
about 7:1. In iodine deficiency, this ratio decreases, as
modulator. DBH is in the particulate fraction of the
does the DIT:MIT ratio. Thyroglobulin, a large mole-
medullary cells, probably in the secretion granule; thus,
cule of about 5000 amino acids, provides the confor-
448
/
CHAPTER 42
FOLLICULAR SPACE WITH COLLOID
MIT
MIT
T3
MIT
DIT
DIT
DIT
DIT
Oxidation
Iodination*
Coupling*
DIT
T4
I +
+
Tgb
Tgb
Tgb
PEROXIDASE
MIT
MIT
MIT
DIT
H2O2
DIT
DIT
DIT
T4
-
I
Phagocytosis
O2
and
Tgb
pinocytosis
NADPH
NADP+
H+
Lysosomes
THYROID CELL
Tgb
Secondary
Tgb
lysosome
Tyrosine
Hydrolysis
Deiodination*
MIT
-
I
DEIODINASE
DIT
-
I
Concentration
T3, T4
Na+-K+ ATPase
Release
Trans-
porter
EXTRACELLULAR SPACE
-
T3, T4
I
Figure 42-11. Model of iodide metabolism in the thyroid follicle. A follicular cell is shown facing the follicular
lumen (top) and the extracellular space (at bottom). Iodide enters the thyroid primarily through a transporter
(bottom left). Thyroid hormone synthesis occurs in the follicular space through a series of reactions, many of
which are peroxidase-mediated. Thyroid hormones, stored in the colloid in the follicular space, are released from
thyroglobulin by hydrolysis inside the thyroid cell. (Tgb, thyroglobulin; MIT, monoiodotyrosine; DIT, diiodotyro-
sine; T3, triiodothyronine; T4, tetraiodothyronine.) Asterisks indicate steps or processes that are inherited enzyme
deficiencies which cause congenital goiter and often result in hypothyroidism.
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
449
mation required for tyrosyl coupling and iodide organi-
mones remain as integral parts of thyroglobulin until
fication necessary in the formation of the diaminoacid
the latter is degraded, as described above.
thyroid hormones. It is synthesized in the basal portion
A deiodinase removes I− from the inactive mono-
of the cell and moves to the lumen, where it is a storage
and diiodothyronine molecules in the thyroid. This
form of T3 and T4 in the colloid; several weeks’ supply
mechanism provides a substantial amount of the I− used
of these hormones exist in the normal thyroid. Within
in T3 and T4 biosynthesis. A peripheral deiodinase in
minutes after stimulation of the thyroid by TSH, col-
target tissues such as pituitary, kidney, and liver selec-
loid reenters the cell and there is a marked increase of
tively removes I− from the 5′ position of T4 to make T3
phagolysosome activity. Various acid proteases and
(see Figure 42-2), which is a much more active mole-
peptidases hydrolyze the thyroglobulin into its con-
cule. In this sense, T4 can be thought of as a prohor-
stituent amino acids, including T4 and T3, which are
mone, though it does have some intrinsic activity.
discharged from the basal portion of the cell (see Figure
42-11). Thyroglobulin is thus a very large prohor-
SEVERAL HORMONES ARE MADE FROM
mone.
LARGER PEPTIDE PRECURSORS
Formation of the critical disulfide bridges in insulin re-
Iodide Metabolism Involves
quires that this hormone be first synthesized as part of a
Several Discrete Steps
larger precursor molecule, proinsulin. This is conceptu-
The thyroid is able to concentrate I− against a strong
ally similar to the example of the thyroid hormones,
electrochemical gradient. This is an energy-dependent
which can only be formed in the context of a much
process and is linked to the Na+-K+ ATPase-dependent
larger molecule. Several other hormones are synthesized
thyroidal I− transporter. The ratio of iodide in thyroid
as parts of large precursor molecules, not because of
to iodide in serum (T:S ratio) is a reflection of the ac-
some special structural requirement but rather as a
tivity of this transporter. This activity is primarily con-
mechanism for controlling the available amount of the
trolled by TSH and ranges from 500:1 in animals
active hormone. PTH and angiotensin II are examples
chronically stimulated with TSH to 5:1 or less in hy-
of this type of regulation. Another interesting example
pophysectomized animals (no TSH). The T:S ratio in
is the POMC protein, which can be processed into
humans on a normal iodine diet is about 25:1.
many different hormones in a tissue-specific manner.
The thyroid is the only tissue that can oxidize I− to a
These examples are discussed in detail below.
higher valence state, an obligatory step in I− organifica-
tion and thyroid hormone biosynthesis. This step in-
Insulin Is Synthesized as a Preprohormone
volves a heme-containing peroxidase and occurs at the
& Modified Within the Cell
luminal surface of the follicular cell. Thyroperoxidase, a
tetrameric protein with a molecular mass of 60 kDa, re-
Insulin has an AB heterodimeric structure with one in-
quires hydrogen peroxide as an oxidizing agent. The
trachain (A6-A11) and two interchain disulfide bridges
H2O2 is produced by an NADPH-dependent enzyme
(A7-B7 and A20-B19) (Figure 42-12). The A and B
resembling cytochrome c reductase. A number of com-
chains could be synthesized in the laboratory, but at-
pounds inhibit I− oxidation and therefore its subse-
tempts at a biochemical synthesis of the mature insulin
quent incorporation into MIT and DIT. The most im-
molecule yielded very poor results. The reason for this
portant of these are the thiourea drugs. They are used as
became apparent when it was discovered that insulin is
antithyroid drugs because of their ability to inhibit thy-
synthesized as a preprohormone (molecular weight ap-
roid hormone biosynthesis at this step. Once iodination
proximately 11,500), which is the prototype for peptides
occurs, the iodine does not readily leave the thyroid.
that are processed from larger precursor molecules. The
Free tyrosine can be iodinated, but it is not incorpo-
hydrophobic 23-amino-acid pre-, or leader, sequence di-
rated into proteins since no tRNA recognizes iodinated
rects the molecule into the cisternae of the endoplasmic
tyrosine.
reticulum and then is removed. This results in the 9000-
The coupling of two DIT molecules to form T4—or
MW proinsulin molecule, which provides the conforma-
of an MIT and DIT to form T3—occurs within the
tion necessary for the proper and efficient formation of
thyroglobulin molecule. A separate coupling enzyme
the disulfide bridges. As shown in Figure 42-12, the se-
has not been found, and since this is an oxidative
quence of proinsulin, starting from the amino terminal,
process it is assumed that the same thyroperoxidase cat-
is B chain—connecting
(C) peptide—A chain. The
alyzes this reaction by stimulating free radical forma-
proinsulin molecule undergoes a series of site-specific
tion of iodotyrosine. This hypothesis is supported by
peptide cleavages that result in the formation of equimo-
the observation that the same drugs which inhibit I− ox-
lar amounts of mature insulin and C peptide. These en-
idation also inhibit coupling. The formed thyroid hor-
zymatic cleavages are summarized in Figure 42-12.
450
/
CHAPTER 42
20
Leu Ser
Gly Ala Gly
Pro
Gln
Pro
Leu
Gly
Gly
Ala
Gly
Leu
Connecting peptide
Leu
Glu
Glu
10
Gly
Val
Ser
Gln
Leu
31
Gly
Gln
Val
Lys
Gln
Arg
Leu
1
Gly
Asp
Ile
Glu
Val
Asn
21
Glu
Cys
Ala
1
Phe
S
Gln
S
Tyr
Glu
1
Val
Asn
Cys
A chain
Glu
Arg
Asn
Cys
Leu
Thr
Ser
Gln
S
Arg
Gln
Ile
Tyr
Cys Ser Leu
S
Thr
Mis
10
30
Leu
S
Lys
Cys
Pro
Insulin
S
Thr
Gly
Tyr
Ser
Phe
Mis
B chain
Phe
Leu
Gly
Arg
10
Val
Glu
Glu
Ala
Cys
Gly
Leu Tyr Leu
Val
20
Figure 42-12. Structure of human proinsulin. Insulin and C-peptide molecules are connected at two sites by
dipeptide links. An initial cleavage by a trypsin-like enzyme (open arrows) followed by several cleavages by a car-
boxypeptidase-like enzyme (solid arrows) results in the production of the heterodimeric (AB) insulin molecule
(light blue) and the C-peptide.
Parathyroid Hormone (PTH) Is Secreted as
mRNA, and this is followed by an increased rate of
an 84-Amino-Acid Peptide
PTH synthesis and secretion. However, about 80-90%
of the proPTH synthesized cannot be accounted for as
The immediate precursor of PTH is proPTH, which
intact PTH in cells or in the incubation medium of ex-
differs from the native 84-amino-acid hormone by hav-
perimental systems. This finding led to the conclusion
ing a highly basic hexapeptide amino terminal exten-
that most of the proPTH synthesized is quickly de-
sion. The primary gene product and the immediate pre-
graded. It was later discovered that this rate of degrada-
cursor for proPTH is the 115-amino-acid preproPTH.
tion decreases when Ca2+ concentrations are low, and it
This differs from proPTH by having an additional 25-
increases when Ca2+ concentrations are high. Very spe-
amino-acid amino terminal extension that, in common
cific fragments of PTH are generated during its prote-
with the other leader or signal sequences characteristic
olytic digestion (Figure 42-13). A number of prote-
of secreted proteins, is hydrophobic. The complete
olytic enzymes, including cathepsins B and D, have
structure of preproPTH and the sequences of proPTH
been identified in parathyroid tissue. Cathepsin B
and PTH are illustrated in Figure 42-13. PTH1-34 has
cleaves PTH into two fragments: PTH1-36
and
full biologic activity, and the region 25-34 is primarily
PTH37-84. PTH37-84 is not further degraded; however,
responsible for receptor binding.
PTH1-36 is rapidly and progressively cleaved into di-
The biosynthesis of PTH and its subsequent secre-
and tripeptides. Most of the proteolysis of PTH occurs
tion are regulated by the plasma ionized calcium (Ca2+)
within the gland, but a number of studies confirm that
concentration through a complex process. An acute de-
PTH, once secreted, is proteolytically degraded in other
crease of Ca2+ results in a marked increase of PTH
tissues, especially the liver, by similar mechanisms.
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
451
Leader (pre) sequence
–6
-10
-20
-31
Gly Asp Ser Arg Ala Leu Phe Cys Ile Ala Leu Met Val Ile Met Val Lys Val Met Asp Lys Ala Ser Met Met
NH2
Lys
Pro
Ser
sequence
Val
Lys
(2)
(1)
Lys
-1 Arg
(3)
1
Ala
Val
Ser
10
20
Glu
Gln Phe Met His Asn Leu Gly Lys His Leu Ser Ser Met Glu Arg Val Glu Trp Leu Arg
Ile
Lys
Lys
Leu
Full biologic activity sequence
Gln
30
Asp
Val
His
Asn
C-fragment sequence
Phe
50
40
Val
Ala
Asp Glu Lys Lys Arg Pro Arg Gln Ser Ser Gly Asp Arg Tyr Ala
Ile
Ser Ala
Gly Leu
Asn
Val
(4)
Leu
60
Val
(5)
Glu
Ser
His
Gln
Lys
O
70
80
Ser
Gly Glu Ala Asp Lys Ala Asp Val Asp Val Leu Ile Lys Ala Lys Pro Gln
Leu
C
OH
Figure 42-13. Structure of bovine preproparathyroid hormone. Arrows indicate sites cleaved by pro-
cessing enzymes in the parathyroid gland (1-5) and in the liver after secretion of the hormone (4-5). The
biologically active region of the molecule is flanked by sequence not required for activity on target re-
ceptors. (Slightly modified and reproduced, with permission, from Habener JF: Recent advances in parathy-
roid hormone research. Clin Biochem 1981;14:223.)
Angiotensin II Is Also Synthesized
tors that decreases fluid volume (dehydration, decreased
From a Large Precursor
blood pressure, fluid or blood loss) or decreases NaCl
concentration stimulates renin release. Renal sympa-
The renin-angiotensin system is involved in the regula-
thetic nerves that terminate in the juxtaglomerular cells
tion of blood pressure and electrolyte metabolism
mediate the central nervous system and postural effects
(through production of aldosterone). The primary hor-
on renin release independently of the baroreceptor and
mone involved in these processes is angiotensin II, an
salt effects, a mechanism that involves the β-adrenergic
octapeptide made from angiotensinogen
(Figure
receptor. Renin acts upon the substrate angiotensino-
42-14). Angiotensinogen, a large α2-globulin made in
gen to produce the decapeptide angiotensin I.
liver, is the substrate for renin, an enzyme produced in
Angiotensin-converting enzyme, a glycoprotein
the juxtaglomerular cells of the renal afferent arteriole.
found in lung, endothelial cells, and plasma, removes
The position of these cells makes them particularly sen-
two carboxyl terminal amino acids from the decapep-
sitive to blood pressure changes, and many of the physi-
tide angiotensin I to form angiotensin II in a step that
ologic regulators of renin release act through renal
is not thought to be rate-limiting. Various nonapeptide
baroreceptors. The juxtaglomerular cells are also sensi-
analogs of angiotensin I and other compounds act as
tive to changes of Na+ and Cl− concentration in the
competitive inhibitors of converting enzyme and are
renal tubular fluid; therefore, any combination of fac-
used to treat renin-dependent hypertension. These are
452
/
CHAPTER 42
Angiotensinogen
Asp-Arg-Val-Tyr-Ile-His-Pro-Phe-His-Leu-Leu (~ 400 more amino acids)
RENIN
Angiotensin I
Asp-Arg-Val-Tyr-Ile-His-Pro-Phe-His-Leu
CONVERTING ENZYME
ANGIOTENSIN II
Asp-Arg-Val-Tyr-Ile-His-Pro-Phe
AMINOPEPTIDASE
Angiotensin III
Arg-Val-Tyr-Ile-His-Pro-Phe
ANGIOTENSINASES
Degradation products
Figure 42-14. Formation and metabolism of angiotensins. Small arrows in-
dicate cleavage sites.
referred to as angiotensin-converting enzyme (ACE)
Complex Processing Generates
inhibitors. Angiotensin II increases blood pressure by
the Pro-opiomelanocortin (POMC)
causing vasoconstriction of the arteriole and is a very
Peptide Family
potent vasoactive substance. It inhibits renin release
from the juxtaglomerular cells and is a potent stimula-
The POMC family consists of peptides that act as hor-
tor of aldosterone production. This results in Na+ re-
mones (ACTH, LPH, MSH) and others that may serve
tention, volume expansion, and increased blood pres-
as neurotransmitters or neuromodulators (endorphins)
sure.
(see Figure 42-15). POMC is synthesized as a precur-
In some species, angiotensin II is converted to the
sor molecule of 285 amino acids and is processed differ-
heptapeptide angiotensin III (Figure 42-14), an equally
ently in various regions of the pituitary.
potent stimulator of aldosterone production. In hu-
The POMC gene is expressed in the anterior and in-
mans, the plasma level of angiotensin II is four times
termediate lobes of the pituitary. The most conserved
greater than that of angiotensin III, so most effects are
sequences between species are within the amino termi-
exerted by the octapeptide. Angiotensins II and III are
nal fragment, the ACTH region, and the β-endorphin
rapidly inactivated by angiotensinases.
region. POMC or related products are found in several
Angiotensin II binds to specific adrenal cortex
other vertebrate tissues, including the brain, placenta,
glomerulosa cell receptors. The hormone-receptor in-
gastrointestinal tract, reproductive tract, lung, and lym-
teraction does not activate adenylyl cyclase, and cAMP
phocytes.
does not appear to mediate the action of this hormone.
The POMC protein is processed differently in the an-
The actions of angiotensin II, which are to stimulate
terior lobe than in the intermediate lobe. The intermedi-
the conversion of cholesterol to pregnenolone and of
ate lobe of the pituitary is rudimentary in adult humans,
corticosterone to 18-hydroxycorticosterone and aldos-
but it is active in human fetuses and in pregnant women
terone, may involve changes in the concentration of in-
during late gestation and is also active in many animal
tracellular calcium and of phospholipid metabolites by
species. Processing of the POMC protein in the periph-
mechanisms similar to those described in Chapter 43.
eral tissues (gut, placenta, male reproductive tract) resem-
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
453
POMC (1-134)
ACTH (1- 39)
β-LPH (42-134)
α-MSH
CLIP
γ-LPH
β-Endorphin
(1-13)
(18-39)
(42-101)
(104 -134)
β-MSH
γ-Endorphin
(84 -101)
(104 -118)
α-Endorphin
(104 -117)
Figure 42-15. Products of pro-opiomelanocortin (POMC) cleavage.
(MSH, melanocyte-stimulating hormone; CLIP, corticotropin-like inter-
mediate lobe peptide; LPH, lipotropin.)
bles that in the intermediate lobe. There are three basic
there is no intracellular reservoir of these hormones.
peptide groups:
(1) ACTH, which can give rise to
The catecholamines, also synthesized in active form, are
α-MSH and corticotropin-like intermediate lobe peptide
stored in granules in the chromaffin cells in the adrenal
(CLIP);
(2) β-lipotropin
(β-LPH), which can yield
medulla. In response to appropriate neural stimulation,
γ-LPH, β-MSH, and β-endorphin (and thus α- and
these granules are released from the cell through exocy-
γ-endorphins); and (3) a large amino terminal peptide,
tosis, and the catecholamines are released into the circu-
which generates γ-MSH. The diversity of these products
lation. A several-hour reserve supply of catecholamines
is due to the many dibasic amino acid clusters that are
exists in the chromaffin cells.
potential cleavage sites for trypsin-like enzymes. Each of
Parathyroid hormone also exists in storage vesicles.
the peptides mentioned is preceded by Lys-Arg, Arg-Lys,
As much as 80-90% of the proPTH synthesized is de-
Arg-Arg, or Lys-Lys residues. After the prehormone seg-
graded before it enters this final storage compartment,
ment is cleaved, the next cleavage, in both anterior and
especially when Ca2+ levels are high in the parathyroid
intermediate lobes, is between ACTH and β-LPH, re-
cell (see above). PTH is secreted when Ca2+ is low in
sulting in an amino terminal peptide with ACTH and a
the parathyroid cells, which contain a several-hour sup-
β-LPH segment (Figure 42-15). ACTH1-39 is subse-
ply of the hormone.
quently cleaved from the amino terminal peptide, and in
The human pancreas secretes about 40-50 units of in-
the anterior lobe essentially no further cleavages occur. In
sulin daily, which represents about 15-20% of the hor-
the intermediate lobe, ACTH1-39 is cleaved into α-MSH
mone stored in the B cells. Insulin and the C-peptide (see
(residues 1-13) and CLIP (18-39); β-LPH (42-134) is
Figure
42-12) are normally secreted in equimolar
converted to γ-LPH (42-101) and β-endorphin (104-
amounts. Stimuli such as glucose, which provokes insulin
134). β-MSH (84-101) is derived from γ-LPH.
secretion, therefore trigger the processing of proinsulin to
There are extensive additional tissue-specific modifi-
insulin as an essential part of the secretory response.
cations of these peptides that affect activity. These
A several-week supply of T3 and T4 exists in the thy-
modifications include phosphorylation, acetylation,
roglobulin that is stored in colloid in the lumen of the
glycosylation, and amidation.
thyroid follicles. These hormones can be released upon
stimulation by TSH. This is the most exaggerated ex-
ample of a prohormone, as a molecule containing ap-
THERE IS VARIATION IN THE STORAGE
proximately 5000 amino acids must be first synthe-
& SECRETION OF HORMONES
sized, then degraded, to supply a few molecules of the
As mentioned above, the steroid hormones and
active hormones T4 and T3.
1,25(OH)2-D3
are synthesized in their final active
The diversity in storage and secretion of hormones
form. They are also secreted as they are made, and thus
is illustrated in Table 42-5.
454
/
CHAPTER 42
Table 42-5. Diversity in the storage of hormones.
lives. A notable exception is IGF-I, which is transported
bound to members of a family of binding proteins.
Hormone
Supply Stored in Cell
Thyroid Hormones Are Transported
Steroids and 1,25(OH)2-D3
None
by Thyroid-Binding Globulin
Catecholamines and PTH
Hours
Many of the principles discussed above are illustrated in
Insulin
Days
a discussion of thyroid-binding proteins. One-half to
T3 and T4
Weeks
two-thirds of T4 and T3 in the body is in an extrathy-
roidal reservoir. Most of this circulates in bound form,
ie, bound to a specific binding protein, thyroxine-
binding globulin (TBG). TBG, a glycoprotein with a
SOME HORMONES HAVE PLASMA
molecular mass of 50 kDa, binds T4 and T3 and has the
TRANSPORT PROTEINS
capacity to bind 20 µg/dL of plasma. Under normal
The class I hormones are hydrophobic in chemical na-
circumstances, TBG binds—noncovalently—nearly all
ture and thus are not very soluble in plasma. These hor-
of the T4 and T3 in plasma, and it binds T4 with greater
mones, principally the steroids and thyroid hormones,
affinity than T3 (Table 42-7). The plasma half-life of
have specialized plasma transport proteins that serve sev-
T4 is correspondingly four to five times that of T3. The
eral purposes. First, these proteins circumvent the solu-
small, unbound (free) fraction is responsible for the bi-
bility problem and thereby deliver the hormone to the
ologic activity. Thus, in spite of the great difference in
target cell. They also provide a circulating reservoir of
total amount, the free fraction of T3 approximates that
the hormone that can be substantial, as in the case of the
of T4, and given that T3 is intrinsically more active than
thyroid hormones. Hormones, when bound to the trans-
T4, most biologic activity is attributed to T3. TBG does
port proteins, cannot be metabolized, thereby prolonging
not bind any other hormones.
their plasma half-life
(t1/2). The binding affinity of a
given hormone to its transporter determines the bound
Glucocorticoids Are Transported
versus free ratio of the hormone. This is important be-
by Corticosteroid-Binding Globulin
cause only the free form of a hormone is biologically ac-
Hydrocortisone (cortisol) also circulates in plasma in
tive. In general, the concentration of free hormone in
protein-bound and free forms. The main plasma bind-
plasma is very low, in the range of 10-15 to 10-9 mol/L. It
ing protein is an α-globulin called transcortin, or cor-
is important to distinguish between plasma transport
ticosteroid-binding globulin
(CBG). CBG is pro-
proteins and hormone receptors. Both bind hormones
duced in the liver, and its synthesis, like that of TBG, is
but with very different characteristics (Table 42-6).
increased by estrogens. CBG binds most of the hor-
The hydrophilic hormones—generally class II and
mone when plasma cortisol levels are within the normal
of peptide structure—are freely soluble in plasma and
range; much smaller amounts of cortisol are bound to
do not require transport proteins. Hormones such as
albumin. The avidity of binding helps determine the
insulin, growth hormone, ACTH, and TSH circulate
biologic half-lives of various glucocorticoids. Cortisol
in the free, active form and have very short plasma half-
binds tightly to CBG and has a t1/2 of 1.5-2 hours,
while corticosterone, which binds less tightly, has a t1/2
Table 42-6. Comparison of receptors with
of less than 1 hour (Table 42-8). The unbound (free)
cortisol constitutes about 8% of the total and represents
transport proteins.
the biologically active fraction. Binding to CBG is not
restricted to glucocorticoids. Deoxycorticosterone and
Feature
Receptors
Transport Proteins
Concentration
Very low
Very high
(thousands/cell)
(billions/µL)
Table 42-7. Comparison of T4 and T3 in plasma.
Binding affinity
High (pmol/L to
Low (µmol/L range)
nmol/L range)
Free Hormone
Binding specificity
Very high
Low
Total
t1⁄2
Hormone
Percent
in Blood
Saturability
Yes
No
(µg/dL)
of Total
ng/dL
Molarity
(days)
Reversibility
Yes
Yes
T4
8
0.03
~2.24
3.0 × 10−11
6.5
Signal transduction
Yes
No
T3
0.15
0.3
~0.4
~0.6 × 10−11
1.5
THE DIVERSITY OF THE ENDOCRINE SYSTEM
/
455
Table 42-8. Approximate affinities of steroids for
binding capacity they probably buffer against sudden
serum-binding proteins.
changes in the plasma level. Because the metabolic
clearance rates of these steroids are inversely related to
the affinity of their binding to SHBG, estrone is cleared
SHBG1
CBG1
more rapidly than estradiol, which in turn is cleared
Dihydrotestosterone
1
> 100
more rapidly than testosterone or DHT.
Testosterone
2
> 100
Estradiol
5
> 10
SUMMARY
Estrone
> 10
> 100
Progesterone
> 100
~ 2
• The presence of a specific receptor defines the target
Cortisol
> 100
~ 3
cells for a given hormone.
Corticosterone
> 100
~ 5
• Receptors are proteins that bind specific hormones
1Affinity expressed as Kd (nmol/L).
and generate an intracellular signal (receptor-effector
coupling).
• Some hormones have intracellular receptors; others
progesterone interact with CBG with sufficient affinity
bind to receptors on the plasma membrane.
to compete for cortisol binding. Aldosterone, the most
• Hormones are synthesized from a number of precur-
potent natural mineralocorticoid, does not have a spe-
sor molecules, including cholesterol, tyrosine per se,
cific plasma transport protein. Gonadal steroids bind
and all the constituent amino acids of peptides and
very weakly to CBG (Table 42-8).
proteins.
• A number of modification processes alter the activity
Gonadal Steroids Are Transported
of hormones. For example, many hormones are syn-
by Sex Hormone-Binding Globulin
thesized from larger precursor molecules.
Most mammals, humans included, have a plasma β-
• The complement of enzymes in a particular cell type
globulin that binds testosterone with specificity, rela-
allows for the production of a specific class of steroid
tively high affinity, and limited capacity (Table 42-8).
hormone.
This protein, usually called sex hormone-binding
• Most of the lipid-soluble hormones are bound to
globulin
(SHBG) or testosterone-estrogen-binding
rather specific plasma transport proteins.
globulin (TEBG), is produced in the liver. Its produc-
tion is increased by estrogens (women have twice the
serum concentration of SHBG as men), certain types of
REFERENCES
liver disease, and hyperthyroidism; it is decreased by
Bartalina L: Thyroid hormone-binding proteins: update 1994. En-
androgens, advancing age, and hypothyroidism. Many
docr Rev 1994;13:140.
of these conditions also affect the production of CBG
Beato M et al: Steroid hormone receptors: many actors in search of
and TBG. Since SHBG and albumin bind 97-99% of
a plot. Cell 1995;83:851.
circulating testosterone, only a small fraction of the
Dai G, Carrasco L, Carrasco N: Cloning and characterization of
hormone in circulation is in the free (biologically ac-
the thyroid iodide transporter. Nature 1996;379:458.
tive) form. The primary function of SHBG may be to
DeLuca HR: The vitamin D story: a collaborative effort of basic
restrict the free concentration of testosterone in the
science and clinical medicine. FASEB J 1988;2:224.
serum. Testosterone binds to SHBG with higher affin-
Douglass J, Civelli O, Herbert E: Polyprotein gene expression:
ity than does estradiol
(Table
42-8). Therefore, a
Generation of diversity of neuroendocrine peptides. Annu
change in the level of SHBG causes a greater change in
Rev Biochem 1984;53:665.
the free testosterone level than in the free estradiol level.
Miller WL: Molecular biology of steroid hormone biosynthesis.
Endocr Rev 1988;9:295.
Estrogens are bound to SHBG and progestins to
CBG. SHBG binds estradiol about five times less avidly
Nagatsu T: Genes for human catecholamine-synthesizing enzymes.
Neurosci Res 1991;12:315.
than it binds testosterone or DHT, while progesterone
Russell DW, Wilson JD: Steroid 5 alpha-reductase: two genes/two
and cortisol have little affinity for this protein (Table
enzymes. Annu Rev Biochem 1994;63:25.
42-8). In contrast, progesterone and cortisol bind with
Russell J et al: Interaction between calcium and 1,25-dihydroxy-
nearly equal affinity to CBG, which in turn has little
vitamin D3 in the regulation of preproparathyroid hormone
avidity for estradiol and even less for testosterone,
and vitamin D receptor mRNA in avian parathyroids. En-
DHT, or estrone.
docrinology 1993;132:2639.
These binding proteins also provide a circulating
Steiner DF et al: The new enzymology of precursor processing en-
reservoir of hormone, and because of the relatively large
doproteases. J Biol Chem 1992;267:23435.
Hormone Action &
43
Signal Transduction
Daryl K. Granner, MD
BIOMEDICAL IMPORTANCE
as described in Chapter 42, ie, based on the location of
their specific cellular receptors and the type of signals
The homeostatic adaptations an organism makes to a
generated. Group I hormones interact with an intracel-
constantly changing environment are in large part ac-
lular receptor and group II hormones with receptor
complished through alterations of the activity and
recognition sites located on the extracellular surface of
amount of proteins. Hormones provide a major means
the plasma membrane of target cells. The cytokines, in-
of facilitating these changes. A hormone-receptor inter-
terleukins, and growth factors should also be considered
action results in generation of an intracellular signal
in this latter category. These molecules, of critical im-
that can either regulate the activity of a select set of
portance in homeostatic adaptation, are hormones in
genes, thereby altering the amount of certain proteins
the sense that they are produced in specific cells, have
in the target cell, or affect the activity of specific pro-
the equivalent of autocrine, paracrine, and endocrine
teins, including enzymes and transporter or channel
actions, bind to cell surface receptors, and activate
proteins. The signal can influence the location of pro-
many of the same signal transduction pathways em-
teins in the cell and can affect general processes such as
ployed by the more traditional group II hormones.
protein synthesis, cell growth, and replication, perhaps
through effects on gene expression. Other signaling
molecules—including cytokines, interleukins, growth
SIGNAL GENERATION
factors, and metabolites—use some of the same general
mechanisms and signal transduction pathways. Exces-
The Ligand-Receptor Complex Is the
sive, deficient, or inappropriate production and release
Signal for Group I Hormones
of hormones and of these other regulatory molecules
The lipophilic group I hormones diffuse through the
are major causes of disease. Many pharmacotherapeutic
plasma membrane of all cells but only encounter their
agents are aimed at correcting or otherwise influencing
specific, high-affinity intracellular receptors in target
the pathways discussed in this chapter.
cells. These receptors can be located in the cytoplasm or
in the nucleus of target cells. The hormone-receptor
complex first undergoes an activation reaction. As
HORMONES TRANSDUCE SIGNALS TO
shown in Figure 43-2, receptor activation occurs by at
AFFECT HOMEOSTATIC MECHANISMS
least two mechanisms. For example, glucocorticoids
The general steps involved in producing a coordinated
diffuse across the plasma membrane and encounter
response to a particular stimulus are illustrated in
their cognate receptor in the cytoplasm of target cells.
Figure 43-1. The stimulus can be a challenge or a
Ligand-receptor binding results in the dissociation of
threat to the organism, to an organ, or to the integrity
heat shock protein 90 (hsp90) from the receptor. This
of a single cell within that organism. Recognition of the
step appears to be necessary for subsequent nuclear lo-
stimulus is the first step in the adaptive response. At the
calization of the glucocorticoid receptor. This receptor
organismic level, this generally involves the nervous sys-
also contains nuclear localization sequences that assist
tem and the special senses (sight, hearing, pain, smell,
in the translocation from cytoplasm to nucleus. The
touch). At the organismic or cellular level, recognition
now activated receptor moves into the nucleus (Figure
involves physicochemical factors such as pH, O2 ten-
43-2) and binds with high affinity to a specific DNA
sion, temperature, nutrient supply, noxious metabo-
sequence called the hormone response element
lites, and osmolarity. Appropriate recognition results in
(HRE). In the case illustrated, this is a glucocorticoid
the release of one or more hormones that will govern
response element, or GRE. Consensus sequences for
generation of the necessary adaptive response. For pur-
HREs are shown in Table 43-1. The DNA-bound, lig-
poses of this discussion, the hormones are categorized
anded receptor serves as a high-affinity binding site for
456
HORMONE ACTION & SIGNAL TRANSDUCTION
/
457
STIMULUS
Recognition
Group I hormones
Group II hormones
Hormone release
Hormone•receptor complex
Many different signals
Signal generation
Effects
Gene
Transporters
Protein
Protein
transcription
Channels
translocation
modification
COORDINATED RESPONSE TO STIMULUS
Figure 43-1. Hormonal involvement in responses to a stimulus. A challenge to the in-
tegrity of the organism elicits a response that includes the release of one or more hormones.
These hormones generate signals at or within target cells, and these signals regulate a vari-
ety of biologic processes which provide for a coordinated response to the stimulus or chal-
lenge. See Figure 43-8 for a specific example.
one or more coactivator proteins, and accelerated gene
gests that these hormones exert their dominant effect
transcription typically ensues when this occurs. By con-
on modulating gene transcription, but they—and many
trast, certain hormones such as the thyroid hormones
of the hormones in the other classes discussed below—
and retinoids diffuse from the extracellular fluid across
can act at any step of the “information pathway” illus-
the plasma membrane and go directly into the nucleus.
trated in Figure 43-3. Direct actions of steroids in the
In this case, the cognate receptor is already bound to
cytoplasm and on various organelles and membranes
the HRE
(the thyroid hormone response element
have also been described.
[TRE], in this example). However, this DNA-bound
receptor fails to activate transcription because it is com-
plexed with a corepressor. Indeed, this receptor-
GROUP II (PEPTIDE &
corepressor complex serves as an active repressor of gene
CATECHOLAMINE) HORMONES
transcription. The association of ligand with these re-
HAVE MEMBRANE RECEPTORS
ceptors results in dissociation of the corepressor. The
& USE INTRACELLULAR MESSENGERS
liganded receptor is now capable of binding one or
more coactivators with high affinity, resulting in the ac-
Many hormones are water-soluble, have no transport
tivation of gene transcription. The relationship of hor-
proteins (and therefore have a short plasma half-life),
mone receptors to other nuclear receptors and to coreg-
and initiate a response by binding to a receptor located
ulators is discussed in more detail below.
in the plasma membrane (see Tables 42-3 and 42-4).
By selectively affecting gene transcription and the
The mechanism of action of this group of hormones
consequent production of appropriate target mRNAs,
can best be discussed in terms of the intracellular sig-
the amounts of specific proteins are changed and meta-
nals they generate. These signals include cAMP (cyclic
bolic processes are influenced. The influence of each of
AMP; 3′,5′-adenylic acid; see Figure
18-5), a nu-
these hormones is quite specific; generally, the hor-
cleotide derived from ATP through the action of
mone affects less than 1% of the genes, mRNA, or pro-
adenylyl cyclase; cGMP, a nucleotide formed by gua-
teins in a target cell; sometimes only a few are affected.
nylyl cyclase; Ca2+; and phosphatidylinositides. Many
The nuclear actions of steroid, thyroid, and retinoid
of these second messengers affect gene transcription, as
hormones are quite well defined. Most evidence sug-
described in the previous paragraph; but they also influ-
458
/
CHAPTER 43
−
Cytoplasm
+
+
−
TRE
TRE
TRE
GRE
GRE
GRE
+
hsp
+
hsp
Nucleus
Figure 43-2. Regulation of gene expression by class I hormones.
Steroid hormones readily gain access to the cytoplasmic compartment
of target cells. Glucocorticoid hormones (solid triangles) encounter
their cognate receptor in the cytoplasm, where it exists in a complex
with heat shock protein 90 (hsp). Ligand binding causes dissociation of
hsp and a conformational change of the receptor. The receptor•ligand
complex then traverses the nuclear membrane and binds to DNA with
specificity and high affinity at a glucocorticoid response element (GRE).
This event triggers the assembly of a number of transcription coregula-
tors ( + ), and enhanced transcription ensues. By contrast, thyroid hor-
mones and retinoic acid (
) directly enter the nucleus, where their
cognate receptors are already bound to the appropriate response ele-
ments with an associated transcription repressor complex ( − ). This
complex, which consists of molecules such as N-CoR or SMRT (see
Table 43-6) in the absence of ligand, actively inhibits transcription. Lig-
and binding results in dissociation of the repressor complex from the
receptor, allowing an activator complex to assemble. The gene is then
actively transcribed.
ence a variety of other biologic processes, as shown in
cloned from various mammalian species. A wide variety
Figure 43-1.
of responses are mediated by the GPCRs.
G Protein-Coupled Receptors (GPCR)
cAMP Is the Intracellular Signal
Many of the group II hormones bind to receptors that
for Many Responses
couple to effectors through a GTP-binding protein in-
Cyclic AMP was the first intracellular signal identified
termediary. These receptors typically have seven hy-
in mammalian cells. Several components comprise a
drophobic plasma membrane-spanning domains. This
system for the generation, degradation, and action of
is illustrated by the seven interconnected cylinders ex-
cAMP.
tending through the lipid bilayer in Figure 43-4. Re-
ceptors of this class, which signal through guanine nu-
A. ADENYLYL CYCLASE
cleotide-bound protein intermediates, are known as
G protein-coupled receptors, or GPCRs. To date,
Different peptide hormones can either stimulate (s) or
over 130 G protein-linked receptor genes have been
inhibit (i) the production of cAMP from adenylyl cy-
HORMONE ACTION & SIGNAL TRANSDUCTION
/
459
Table 43-1. The DNA sequences of several
Gene
hormone response elements (HREs).1
TRANSCRIPTION
Hormone or Effector
HRE
DNA Sequence
Primary transcript
Degradation
Glucocorticoids
GRE
Progestins
PRE
GGTACA NNN TGTTCT
←
NUCLEUS
MODIFICATION/PROCESSING
Mineralocorticoids
MRE
Androgens
ARE
mRNA
Degradation
Estrogens
ERE
AGGTCA --- TGA/TCCT
←
Transport
Thyroid hormone
TRE
Retinoic acid
RARE
AGGTCA N3,4,5, AGGTCA
Active
inactive
Vitamin D
VDRE
mRNA
degradation
cAMP
CRE
TGACGTCA
CYTOPLASM
TRANSLATION
1Letters indicate nucleotide; N means any one of the four can be
used in that position. The arrows pointing in opposite directions
Protein
Modification
degradation
illustrate the slightly imperfect inverted palindromes present in
many HREs; in some cases these are called “half binding sites” be-
Figure 43-3. The “information pathway.” Informa-
cause each binds one monomer of the receptor. The GRE, PRE,
tion flows from the gene to the primary transcript to
MRE, and ARE consist of the same DNA sequence. Specificity may
mRNA to protein. Hormones can affect any of the steps
be conferred by the intracellular concentration of the ligand or
hormone receptor, by flanking DNA sequences not included in
involved and can affect the rates of processing, degra-
the consensus, or by other accessory elements. A second group of
dation, or modification of the various products.
HREs includes those for thyroid hormones, estrogens, retinoic
acid, and vitamin D. These HREs are similar except for the orienta-
tion and spacing between the half palindromes. Spacing deter-
mines the hormone specificity. VDRE (N=3), TRE (N=4), and RARE
tively. In the case of αs, this modification disrupts the
(N=5) bind to direct repeats rather than to inverted repeats. An-
intrinsic GTP-ase activity; thus, αs cannot reassociate
other member of the steroid receptor superfamily, the retinoid X
with βγ and is therefore irreversibly activated. ADP-
receptor (RXR), forms heterodimers with VDR, TR, and RARE, and
these constitute the trans-acting factors. cAMP affects gene tran-
ribosylation of αi-2
prevents the dissociation of αi-2
scription through the CRE.
from βγ, and free αi-2 thus cannot be formed. αs activ-
ity in such cells is therefore unopposed.
There is a large family of G proteins, and these are
clase, which is encoded by at least nine different genes
part of the superfamily of GTPases. The G protein
(Table 43-2). Two parallel systems, a stimulatory (s)
family is classified according to sequence homology
one and an inhibitory (i) one, converge upon a single
into four subfamilies, as illustrated in Table
43-3.
catalytic molecule (C). Each consists of a receptor, Rs or
There are 21 α, 5 β, and 8 γ subunit genes. Various
Ri, and a regulatory complex, Gs and Gi. Gs and Gi are
combinations of these subunits provide a large number
each trimers composed of α, β, and γ subunits. Because
of possible αβγ and cyclase complexes.
the α subunit in Gs differs from that in Gi, the pro-
The α subunits and the βγ complex have actions in-
teins, which are distinct gene products, are designated
dependent of those on adenylyl cyclase
(see Figure
αs and αi. The α subunits bind guanine nucleotides.
43-4 and Table 43-3). Some forms of αi stimulate K+
The β and γ subunits are always associated (βγ) and ap-
channels and inhibit Ca2+ channels, and some αs mole-
pear to function as a heterodimer. The binding of a hor-
cules have the opposite effects. Members of the Gq fam-
mone to Rs or Ri results in a receptor-mediated activa-
ily activate the phospholipase C group of enzymes. The
tion of G, which entails the exchange of GDP by GTP
βγ complexes have been associated with K+ channel
on α and the concomitant dissociation of βγ from α.
stimulation and phospholipase C activation. G proteins
The αs protein has intrinsic GTPase activity. The
are involved in many important biologic processes in
active form, αs•GTP, is inactivated upon hydrolysis of
addition to hormone action. Notable examples include
the GTP to GDP; the trimeric Gs complex (αβγ) is
olfaction (αOLF) and vision (αt). Some examples are
then re-formed and is ready for another cycle of activa-
listed in Table 43-3. GPCRs are implicated in a num-
tion. Cholera and pertussis toxins catalyze the ADP-
ber of diseases and are major targets for pharmaceutical
ribosylation of αs and αi-2 (see Table 43-3), respec-
agents.
460
/
CHAPTER 43
N
N
H
E
E
γ
γ
αs
β
β
αs
C
C
No hormone: inactive effector
Bound hormone (H): active effector
Figure 43-4. Components of the hormone receptor-G protein effector system. Receptors that
couple to effectors through G proteins (GPCR) typically have seven membrane-spanning domains. In
the absence of hormone (left), the heterotrimeric G-protein complex (α, β, γ) is in an inactive guano-
sine diphosphate (GDP)-bound form and is probably not associated with the receptor. This complex is
anchored to the plasma membrane through prenylated groups on the βγ subunits (wavy lines) and
perhaps by myristoylated groups on α subunits (not shown). On binding of hormone ( H ) to the re-
ceptor, there is a presumed conformational change of the receptor—as indicated by the tilted mem-
brane spanning domains—and activation of the G-protein complex. This results from the exchange of
GDP with guanosine triphosphate (GTP) on the α subunit, after which α and βγ dissociate. The α sub-
unit binds to and activates the effector (E). E can be adenylyl cyclase, Ca2+, Na+, or Cl− channels (αs), or
it could be a K+ channel (αi), phospholipase Cβ (αq), or cGMP phosphodiesterase (αt). The βγ subunit
can also have direct actions on E. (Modified and reproduced, with permission, from Granner DK in: Princi-
ples and Practice of Endocrinology and Metabolism, 3rd ed. Becker KL [editor]. Lippincott, 2000.)
B. PROTEIN KINASE
In prokaryotic cells, cAMP binds to a specific protein
called catabolite regulatory protein (CRP) that binds
Table 43-2. Subclassification of group II.A
directly to DNA and influences gene expression. In eu-
hormones.
karyotic cells, cAMP binds to a protein kinase called
protein kinase A (PKA) that is a heterotetrameric mol-
ecule consisting of two regulatory subunits (R) and two
Hormones That Stimulate
Hormones That Inhibit
catalytic subunits (C). cAMP binding results in the fol-
Adenylyl Cyclase
Adenylyl Cyclase
lowing reaction:
(Hs)
(Hl)
ACTH
Acetylcholine
4
cAMP + R
C
a
R
⋅(
4
cAMP) + 2C
ADH
α
2
2
2
2-Adrenergics
β-Adrenergics
Angiotensin II
Calcitonin
Somatostatin
The R2C2 complex has no enzymatic activity, but
CRH
the binding of cAMP by R dissociates R from C,
FSH
thereby activating the latter (Figure 43-5). The active
Glucagon
C subunit catalyzes the transfer of the γ phosphate of
hCG
ATP to a serine or threonine residue in a variety of pro-
LH
teins. The consensus phosphorylation sites are -Arg-
LPH
Arg/Lys-X-Ser/Thr- and -Arg-Lys-X-X-Ser-, where X
MSH
can be any amino acid.
PTH
Protein kinase activities were originally described as
TSH
being “cAMP-dependent” or “cAMP-independent.” This
HORMONE ACTION & SIGNAL TRANSDUCTION
/
461
Table 43-3. Classes and functions of selected G proteins.1,2
Class or Type
Stimulus
Effector
Effect
Gs αs
Glucagon, β-adrenergics
↑ Adenylyl cyclase
Gluconeogenesis, lipolysis,
↑ Cardiac Ca2+, Cl−, and Na+ channels
glycogenolysis
αolf
Odorant
↑ Adenylyl cyclase
Olfaction
Gi αi
-1,2,3
Acetylcholine,
↓ Adenylyl cyclase
Slowed heart rate
α2-adrenergics
↑ Potassium channels
M2 cholinergics
↓ Calcium channels
α0
Opioids, endorphins
↑ Potassium channels
Neuronal electrical activity
αt
Light
↑ cGMP phosphodiesterase
Vision
Gqαq
M1 cholinergics
α1-Adrenergics
↑ Phospholipase C-β1
↑ Muscle contraction
and
α11
α1-Adrenergics
↑ Phospholipase c-β2
↑ Blood pressure
G12
α12
?
Cl− channel
?
1Modified and reproduced, with permission, from Granner DK in: Principles and Practice of Endocrinology and Metabolism, 3rd
ed. Becker KL (editor). Lippincott, 2000.
2The four major classes or families of mammalian G proteins (Gs , Gi , Gq , and G12) are based on protein sequence homology.
Representative members of each are shown, along with known stimuli, effectors, and well-defined biologic effects. Nine iso-
forms of adenylyl cyclase have been identified (isoforms I-IX). All isoforms are stimulated by αs ; αi isoforms inhibit types V
and VI, and α0 inhibits types I and V. At least 16 different α subunits have been identified.
classification has changed, as protein phosphorylation is
ment binding protein (CREB). CREB binds
to
a
now recognized as being a major regulatory mecha-
cAMP responsive element (CRE) (see Table 43-1) in
nism. Several hundred protein kinases have now been
its nonphosphorylated state and is a weak activator of
described. The kinases are related in sequence and
transcription. When phosphorylated by PKA, CREB
structure within the catalytic domain, but each is a
binds the coactivator CREB-binding protein CBP/
unique molecule with considerable variability with re-
p300 (see below) and as a result is a much more potent
spect to subunit composition, molecular weight, au-
transcription activator.
tophosphorylation, Km for ATP, and substrate speci-
D. PHOSPHODIESTERASES
ficity.
Actions caused by hormones that increase cAMP con-
C. PHOSPHOPROTEINS
centration can be terminated in a number of ways, in-
The effects of cAMP in eukaryotic cells are all thought
cluding the hydrolysis of cAMP to 5′-AMP by phos-
to be mediated by protein phosphorylation-dephosphor-
phodiesterases (see Figure 43-5). The presence of these
ylation, principally on serine and threonine residues.
hydrolytic enzymes ensures a rapid turnover of the sig-
The control of any of the effects of cAMP, including
nal (cAMP) and hence a rapid termination of the bio-
such diverse processes as steroidogenesis, secretion, ion
logic process once the hormonal stimulus is removed.
transport, carbohydrate and fat metabolism, enzyme in-
There are at least 11 known members of the phospho-
duction, gene regulation, synaptic transmission, and
diesterase family of enzymes. These are subject to regu-
cell growth and replication, could be conferred by a
lation by their substrates, cAMP and cGMP; by hor-
specific protein kinase, by a specific phosphatase, or by
mones; and by intracellular messengers such as calcium,
specific substrates for phosphorylation. These substrates
probably acting through calmodulin. Inhibitors of
help define a target tissue and are involved in defining
phosphodiesterase, most notably methylated xanthine
the extent of a particular response within a given cell.
derivatives such as caffeine, increase intracellular cAMP
For example, the effects of cAMP on gene transcription
and mimic or prolong the actions of hormones through
are mediated by the protein cyclic AMP response ele-
this signal.
462
/
CHAPTER 43
ATP • Mg2+
R2C2
Inactive PKA
Active
adenylyl
cyclase
cAMP
Phosphodiesterase
C2
+ R2
Cell
Active PKA
5′-AMP
membrane
Mg2+ • ATP
Protein
Phosphoprotein
Phosphatase
Physiologic
effects
Figure 43-5. Hormonal regulation of cellular processes through
cAMP-dependent protein kinase (PKA). PKA exists in an inactive form as
an R2C2 heterotetramer consisting of two regulatory and two catalytic
subunits. The cAMP generated by the action of adenylyl cyclase (acti-
vated as shown in Figure 43-4) binds to the regulatory (R) subunit of
PKA. This results in dissociation of the regulatory and catalytic subunits
and activation of the latter. The active catalytic subunits phosphorylate
a number of target proteins on serine and threonine residues. Phos-
phatases remove phosphate from these residues and thus terminate
the physiologic response. A phosphodiesterase can also terminate the
response by converting cAMP to 5′-AMP.
E. PHOSPHOPROTEIN PHOSPHATASES
heat-stable protein inhibitors regulate type I phos-
phatase activity. Inhibitor-1 is phosphorylated and acti-
Given the importance of protein phosphorylation, it is
vated by cAMP-dependent protein kinases; and in-
not surprising that regulation of the protein dephos-
hibitor-2, which may be a subunit of the inactive
phorylation reaction is another important control
phosphatase, is also phosphorylated, possibly by glyco-
mechanism (see Figure
43-5). The phosphoprotein
gen synthase kinase-3.
phosphatases are themselves subject to regulation by
phosphorylation-dephosphorylation reactions and by a
variety of other mechanisms, such as protein-protein
cGMP Is Also an Intracellular Signal
interactions. In fact, the substrate specificity of the
phosphoserine-phosphothreonine phosphatases may be
Cyclic GMP is made from GTP by the enzyme gua-
dictated by distinct regulatory subunits whose binding
nylyl cyclase, which exists in soluble and membrane-
is regulated hormonally. The best-studied role of regu-
bound forms. Each of these isozymes has unique physi-
lation by the dephosphorylation of proteins is that of
ologic properties. The atriopeptins, a family of peptides
glycogen metabolism in muscle. Two major types of
produced in cardiac atrial tissues, cause natriuresis, di-
phosphoserine-phosphothreonine phosphatases have
uresis, vasodilation, and inhibition of aldosterone secre-
been described. Type I preferentially dephosphorylates
tion. These peptides (eg, atrial natriuretic factor) bind
the β subunit of phosphorylase kinase, whereas type II
to and activate the membrane-bound form of guanylyl
dephosphorylates the α subunit. Type I phosphatase is
cyclase. This results in an increase of cGMP by as much
implicated in the regulation of glycogen synthase, phos-
as 50-fold in some cases, and this is thought to mediate
phorylase, and phosphorylase kinase. This phosphatase
the effects mentioned above. Other evidence links
is itself regulated by phosphorylation of certain of its
cGMP to vasodilation. A series of compounds, includ-
subunits, and these reactions are reversed by the action
ing nitroprusside, nitroglycerin, nitric oxide, sodium
of one of the type II phosphatases. In addition, two
nitrite, and sodium azide, all cause smooth muscle re-
HORMONE ACTION & SIGNAL TRANSDUCTION
/
463
laxation and are potent vasodilators. These agents in-
B. CALMODULIN
crease cGMP by activating the soluble form of guanylyl
The calcium-dependent regulatory protein is calmod-
cyclase, and inhibitors of cGMP phosphodiesterase (the
ulin, a 17-kDa protein that is homologous to the mus-
drug sildenafil [Viagra], for example) enhance and pro-
cle protein troponin C in structure and function.
long these responses. The increased cGMP activates
Calmodulin has four Ca2+ binding sites, and full occu-
cGMP-dependent protein kinase (PKG), which in turn
pancy of these sites leads to a marked conformational
phosphorylates a number of smooth muscle proteins.
change, which allows calmodulin to activate enzymes
Presumably, this is involved in relaxation of smooth
and ion channels. The interaction of Ca2+ with calmod-
muscle and vasodilation.
ulin (with the resultant change of activity of the latter)
is conceptually similar to the binding of cAMP to PKA
Several Hormones Act Through
and the subsequent activation of this molecule.
Calcium or Phosphatidylinositols
Calmodulin can be one of numerous subunits of com-
plex proteins and is particularly involved in regulating
Ionized calcium is an important regulator of a variety of
various kinases and enzymes of cyclic nucleotide gener-
cellular processes, including muscle contraction, stimu-
ation and degradation. A partial list of the enzymes reg-
lus-secretion coupling, the blood clotting cascade, en-
ulated directly or indirectly by Ca2+, probably through
zyme activity, and membrane excitability. It is also an
calmodulin, is presented in Table 43-4.
intracellular messenger of hormone action.
In addition to its effects on enzymes and ion trans-
port, Ca2+/calmodulin regulates the activity of many
A. CALCIUM METABOLISM
structural elements in cells. These include the actin-
The extracellular calcium (Ca2+) concentration is about
myosin complex of smooth muscle, which is under β-
5 mmol/L and is very rigidly controlled. Although sub-
adrenergic control, and various microfilament-medi-
stantial amounts of calcium are associated with intracel-
ated processes in noncontractile cells, including cell
lular organelles such as mitochondria and the endoplas-
motility, cell conformation changes, mitosis, granule re-
mic reticulum, the intracellular concentration of free or
lease, and endocytosis.
ionized calcium (Ca2+) is very low: 0.05-10 µmol/L. In
spite of this large concentration gradient and a favor-
C. CALCIUM IS A MEDIATOR OF HORMONE ACTION
able transmembrane electrical gradient, Ca2+ is re-
A role for Ca2+ in hormone action is suggested by the
strained from entering the cell. A considerable amount
observations that the effect of many hormones is (1)
of energy is expended to ensure that the intracellular
blunted by Ca2+-free media or when intracellular cal-
Ca2+ is controlled, as a prolonged elevation of Ca2+ in
cium is depleted; (2) can be mimicked by agents that
the cell is very toxic. A Na+/Ca2+ exchange mechanism
increase cytosolic Ca2+, such as the Ca2+ ionophore
that has a high capacity but low affinity pumps Ca2+
A23187; and (3) influences cellular calcium flux. The
out of cells. There also is a Ca2+/proton ATPase-depen-
regulation of glycogen metabolism in liver by vaso-
dent pump that extrudes Ca2+ in exchange for H+. This
pressin and α-adrenergic catecholamines provides a
has a high affinity for Ca2+ but a low capacity and is
good example. This is shown schematically in Figures
probably responsible for fine-tuning cytosolic Ca2+.
18-6 and 18-7.
Furthermore, Ca2+ ATPases pump Ca2+ from the cy-
tosol to the lumen of the endoplasmic reticulum. There
are three ways of changing cytosolic Ca2+: (1) Certain
hormones (class II.C, Table 42-3) by binding to recep-
Table 43-4. Enzymes and proteins regulated by
tors that are themselves Ca2+ channels, enhance mem-
brane permeability to Ca2+ and thereby increase Ca2+
calcium or calmodulin.
influx. (2) Hormones also indirectly promote Ca2+ in-
flux by modulating the membrane potential at the
Adenylyl cyclase
plasma membrane. Membrane depolarization opens
Ca2+-dependent protein kinases
voltage-gated Ca2+ channels and allows for Ca2+ influx.
Ca2+-Mg2+ ATPase
(3) Ca2+ can be mobilized from the endoplasmic reticu-
Ca2+-phospholipid-dependent protein kinase
lum, and possibly from mitochondrial pools.
Cyclic nucleotide phosphodiesterase
An important observation linking Ca2+ to hormone
Some cytoskeletal proteins
action involved the definition of the intracellular targets
Some ion channels (eg, L-type calcium channels)
Nitric oxide synthase
of Ca2+ action. The discovery of a Ca2+-dependent reg-
Phosphorylase kinase
ulator of phosphodiesterase activity provided the basis
Phosphoprotein phosphatase 2B
for a broad understanding of how Ca2+ and cAMP in-
Some receptors (eg, NMDA-type glutamate receptor)
teract within cells.
464
/
CHAPTER 43
A number of critical metabolic enzymes are regu-
face receptors such as those for acetylcholine, antidi-
lated by Ca2+, phosphorylation, or both, including
uretic hormone, and α1-type catecholamines are, when
glycogen synthase, pyruvate kinase, pyruvate carboxy-
occupied by their respective ligands, potent activators
lase, glycerol-3-phosphate dehydrogenase, and pyruvate
of phospholipase C. Receptor binding and activation of
dehydrogenase.
phospholipase C are coupled by the Gq isoforms (Table
43-3 and Figure 43-6). Phospholipase C catalyzes the
D. PHOSPHATIDYLINOSITIDE METABOLISM AFFECTS
hydrolysis of phosphatidylinositol 4,5-bisphosphate to
CA2+-DEPENDENT HORMONE ACTION
inositol trisphosphate (IP3) and 1,2-diacylglycerol (Fig-
Some signal must provide communication between the
ure 43-7). Diacylglycerol is itself capable of activating
hormone receptor on the plasma membrane and the in-
protein kinase C (PKC), the activity of which also de-
tracellular Ca2+ reservoirs. This is accomplished by
pends upon Ca2+. IP3, by interacting with a specific in-
products of phosphatidylinositol metabolism. Cell sur-
tracellular receptor, is an effective releaser of Ca2+ from
Ca2+
Receptor
G protein
Phospholipase C
PIP2
Diacylglycerol
+
Endoplasmic reticulum
Protein kinase C
Inositol-P3
(PKC)
(IP
3)
Mitochondrion
Ca2+
Calmodulin
Ca2+-Calmodulin
+
+
Specific
Multifunctional
calmodulin kinase calmodulin kinase
Proteins
Phosphoproteins
Other
proteins
Physiologic responses
Figure 43-6. Certain hormone-receptor interactions result in the activation of phospholipase C. This ap-
pears to involve a specific G protein, which also may activate a calcium channel. Phospholipase C results in
generation of inositol trisphosphate (IP3), which liberates stored intracellular Ca2+, and diacylglycerol (DAG),
a potent activator of protein kinase C (PKC). In this scheme, the activated PKC phosphorylates specific sub-
strates, which then alter physiologic processes. Likewise, the Ca2+-calmodulin complex can activate specific
kinases, two of which are shown here. These actions result in phosphorylation of substrates, and this leads to
altered physiologic responses. This figure also shows that Ca2+ can enter cells through voltage- or ligand-
gated Ca2+ channels. The intracellular Ca2+ is also regulated through storage and release by the mitochon-
dria and endoplasmic reticulum. (Courtesy of JH Exton.)
HORMONE ACTION & SIGNAL TRANSDUCTION
/
465
R1
R2
P
OH
P
OH
R1
R2
OH
Phospholipase C
1,2-Diacylglycerol
OH
(DAG)
P
P
Phosphatidylinositol 4,5-bisphosphate
OH
P
OH
(PIP2)
Figure 43-7. Phospholipase C cleaves PIP2
into diacylglycerol and inositol trisphosphate. R
1
OH
generally is stearate, and R2 is usually arachido-
nate. IP3
can be dephosphorylated (to the inac-
P
tive I-1,4-P2) or phosphorylated (to the potentially
Inositol 1,4,5-trisphosphate
(IP3)
active I-1,3,4,5-P4).
intracellular storage sites in the endoplasmic reticulum.
ligand-activated tyrosine kinase activity. Several recep-
Thus, the hydrolysis of phosphatidylinositol 4,5-bis-
tors—generally those involved in binding ligands in-
phosphate leads to activation of PKC and promotes an
volved in growth control, differentiation, and the in-
increase of cytoplasmic Ca2+. As shown in Figure 43-4,
flammatory response—either have intrinsic tyrosine
the activation of G proteins can also have a direct ac-
kinase activity or are associated with proteins that are
tion on Ca2+ channels. The resulting elevations of cy-
tyrosine kinases. Another distinguishing feature of this
tosolic Ca2+ activate Ca2+-calmodulin-dependent kinases
class of hormone action is that these kinases preferen-
and many other Ca2+-calmodulin-dependent enzymes.
tially phosphorylate tyrosine residues, and tyrosine
Steroidogenic agents—including ACTH and cAMP
phosphorylation is infrequent (< 0.03% of total amino
in the adrenal cortex; angiotensin II, K+, serotonin,
acid phosphorylation) in mammalian cells. A third dis-
ACTH, and cAMP in the zona glomerulosa of the
tinguishing feature is that the ligand-receptor interac-
adrenal; LH in the ovary; and LH and cAMP in the
tion that results in a tyrosine phosphorylation event ini-
Leydig cells of the testes—have been associated with in-
tiates a cascade that may involve several protein kinases,
creased amounts of phosphatidic acid, phosphatidyl-
phosphatases, and other regulatory proteins.
inositol, and polyphosphoinositides (see Chapter 14) in
A. INSULIN TRANSMITS SIGNALS
the respective target tissues. Several other examples
BY SEVERAL KINASE CASCADES
could be cited.
The roles that Ca2+ and polyphosphoinositide break-
The insulin, epidermal growth factor (EGF), and IGF-I
down products might play in hormone action are pre-
receptors have intrinsic protein tyrosine kinase activities
sented in Figure 43-6. In this scheme the activated pro-
located in their cytoplasmic domains. These activities
tein kinase C can phosphorylate specific substrates,
are stimulated when the receptor binds ligand. The re-
which then alter physiologic processes. Likewise, the
ceptors are then autophosphorylated on tyrosine
Ca2+-calmodulin complex can activate specific kinases.
residues, and this initiates a complex series of events
These then modify substrates and thereby alter physio-
(summarized in simplified fashion in Figure 43-8). The
logic responses.
phosphorylated insulin receptor next phosphorylates
insulin receptor substrates (there are at least four of
these molecules, called IRS 1-4) on tyrosine residues.
Some Hormones Act Through
Phosphorylated IRS binds to the Src homology
2
a Protein Kinase Cascade
(SH2) domains of a variety of proteins that are directly
Single protein kinases such as PKA, PKC, and Ca2+-
involved in mediating different effects of insulin. One
calmodulin (CaM)-kinases, which result in the phos-
of these proteins, PI-3 kinase, links insulin receptor ac-
phorylation of serine and threonine residues in target
tivation to insulin action through activation of a num-
proteins, play a very important role in hormone action.
ber of molecules, including the kinase PDK1 (phospho-
The discovery that the EGF receptor contains an intrin-
inositide-dependent kinase-1). This enzyme propagates
sic tyrosine kinase activity that is activated by the bind-
the signal through several other kinases, including PKB
ing of the ligand EGF was an important breakthrough.
(akt), SKG, and aPKC (see legend to Figure 43-8 for
The insulin and IGF-I receptors also contain intrinsic
definitions and expanded abbreviations). An alternative
466
/
CHAPTER 43
RECOGNITION
(HYPERGLYCEMIA)
INSULIN
P-Y
Y-P
Y
Y
IRS 1-4
IRS 1-4
Y
Y
SIGNAL
P-Y
Y-P
GRB2/mSOS
GENERATION
PI3 -
+
PTEN
+
kinase
mTOR
p21Ras
+
Raf-1
PKB
MEK
SGK
?
p70S6K
MAP
aPKC
kinase
Protein translocation
Enzyme activity
Gene transcription
Cell growth
DNA synthesis
EFFECTS
Glucose transporter
Insulin receptor
PEPCK
HKII
Early response
Insulin receptor
Protein phosphatases
Glucagon
Glucokinase
genes
IGF-II receptor
Phosphodiesterases*
IGFBP1
> 100 others
Others
Figure 43-8. Insulin signaling pathways. The insulin signaling pathways provide an excellent example of the
“recognition → hormone release → signal generation → effects” paradigm outlined in Figure 43-1. Insulin is re-
leased in response to hyperglycemia. Binding of insulin to a target cell-specific plasma membrane receptor results in
a cascade of intracellular events. Stimulation of the intrinsic tyrosine kinase activity of the insulin receptor marks the
initial event, resulting in increased tyrosine (Y) phosphorylation (Y → Y-P) of the receptor and then one or more of
the insulin receptor substrate molecules (IRS 1-4). This increase in phosphotyrosine stimulates the activity of many
intracellular molecules such as GTPases, protein kinases, and lipid kinases, all of which play a role in certain meta-
bolic actions of insulin. The two best-described pathways are shown. First, phosphorylation of an IRS molecule
(probably IRS-2) results in docking and activation of the lipid kinase, PI-3 kinase, which generates novel inositol lipids
that may act as “second messenger” molecules. These, in turn, activate PDK1 and then a variety of downstream sig-
naling molecules, including protein kinase B (PKB or akt), SGK, and aPKC. An alternative pathway involves the activa-
tion of p70S6K and perhaps other as yet unidentified kinases. Second, phosphorylation of IRS (probably IRS-1) re-
sults in docking of GRB2/mSOS and activation of the small GTPase, p21RAS, which initiates a protein kinase cascade
that activates Raf-1, MEK, and the p42/p44 MAP kinase isoforms. These protein kinases are important in the regula-
tion of proliferation and differentiation of several cell types. The mTOR pathway provides an alternative way of acti-
vating p70S6K and appears to be involved in nutrient signaling as well as insulin action. Each of these cascades may
influence different physiologic processes, as shown. Each of the phosphorylation events is reversible through the
action of specific phosphatases. For example, the lipid phosphatase PTEN dephosphorylates the product of the PI-3
kinase reaction, thereby antagonizing the pathway and terminating the signal. Representative effects of major ac-
tions of insulin are shown in each of the boxes. The asterisk after phosphodiesterase indicates that insulin indirectly
affects the activity of many enzymes by activating phosphodiesterases and reducing intracellular cAMP levels.
(IGFBP, insulin-like growth factor binding protein; IRS 1-4, insulin receptor substrate isoforms 1-4); PI-3 kinase, phos-
phatidylinositol 3-kinase; PTEN, phosphatase and tensin homolog deleted on chromosome 10; PKD1, phosphoinosi-
tide-dependent kinase; PKB, protein kinase B; SGK, serum and glucocorticoid-regulated kinase; aPKC, atypical pro-
tein kinase C; p70S6K, p70 ribosomal protein S6 kinase; mTOR, mammalian target of rapamycin; GRB2, growth factor
receptor binding protein 2; mSOS, mammalian son of sevenless; MEK, MAP kinase kinase and ERK kinase; MAP
kinase, mitogen-activated protein kinase.)
HORMONE ACTION & SIGNAL TRANSDUCTION
/
467
pathway downstream from PKD1 involves p70S6K and
balancing actions of phosphatases. Two mechanisms
perhaps other as yet unidentified kinases. A second major
are employed to initiate this cascade. Some hormones,
pathway involves mTOR. This enzyme is directly regu-
such as growth hormone, prolactin, erythropoietin, and
lated by amino acids and insulin and is essential for
the cytokines, initiate their action by activating a tyro-
p70S6K activity. This pathway provides a distinction
sine kinase, but this activity is not an integral part of
between the PKB and p70S6K branches downstream
the hormone receptor. The hormone-receptor interac-
from PKD1. These pathways are involved in protein
tion promotes binding and activation of cytoplasmic
translocation, enzyme activity, and the regulation, by
protein tyrosine kinases, such as Tyk-2, Jak1, or
insulin, of genes involved in metabolism (Figure 43-8).
Jak2. These kinases phosphorylate one or more cyto-
Another SH2 domain-containing protein is GRB2,
plasmic proteins, which then associate with other dock-
which binds to IRS-1 and links tyrosine phosphoryla-
ing proteins through binding to SH2 domains. One
tion to several proteins, the result of which is activation
such interaction results in the activation of a family of
of a cascade of threonine and serine kinases. A pathway
cytosolic proteins called signal transducers and activa-
showing how this insulin-receptor interaction activates
tors of transcription (STATs). The phosphorylated
the mitogen-activated protein (MAP) kinase pathway
STAT protein dimerizes and translocates into the nu-
and the anabolic effects of insulin is illustrated in Fig-
cleus, binds to a specific DNA element such as the in-
ure 43-8. The exact roles of many of these docking
terferon response element, and activates transcription.
proteins, kinases, and phosphatases remain to be estab-
This is illustrated in Figure 43-9. Other SH2 docking
lished.
events may result in the activation of PI 3-kinase, the
MAP kinase pathway (through SHC or GRB2), or G
B. THE JAK/STAT PATHWAY IS USED
protein-mediated activation of phospholipase C (PLCγ)
BY HORMONES AND CYTOKINES
with the attendant production of diacylglycerol and ac-
Tyrosine kinase activation can also initiate a phosphor-
tivation of protein kinase C. It is apparent that there is
ylation and dephosphorylation cascade that involves the
a potential for “cross-talk” when different hormones ac-
action of several other protein kinases and the counter-
tivate these various signal transduction pathways.
Ligand
R
R
R
R
R
R
JAK
JAK
P JAK
JAK
P P
JAK
JAK
P
P
P
P
P
STAT
P
P
P
P
SH2
P
P
X
x =
SHC
Dimerization
GRB2
and
PLCγ
nuclear
PI-3K
translocation
GAP
Figure 43-9. Initiation of signal transduction by receptors linked to Jak ki-
nases. The receptors (R) that bind prolactin, growth hormone, interferons, and cy-
tokines lack endogenous tyrosine kinase. Upon ligand binding, these receptors
dimerize and an associated protein (Jak1, Jak2, or TYK) is phosphorylated. Jak-P,
an active kinase, phosphorylates the receptor on tyrosine residues. The STAT pro-
teins associate with the phosphorylated receptor and then are themselves phos-
phorylated by Jak-P. STAT P dimerizes, translocates to the nucleus, binds to spe-
cific DNA elements, and regulates transcription. The phosphotyrosine residues of
the receptor also bind to several SH2 domain-containing proteins. This results in
activation of the MAP kinase pathway (through SHC or GRB2), PLCγ, or PI-3 kinase.
468
/
CHAPTER 43
C. THE NF- B PATHWAY IS
Glucocorticoid hormones are therapeutically useful
agents for the treatment of a variety of inflammatory and
REGULATED BY GLUCOCORTICOIDS
immune diseases. Their anti-inflammatory and im-
The transcription factor NF-κB is a heterodimeric
munomodulatory actions are explained in part by the in-
complex typically composed of two subunits termed
hibition of NF-κB and its subsequent actions. Evidence
p50 and p65 (Figure 43-10). Normally, NF-κB is kept
for three mechanisms for the inhibition of NF-κB by
sequestered in the cytoplasm in a transcriptionally inac-
glucocorticoids has been presented: (1) Glucocorticoids
tive form by members of the inhibitor of NF-κB (IκB)
increase IκB mRNA, which leads to an increase of IκB
family. Extracellular stimuli such as proinflammatory
protein and more efficient sequestration of NF-κB in the
cytokines, reactive oxygen species, and mitogens lead to
cytoplasm.
(2) The glucocorticoid receptor competes
activation of the IκB kinase complex, IKK, which is a
with NF-κB for binding to coactivators. (3) The gluco-
heterohexameric structure consisting of α, β, and γ sub-
corticoid receptor directly binds to the p65 subunit of
units. IKK phosphorylates IκB on two serine residues,
NF-κB and inhibits its activation (Figure 43-10).
and this targets IκB for ubiquitination and subsequent
degradation by the proteasome. Following IκB degra-
dation, free NF-κB can now translocate to the nucleus,
HORMONES CAN INFLUENCE
where it binds to a number of gene promoters and acti-
SPECIFIC BIOLOGIC EFFECTS BY
vates transcription, particularly of genes involved in the
MODULATING TRANSCRIPTION
inflammatory response. Transcriptional regulation by
NF-κB is mediated by a variety of coactivators such as
The signals generated as described above have to be
CREB binding protein (CBP), as described below (Fig-
translated into an action that allows the cell to effec-
ure 43-13).
tively adapt to a challenge (Figure 43-1). Much of this
NF-κB Activators
Proinflammatory cytokines
Bacterial and viral infection
Reactive oxygen species
Mitogens
IKK
complex
γ
γ
P P
Proteasome
α
α
IκB
β β
Ubiquitin
1
IκB
p50
p65
Cytoplasm
p50
p65
Nucleus
2
Coactivators
3
p50
p65
Target gene
Figure 43-10. Regulation of the NF-κB pathway. NF-κB consists of two sub-
units, p50 and p65, which regulate transcription of many genes when in the
nucleus. NF-κB is restricted from entering the nucleus by IκB, an inhibitor of
NF-κB. IκB binds to—and masks—the nuclear localization signal of NF-κB.
This cytoplasmic protein is phosphorylated by an IKK complex which is acti-
vated by cytokines, reactive oxygen species, and mitogens. Phosphorylated
IκB can be ubiquitinylated and degraded, thus releasing its hold on NF-κB. Glu-
cocorticoids affect many steps in this process, as described in the text.
HORMONE ACTION & SIGNAL TRANSDUCTION
/
469
Figure 43-11. The hormone response transcrip-
tion unit. The hormone response transcription unit
is an assembly of DNA elements and bound pro-
teins that interact, through protein-protein interac-
tions, with a number of coactivator or corepressor
molecules. An essential component is the hormone
response element which binds the ligand (
)-
bound receptor (R). Also important are the acces-
sory factor elements (AFEs) with bound transcrip-
AF
tion factors. More than two dozen of these
R
accessory factors (AFs), which are often members of
R
the nuclear receptor superfamily, have been linked
AF
p160
to hormone effects on transcription. The AFs can in-
teract with each other, with the liganded nuclear
p300
receptors, or with coregulators. These components
communicate with the basal transcription complex
through a coregulator complex that can consist of
BTC
one or more members of the p160, corepressor,
mediator-related, or CBP/p300 families (see
Table 43-6).
adaptation is accomplished through alterations in the
tion initiation site, but they may be located within the
rates of transcription of specific genes. Many different
coding region of the gene, in introns. HREs were de-
observations have led to the current view of how hor-
fined by the strategy illustrated in Figure 39-11. The
mones affect transcription. Some of these are as follows:
consensus sequences illustrated in Table 43-1 were ar-
(1) Actively transcribed genes are in regions of “open”
rived at through analysis of several genes regulated by a
chromatin (defined by a susceptibility to the enzyme
given hormone using simple, heterologous reporter sys-
DNase I), which allows for the access of transcription
tems (see Figure 39-10). Although these simple HREs
factors to DNA. (2) Genes have regulatory regions, and
bind the hormone-receptor complex more avidly than
transcription factors bind to these to modulate the fre-
surrounding DNA—or DNA from an unrelated
quency of transcription initiation. (3) The hormone-
source—and confer hormone responsiveness to a re-
receptor complex can be one of these transcription fac-
porter gene, it soon became apparent that the regula-
tors. The DNA sequence to which this binds is called a
tory circuitry of natural genes must be much more
hormone response element (HRE; see Table 43-1 for
complicated. Glucocorticoids, progestins, mineralocor-
examples). (4) Alternatively, other hormone-generated
ticoids, and androgens have vastly different physiologic
signals can modify the location, amount, or activity of
actions. How could the specificity required for these ef-
transcription factors and thereby influence binding to
fects be achieved through regulation of gene expression
the regulatory or response element. (5) Members of a
by the same HRE (Table 43-1)? Questions like this
large superfamily of nuclear receptors act with—or in a
have led to experiments which have allowed for elabora-
manner analogous to—hormone receptors. (6) These
tion of a very complex model of transcription regula-
nuclear receptors interact with another large group of
tion. For example, the HRE must associate with other
coregulatory molecules to effect changes in the tran-
DNA elements (and associated binding proteins) to
scription of specific genes.
function optimally. The extensive sequence similarity
noted between steroid hormone receptors, particularly
in their DNA-binding domains, led to discovery of the
Several Hormone Response Elements
nuclear receptor superfamily of proteins. These—and
(HREs) Have Been Defined
a large number of coregulator proteins—allow for a
Hormone response elements resemble enhancer ele-
wide variety of DNA-protein and protein-protein inter-
ments in that they are not strictly dependent on posi-
actions and the specificity necessary for highly regulated
tion and location. They generally are found within a
physiologic control. A schematic of such an assembly is
few hundred nucleotides upstream (5′) of the transcrip-
illustrated in Figure 43-11.
470
/
CHAPTER 43
A/B
C
D
E
F
N
AF-1
DBD
Hinge
LBD
AF-2
C
×
×
GR, MR, PR
TR, RAR, VDR
COUP-TF, TR2, GEN8
AR, ER
PPARα, β, γ
HNF-4, TLX
FXR, CAR, LXR,
PXR/SXR
Receptors:
Steroid class
RXR partnered
Orphans
Binding:
Homodimers
Heterodimers
Homodimers
Ligand:
Steroids
9-Cis RA + (x)
?
DNA element:
Inverted
Direct repeats
Direct repeats
repeat
Figure 43-12. The nuclear receptor superfamily. Members of this family are
divided into six structural domains (A-F). Domain A/B is also called AF-1, or the
modulator region, because it is involved in activating transcription. The C do-
main consists of the DNA-binding domain (DBD). The D region contains the
hinge, which provides flexibility between the DBD and the ligand-binding do-
main (LBD, region E). The amino (N) terminal part of region E contains AF-2, an-
other domain important for transactivation. The F region is poorly defined. The
functions of these domains are discussed in more detail in the text. Receptors
with known ligands, such as the steroid hormones, bind as homodimers on in-
verted repeat half-sites. Other receptors form heterodimers with the partner
RXR on direct repeat elements. There can be nucleotide spacers of one to five
bases between these direct repeats (DR1-5). Another class of receptors for
which ligands have not been determined (orphan receptors) bind as homo-
dimers to direct repeats and occasionally as monomers to a single half-site.
receptor [RXR] partner), or as monomers. The target
There Is a Large Family of Nuclear
response element consists of one or two half-site con-
Receptor Proteins
sensus sequences arranged as an inverted or direct re-
The nuclear receptor superfamily consists of a diverse set
peat. The spacing between the latter helps determine
of transcription factors that were discovered because of a
binding specificity. Thus, a direct repeat with three,
sequence similarity in their DNA-binding domains. This
four, or five nucleotide spacer regions specifies the
family, now with more than 50 members, includes the
binding of the vitamin D, thyroid, and retinoic acid re-
nuclear hormone receptors discussed above, a number of
ceptors, respectively, to the same consensus response
other receptors whose ligands were discovered after the
element
(Table
43-1). A multifunctional ligand-
receptors were identified, and many putative or orphan
binding domain (LBD) is located in the carboxyl ter-
receptors for which a ligand has yet to be discovered.
minal half of the receptor. The LBD binds hormones
These nuclear receptors have several common struc-
or metabolites with selectivity and thus specifies a par-
tural features (Figure 43-12). All have a centrally lo-
ticular biologic response. The LBD also contains do-
cated DNA-binding domain (DBD) that allows the
mains that mediate the binding of heat shock proteins,
receptor to bind with high affinity to a response ele-
dimerization, nuclear localization, and transactivation.
ment. The DBD contains two zinc finger binding mo-
The latter function is facilitated by the carboxyl termi-
tifs (see Figure 39-14) that direct binding either as ho-
nal transcription activation function (AF-2 domain),
modimers, as heterodimers (usually with a retinoid X
which forms a surface required for the interaction with
HORMONE ACTION & SIGNAL TRANSDUCTION
/
471
Group I
GH, Prl,
Insulin,
GPCR
Hormones
Cytokines, etc
EGF, etc
TNF, etc
Jak
cAMP
Retinoic acid,
RAS
Plasma
IRS
estrogen,
membrane
vitamin D,
NFκB•IκB
glucocorticoids,
MEK
etc
STATs
PKA
NFκB
MAPK
Nuclear
CREB
receptors
Nuclear
STATs
membrane
AP-1
NFκB
CBP
p300
Figure 43-13. Several signal transduction pathways converge on
CBP/p300. Ligands that associate with membrane or nuclear receptors even-
tually converge on CBP/p300. Several different signal transduction pathways
are employed. EGF, epidermal growth factor; GH, growth hormone; Prl, pro-
lactin; TNF, tumor necrosis factor; other abbreviations are expanded in the
text.
coactivators. A highly variable hinge region separates
Another group of orphan receptors that as yet have no
the DBD from the LBD. This region provides flexibil-
known ligand bind as homodimers or monomers to di-
ity to the receptor, so it can assume different DNA-
rect repeat sequences.
binding conformations. Finally, there is a highly vari-
As illustrated in Table 43-5, the discovery of the nu-
able amino terminal region that contains another trans-
clear receptor superfamily has led to an important un-
activation domain referred to as AF-1. Less well de-
derstanding of how a variety of metabolites and xenobi-
fined, the AF-1 domain may provide for distinct
otics regulate gene expression and thus the metabolism,
physiologic functions through the binding of different
detoxification, and elimination of normal body prod-
coregulator proteins. This region of the receptor,
ucts and exogenous agents such as pharmaceuticals.
through the use of different promoters, alternative
Not surprisingly, this area is a fertile field for investiga-
splice sites, and multiple translation initiation sites,
tion of new therapeutic interventions.
provides for receptor isoforms that share DBD and
LBD identity but exert different physiologic responses
A Large Number of Nuclear Receptor
because of the association of various coregulators with
Coregulators Also Participate
this variable amino terminal AF-1 domain.
in Regulating Transcription
It is possible to sort this large number of receptors
into groups in a variety of ways. Here they are discussed
Chromatin remodeling, transcription factor modification
according to the way they bind to their respective DNA
by various enzyme activities, and the communication
elements (Figure 43-12). Classic hormone receptors for
between the nuclear receptors and the basal transcrip-
glucocorticoids (GR), mineralocorticoids (MR), estro-
tion apparatus are accomplished by protein-protein in-
gens (ER), androgens (AR), and progestins (PR) bind
teractions with one or more of a class of coregulator
as homodimers to inverted repeat sequences. Other
molecules. The number of these coregulator molecules
hormone receptors such as thyroid (TR), retinoic acid
now exceeds 100, not counting species variations and
(RAR), and vitamin D (VDR) and receptors that bind
splice variants. The first of these to be described was the
various metabolite ligands such as PPAR α β, and γ,
CREB-binding protein, CBP. CBP, through an
FXR, LXR, PXR/SXR, and CAR bind as heterodimers,
amino terminal domain, binds to phosphorylated serine
with retinoid X receptor (RXR) as a partner, to direct
137 of CREB and mediates transactivation in response
repeat sequences (see Figure 43-12 and Table 43-5).
to cAMP. It thus is described as a coactivator. CBP and
472
/
CHAPTER 43
Table 43-5. Nuclear receptors with special ligands.1
Receptor
Partner
Ligand
Process Affected
Peroxisome PPARα
RXR (DR1)
Fatty acids
Peroxisome proliferation
Proliferator- PPARβ
Fatty acids
activated PPARγ
Fatty acids
Lipid and carbohydrate metabolism
Eicosanoids,
thiazolidinediones
Farnesoid X FXR
RXR (DR4)
Farnesol, bile acids
Bile acid metabolism
Liver X
LXR
RXR (DR4)
Oxysterols
Cholesterol metabolism
Xenobiotic X CAR
RXR (DR5)
Androstanes
Phenobarbital
Protection against certain drugs, toxic
Xenobiotics
metabolites, and xenobiotics
PXR
RXR (DR3)
Pregnanes
Xenobiotics
1Many members of the nuclear receptor superfamily were discovered by cloning, and the corresponding
ligands were then identified. These ligands are not hormones in the classic sense, but they do have a
similar function in that they activate specific members of the nuclear receptor superfamily. The recep-
tors described here form heterodimers with RXR and have variable nucleotide sequences separating the
direct repeat binding elements (DR1-5). These receptors regulate a variety of genes encoding cy-
tochrome p450s (CYP), cytosolic binding proteins, and ATP-binding cassette (ABC) transporters to influ-
ence metabolism and protect cells against drugs and noxious agents.
its
close
relative, p300, interact directly or indirectly
with a number of signaling molecules, including activa-
tor protein-1 (AP-1), signal transducers and activators
of transcription (STATs), nuclear receptors, and CREB
(Figure
39-11). CBP/p300 also binds to the p160
family of coactivators described below and to a number
Table 43-6. Some mammalian coregulator
of other proteins, including viral transcription factor
proteins.
Ela, the p90rsk protein kinase, and RNA helicase A. It is
important to note that CBP/p300 also has intrinsic
I.
300-kDa family of coactivators
histone acetyltransferase (HAT) activity. The impor-
CBP
CREB-binding protein
tance of this is described below. Some of the many ac-
p300
Protein of 300 kDa
tions of CBP/p300, which appear to depend on intrin-
II.
160-kDa family of coactivators
sic enzyme activities and its ability to serve as a scaffold
A. SRC-1
Steroid receptor coactivator 1
for the binding of other proteins, are illustrated in Fig-
NCoA-1
Nuclear receptor coactivator 1
ure 43-11. Other coregulators may serve similar func-
B. TIF2
Transcriptional intermediary factor 2
tions.
GRIP1
Glucocorticoid receptor-interacting protein
Several other families of coactivator molecules have
NCoA-2
Nuclear receptor coactivator 2
been described. Members of the p160 family of coac-
C. p/CIP
p300/CBP cointegrator-associated protein 1
tivators, all of about 160 kDa, include (1) SRC-1 and
ACTR Activator of the thyroid and retinoic acid
NCoA-1; (2) GRIP 1, TIF2, and NCoA-2; and (3)
receptors
p/CIP, ACTR, AIB1, RAC3, and TRAM-1 (Table
AIB
Amplified in breast cancer
43-6). The different names for members within a sub-
RAC3
Receptor-associated coactivator 3
family often represent species variations or minor splice
TRAM-1
TR activator molecule 1
variants. There is about 35% amino acid identity be-
III. Corepressors
tween members of the different subfamilies. The p160
NCoR
Nuclear receptor corepressor
coactivators share several properties. They
(1) bind
SMRT
Silencing mediator for RXR and TR
IV. Mediator-related proteins
nuclear receptors in an agonist and AF-2 transactiva-
TRAPs
Thyroid hormone receptor-associated
tion domain-dependent manner; (2) have a conserved
proteins
amino terminal basic helix-loop-helix (bHLH) motif
DRIPs
Vitamin D receptor-interacting proteins
(see Chapter 39); (3) have a weak carboxyl terminal
ARC
Activator-recruited cofactor
transactivation domain and a stronger amino terminal
HORMONE ACTION & SIGNAL TRANSDUCTION
/
473
transactivation domain in a region that is required for
• Many hormone responses are accomplished through
the CBP/p16O interaction; (4) contain at least three of
alterations in the rate of transcription of specific
the LXXLL motifs required for protein-protein inter-
genes.
action with other coactivators; and (5) often have HAT
• The nuclear receptor superfamily of proteins plays a
activity. The role of HAT is particularly interesting, as
central role in the regulation of gene transcription.
mutations of the HAT domain disable many of these
• These receptors, which may have hormones, metabo-
transcription factors. Current thinking holds that these
lites, or drugs as ligands, bind to specific DNA ele-
HAT activities acetylate histones and result in remodel-
ments as homodimers or as heterodimers with RXR.
ing of chromatin into a transcription-efficient environ-
Some—orphan receptors—have no known ligand
ment; however, other protein substrates for HAT-
but bind DNA and influence transcription.
mediated acetylation have been reported. Histone
• Another large family of coregulator proteins remodel
acetylation/deacetylation is proposed to play a critical
chromatin, modify other transcription factors, and
role in gene expression.
bridge the nuclear receptors to the basal transcription
A small number of proteins, including NCoR and
apparatus.
SMRT, comprise the corepressor family. They func-
tion, at least in part, as described in Figure 43-2. An-
other family includes the TRAPs, DRIPs, and ARC
REFERENCES
(Table 43-6). These so-called mediator-related pro-
teins range in size from 80 kDa to 240 kDa and are
Arvanitakis L et al: Constitutively signaling G-protein-coupled re-
ceptors and human disease. Trends Endocrinol Metab
thought to be involved in linking the nuclear receptor-
1998;9:27.
coactivator complex to RNA polymerase II and the
Berridge M: Inositol triphosphate and calcium signalling. Nature
other components of the basal transcription apparatus.
1993;361:315.
The exact role of these coactivators is presently
Chawla A et al: Nuclear receptors and lipid physiology: opening the
under intensive investigation. Many of these proteins
X files. Science 2001;294:1866.
have intrinsic enzymatic activities. This is particularly
Darnell JE Jr, Kerr IM, Stark GR: Jak-STAT pathways and trans-
interesting in view of the fact that acetylation, phos-
criptional activation in response to IFNs and other extracellu-
phorylation, methylation, and ubiquitination—as well
lar signaling proteins. Science 1994;264:1415.
as proteolysis and cellular translocation—have been
Fantl WJ, Johnson DE, Williams LT: Signalling by receptor tyro-
proposed to alter the activity of some of these coregula-
sine kinases. Annu Rev Biochem 1993;62:453.
tors and their targets.
Giguère V: Orphan nuclear receptors: from gene to function. En-
It appears that certain combinations of coregula-
docr Rev 1999;20:689.
tors—and thus different combinations of activators and
Grunstein M: Histone acetylation in chromatin structure and trans-
inhibitors—are responsible for specific ligand-induced
cription. Nature 1997;389:349.
actions through various receptors.
Hanoune J, Defer N: Regulation and role of adenylyl cyclase iso-
forms. Annu Rev Pharmacol Toxicol 2001;41:145.
Hermanson O, Glass CK, Rosenfeld MG: Nuclear receptor coregu-
SUMMARY
lators: multiple modes of receptor modification. Trends En-
• Hormones, cytokines, interleukins, and growth fac-
docrinol Metab 2002;13:55.
tors use a variety of signaling mechanisms to facilitate
Jaken S: Protein kinase C isozymes and substrates. Curr Opin Cell
cellular adaptive responses.
Biol 1996;8:168.
Lucas P, Granner D: Hormone response domains in gene transcrip-
• The ligand-receptor complex serves as the initial sig-
tion. Annu Rev Biochem 1992;61:1131.
nal for members of the nuclear receptor family.
Montminy M: Transcriptional regulation by cyclic AMP. Annu
• Class II hormones, which bind to cell surface recep-
Rev Biochem 1997;66:807
tors, generate a variety of intracellular signals. These
Morris AJ, Malbon CC: Physiological regulation of G protein-
include cAMP, cGMP, Ca2+, phosphatidylinositides,
linked signaling. Physiol Rev 1999;79:1373.
and protein kinase cascades.
Walton KM, Dixon JE: Protein tyrosine phosphatases. Annu Rev
Biochem 1993;62:101.
SECTION VI
Special Topics
Nutrition, Digestion, & Absorption
44
David A. Bender, PhD, & Peter A. Mayes, PhD, DSc
BIOMEDICAL IMPORTANCE
and steatorrhea. Lactose intolerance is due to lactase
deficiency leading to diarrhea and intestinal discomfort.
Besides water, the diet must provide metabolic fuels
Absorption of intact peptides that stimulate antibody
(mainly carbohydrates and lipids), protein (for growth
responses causes allergic reactions, and celiac disease
and turnover of tissue proteins), fiber (for roughage),
is an allergic reaction to wheat gluten.
minerals (elements with specific metabolic functions),
and vitamins and essential fatty acids (organic com-
pounds needed in small amounts for essential metabolic
DIGESTION & ABSORPTION
and physiologic functions). The polysaccharides, tri-
OF CARBOHYDRATES
acylglycerols, and proteins that make up the bulk of the
diet must be hydrolyzed to their constituent monosac-
The digestion of complex carbohydrates is by hydroly-
charides, fatty acids, and amino acids, respectively, be-
sis to liberate oligosaccharides, then free mono- and di-
fore absorption and utilization. Minerals and vitamins
saccharides. The increase in blood glucose after a test
must be released from the complex matrix of food be-
dose of a carbohydrate compared with that after an
fore they can be absorbed and utilized.
equivalent amount of glucose is known as the glycemic
Globally, undernutrition is widespread, leading to
index. Glucose and galactose have an index of 1, as do
impaired growth, defective immune systems, and re-
lactose, maltose, isomaltose, and trehalose, which give
duced work capacity. By contrast, in developed coun-
rise to these monosaccharides on hydrolysis. Fructose
tries, there is often excessive food consumption (espe-
and the sugar alcohols are absorbed less rapidly and
cially of fat), leading to obesity and to the development
have a lower glycemic index, as does sucrose. The
of cardiovascular disease and some forms of cancer. De-
glycemic index of starch varies between near 1 to near
ficiencies of vitamin A, iron, and iodine pose major
zero due to variable rates of hydrolysis, and that of non-
health concerns in many countries, and deficiencies of
starch polysaccharides is zero. Foods that have a low
other vitamins and minerals are a major cause of ill
glycemic index are considered to be more beneficial
health. In developed countries, nutrient deficiency is
since they cause less fluctuation in insulin secretion.
rare, though there are vulnerable sections of the popula-
tion at risk. Intakes of minerals and vitamins that are
adequate to prevent deficiency may be inadequate to
Amylases Catalyze
promote optimum health and longevity.
the Hydrolysis of Starch
Excessive secretion of gastric acid, associated with
Helicobacter pylori infection, can result in the develop-
The hydrolysis of starch by salivary and pancreatic
ment of gastric and duodenal ulcers; small changes in
amylases catalyze random hydrolysis of α(1→4) glyco-
the composition of bile can result in crystallization of
side bonds, yielding dextrins, then a mixture of glucose,
cholesterol as gallstones; failure of exocrine pancreatic
maltose, and isomaltose (from the branch points in
secretion (as in cystic fibrosis) leads to undernutrition
amylopectin).
474
NUTRITION, DIGESTION, & ABSORPTION
/
475
Disaccharidases Are Brush
SGLT 1
transporter
Border Enzymes
Glucose
protein
Glucose
Na+
The disaccharidases—maltase, sucrase-isomaltase
(a
Galactose
Glucose
bifunctional enzyme catalyzing hydrolysis of sucrose and
Fructose
Galactose
isomaltose), lactase, and trehalase—are located on the
brush border of the intestinal mucosal cells where the re-
GLUT 5
sultant monosaccharides and others arising from the diet
are absorbed. In most people, apart from those of north-
ern European genetic origin, lactase is gradually lost
through adolescence, leading to lactose intolerance.
Brush
border
Lactose remains in the intestinal lumen, where it is a
substrate for bacterial fermentation to lactate, resulting
in discomfort and diarrhea.
Na+ -K+
Intestinal
pump
There Are Two Separate Mechanisms
epithelium
for the Absorption of Monosaccharides
Na+
ATP
in the Small Intestine
3Na+
Glucose
Glucose and galactose are absorbed by a sodium-depen-
Fructose
2K+
dent process. They are carried by the same transport
Galactose
2K+
ADP
protein (SGLT 1) and compete with each other for in-
+ Pi
testinal absorption (Figure 44-1). Other monosaccha-
rides are absorbed by carrier-mediated diffusion. Be-
cause they are not actively transported, fructose and
To capillaries
sugar alcohols are only absorbed down their concentra-
GLUT 2
tion gradient, and after a moderately high intake some
Figure 44-1. Transport of glucose, fructose, and
may remain in the intestinal lumen, acting as a sub-
galactose across the intestinal epithelium. The SGLT 1
strate for bacterial fermentation.
transporter is coupled to the Na+-K+ pump, allowing
glucose and galactose to be transported against their
DIGESTION & ABSORPTION OF LIPIDS
concentration gradients. The GLUT 5 Na+-independent
The major lipids in the diet are triacylglycerols and, to a
facilitative transporter allows fructose as well as glu-
lesser extent, phospholipids. These are hydrophobic
cose and galactose to be transported with their con-
molecules and must be hydrolyzed and emulsified to
centration gradients. Exit from the cell for all the sugars
very small droplets (micelles) before they can be ab-
is via the GLUT 2 facilitative transporter.
sorbed. The fat-soluble vitamins—A, D, E, and K—
and a variety of other lipids (including cholesterol) are
absorbed dissolved in the lipid micelles. Absorption of
the fat-soluble vitamins is impaired on a very low fat
of the products of lipid digestion into micelles and lipo-
diet.
somes together with phospholipids and cholesterol
Hydrolysis of triacylglycerols is initiated by lingual
from the bile. Because the micelles are soluble, they
and gastric lipases that attack the sn-3 ester bond, form-
allow the products of digestion, including the fat-
ing 1,2-diacylglycerols and free fatty acids, aiding emul-
soluble vitamins, to be transported through the aqueous
sification. Pancreatic lipase is secreted into the small
environment of the intestinal lumen and permit close
intestine and requires a further pancreatic protein, coli-
contact with the brush border of the mucosal cells, al-
pase, for activity. It is specific for the primary ester
lowing uptake into the epithelium, mainly of the je-
links—ie, positions 1 and 3 in triacylglycerols—result-
junum. The bile salts pass on to the ileum, where
ing in 2-monoacylglycerols and free fatty acids as the
most are absorbed into the enterohepatic circula-
major end-products of luminal triacylglycerol digestion.
tion (Chapter 26). Within the intestinal epithelium,
Monoacylglycerols are hydrolyzed with difficulty to
1-monoacylglycerols are hydrolyzed to fatty acids and
glycerol and free fatty acids, so that less than 25% of in-
glycerol and 2-monoacylglycerols are re-acylated to tri-
gested triacylglycerol is completely hydrolyzed to glyc-
acylglycerols via the monoacylglycerol pathway. Glyc-
erol and fatty acids (Figure 44-2). Bile salts, formed in
erol released in the intestinal lumen is not reutilized but
the liver and secreted in the bile, enable emulsification
passes into the portal vein; glycerol released within the
NUTRITION, DIGESTION, & ABSORPTION
/
477
epithelium is reutilized for triacylglycerol synthesis via
several different amino acid transporters, with specificity
the normal phosphatidic acid pathway (Chapter 24).
for the nature of the amino acid side chain (large or
All long-chain fatty acids absorbed are converted to tri-
small; neutral, acidic, or basic). The various amino acids
acylglycerol in the mucosal cells and, together with the
carried by any one transporter compete with each other
other products of lipid digestion, secreted as chylomi-
for absorption and tissue uptake. Dipeptides and tripep-
crons into the lymphatics, entering the blood stream via
tides enter the brush border of the intestinal mucosal
the thoracic duct (Chapter 25).
cells, where they are hydrolyzed to free amino acids,
which are then transported into the hepatic portal vein.
Relatively large peptides may be absorbed intact, either
DIGESTION & ABSORPTION OF PROTEINS
by uptake into mucosal epithelial cells (transcellular) or
Few peptide bonds that are hydrolyzed by proteolytic
by passing between epithelial cells (paracellular). Many
enzymes are accessible without prior denaturation of di-
such peptides are large enough to stimulate antibody for-
etary proteins (by heat in cooking and by the action of
mation—this is the basis of allergic reactions to foods.
gastric acid).
DIGESTION & ABSORPTION
Several Groups of Enzymes Catalyze
OF VITAMINS & MINERALS
the Digestion of Proteins
Vitamins and minerals are released from food during
There are two main classes of proteolytic digestive en-
digestion—though this is not complete—and the avail-
zymes (proteases), with different specificities for the
ability of vitamins and minerals depends on the type of
amino acids forming the peptide bond to be hydrolyzed.
food and, especially for minerals, the presence of chelat-
Endopeptidases hydrolyze peptide bonds between spe-
ing compounds. The fat-soluble vitamins are absorbed
cific amino acids throughout the molecule. They are the
in the lipid micelles that result from fat digestion;
first enzymes to act, yielding a larger number of smaller
water-soluble vitamins and most mineral salts are
fragments, eg, pepsin in the gastric juice and trypsin,
absorbed from the small intestine either by active trans-
chymotrypsin, and elastase secreted into the small in-
port or by carrier-mediated diffusion followed by bind-
testine by the pancreas. Exopeptidases catalyze the hy-
ing to intracellular binding proteins to achieve concen-
drolysis of peptide bonds, one at a time, from the ends
tration upon uptake. Vitamin B12 absorption requires a
of polypeptides. Carboxypeptidases, secreted in the
specific transport protein, intrinsic factor; calcium ab-
pancreatic juice, release amino acids from the free car-
sorption is dependent on vitamin D; zinc absorption
boxyl terminal, and aminopeptidases, secreted by the
probably requires a zinc-binding ligand secreted by the
intestinal mucosal cells, release amino acids from the
exocrine pancreas; and the absorption of iron is limited.
amino terminal. Dipeptides, which are not substrates for
exopeptidases, are hydrolyzed in the brush border of in-
Calcium Absorption Is Dependent
testinal mucosal cells by dipeptidases.
on Vitamin D
The proteases are secreted as inactive zymogens; the
active site of the enzyme is masked by a small region of
In addition to its role in regulating calcium homeosta-
its peptide chain, which is removed by hydrolysis of a
sis, vitamin D is required for the intestinal absorption
specific peptide bond. Pepsinogen is activated to pepsin
of calcium. Synthesis of the intracellular calcium-
by gastric acid and by activated pepsin (autocatalysis). In
binding protein, calbindin, required for calcium ab-
the small intestine, trypsinogen, the precursor of
sorption, is induced by vitamin D, which also affects
trypsin, is activated by enteropeptidase, which is se-
the permeability of the mucosal cells to calcium, an ef-
creted by the duodenal epithelial cells; trypsin can then
fect that is rapid and independent of protein synthesis.
activate chymotrypsinogen to chymotrypsin, proelas-
Phytic acid (inositol hexaphosphate) in cereals binds
tase to elastase, procarboxypeptidase to carboxypepti-
calcium in the intestinal lumen, preventing its absorp-
dase, and proaminopeptidase to aminopeptidase.
tion. Other minerals, including zinc, are also chelated
by phytate. This is mainly a problem among people
who consume large amounts of unleavened whole
Free Amino Acids & Small Peptides Are
wheat products; yeast contains an enzyme, phytase,
Absorbed by Different Mechanisms
which dephosphorylates phytate, so rendering it inac-
The end product of the action of endopeptidases and
tive. High concentrations of fatty acids in the intestinal
exopeptidases is a mixture of free amino acids, di- and
lumen—as a result of impaired fat absorption—can
tripeptides, and oligopeptides, all of which are absorbed.
also reduce calcium absorption by forming insoluble
Free amino acids are absorbed across the intestinal mu-
calcium salts; a high intake of oxalate can sometimes
cosa by sodium-dependent active transport. There are
cause deficiency, since calcium oxalate is insoluble.
478
/
CHAPTER 44
Iron Absorption Is Limited
lized is carbohydrate, fat, or protein. Measurement of
& Strictly Controlled but Is
the ratio of the volume of carbon dioxide produced to
Enhanced by Vitamin C & Ethanol
volume of oxygen consumed (respiratory quotient; RQ)
is an indication of the mixture of metabolic fuels being
Although iron deficiency is a common problem, about
oxidized (Table 27-1). A more recent technique per-
10% of the population are genetically at risk of iron
mits estimation of total energy expenditure over a pe-
overload (hemochromatosis), and elemental iron can
riod of 1-2 weeks using dual isotopically labeled water,
lead to nonenzymic generation of free radicals. Absorp-
2H218O. 2H is lost from the body only in water, while
tion of iron is strictly regulated. Inorganic iron is accu-
18O is lost in both water and carbon dioxide; the differ-
mulated in intestinal mucosal cells bound to an intra-
ence in the rate of loss of the two labels permits estima-
cellular protein, ferritin. Once the ferritin in the cell is
tion of total carbon dioxide production and thus oxy-
saturated with iron, no more can enter. Iron can only
gen consumption and energy expenditure.
leave the mucosal cell if there is transferrin in plasma
Basal metabolic rate (BMR) is the energy expendi-
to bind to. Once transferrin is saturated with iron, any
ture by the body when at rest—but not asleep—under
that has accumulated in the mucosal cells will be lost
controlled conditions of thermal neutrality, measured
when the cells are shed. As a result of this mucosal bar-
at about 12 hours after the last meal, and depends on
rier, only about 10% of dietary iron is normally ab-
weight, age, and gender. Total energy expenditure de-
sorbed and only 1-5% from many plant foods.
pends on the basal metabolic rate, the energy required
Inorganic iron is absorbed only in the Fe2+ (reduced)
for physical activity, and the energy cost of synthesizing
state, and for that reason the presence of reducing agents
reserves in the fed state. It is therefore possible to calcu-
will enhance absorption. The most effective compound
late an individual’s energy requirement from body
is vitamin C, and while intakes of 40-60 mg of vitamin
weight, age, gender, and level of physical activity. Body
C per day are more than adequate to meet requirements,
weight affects BMR because there is a greater amount
an intake of 25-50 mg per meal will enhance iron ab-
of active tissue in a larger body. The decrease in BMR
sorption, especially when iron salts are used to treat iron
with increasing age, even when body weight remains
deficiency anemia. Ethanol and fructose also enhance
constant, is due to muscle tissue replacement by adi-
iron absorption. Heme iron from meat is absorbed sepa-
pose tissue, which is metabolically much less active.
rately and is considerably more available than inorganic
Similarly, women have a significantly lower BMR than
iron. However, the absorption of both inorganic and
do men of the same body weight because women’s bod-
heme iron is impaired by calcium—a glass of milk with
ies have proportionately more adipose tissue than men.
a meal significantly reduces availability.
Energy Requirements Increase
ENERGY BALANCE:
With Activity
OVER- & UNDERNUTRITION
The most useful way of expressing the energy cost of
After the provision of water, the body’s first requirement
physical activities is as a multiple of BMR. Sedentary
is for metabolic fuels—fats, carbohydrates, and amino
activities use only about 1.1-1.2 × BMR. By contrast,
acids from proteins (and ethanol) (Table 27-1). Food in-
vigorous exertion, such as climbing stairs, cross-country
take in excess of energy expenditure leads to obesity,
skiing, walking uphill, etc, may use 6-8 × BMR.
while intake less than expenditure leads to emaciation
and wasting, as in marasmus and kwashiorkor. Both
Ten Percent of the Energy Yield of a Meal
obesity and severe undernutrition are associated with in-
May Be Expended in Forming Reserves
creased mortality. The body mass index, defined as
weight in kilograms divided by height in meters squared,
There is a considerable increase in metabolic rate after a
is commonly used as a way of expressing relative obesity
meal, a phenomenon known as diet-induced thermo-
to height. A desirable range is between 20 and 25.
genesis. A small part of this is the energy cost of secret-
ing digestive enzymes and of active transport of the
Energy Requirements Are Estimated by
products of digestion; the major part is due to synthe-
sizing reserves of glycogen, triacylglycerol, and protein.
Measurement of Energy Expenditure
Energy expenditure can be determined directly by mea-
There Are Two Extreme Forms
suring heat output from the body but is normally esti-
of Undernutrition
mated indirectly from the consumption of oxygen.
There is an energy expenditure of 20 kJ/L of oxygen
Marasmus can occur in both adults and children and
consumed regardless of whether the fuel being metabo-
occurs in vulnerable groups of all populations. Kwash-
NUTRITION, DIGESTION, & ABSORPTION
/
479
iorkor only affects children and has only been reported
the liver due to accumulation of fat. It was formerly be-
in developing countries. The distinguishing feature of
lieved that the cause of kwashiorkor was a lack of pro-
kwashiorkor is that there is fluid retention, leading to
tein, with a more or less adequate energy intake; how-
edema. Marasmus is a state of extreme emaciation; it is
ever, analysis of the diets of affected children shows that
the outcome of prolonged negative energy balance. Not
this is not so. Children with kwashiorkor are less
only have the body’s fat reserves been exhausted, but
stunted than those with marasmus, and the edema be-
there is wastage of muscle as well, and as the condition
gins to improve early in treatment, when the child is
progresses there is loss of protein from the heart, liver,
still receiving a low-protein diet. Very commonly, an
and kidneys. The amino acids released by the catabo-
infection precipitates kwashiorkor. Superimposed on
lism of tissue proteins are used as a source of metabolic
general food deficiency, there is probably a deficiency
fuel and as substrates for gluconeogenesis to maintain a
of the antioxidant nutrients such as zinc, copper,
supply of glucose for the brain and red blood cells. As a
carotene, and vitamins C and E. The respiratory burst
result of the reduced synthesis of proteins, there is im-
in response to infection leads to the production of oxy-
paired immune response and more risk from infections.
gen and halogen free radicals as part of the cytotoxic
Impairment of cell proliferation in the intestinal mu-
action of stimulated macrophages. This added oxidant
cosa occurs, resulting in reduction in surface area of the
stress may well trigger the development of kwashiorkor.
intestinal mucosa and reduction in absorption of such
nutrients as are available.
PROTEIN & AMINO ACID REQUIREMENTS
Patients With Advanced Cancer
Protein Requirements Can Be Determined
& AIDS Are Malnourished
by Measuring Nitrogen Balance
Patients with advanced cancer, HIV infection and
The state of protein nutrition can be determined by
AIDS, and a number of other chronic diseases are fre-
measuring the dietary intake and output of nitrogenous
quently undernourished—the condition is called
compounds from the body. Although nucleic acids also
cachexia. Physically, they show all the signs of maras-
contain nitrogen, protein is the major dietary source of
mus, but there is considerably more loss of body pro-
nitrogen and measurement of total nitrogen intake
tein than occurs in starvation. The secretion of cy-
gives a good estimate of protein intake (mg N × 6.25 =
tokines in response to infection and cancer increases the
mg protein, as nitrogen is 16% of most proteins). The
catabolism of tissue protein. This differs from maras-
output of nitrogen from the body is mainly in urea and
mus, in which protein synthesis is reduced but catabo-
smaller quantities of other compounds in urine and
lism in unaffected. Patients are hypermetabolic, ie,
undigested protein in feces, and significant amounts
there is a considerable increase in basal metabolic rate.
may also be lost in sweat and shed skin.
Many tumors metabolize glucose anaerobically to re-
The difference between intake and output of nitroge-
lease lactate. This is used for gluconeogenesis in the
nous compounds is known as nitrogen balance. Three
liver, which is energy-consuming with a net cost of six
states can be defined: In a healthy adult, nitrogen bal-
ATP for each mole of glucose cycled (Chapter 19).
ance is in equilibrium when intake equals output, and
There is increased stimulation of uncoupling proteins
there is no change in the total body content of protein.
by cytokines, leading to thermogenesis and increased
In a growing child, a pregnant woman, or in recovery
oxidation of metabolic fuels. Futile cycling of lipids oc-
from protein loss, the excretion of nitrogenous com-
curs because hormone-sensitive lipase is activated by a
pounds is less than the dietary intake and there is net re-
proteoglycan secreted by tumors, resulting in liberation
tention of nitrogen in the body as protein, ie, positive
of fatty acids from adipose tissue and ATP-expensive
nitrogen balance. In response to trauma or infection—
reesterification in the liver to triacylglycerols, which are
or if the intake of protein is inadequate to meet require-
exported in VLDL.
ments—there is net loss of protein nitrogen from the
body, ie, negative nitrogen balance. The continual ca-
tabolism of tissue proteins creates the requirement for
Kwashiorkor Affects
dietary protein even in an adult who is not growing,
Undernourished Children
though some of the amino acids released can be reuti-
In addition to the wasting of muscle tissue, loss of in-
lized. Nitrogen balance studies show that the average
testinal mucosa, and impaired immune responses seen
daily requirement is 0.6 g of protein per kilogram of
in marasmus, children with kwashiorkor show a num-
body weight (the factor 0.75 should be used to allow for
ber of characteristic features. The defining characteristic
individual variation), or approximately 50 g/d. Average
is edema, associated with a decreased concentration of
intakes of protein in developed countries are about
plasma proteins. In addition, there is enlargement of
80-100 g/d, ie,
14-15% of energy intake. Because
480
/
CHAPTER 44
growing children are increasing the protein in the body,
aloacetate, and α-ketoglutarate, respectively). The re-
they have a proportionately greater requirement than
maining amino acids are considered as nonessential, but
adults and should be in positive nitrogen balance. Even
under some circumstances the requirement for them
so, the need is relatively small compared with the re-
may outstrip the organism’s capacity for synthesis.
quirement for protein turnover. In some countries, pro-
tein intake may be inadequate to meet these require-
SUMMARY
ments, resulting in stunting of growth.
•
Digestion involves hydrolyzing food molecules into
There Is a Loss of Body Protein in
smaller molecules for absorption through the
Response to Trauma & Infection
gastrointestinal epithelium. Polysaccharides are
absorbed as monosaccharides; triacylglycerols as
One of the metabolic reactions to major trauma, such
2-monoacylglycerols, fatty acids, and glycerol; and
as a burn, a broken limb, or surgery, is an increase in
proteins as amino acids.
the net catabolism of tissue proteins. As much as 6-7%
•
Digestive disorders arise as a result of (1) enzyme de-
of the total body protein may be lost over 10 days. Pro-
ficiency, eg, lactase and sucrase; (2) malabsorption,
longed bed rest results in considerable loss of protein
eg, of glucose and galactose due to defects in the
because of atrophy of muscles. Protein is catabolized as
Na+-glucose cotransporter (SGLT 1); (3) absorption
normal, but without the stimulus of exercise it is not
of unhydrolyzed polypeptides, leading to immuno-
completely replaced. Lost protein is replaced during
logic responses, eg, as in celiac disease; and (4) pre-
convalescence, when there is positive nitrogen balance.
cipitation of cholesterol from bile as gallstones.
A normal diet is adequate to permit this replacement.
•
Besides water, the diet must provide metabolic fuels
(carbohydrate and fat) for bodily growth and activity;
The Requirement Is Not for Protein Itself
protein for synthesis of tissue proteins; fiber for
but for Specific Amino Acids
roughage; minerals for specific metabolic functions;
Not all proteins are nutritionally equivalent. More of
certain polyunsaturated fatty acids of the n-3 and n-6
some than of others is needed to maintain nitrogen
families for eicosanoid synthesis and other functions;
balance because different proteins contain different
and vitamins, organic compounds needed in small
amounts of the various amino acids. The body’s re-
amounts for many varied essential functions.
quirement is for specific amino acids in the correct
•
Twenty different amino acids are required for pro-
proportions to replace the body proteins. The amino
tein synthesis, of which nine are essential in the
acids can be divided into two groups: essential and
human diet. The quantity of protein required is af-
nonessential. There are nine essential or indispensable
fected by protein quality, energy intake, and physical
amino acids, which cannot be synthesized in the body:
activity.
histidine, isoleucine, leucine, lysine, methionine, phen-
•
Undernutrition occurs in two extreme forms: maras-
ylalanine, threonine, tryptophan, and valine. If one of
mus in adults and children and kwashiorkor in chil-
these is lacking or inadequate, then—regardless of the
dren. Overnutrition from excess energy intake is as-
total intake of protein—it will not be possible to main-
sociated with diseases such as obesity, type 2 diabetes
tain nitrogen balance since there will not be enough of
mellitus, atherosclerosis, cancer, and hypertension.
that amino acid for protein synthesis.
Two amino acids—cysteine and tyrosine—can be
synthesized in the body, but only from essential amino
REFERENCES
acid precursors (cysteine from methionine and tyrosine
Bender DA, Bender AE: Nutrition: A Reference Handbook. Oxford
from phenylalanine). The dietary intakes of cysteine
Univ Press, 1997.
and tyrosine thus affect the requirements for methio-
Büller HA, Grand RJ: Lactose intolerance. Annu Rev Med
nine and phenylalanine. The remaining 11 amino acids
1990;41:141.
in proteins are considered to be nonessential or dispens-
Fuller MF, Garlick PJ: Human amino acid requirements. Annu
able, since they can be synthesized as long as there is
Rev Nutr 1994;14:217.
enough total protein in the diet—ie, if one of these
Garrow JS, James WPT, Ralph A: Human Nutrition and Dietetics,
amino acids is omitted from the diet, nitrogen balance
10th ed. Churchill-Livingstone, 2000.
can still be maintained. However, only three amino
National Academy of Sciences report on diet and health. Nutr Rev
acids—alanine, aspartate, and glutamate—can be con-
1989;47:142.
sidered to be truly dispensable; they are synthesized
Nielsen FH: Nutritional significance of the ultratrace elements.
from common metabolic intermediates (pyruvate, ox-
Nutr Rev 1988;46:337.
Vitamins & Minerals
45
David A. Bender, PhD, & Peter A. Mayes, PhD, DSc
BIOMEDICAL IMPORTANCE
mia (iron), cretinism and goiter (iodine). If present in
excess as with selenium, toxicity symptoms may occur.
Vitamins are a group of organic nutrients required in
small quantities for a variety of biochemical functions
THE DETERMINATION OF NUTRIENT
and which, generally, cannot be synthesized by the
body and must therefore be supplied in the diet.
REQUIREMENTS DEPENDS ON THE
The lipid-soluble vitamins are apolar hydrophobic
CRITERIA OF ADEQUACY CHOSEN
compounds that can only be absorbed efficiently when
For any nutrient, particularly minerals and vitamins,
there is normal fat absorption. They are transported in
there is a range of intakes between that which is clearly
the blood, like any other apolar lipid, in lipoproteins or
inadequate, leading to clinical deficiency disease, and
attached to specific binding proteins. They have diverse
that which is so much in excess of the body’s metabolic
functions, eg, vitamin A, vision; vitamin D, calcium
capacity that there may be signs of toxicity. Between
and phosphate metabolism; vitamin E, antioxidant; vi-
these two extremes is a level of intake that is adequate
tamin K, blood clotting. As well as dietary inadequacy,
for normal health and the maintenance of metabolic in-
conditions affecting the digestion and absorption of the
tegrity. Individuals do not all have the same require-
lipid-soluble vitamins—such as steatorrhea and disor-
ment for nutrients even when calculated on the basis of
ders of the biliary system—can all lead to deficiency
body size or energy expenditure. There is a range of in-
syndromes, including: night blindness and xeroph-
dividual requirements of up to 25% around the mean.
thalmia (vitamin A); rickets in young children and os-
Therefore, in order to assess the adequacy of diets, it is
teomalacia in adults (vitamin D); neurologic disorders
necessary to set a reference level of intake high enough
and anemia of the newborn (vitamin E); and hemor-
to ensure that no one will either suffer from deficiency
rhage of the newborn (vitamin K). Toxicity can result
or be at risk of toxicity. If it is assumed that individual
from excessive intake of vitamins A and D. Vitamin A
requirements are distributed in a statistically normal
and β-carotene (provitamin A), as well as vitamin E, are
fashion around the observed mean requirement, then a
antioxidants and have possible roles in atherosclerosis
range of +/− 2 × the standard deviation (SD) around
and cancer prevention.
the mean will include the requirements of 95% of the
The water-soluble vitamins comprise the B complex
population.
and vitamin C and function as enzyme cofactors. Folic
acid acts as a carrier of one-carbon units. Deficiency of
a single vitamin of the B complex is rare, since poor
THE VITAMINS ARE A DISPARATE GROUP
diets are most often associated with multiple deficiency
OF COMPOUNDS WITH A VARIETY
states. Nevertheless, specific syndromes are characteris-
OF METABOLIC FUNCTIONS
tic of deficiencies of individual vitamins, eg, beriberi
(thiamin); cheilosis, glossitis, seborrhea
(riboflavin);
A vitamin is defined as an organic compound that is re-
pellagra
(niacin); peripheral neuritis
(pyridoxine);
quired in the diet in small amounts for the maintenance
megaloblastic anemia, methylmalonic aciduria, and
of normal metabolic integrity. Deficiency causes a spe-
pernicious anemia
(vitamin B12); and megaloblastic
cific disease, which is cured or prevented only by restor-
anemia (folic acid). Vitamin C deficiency leads to
ing the vitamin to the diet (Table 45-1). However, vit-
scurvy.
amin D, which can be made in the skin after exposure
Inorganic mineral elements that have a function in
to sunlight, and niacin, which can be formed from the
the body must be provided in the diet. When the intake
essential amino acid tryptophan, do not strictly con-
is insufficient, deficiency symptoms may arise, eg, ane-
form to this definition.
481
482
/
CHAPTER 45
Table 45-1. The vitamins.
Vitamin
Functions
Deficiency Disease
A
Retinol, β-carotene
Visual pigments in the retina; regulation of
Night blindness, xerophthalmia;
gene expression and cell differentiation;
keratinization of skin
β-carotene is an antioxidant
D
Calciferol
Maintenance of calcium balance; enhances
Rickets = poor mineralization of bone;
intestinal absorption of Ca2+ and mobilizes
osteomalacia = bone demineralization
bone mineral
E
Tocopherols, tocotrienols
Antioxidant, especially in cell membranes
Extremely rare—serious neurologic
dysfunction
K
Phylloquinone,
Coenzyme in formation of γ -carboxyglutamate
Impaired blood clotting, hemor-
menaquinones
in enzymes of blood clotting and bone matrix
rhagic disease
B1
Thiamin
Coenzyme in pyruvate and α−ketoglutarate,
Peripheral nerve damage (beriberi) or
dehydrogenases, and transketolase; poorly
central nervous system lesions
defined function in nerve conduction
(Wernicke-Korsakoff syndrome)
B2
Riboflavin
Coenzyme in oxidation and reduction reactions;
Lesions of corner of mouth, lips, and
prosthetic group of flavoproteins
tongue; seborrheic dermatitis
Niacin
Nicotinic acid,
Coenzyme in oxidation and reduction reactions,
Pellagra—photosensitive dermatitis,
nicotinamide
functional part of NAD and NADP
depressive psychosis
B6
Pyridoxine, pyridoxal,
Coenzyme in transamination and decarboxy-
Disorders of amino acid metabolism,
pyridoxamine
lation of amino acids and glycogen
convulsions
phosphorylase; role in steroid hormone action
Folic acid
Coenzyme in transfer of one-carbon fragments
Megaloblastic anemia
B12
Cobalamin
Coenzyme in transfer of one-carbon fragments
Pernicious anemia = megaloblastic
and metabolism of folic acid
anemia with degeneration of the
spinal cord
Pantothenic acid
Functional part of CoA and acyl carrier protein:
fatty acid synthesis and metabolism
H
Biotin
Coenzyme in carboxylation reactions in gluco-
Impaired fat and carbohydrate metab-
neogenesis and fatty acid synthesis
olism, dermatitis
C
Ascorbic acid
Coenzyme in hydroxylation of proline and
Scurvy—impaired wound healing,
lysine in collagen synthesis; antioxidant;
loss of dental cement, subcutaneous
enhances absorption of iron
hemorrhage
provitamin A, as they can be cleaved to yield retinalde-
hyde and thence retinol and retinoic acid. The α-, β-,
LIPID-SOLUBLE VITAMINS
and γ-carotenes and cryptoxanthin are quantitatively
the most important provitamin A carotenoids. Al-
RETINOIDS & CAROTENOIDS
though it would appear that one molecule of β-
carotene should yield two of retinol, this is not so in
HAVE VITAMIN A ACTIVITY
practice; 6 µg of β-carotene is equivalent to 1 µg of
(Figure 45-1)
preformed retinol. The total amount of vitamin A in
foods is therefore expressed as micrograms of retinol
Retinoids comprise retinol, retinaldehyde, and
equivalents. Beta-carotene and other provitamin A
retinoic acid (preformed vitamin A, found only in
carotenoids are cleaved in the intestinal mucosa by
foods of animal origin); carotenoids, found in plants,
carotene dioxygenase, yielding retinaldehyde, which is
comprise carotenes and related compounds, known as
reduced to retinol, esterified, and secreted in chylomi-
VITAMINS & MINERALS
/
483
H3C
CH3
CH3
H3C
CH3
H3C
CH3
CH3
CH3
CH3
β
-Carotene
CH3
CH3
CH3
CH3
H3C
CH3
H3C
CH3
CH2OH
CHO
CH3
Retinol
CH3
Retinaldehyde
CH3
CH3
CH3
H3C
CH3
H3C
CH3
COOH
CH3
CH3
Figure 45-1. β-Carotene and the major vita-
min A vitamers. * Shows the site of cleavage of
H3C
All-trans-retinoic acid
β-carotene into two molecules of retinaldehyde
COOH
by carotene dioxygenase.
9-cis-retinoic acid
crons together with esters formed from dietary retinol.
ciency, both the time taken to adapt to darkness and the
The intestinal activity of carotene dioxygenase is low,
ability to see in poor light are impaired.
so that a relatively large proportion of ingested β-
carotene may appear in the circulation unchanged.
Retinoic Acid Has a Role
While the principal site of carotene dioxygenase attack
in the Regulation of Gene
is the central bond of β-carotene, asymmetric cleavage
Expression & Tissue Differentiation
may also occur, leading to the formation of 8′-, 10′-,
and 12′-apo-carotenals, which are oxidized to retinoic
A most important function of vitamin A is in the con-
acid but cannot be used as sources of retinol or retin-
trol of cell differentiation and turnover. All-trans-
aldehyde.
retinoic acid and 9-cis-retinoic acid (Figure 45-1) regu-
late growth, development, and tissue differentiation;
they have different actions in different tissues. Like the
Vitamin A Has a Function in Vision
steroid hormones and vitamin D, retinoic acid binds to
In the retina, retinaldehyde functions as the prosthetic
nuclear receptors that bind to response elements of
group of the light-sensitive opsin proteins, forming
DNA and regulate the transcription of specific genes.
rhodopsin (in rods) and iodopsin (in cones). Any one
There are two families of nuclear retinoid receptors: the
cone cell contains only one type of opsin and is sensitive
retinoic acid receptors
(RARs) bind all-trans-retinoic
to only one color. In the pigment epithelium of the
acid or 9-cis-retinoic acid, and the retinoid X receptors
retina, all-trans-retinol is isomerized to
11-cis-retinol
(RXRs) bind 9-cis-retinoic acid.
and oxidized to 11-cis-retinaldehyde. This reacts with a
lysine residue in opsin, forming the holoprotein
Vitamin A Deficiency Is a Major Public
rhodopsin. As shown in Figure 45-2, the absorption of
Health Problem Worldwide
light by rhodopsin causes isomerization of the retinalde-
hyde from 11-cis to all-trans, and a conformational
Vitamin A deficiency is the most important preventable
change in opsin. This results in the release of retinalde-
cause of blindness. The earliest sign of deficiency is a
hyde from the protein and the initiation of a nerve im-
loss of sensitivity to green light, followed by impair-
pulse. The formation of the initial excited form of
ment of adaptation to dim light, followed by night
rhodopsin, bathorhodopsin, occurs within picoseconds
blindness. More prolonged deficiency leads to xeroph-
of illumination. There is then a series of conformational
thalmia: keratinization of the cornea and skin and
changes leading to the formation of metarhodopsin II,
blindness. Vitamin A also has an important role in dif-
which initiates a guanine nucleotide amplification cas-
ferentiation of immune system cells, and mild defi-
cade and then a nerve impulse. The final step is hydroly-
ciency leads to increased susceptibility to infectious dis-
sis to release all-trans-retinaldehyde and opsin. The key
eases. Furthermore, the synthesis of retinol-binding
to initiation of the visual cycle is the availability of
protein in response to infection is reduced (it is a nega-
11-cis-retinaldehyde, and hence vitamin A. In defi-
tive acute phase protein), decreasing the circulating vi-
484
/
CHAPTER 45
CH3
CH
ataxia, and anorexia, all associated with increased cere-
H3C
CH3
3
CH2OH
brospinal fluid pressure), the liver (hepatomegaly with
histologic changes and hyperlipidemia), calcium ho-
CH3
All-trans-retinol
meostasis (thickening of the long bones, hypercalcemia
CH3
and calcification of soft tissues), and the skin (excessive
H3C
CH3
dryness, desquamation, and alopecia).
11-cis-Retinol
CH3 H3C
CH2OH
VITAMIN D IS REALLY A HORMONE
CH3
H3C
CH3
Vitamin D is not strictly a vitamin since it can be syn-
11-cis-Retinaldehyde
thesized in the skin, and under most conditions that is
CH3 H3C
its major source. Only when sunlight is inadequate is a
HC=O
C=O
dietary source required. The main function of vitamin
H2N
D is in the regulation of calcium absorption and ho-
CH3
Lysine residue
H3C
CH3
meostasis; most of its actions are mediated by way
in opsin
NH
of nuclear receptors that regulate gene expression.
CH3 H3C
Deficiency—leading to rickets in children and osteo-
C=O
HC=N
malacia in adults—continues to be a problem in north-
Rhodopsin (visual purple)
ern latitudes, where sunlight exposure is poor.
NH
LIGHT
10-15sec
CH3
CH3
C=O
Vitamin D Is Synthesized in the Skin
H3C
CH3
C=N
7-Dehydrocholesterol (an intermediate in the synthesis
CH3
NH
of cholesterol that accumulates in the skin), undergoes
Photorhodopsin
a nonenzymic reaction on exposure to ultraviolet light,
45 psec
yielding previtamin D (Figure 45-3). This undergoes a
5'GMP
Bathorhodopsin
further reaction over a period of hours to form the vita-
+
cGMP
Na channel closed
30 nsec
min itself, cholecalciferol, which is absorbed into the
+
Na channel open
Lumirhodopsin
bloodstream. In temperate climates, the plasma concen-
75 µsec
Inactive
Active
tration of vitamin D is highest at the end of summer
phosphodiesterase
Metarhodopsin I
and lowest at the end of winter. Beyond about 40 de-
GDP
Transducin-GTP
grees north or south in winter, there is very little ultra-
10 msec
Metarhodopsin II
violet radiation of appropriate wavelength.
minutes
GTP
Transducin-GDP
Pi
Metarhodopsin III
Vitamin D Is Metabolized to the Active
Metabolite, Calcitriol, in Liver & Kidney
CH3
CH3
H
H3C
CH3
C=O
In the liver, cholecalciferol, which has been synthesized
in the skin or derived from food, is hydroxylated to
All-trans-retinaldehyde + opsin
CH3
form the 25-hydroxy derivative calcidiol (Figure 45-4).
This is released into the circulation bound to a vitamin
The role of retinaldehyde in vision.
Figure 45-2.
D-binding globulin which is the main storage form of
the vitamin. In the kidney, calcidiol undergoes either
1-hydroxylation to yield the active metabolite 1,25-dihy-
tamin, and therefore there is further impairment of im-
droxyvitamin D (calcitriol) or 24-hydroxylation to yield
an inactive metabolite, 24,25-dihydroxyvitamin D (24-
mune responses.
hydroxycalcidiol). Ergocalciferol from fortified foods
undergoes similar hydroxylations to yield ercalcitriol.
Vitamin A Is Toxic in Excess
There is only a limited capacity to metabolize vitamin
Vitamin D Metabolism Both Regulates
A, and excessive intakes lead to accumulation beyond
& Is Regulated by Calcium Homeostasis
the capacity of binding proteins, so that unbound vita-
min A causes tissue damage. Symptoms of toxicity af-
The main function of vitamin D is in the control of cal-
fect the central nervous system
(headache, nausea,
cium homeostasis, and in turn vitamin D metabolism is
VITAMINS & MINERALS
/
485
OH
Thermal isomerization
LIGHT
Cholecalciferol
(calciol;vitamin D3)
HO
CH3
CH2
7-Dehydrocholesterol
Previtamin D
HO
Figure 45-3. Synthesis of vitamin D in the skin.
regulated by factors that respond to plasma concentra-
Vitamin D Deficiency Affects
tions of calcium and phosphate. Calcitriol acts to reduce
Children & Adults
its own synthesis by inducing the 24-hydroxylase and
In the vitamin D deficiency disease rickets, the bones
repressing the 1-hydroxylase in the kidney. Its principal
of children are undermineralized as a result of poor ab-
function is to maintain the plasma calcium concentra-
sorption of calcium. Similar problems occur in adoles-
tion. Calcitriol achieves this in three ways: it increases
cents who are deficient during their growth spurt. Os-
intestinal absorption of calcium, reduces excretion of
teomalacia in adults results from demineralization of
calcium (by stimulating resorption in the distal renal
bone in women who have little exposure to sunlight,
tubules), and mobilizes bone mineral. In addition, cal-
often after several pregnancies. Although vitamin D is
citriol is involved in insulin secretion, synthesis and se-
essential for prevention and treatment of osteomalacia
cretion of parathyroid and thyroid hormones, inhibition
in the elderly, there is little evidence that it is beneficial
of production of interleukin by activated T lymphocytes
in treating osteoporosis.
and of immunoglobulin by activated B lymphocytes,
differentiation of monocyte precursor cells, and mod-
Vitamin D Is Toxic in Excess
ulation of cell proliferation. In its actions, it behaves like
a steroid hormone, binding to a nuclear receptor
Some infants are sensitive to intakes of vitamin D as
protein.
low as 50 µg/d, resulting in an elevated plasma concen-
OH
OH
Calciol-25-hydroxylase
Calcidiol-1-hydroxylase
CH2
CH2
CH2
Calcitriol
Calcidiol
(1,25-hydroxycholecalciferol)
Cholecalciferol
(25-hydroxycholecalciferol)
HO
HO
HO
OH
(calciol;vitamin D3)
Calcidiol-24-hydroxylase
Calcidiol-24-hydroxylase
OH
OH
OH
OH
Calcidiol-1-hydroxylase
CH2
CH2
24-hydroxycalcidiol
Calcitetrol
HO
HO
OH
Figure 45-4. Metabolism of vitamin D.
486
/
CHAPTER 45
tration of calcium. This can lead to contraction of
reduced back to tocopherol by reaction with vitamin
blood vessels, high blood pressure, and calcinosis—the
C from plasma (Figure 45-6). The resultant monode-
calcification of soft tissues. Although excess dietary vita-
hydroascorbate free radical then undergoes enzymic or
min D is toxic, excessive exposure to sunlight does not
nonenzymic reaction to yield ascorbate and dehy-
lead to vitamin D poisoning because there is a limited
droascorbate, neither of which is a free radical. The
capacity to form the precursor
7-dehydrocholesterol
stability of the tocopheroxyl free radical means that it
and to take up cholecalciferol from the skin.
can penetrate farther into cells and, potentially, propa-
gate a chain reaction. Therefore, vitamin E may, like
other antioxidants, also have pro-oxidant actions, es-
VITAMIN E DOES NOT HAVE A PRECISELY
pecially at high concentrations. This may explain why,
DEFINED METABOLIC FUNCTION
although studies have shown an association between
No unequivocal unique function for vitamin E has
high blood concentrations of vitamin E and a lower
been defined. However, it does act as a lipid-soluble an-
incidence of atherosclerosis, the effect of high doses of
tioxidant in cell membranes, where many of its func-
vitamin E have been disappointing.
tions can be provided by synthetic antioxidants. Vita-
min E is the generic descriptor for two families of
Dietary Vitamin E Deficiency
compounds, the tocopherols and the tocotrienols (Fig-
in Humans Is Unknown
ure 45-5). The different vitamers (compounds having
similar vitamin activity) have different biologic poten-
In experimental animals, vitamin E deficiency results in
cies; the most active is D-α-tocopherol, and it is usual
resorption of fetuses and testicular atrophy. Dietary de-
to express vitamin E intake in milligrams of D-α-tocoph-
ficiency of vitamin E in humans is unknown, though
erol equivalents. Synthetic DL-α-tocopherol does not
patients with severe fat malabsorption, cystic fibrosis,
have the same biologic potency as the naturally occur-
and some forms of chronic liver disease suffer defi-
ring compound.
ciency because they are unable to absorb the vitamin or
transport it, exhibiting nerve and muscle membrane
Vitamin E Is the Major Lipid-Soluble
damage. Premature infants are born with inadequate re-
Antioxidant in Cell Membranes
serves of the vitamin. Their erythrocyte membranes are
& Plasma Lipoproteins
abnormally fragile as a result of peroxidation, which
leads to hemolytic anemia.
The main function of vitamin E is as a chain-break-
ing, free radical trapping antioxidant in cell mem-
branes and plasma lipoproteins. It reacts with the lipid
VITAMIN K IS REQUIRED FOR SYNTHESIS
peroxide radicals formed by peroxidation of polyun-
OF BLOOD-CLOTTING PROTEINS
saturated fatty acids before they can establish a chain
Vitamin K was discovered as a result of investigations
reaction. The tocopheroxyl free radical product is rela-
into the cause of a bleeding disorder—hemorrhagic
tively unreactive and ultimately forms nonradical
(sweet clover) disease—of cattle, and of chickens fed on
compounds. Commonly, the tocopheroxyl radical is
a fat-free diet. The missing factor in the diet of the
chickens was vitamin K, while the cattle feed contained
dicumarol, an antagonist of the vitamin. Antagonists
of vitamin K are used to reduce blood coagulation in
R1
patients at risk of thrombosis—the most widely used
HO
agent is warfarin.
R2
O
Three compounds have the biologic activity of vita-
CH3
R3
Tocopherol
min K (Figure 45-7): phylloquinone, the normal di-
etary source, found in green vegetables; menaqui-
R1
nones, synthesized by intestinal bacteria, with differing
HO
lengths of side-chain; menadione, menadiol, and
R2
O
menadiol diacetate, synthetic compounds that can be
CH3
R3
Tocotrienol
metabolized to phylloquinone. Menaquinones are ab-
sorbed to some extent but it is not clear to what extent
Figure 45-5. The vitamin E vitamers. In α-tocoph-
they are biologically active as it is possible to induce
erol and tocotrienol R1, R2, and R3 are all CH3 groups.
signs of vitamin K deficiency simply by feeding a phyl-
In the β-vitamers R2 is H; in the γ-vitamers R1 is H, and in
loquinone deficient diet, without inhibiting intestinal
the δ-vitamers R1 and R2 are both H.
bacterial action.
VITAMINS & MINERALS
/
487
Free radical
chain reaction
PUFA
OO
PUFA OOH
R
TocOH
TocO
PHOSPHOLIPASE
R
O2
A2
PUFA H
(in phospholipid)
MEMBRANES
CYTOSOL
Vitamin Cox
,
Vitamin Cred,
PUFA OOH,
GS SG
GSH
H2O2
GSH
GLUTATHIONE
SUPEROXIDE
CATALASE
Se
PEROXIDASE
DISMUTASE
–
O2
H2O,
GS SG
Superoxide
PUFA OH
Figure 45-6. Interaction and synergism between antioxidant systems operating in the lipid
phase (membranes) of the cell and the aqueous phase (cytosol). (R•, free radical; PUFA-OO•, peroxyl
free radical of polyunsaturated fatty acid in membrane phospholipid; PUFA-OOH, hydroperoxy
polyunsaturated fatty acid in membrane phospholipid released as hydroperoxy free fatty acid into
cytosol by the action of phospholipase A2; PUFA-OH, hydroxy polyunsaturated fatty acid; TocOH,
vitamin E (α-tocopherol); TocO•, free radical of α-tocopherol; Se, selenium; GSH, reduced glu-
tathione; GS-SG, oxidized glutathione, which is returned to the reduced state after reaction with
NADPH catalyzed by glutathione reductase; PUFA-H, polyunsaturated fatty acid.)
Vitamin K Is the Coenzyme
quinone reductase. In the presence of warfarin, vitamin
for Carboxylation of Glutamate
K epoxide cannot be reduced but accumulates, and is
in the Postsynthetic Modification
excreted. If enough vitamin K (a quinone) is provided
in the diet, it can be reduced to the active hydro-
of Calcium-Binding Proteins
quinone by the warfarin-insensitive enzyme, and car-
Vitamin K is the cofactor for the carboxylation of gluta-
boxylation can continue, with stoichiometric utilization
mate residues in the post-synthetic modification of pro-
of vitamin K and excretion of the epoxide. A high dose
teins to form the unusual amino acid γ-carboxygluta-
of vitamin K is the antidote to an overdose of warfarin.
mate (Gla), which chelates the calcium ion. Initially,
Prothrombin and several other proteins of the blood
vitamin K hydroquinone is oxidized to the epoxide
clotting system (Factors VII, IX and X, and proteins C
(Figure 45-8), which activates a glutamate residue in
and S) each contain between four and six γ-carboxygluta-
the protein substrate to a carbanion, that reacts non-
mate residues which chelate calcium ions and so permit
enzymically with carbon dioxide to form γ-carboxyglut-
the binding of the blood clotting proteins to membranes.
amate. Vitamin K epoxide is reduced to the quinone by
In vitamin K deficiency or in the presence of warfarin, an
a warfarin-sensitive reductase, and the quinone is re-
abnormal precursor of prothrombin (preprothrombin)
duced to the active hydroquinone by either the same
containing little or no γ-carboxyglutamate, and incapable
warfarin-sensitive reductase or a warfarin-insensitive
of chelating calcium, is released into the circulation.
488
/
CHAPTER 45
O
Vitamin K Is Also Important
CH3
in the Synthesis of Bone
Calcium-Binding Proteins
H
O
3
Treatment of pregnant women with warfarin can lead to
Phylloquinone
fetal bone abnormalities (fetal warfarin syndrome). Two
proteins are present in bone that contain γ-carboxygluta-
O
mate, osteocalcin and bone matrix Gla protein. Osteocal-
CH3
cin also contains hydroxyproline, so its synthesis is depen-
dent on both vitamins K and C; in addition, its synthesis
H
is induced by vitamin D. The release into the circulation
O
n
Menaquinone
CH3
of osteocalcin provides an index of vitamin D status.
C
O
OH
O
CH3
CH3
WATER-SOLUBLE VITAMINS
OH
O
Menadiol
C
O
VITAMIN B1 (THIAMIN) HAS A KEY ROLE
CH3
IN CARBOHYDRATE METABOLISM
Menadiolo diacetate
Thiamin has a central role in energy-yielding metabo-
(acetomenaphthone)
lism, and especially the metabolism of carbohydrate
Figure 45-7. The vitamin K vitamers. Menadiol (or
(Figure 45-9). Thiamin diphosphate is the coenzyme
menadione) and menadiol diacetate are synthetic com-
for three multi-enzyme complexes that catalyze oxida-
pounds that are converted to menaquinone in the liver
tive decarboxylation reactions: pyruvate dehydrogenase
and have vitamin K activity.
in carbohydrate metabolism; α-ketoglutarate dehydro-
OOC
CH
COO
CH2
HN
CH C
O
Carboxyglutamate residue
CO 2
non-
enzymic
CH COO
O2
CH COO
2
CH2
CH2
HN CH C O
HN CH C O
VITAMIN K
EPOXIDASE
Glutamate residue
Glutamate carbanion
OH
O
CH3
CH3
O
R
R
OH
O
Vitamin K hydroquinone
Vitamin K epoxide
Disulfide
Sulfhydryl
NADP+
VITAMIN K QUINONE
VITAMIN K EPOXIDE
QUINONE
REDUCTASE
O
REDUCTASE
REDUCTASE
Sulfhydryl
NADPH
CH3
Disulfide
R
O
Figure 45-8.
The role of vitamin K in the
Vitamin K quinone
biosynthesis of γ-carboxyglutamate.
VITAMINS & MINERALS
/
489
O
O
H3C
H3C
N
NH2
H3C
CH2
CH2
CH2OH
CH2
CH2
O
P O P O
N
N
S
CH2
N
O
O
S
H+
Thiamin
Thiamin diphosphate
Carbanion
Figure 45-9. Thiamin, thiamin diphosphate, and the carbanion form.
genase in the citric acid cycle; and the branched-chain
pyruvate dehydrogenase means that in deficiency there is
keto-acid dehydrogenase involved in the metabolism of
impaired conversion of pyruvate to acetyl CoA. In sub-
leucine, isoleucine, and valine. It is also the coenzyme
jects on a relatively high carbohydrate diet, this results in
for transketolase, in the pentose phosphate pathway. In
increased plasma concentrations of lactate and pyruvate,
each case, the thiamin diphosphate provides a reactive
which may cause life-threatening lactic acidosis.
carbon on the thiazole moiety that forms a carbanion,
which then adds to the carbonyl group of, for instance,
Thiamin Nutritional Status Can
pyruvate. The addition compound then decarboxylates,
Be Assessed by Erythrocyte
eliminating CO2. Electrical stimulation of nerve leads
to a fall in membrane thiamin triphosphate and release
Transketolase Activation
of free thiamin. It is likely that thiamin triphosphate
The activation of apo-transketolase(the enzyme pro-
acts as a phosphate donor for phosphorylation of the
tein) in erythrocyte lysate by thiamin diphosphate
nerve membrane sodium transport channel.
added in vitro has become the accepted index of thi-
amin nutritional status.
Thiamin Deficiency Affects
the Nervous System & Heart
VITAMIN B2 (RIBOFLAVIN) HAS
Thiamin deficiency can result in three distinct syn-
A CENTRAL ROLE IN ENERGY-
dromes: a chronic peripheral neuritis, beriberi, which
YIELDING METABOLISM
may or may not be associated with heart failure and
edema; acute pernicious (fulminating) beriberi (shoshin
Riboflavin fulfills its role in metabolism as the coenzymes
beriberi), in which heart failure and metabolic abnor-
flavin mononucleotide
(FMN) and flavin adenine
malities predominate, without peripheral neuritis; and
dinucleotide (FAD) (Figure 45-10). FMN is formed by
Wernicke’s encephalopathy with Korsakoff’s psy-
ATP-dependent phosphorylation of riboflavin, whereas
chosis, which is associated especially with alcohol and
FAD is synthesized by further reaction of FMN with
drug abuse. The central role of thiamin diphosphate in
ATP in which its AMP moiety is transferred to the
OH OH OH
OH OH OH
O
CH2
CH CH CH
CH2OH
CH2
CH CH CH
CH2
O P O
H3C
N N
H3C
N N
O
N
N
H3C
N
H3C
N
O
O
Riboflavin
FMN
NH2
OH OH OH
O
O
N
N
CH2
CH CH CH
CH2
O P O
P
O
CH2
O
N N
H3C
N N
O
O
N
H3C
N
O
FAD
Figure 45-10. Riboflavin and the coenzymes flavin mononucleotide (FMN) and flavin
adenine dinucleotide (FAD).
490
/
CHAPTER 45
FMN. The main dietary sources of riboflavin are milk
COO
CONH2
and dairy products. In addition, because of its intense
N
N
yellow color, riboflavin is widely used as a food additive.
Nicotinic acid
Nicotinamide
Flavin Coenzymes Are Electron Carriers
CONH2
NH2
in Oxidoreduction Reactions
+
N
O
O
N
N
These include the mitochondrial respiratory chain, key
CH2
O P O
P
O
CH2
O
N N
enzymes in fatty acid and amino acid oxidation, and
O
O
the citric acid cycle. Reoxidation of the reduced flavin
O
in oxygenases and mixed-function oxidases proceeds by
way of formation of the flavin radical and flavin hy-
NAD
droperoxide, with the intermediate generation of super-
oxide and perhydroxyl radicals and hydrogen peroxide.
Figure 45-11. Niacin (nicotinic acid and nicotin-
Because of this, flavin oxidases make a significant con-
amide) and nicotinamide adenine dinucleotide (NAD).
tribution to the total oxidant stress of the body.
* Shows the site of phosphorylation in NADP.
Riboflavin Deficiency Is
Widespread But Not Fatal
diarrhea, and, if untreated, death. Although the nutri-
Although riboflavin is fundamentally involved in me-
tional etiology of pellagra is well established and trypto-
tabolism, and deficiencies are found in most countries,
phan or niacin will prevent or cure the disease, addi-
it is not fatal as there is very efficient conservation of
tional factors, including deficiency of riboflavin or
tissue riboflavin. Riboflavin deficiency is characterized
vitamin B6, both of which are required for synthesis of
by cheilosis, lingual desquamation and a seborrheic der-
nicotinamide from tryptophan, may be important. In
matitis. Riboflavin nutritional status is assessed by mea-
most outbreaks of pellagra twice as many women as
surement of the activation of erythrocyte glutathione
men are affected, probably the result of inhibition of
reductase by FAD added in vitro.
tryptophan metabolism by estrogen metabolites.
NIACIN IS NOT STRICTLY A VITAMIN
Pellagra Can Occur as a Result
Niacin was discovered as a nutrient during studies of pel-
of Disease Despite an Adequate
lagra. It is not strictly a vitamin since it can be syn-
Intake of Tryptophan & Niacin
thesized in the body from the essential amino acid
A number of genetic diseases that result in defects of
tryptophan. Two compounds, nicotinic acid and nico-
tinamide, have the biologic activity of niacin; its meta-
tryptophan metabolism are associated with the develop-
ment of pellagra despite an apparently adequate intake
bolic function is as the nicotinamide ring of the coen-
zymes NAD and NADP in oxidation-reduction reactions
of both tryptophan and niacin. Hartnup disease is a
rare genetic condition in which there is a defect of the
(Figure 45-11). About 60 mg of tryptophan is equivalent
to 1 mg of dietary niacin. The niacin content of foods is
membrane transport mechanism for tryptophan, result-
ing in large losses due to intestinal malabsorption and
expressed as mg niacin equivalents = mg preformed niacin
+ 1/60 × mg tryptophan. Because most of the niacin in
failure of the renal resorption mechanism. In carcinoid
syndrome there is metastasis of a primary liver tumor
cereals is biologically unavailable, this is discounted.
of enterochromaffin cells which synthesize 5-hydroxy-
tryptamine. Overproduction of
5-hydroxytryptamine
NAD Is the Source of ADP-Ribose
may account for as much as 60% of the body’s trypto-
In addition to its coenzyme role, NAD is the source of
phan metabolism, causing pellagra because of the diver-
ADP-ribose for the ADP-ribosylation of proteins and
sion away from NAD synthesis.
polyADP-ribosylation of nucleoproteins involved in the
DNA repair mechanism.
Niacin Is Toxic in Excess
Nicotinic acid has been used to treat hyperlipidemia
Pellagra Is Caused by Deficiency
when of the order of 1-6 g/d are required, causing dila-
of Tryptophan & Niacin
tion of blood vessels and flushing, with skin irritation.
Pellagra is characterized by a photosensitive dermatitis.
Intakes of both nicotinic acid and nicotinamide in ex-
As the condition progresses, there is dementia, possibly
cess of 500 mg/d can cause liver damage.
VITAMINS & MINERALS
/
491
VITAMIN B6 IS IMPORTANT IN AMINO
Vitamin B6 Deficiency Is Rare
ACID & GLYCOGEN METABOLISM
Although clinical deficiency disease is rare, there is evi-
& IN STEROID HORMONE ACTION
dence that a significant proportion of the population
have marginal vitamin B6 status. Moderate deficiency
Six compounds have vitamin B6 activity (Figure 45-12):
results in abnormalities of tryptophan and methionine
pyridoxine, pyridoxal, pyridoxamine, and their 5′-
metabolism. Increased sensitivity to steroid hormone ac-
phosphates. The active coenzyme is pyridoxal 5′-phos-
tion may be important in the development of hormone-
phate. Approximately 80% of the body’s total vitamin B6
dependent cancer of the breast, uterus, and prostate,
is present as pyridoxal phosphate in muscle, mostly asso-
and vitamin B6 status may affect the prognosis.
ciated with glycogen phosphorylase. This is not available
in B6 deficiency but is released in starvation, when glyco-
gen reserves become depleted, and is then available, espe-
Vitamin B6 Status Is Assessed by Assaying
cially in liver and kidney, to meet increased requirement
Erythrocyte Aminotransferases
for gluconeogenesis from amino acids.
The most widely used method of assessing vitamin B6
status is by the activation of erythrocyte aminotrans-
Vitamin B6 Has Several Roles
ferases by pyridoxal phosphate added in vitro, expressed
in Metabolism
as the activation coefficient.
Pyridoxal phosphate is a coenzyme for many enzymes
involved in amino acid metabolism, especially in
In Excess, Vitamin B6 Causes
transamination and decarboxylation. It is also the co-
Sensory Neuropathy
factor of glycogen phosphorylase, where the phosphate
group is catalytically important. In addition, vitamin B6
The development of sensory neuropathy has been re-
is important in steroid hormone action where it re-
ported in patients taking 2-7 g of pyridoxine per day for
moves the hormone-receptor complex from DNA
a variety of reasons (there is some slight evidence that it
binding, terminating the action of the hormones. In vi-
is effective in treating premenstrual syndrome). There
tamin B6 deficiency, this results in increased sensitivity
was some residual damage after withdrawal of these high
to the actions of low concentrations of estrogens, an-
doses; other reports suggest that intakes in excess of 200
drogens, cortisol, and vitamin D.
mg/d are associated with neurologic damage.
VITAMIN B12 IS FOUND ONLY
IN FOODS OF ANIMAL ORIGIN
O
CH2OH
CH2OH
KINASE
The term “vitamin B12” is used as a generic descriptor
HOCH2
OH
O
POCH2
OH
for the cobalamins—those corrinoids (cobalt con-
PHOSPHATASE
O
N CH3
N CH3
taining compounds possessing the corrin ring) having
Pyridoxine
Pyridoxine phosphate
the biologic activity of the vitamin (Figure 45-13).
Some corrinoids that are growth factors for microor-
OXIDASE
ganisms not only have no vitamin B12 activity but may
also be antimetabolites of the vitamin. Although it is
O
HC=O
HC=O
synthesized exclusively by microorganisms, for practi-
KINASE
HOCH2
OH
O
POCH2
OH
cal purposes vitamin B12 is found only in foods of ani-
PHOSPHATASE
O
mal origin, there being no plant sources of this vita-
N CH3
N CH3
min. This means that strict vegetarians (vegans) are at
Pyridoxal
Pyridoxal phosphate
risk of developing B12 deficiency. The small amounts
of the vitamin formed by bacteria on the surface of
AMINOTRANSFERASES
OXIDASE
fruits may be adequate to meet requirements, but
O
preparations of vitamin B12 made by bacterial fermen-
CH2NH2
CH2NH2
KINASE
tation are available.
HOCH2
OH
O
POCH2
OH
PHOSPHATASE
O
N CH3
N CH3
Vitamin B12 Absorption Requires Two
Pyridoxamine
Pyridoxamine phosphate
Binding Proteins
Figure 45-12. Interconversion of the vitamin B6
Vitamin B12 is absorbed bound to intrinsic factor, a
vitamers.
small glycoprotein secreted by the parietal cells of the
492
/
CHAPTER 45
CH2CONH2
mentation in ruminants. It undergoes vitamin B12-
H3C
CH2CH2CONH2
dependent rearrangement to succinyl-CoA, catalyzed
H3C
by methylmalonyl-CoA isomerase (Figure 19-2). The
H2NCOCH2CH2
N
activity of this enzyme is greatly reduced in vitamin B12
CH3
H2NCOCH2
R
N
CH
deficiency, leading to an accumulation of methyl-
Co
3
H3C
N
malonyl-CoA and urinary excretion of methylmalonic
H3C
N
acid, which provides a means of assessing vitamin B12
CH2CH2CONH2
CH3
nutritional status.
H2NCOCH2
CH
3
CH2
Vitamin B12 Deficiency Causes
CH2
Pernicious Anemia
C
O
Pernicious anemia arises when vitamin B12 deficiency
NH
blocks the metabolism of folic acid, leading to func-
CH
O
tional folate deficiency. This impairs erythropoiesis,
2
causing immature precursors of erythrocytes to be re-
N
CH3
H3C C O
P
O
H
leased into the circulation (megaloblastic anemia). The
N
CH3
commonest cause of pernicious anemia is failure of the
absorption of vitamin B12
rather than dietary defi-
ciency. This can be due to failure of intrinsic factor se-
HOCH2 O
cretion caused by autoimmune disease of parietal cells
or to generation of anti-intrinsic factor antibodies.
Figure 45-13. Vitamin B12 (cobalamin). R may be
varied to give the various forms of the vitamin, eg,
THERE ARE MULTIPLE FORMS
in hydroxocobal-
R = CN- in cyanocobalamin; R = OH-
OF FOLATE IN THE DIET
amin; R = 5′-deoxyadenosyl in 5′-deoxyadenosylcobal-
amin; R = H2O in aquocobalamin; and R = CH3 in
The active form of folic acid (pteroyl glutamate) is
methylcobalamin.
tetrahydrofolate (Figure 45-15). The folates in foods
may have up to seven additional glutamate residues
linked by γ-peptide bonds. In addition, all of the one-
carbon substituted folates in Figure 45-15 may also be
gastric mucosa. Gastric acid and pepsin release the vita-
present in foods.
min from protein binding in food and make it available
The extent to which the different forms of folate can
to bind to cobalophilin, a binding protein secreted in
be absorbed varies, and this must be allowed for in cal-
the saliva. In the duodenum, cobalophilin is hy-
culating folate intakes.
drolyzed, releasing the vitamin for binding to intrinsic
factor. Pancreatic insufficiency can therefore be a fac-
tor in the development of vitamin B12 deficiency, re-
sulting in the excretion of cobalophilin-bound vitamin
B12. Intrinsic factor binds the various vitamin B12
vita-
SH
H3C S
mers, but not other corrinoids. Vitamin B12 is absorbed
(CH2 )2
(CH2 )2
from the distal third of the ileum via receptors that
bind the intrinsic factor-vitamin B12 complex but not
+
+
H C NH
3
H C NH
3
free intrinsic factor or free vitamin.
-
COO
COO-
Homocysteine
Methionine
There Are Three Vitamin
METHIONINE
B12-Dependent Enzymes
SYNTHASE
Methylmalonyl CoA mutase, leucine aminomutase,
and methionine synthase (Figure 45-14) are vitamin
Methylcobalamin
Methyl
H
B12
H4
folate
B12-dependent enzymes. Methylmalonyl CoA is formed
4folate
as an intermediate in the catabolism of valine and by
the carboxylation of propionyl CoA arising in the ca-
Figure 45-14. Homocysteinuria and the folate trap.
tabolism of isoleucine, cholesterol, and, rarely, fatty
Vitamin B12 deficiency leads to inhibition of methionine
acids with an odd number of carbon atoms—or directly
synthase activity causing homocysteinuria and the
from propionate, a major product of microbial fer-
trapping of folate as methyltetrahydrofolate.
VITAMINS & MINERALS
/
493
O
COO
OH
H
H
N
CH2
N
C
N
CH
N
10
H
5
CH2
H2N
N N
H
Tetrahydrofolate (THF)
CH
2
C
O
(Glu)
n
HC
O
HC
O
OH
H
OH
H
H
N
CH2
N
N
CH2
N
N
N
5-Formyl THF
10-Formyl THF
H2N
N N
H2N N N
H
H
HC NH
OH
OH
CH2
H
H
N
CH2
N
N
CH2
N
N
N
5-Formimino THF
5,10-Methylene THF
H2N
N N
H2N N N
H
H
CH3
OH
OH
CH
H
H
N
CH2
N
N
CH2
N
N
N
+
Figure 45-15.
Tetrahydrofolic acid and the
5-Methyl THF
5,10-Methenyl THF
H2N
N N
H2N N N
one-carbon substituted folates.
H
H
Tetrahydrofolate Is a Carrier
ceutically in the agent known as folinic acid and in the
of One-Carbon Units
synthetic (racemic) compound leucovorin.
The major point of entry for one-carbon fragments
Tetrahydrofolate can carry one-carbon fragments at-
into substituted folates is methylene tetrahydrofolate
tached to N-5 (formyl, formimino, or methyl groups),
(Figure 45-16), which is formed by the reaction of
N-10 (formyl group), or bridging N-5 to N-10 (meth-
glycine, serine, and choline with tetrahydrofolate. Serine
ylene or methenyl groups). 5-Formyl-tetrahydrofolate is
is the most important source of substituted folates
more stable than folate and is therefore used pharma-
for biosynthetic reactions, and the activity of serine hy-
Sources of one-carbon units
Synthesis using one-carbon units
Serine
Serine
Glycine
Methylene-THF
Methyl-THF
Methionine
Choline
TMP + dihydrofolate
Histidine
Formimino-THF
Methenyl-THF
DNA
Formyl-methionine
Formate
Formyl-THF
Purines
CO2
Figure 45-16. Sources and utilization of one-carbon substituted folates.
494
/
CHAPTER 45
droxymethyltransferase is regulated by the state of folate
Folic Acid Supplements Reduce
substitution and the availability of folate. The reaction is
the Risk of Neural Tube Defects
reversible, and in liver it can form serine from glycine as
& Hyperhomocysteinemia
a substrate for gluconeogenesis. Methylene, methenyl,
Supplements of 400 µg/d of folate begun before con-
and 10-formyl tetrahydrofolates are interconvertible.
When one-carbon folates are not required, the oxidation
ception result in a significant reduction in the incidence
of neural tube defects as found in spina bifida. Ele-
of formyl tetrahydrofolate to yield carbon dioxide pro-
vides a means of maintaining a pool of free folate.
vated blood homocysteine is an associated risk factor
for atherosclerosis, thrombosis, and hypertension.
The condition is due to impaired ability to form
Inhibitors of Folate Metabolism
methyl-tetrahydrofolate by methylene-tetrahydrofolate
Provide Cancer Chemotherapy &
reductase, causing functional folate deficiency and re-
Antibacterial & Antimalarial Drugs
sulting in failure to remethylate homocysteine to me-
The methylation of deoxyuridine monophosphate
thionine. People with the causative abnormal variant of
(dUMP) to thymidine monophosphate (TMP), cat-
methylene-tetrahydrofolate reductase do not develop
alyzed by thymidylate synthase, is essential for the syn-
hyperhomocysteinemia if they have a relatively high in-
thesis of DNA. The one-carbon fragment of methy-
take of folate, but it is not yet known whether this af-
lene-tetrahydrofolate is reduced to a methyl group with
fects the incidence of cardiovascular disease.
release of dihydrofolate, which is then reduced back to
tetrahydrofolate by dihydrofolate reductase. Thymi-
Folate Enrichment of Foods
dylate synthase and dihydrofolate reductase are espe-
May Put Some People at Risk
cially active in tissues with a high rate of cell division.
Folate supplements will rectify the megaloblastic anemia
Methotrexate, an analog of 10-methyl-tetrahydrofo-
of vitamin B12 deficiency but may hasten the develop-
late, inhibits dihydrofolate reductase and has been ex-
ment of the (irreversible) nerve damage found in B12 de-
ploited as an anticancer drug. The dihydrofolate reduc-
ficiency. There is also antagonism between folic acid and
tases of some bacteria and parasites differ from the
the anticonvulsants used in the treatment of epilepsy.
human enzyme; inhibitors of these enzymes can be used
as antibacterial drugs, eg, trimethoprim, and anti-
malarial drugs, eg, pyrimethamine.
DIETARY BIOTIN DEFICIENCY
IS UNKNOWN
Vitamin B12 Deficiency Causes Functional
The structures of biotin, biocytin, and carboxy-biotin
Folate Deficiency—the Folate Trap
(the active metabolic intermediate) are shown in Figure
When acting as a methyl donor, S-adenosylmethionine
45-17. Biotin is widely distributed in many foods as
forms homocysteine, which may be remethylated by
biocytin (ε-amino-biotinyl lysine), which is released on
methyltetrahydrofolate catalyzed by methionine syn-
proteolysis. It is synthesized by intestinal flora in excess
thase, a vitamin B12-dependent enzyme (Figure 45-14).
of requirements. Deficiency is unknown except among
The reduction of methylene-tetrahydrofolate to methyl-
people maintained for many months on parenteral nu-
tetrahydrofolate is irreversible, and since the major source
trition and a very small number who eat abnormally
of tetrahydrofolate for tissues is methyl-tetrahydrofolate,
large amounts of uncooked egg white, which contains
the role of methionine synthase is vital and provides a link
avidin, a protein that binds biotin and renders it un-
between the functions of folate and vitamin B12. Impair-
available for absorption.
ment of methionine synthase in B12 deficiency results in
the accumulation of methyl-tetrahydrofolate—the “fo-
Biotin Is a Coenzyme
late trap.” There is therefore functional deficiency of fo-
of Carboxylase Enzymes
late secondary to the deficiency of vitamin B12.
Biotin functions to transfer carbon dioxide in a small
number of carboxylation reactions. A holocarboxylase
Folate Deficiency Causes
synthetase acts on a lysine residue of the apoenzymes of
Megaloblastic Anemia
acetyl-CoA carboxylase, pyruvate carboxylase, propi-
Deficiency of folic acid itself—or deficiency of vitamin
onyl-CoA carboxylase, or methylcrotonyl-CoA carboxy-
B12, which leads to functional folic acid deficiency—af-
lase to react with free biotin to form the biocytin residue
fects cells that are dividing rapidly because they have a
of the holoenzyme. The reactive intermediate is 1-N-
large requirement for thymidine for DNA synthesis.
carboxybiocytin, formed from bicarbonate in an ATP-
Clinically, this affects the bone marrow, leading to
dependent reaction. The carboxyl group is then trans-
megaloblastic anemia.
ferred to the substrate for carboxylation (Figure 21-1).
VITAMINS & MINERALS
/
495
O
the SH prosthetic group of CoA and ACP. CoA
takes part in reactions of the citric acid cycle, fatty acid
HN NH
Biotin
synthesis and oxidation, acetylations, and cholesterol
synthesis. ACP participates in fatty acid synthesis. The
COO
S
vitamin is widely distributed in all foodstuffs, and defi-
ciency has not been unequivocally reported in human
O
beings except in specific depletion studies.
HN NH
Biotinyl-lysine (biocytin)
C O
H
ASCORBIC ACID IS A VITAMIN
C
N
S
CH
FOR ONLY SOME SPECIES
O
NH
Vitamin C (Figure 45-19) is a vitamin for human beings
O
and other primates, the guinea pig, bats, passerine birds,
OOC
N NH
Carboxy-biocytin
and most fishes and invertebrates; other animals synthe-
C O
H
size it as an intermediate in the uronic acid pathway of
C
N
S
CH
glucose metabolism (Chapter 20). In those species for
O
which it is a vitamin, there is a block in that pathway due
NH
to absence of gulonolactone oxidase. Both ascorbic acid
and dehydroascorbic acid have vitamin activity.
Figure 45-17. Biotin, biocytin, and carboxy-biocytin.
Vitamin C Is the Coenzyme
for Two Groups of Hydroxylases
Biotin also has a role in regulation of the cell cycle,
Ascorbic acid has specific roles in the copper-containing
acting to biotinylate key nuclear proteins.
hydroxylases and the α-ketoglutarate-linked iron-con-
taining hydroxylases. It also increases the activity of a
AS PART OF CoA AND ACP,
number of other enzymes in vitro, though this is a non-
specific reducing action. In addition, it has a number of
PANTOTHENIC ACID ACTS AS
nonenzymic effects due to its action as a reducing agent
A CARRIER OF ACYL RADICALS
and oxygen radical quencher.
Pantothenic acid has a central role in acyl group metab-
Dopamine
-hydroxylase is a copper-containing
olism when acting as the pantetheine functional moiety
enzyme involved in the synthesis of the catecholamines
of coenzyme A or acyl carrier protein (ACP) (Figure
norepinephrine and epinephrine from tyrosine in the
45-18). The pantetheine moiety is formed after combi-
adrenal medulla and central nervous system. During hy-
nation of pantothenate with cysteine, which provides
droxylation, the Cu+ is oxidized to Cu2+; reduction back
O C
O C
CH2
CH2
SH
CH2
CH2
CH2
CH2
C
O
C
O
NH2
H3C
CH3
H3C
CH3
O
O
N
N
CH2OH
CH2
O
P
O
P
O
CH2
N N
O
H
O
O
Pantothenic acid
Coenzyme A (CoASH)
Figure 45-18. Pantothenic acid and
coenzyme A. * Shows the site of acylation
O P O
by fatty acids.
O
496
/
CHAPTER 45
CH2OH
CH2OH
CH2OH
HO
CH2
HO
CH2
HO
CH2
O
O
O
O
O
O
Ascorbate
Monodehydroascorbate
Dehydroascorbate
(semidehydroascorbate)
Figure 45-19. Vitamin C.
vitamin C prevent the common cold or reduce the du-
to Cu+ specifically requires ascorbate, which is oxidized
ration of its symptoms.
to monodehydroascorbate. Similar actions of ascorbate
occur in tyrosine degradation at the p-hydroxy-
phenylpyruvate hydroxylase step and at the homogenti-
sate dioxygenase step, which needs Fe2+ (Figure 30-12).
A number of peptide hormones have a carboxyl ter-
MINERALS ARE REQUIRED
minal amide which is derived from a glycine terminal
FOR BOTH PHYSIOLOGIC &
residue. This glycine is hydroxylated on the α-carbon
by a copper-containing enzyme, peptidylglycine hy-
BIOCHEMICAL FUNCTIONS
droxylase, which, again, requires ascorbate for reduc-
tion of Cu2+.
Many of the essential minerals (Table 45-2) are widely
A number of iron-containing, ascorbate-requiring
distributed in foods, and most people eating a normal
hydroxylases share a common reaction mechanism in
mixed diet are likely to receive adequate intakes. The
which hydroxylation of the substrate is linked to decar-
boxylation of α-ketoglutarate (Figure 28-11). Many of
these enzymes are involved in the modification of pre-
cursor proteins. Proline and lysine hydroxylases are
Table 45-2. Classification of essential minerals
required for the postsynthetic modification of procol-
according to their function.
lagen to collagen, and proline hydroxylase is also re-
quired in formation of osteocalcin and the C1q com-
Function
Mineral
ponent of complement. Aspartate β-hydroxylase is
required for the postsynthetic modification of the pre-
Structural function
Calcium, magnesium, phosphate
cursor of protein C, the vitamin K-dependent protease
Involved in membrane
Sodium, potassium
which hydrolyzes activated factor V in the blood clot-
function: principal
ting cascade. Trimethyllysine and γ-butyrobetaine hy-
cations of extracellular-
droxylases are required for the synthesis of carnitine.
and intracellular fluids,
respectively
Vitamin C Deficiency Causes Scurvy
Function as prosthetic
Cobalt, copper, iron, molybde-
Signs of vitamin C deficiency in scurvy include skin
groups in enzymes
num, selenium, zinc
changes, fragility of blood capillaries, gum decay, tooth
Regulatory role or role
Calcium, chromium, iodine,
loss, and bone fracture, many of which can be attrib-
in hormone action
magnesium, manganese, sodium,
uted to deficient collagen synthesis.
potassium
Known to be essential,
Silicon, vanadium, nickel, tin
There May Be Benefits From Higher
but function unknown
Intakes of Vitamin C
Have effects in the
Fluoride, lithium
At intakes above approximately 100 mg/d, the body’s
body, but essentiality is
capacity to metabolize vitamin C is saturated, and any
not established
further intake is excreted in the urine. However, in ad-
Without known nutritional
Aluminum, arsenic, antimony,
dition to its other roles, vitamin C enhances the absorp-
function but toxic in
boron, bromine, cadmium, ce-
tion of iron, and this depends on the presence of the vi-
excess
sium, germanium, lead, mercury,
tamin in the gut. Therefore, increased intakes may be
silver, strontium
beneficial. Evidence is unconvincing that high doses of
VITAMINS & MINERALS
/
497
amounts required vary from the order of grams per day
decarboxylation of α-keto acids and of transketolase
for sodium and calcium, through milligrams per day
in the pentose phosphate pathway. Riboflavin and
(eg, iron) to micrograms per day for the trace elements.
niacin are important cofactors in oxidoreduction re-
In general, mineral deficiencies are encountered when
actions, respectively present in flavoprotein enzymes
foods come from one region, where the soil may be de-
and in NAD and NADP.
ficient in some minerals, eg, iodine deficiency. Where
•
Pantothenic acid is present in coenzyme A and acyl
the diet comes from a variety of different regions, min-
carrier protein, which act as carriers for acyl groups
eral deficiencies are unlikely. However, iron deficiency
in metabolic reactions. Pyridoxine, as pyridoxal
is a general problem because if iron losses from the
phosphate, is the coenzyme for several enzymes of
body are relatively high
(eg, from heavy menstrual
amino acid metabolism, including the aminotrans-
blood loss), it is difficult to achieve an adequate intake
ferases, and of glycogen phosphorylase. Biotin is the
to replace the losses. Foods from soils containing high
coenzyme for several carboxylase enzymes.
levels of selenium cause toxicity, and increased intakes
•
Besides other functions, vitamin B12 and folic acid
of common salt (sodium chloride) cause hypertension
take part in providing one-carbon residues for DNA
in susceptible individuals.
synthesis, deficiency resulting in megaloblastic ane-
mia. Vitamin C is a water-soluble antioxidant that
SUMMARY
maintains vitamin E and many metal cofactors in the
reduced state.
• Vitamins are organic nutrients with essential meta-
•
Inorganic mineral elements that have a function in
bolic functions, generally required in small amounts
the body must be provided in the diet. When insuffi-
in the diet because they cannot be synthesized by the
cient, deficiency symptoms may arise, and if present
body. The lipid-soluble vitamins (A, D, E, and K)
in excess they may be toxic.
are hydrophobic molecules requiring normal fat ab-
sorption for their efficient absorption and the avoid-
ance of deficiency symptoms.
REFERENCES
• Vitamin A (retinol), present in carnivorous diets, and
Bender DA, Bender AE: Nutrition: A Reference Handbook. Oxford
the provitamin (β-carotene), found in plants, form
Univ Press, 1997.
retinaldehyde, utilized in vision, and retinoic acid,
Bender DA: Nutritional Biochemistry of the Vitamins. 2nd ed. Cam-
which acts in the control of gene expression. Vitamin
bridge Univ Press, 2003.
D is a steroid prohormone yielding the active hor-
Garrow JS, James WPT, Ralph A: Human Nutrition and Dietetics,
mone derivative calcitriol, which regulates calcium
10th ed. Churchill-Livingstone, 2000.
and phosphate metabolism. Vitamin D deficiency
Halliwell B, Chirico S: Lipid peroxidation: its mechanism, mea-
leads to rickets and osteomalacia.
surement, and significance. Am J Clin Nutr
1993;57(5
Suppl):715S.
• Vitamin E (tocopherol) is the most important an-
tioxidant in the body, acting in the lipid phase of
Krinsky NI: Actions of carotenoids in biological systems. Annu Rev
Nutr 1993;13:561.
membranes and protecting against the effects of free
Padh H: Vitamin C: newer insights into its biochemical functions.
radicals. Vitamin K functions as cofactor to a car-
Nutr Rev 1991;49:65.
boxylase that acts on glutamate residues of clotting
Shane B: Folylpolyglutamate synthesis and role in the regulation of
factor precursor proteins to enable them to chelate
one-carbon metabolism. Vitam Horm 1989;45:263.
calcium.
Wiseman H, Halliwell B: Damage to DNA by reactive oxygen and
• The water-soluble vitamins of the B complex act as
nitrogen species: role in inflammatory disease and progression
enzyme cofactors. Thiamin is a cofactor in oxidative
to cancer. Biochem J 1996;313:17.
Intracellular Traffic & Sorting
46
of Proteins
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
the signal peptide are given below. Proteins synthesized
on free polyribosomes lack this particular signal pep-
Proteins must travel from polyribosomes to many dif-
tide and are delivered into the cytosol. There they are
ferent sites in the cell to perform their particular func-
directed to mitochondria, nuclei, and peroxisomes by
tions. Some are destined to be components of specific
specific signals—or remain in the cytosol if they lack a
organelles, others for the cytosol or for export, and yet
signal. Any protein that contains a targeting sequence
others will be located in the various cellular mem-
that is subsequently removed is designated as a prepro-
branes. Thus, there is considerable intracellular traffic
tein. In some cases a second peptide is also removed,
of proteins. Many studies have shown that the Golgi
and in that event the original protein is known as a pre-
apparatus plays a major role in the sorting of proteins
proprotein (eg, preproalbumin; Chapter 50).
for their correct destinations. A major insight was the
Proteins synthesized and sorted in the rough ER
recognition that for proteins to attain their proper loca-
branch (Figure 46-2) include many destined for vari-
tions, they generally contain information (a signal or
ous membranes (eg, of the ER, Golgi apparatus, lyso-
coding sequence) that targets them appropriately. Once
somes, and plasma membrane) and for secretion. Lyso-
a number of the signals were defined, it became appar-
somal enzymes are also included. Thus, such proteins
ent that certain diseases result from mutations that af-
may reside in the membranes or lumens of the ER or
fect these signals. In this chapter we discuss the intracel-
follow the major transport route of intracellular pro-
lular traffic of proteins and their sorting and briefly
teins to the Golgi apparatus. Further signal-mediated
consider some of the disorders that result when abnor-
sorting of certain proteins occurs in the Golgi appara-
malities occur.
tus, resulting in delivery to lysosomes, membranes of
the Golgi apparatus, and other sites. Proteins destined
MANY PROTEINS ARE TARGETED
for the plasma membrane or for secretion pass through
the Golgi apparatus but generally are not thought to
BY SIGNAL SEQUENCES TO THEIR
carry specific sorting signals; they are believed to reach
CORRECT DESTINATIONS
their destinations by default.
The protein biosynthetic pathways in cells can be con-
The entire pathway of ER → Golgi apparatus →
sidered to be one large sorting system. Many proteins
plasma membrane is often called the secretory or exo-
carry signals (usually but not always specific sequences
cytotic pathway. Events along this route will be given
of amino acids) that direct them to their destination,
special attention. Most of the proteins reaching the
thus ensuring that they will end up in the appropriate
Golgi apparatus or the plasma membrane are carried in
membrane or cell compartment; these signals are a fun-
transport vesicles; a brief description of the formation
damental component of the sorting system. Usually the
of these important particles will be given subsequently.
signal sequences are recognized and interact with com-
Other proteins destined for secretion are carried in se-
plementary areas of proteins that serve as receptors for
cretory vesicles (Figure 46-2). These are prominent in
the proteins that contain them.
the pancreas and certain other glands. Their mobiliza-
A major sorting decision is made early in protein
tion and discharge are regulated and often referred to as
biosynthesis, when specific proteins are synthesized ei-
“regulated secretion,” whereas the secretory pathway
ther on free or on membrane-bound polyribosomes.
involving transport vesicles is called
“constitutive.”
This results in two sorting branches called the cytosolic
Experimental approaches that have afforded major
branch and the rough endoplasmic reticulum (RER)
insights to the processes described in this chapter in-
branch (Figure 46-1). This sorting occurs because pro-
clude (1) use of yeast mutants; (2) application of re-
teins synthesized on membrane-bound polyribosomes
combinant DNA techniques (eg, mutating or eliminat-
contain a signal peptide that mediates their attach-
ing particular sequences in proteins, or fusing new
ment to the membrane of the ER. Further details on
sequences onto them; and (3) development of in vitro
498
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
499
Proteins
about 20-80 amino acids in length, which is not highly
Mitochondrial
conserved but contains many positively charged amino
acids (eg, Lys or Arg). The presequence is equivalent to
Nuclear
(1) Cytosolic
a signal peptide mediating attachment of polyribosomes
Peroxisomal
to membranes of the ER (see below), but in this in-
Cytosolic
stance targeting proteins to the matrix; if the leader se-
Polyribosomes
quence is cleaved off, potential matrix proteins will not
ER membrane
reach their destination.
GA membrane
Translocation is believed to occur posttranslation-
(2) Rough ER
Plasma membrane
ally, after the matrix proteins are released from the cy-
tosolic polyribosomes. Interactions with a number of
Secretory
cytosolic proteins that act as chaperones (see below)
Lysosomal enzymes
and as targeting factors occur prior to translocation.
Figure 46-1. Diagrammatic representation of the
Two distinct translocation complexes are situated
two branches of protein sorting occurring by synthesis
in the outer and inner mitochondrial membranes, re-
ferred to
(respectively) as TOM
(translocase-of-the-
on (1) cytosolic and (2) membrane-bound polyribo-
outer membrane) and TIM (translocase-of-the-inner
somes. The mitochondrial proteins listed are encoded
membrane). Each complex has been analyzed and
by nuclear genes. Some of the signals used in further
found to be composed of a number of proteins, some of
sorting of these proteins are listed in Table 46-4. (ER,
which act as receptors for the incoming proteins and
endoplasmic reticulum; GA, Golgi apparatus.)
others as components of the transmembrane pores
through which these proteins must pass. Proteins must
be in the unfolded state to pass through the com-
systems (eg, to study translocation in the ER and mech-
plexes, and this is made possible by ATP-dependent
anisms of vesicle formation).
binding to several chaperone proteins. The roles of
The sorting of proteins belonging to the cytosolic
chaperone proteins in protein folding are discussed later
branch referred to above is described next, starting with
in this chapter. In mitochondria, they are involved in
mitochondrial proteins.
translocation, sorting, folding, assembly, and degrada-
tion of imported proteins. A proton-motive force
across the inner membrane is required for import; it is
made up of the electric potential across the membrane
THE MITOCHONDRION BOTH IMPORTS
(inside negative) and the pH gradient (see Chapter
& SYNTHESIZES PROTEINS
12). The positively charged leader sequence may be
Mitochondria contain many proteins. Thirteen pro-
helped through the membrane by the negative charge
teins
(mostly membrane components of the electron
in the matrix. The presequence is split off in the matrix
transport chain) are encoded by the mitochondrial
by a matrix-processing peptidase
(MPP). Contact
genome and synthesized in that organelle using its own
with other chaperones present in the matrix is essential
protein-synthesizing system. However, the majority (at
to complete the overall process of import. Interaction
least several hundred) are encoded by nuclear genes,
with mt-Hsp70 (Hsp = heat shock protein) ensures
are synthesized outside the mitochondria on cytosolic
proper import into the matrix and prevents misfolding
polyribosomes, and must be imported. Yeast cells have
or aggregation, while interaction with the mt-Hsp60-
proved to be a particularly useful system for analyzing
Hsp10 system ensures proper folding. The latter pro-
the mechanisms of import of mitochondrial proteins,
teins resemble the bacterial GroEL chaperonins, a sub-
partly because it has proved possible to generate a vari-
class of chaperones that form complex cage-like
ety of mutants that have illuminated the fundamental
assemblies made up of heptameric ring structures. The
processes involved. Most progress has been made in the
interactions of imported proteins with the above chap-
study of proteins present in the mitochondrial matrix,
erones require hydrolysis of ATP to drive them.
such as the F1 ATPase subunits. Only the pathway of
The details of how preproteins are translocated have
import of matrix proteins will be discussed in any detail
not been fully elucidated. It is possible that the electric
here.
potential associated with the inner mitochondrial mem-
Matrix proteins must pass from cytosolic polyribo-
brane causes a conformational change in the unfolded
somes through the outer and inner mitochondrial
preprotein being translocated and that this helps to pull
membranes to reach their destination. Passage through
it across. Furthermore, the fact that the matrix is more
the two membranes is called translocation. They have
negative than the intermembrane space may “attract”
an amino terminal leader sequence
(presequence),
the positively charged amino terminal of the preprotein
Cytosol
Early
Secretory
endosome
Constitutive
storage
(excretory)
granule
transport
Prelysosome
vesicle
(or late endosome)
Lysosome
TGN
Golgi
trans
apparatus
medial
cis
Endoplasmic
reticulum
Nuclear
envelope
Figure 46-2. Diagrammatic representation of the rough endoplasmic reticu-
lum branch of protein sorting. Newly synthesized proteins are inserted into the
ER membrane or lumen from membrane-bound polyribosomes (small black cir-
cles studding the cytosolic face of the ER). Those proteins that are transported
out of the ER (indicated by solid black arrows) do so from ribosome-free transi-
tional elements. Such proteins may then pass through the various subcompart-
ments of the Golgi until they reach the TGN, the exit side of the Golgi. In the TGN,
proteins are segregated and sorted. Secretory proteins accumulate in secretory
storage granules from which they may be expelled as shown in the upper right-
hand side of the figure. Proteins destined for the plasma membrane or those that
are secreted in a constitutive manner are carried out to the cell surface in trans-
port vesicles, as indicated in the upper middle area of the figure. Some proteins
may reach the cell surface via late and early endosomes. Other proteins enter
prelysosomes (late endosomes) and are selectively transferred to lysosomes. The
endocytic pathway illustrated in the upper left-hand area of the figure is consid-
ered elsewhere in this chapter. Retrieval from the Golgi apparatus to the ER is not
considered in this scheme. (CGN, cis-Golgi network; TGN, trans-Golgi network.)
(Courtesy of E Degen.)
500
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
501
to enter the matrix. Close contact between the mem-
These macromolecules include histones, ribosomal pro-
brane sites in the outer and inner membranes involved
teins and ribosomal subunits, transcription factors, and
in translocation is necessary.
mRNA molecules. The transport is bidirectional and
The above describes the major pathway of proteins
occurs through the nuclear pore complexes (NPCs).
destined for the mitochondrial matrix. However, cer-
These are complex structures with a mass approxi-
tain proteins insert into the outer mitochondrial
mately 30 times that of a ribosome and are composed
membrane facilitated by the TOM complex. Others
of about 100 different proteins. The diameter of an
stop in the intermembrane space, and some insert into
NPC is approximately 9 nm but can increase up to ap-
the inner membrane. Yet others proceed into the ma-
proximately 28 nm. Molecules smaller than about 40
trix and then return to the inner membrane or inter-
kDa can pass through the channel of the NPC by diffu-
membrane space. A number of proteins contain two
sion, but special translocation mechanisms exist for
signaling sequences—one to enter the mitochondrial
larger molecules. These mechanisms are under intensive
matrix and the other to mediate subsequent relocation
investigation, but some important features have already
(eg, into the inner membrane). Certain mitochondrial
emerged.
proteins do not contain presequences (eg, cytochrome
Here we shall mainly describe nuclear import of
c, which locates in the inter membrane space), and oth-
certain macromolecules. The general picture that has
ers contain internal presequences. Overall, proteins
emerged is that proteins to be imported (cargo mole-
employ a variety of mechanisms and routes to attain
cules) carry a nuclear localization signal (NLS). One
their final destinations in mitochondria.
example of an NLS is the amino acid sequence (Pro)2-
General features that apply to the import of proteins
(Lys)4-Ala-Lys-Val, which is markedly rich in basic ly-
into organelles, including mitochondria and some of
sine residues. Depending on which NLS it contains, a
the other organelles to be discussed below, are summa-
cargo molecule interacts with one of a family of soluble
rized in Table 46-1.
proteins called importins, and the complex docks at
the NPC. Another family of proteins called Ran plays a
IMPORTINS & EXPORTINS ARE
critical regulatory role in the interaction of the complex
with the NPC and in its translocation through the
INVOLVED IN TRANSPORT
NPC. Ran proteins are small monomeric nuclear GTP-
OF MACROMOLECULES IN
ases and, like other GTPases, exist in either GTP-
& OUT OF THE NUCLEUS
bound or GDP-bound states. They are themselves reg-
ulated by guanine nucleotide exchange factors
It has been estimated that more than a million macro-
(GEFs; eg, the protein RCC1 in eukaryotes), which are
molecules per minute are transported between the nu-
located in the nucleus, and Ran guanine-activating
cleus and the cytoplasm in an active eukaryotic cell.
proteins (GAPs), which are predominantly cytoplas-
mic. The GTP-bound state of Ran is favored in the nu-
cleus and the GDP-bound state in the cytoplasm. The
Table 46-1. Some general features of protein
conformations and activities of Ran molecules vary de-
import to organelles.1
pending on whether GTP or GDP is bound to them
(the GTP-bound state is active; see discussion of G pro-
• Import of a protein into an organelle usually occurs in three
teins in Chapter 43). The asymmetry between nucleus
stages: recognition, translocation, and maturation.
and cytoplasm—with respect to which of these two nu-
• Targeting sequences on the protein are recognized in the
cleotides is bound to Ran molecules—is thought to be
cytoplasm or on the surface of the organelle.
crucial in understanding the roles of Ran in transferring
• The protein is unfolded for translocation, a state main-
complexes unidirectionally across the NPC. When
tained in the cytoplasm by chaperones.
cargo molecules are released inside the nucleus, the im-
• Threading of the protein through a membrane requires en-
ergy and organellar chaperones on the trans side of the
portins recirculate to the cytoplasm to be used again.
membrane.
Figure 46-3 summarizes some of the principal features
• Cycles of binding and release of the protein to the chaper-
in the above process.
one result in pulling of its polypeptide chain through the
Other small monomeric GTPases (eg, ARF, Rab,
membrane.
Ras, and Rho) are important in various cellular pro-
• Other proteins within the organelle catalyze folding of the
cesses such as vesicle formation and transport (ARF and
protein, often attaching cofactors or oligosaccharides and
Rab; see below), certain growth and differentiation
assembling them into active monomers or oligomers.
processes (Ras), and formation of the actin cytoskele-
ton. A process involving GTP and GDP is also crucial
1Data from McNew JA, Goodman JM: The targeting and assembly
of peroxisomal proteins: some old rules do not apply. Trends
in the transport of proteins across the membrane of the
Biochem Sci 1998;21:54.
ER (see below).
502
/
CHAPTER 46
Targeting
1
β
RanGTP
RanGTP
α
GDP
OFF
GTP
ON
Ran
GAP
Ran
Docking
+
Pi
Ran exchange
GEF
+
2
RanGDP
RanGDP
α
β
Ran
GTP
GDP
3
Ran
GEF ?
RanBP1
RanBP1
GDP
Ran
4
GTP
α
β
Termination
Translocation
Ran
6
GTP
5
Ran
α
+
Ran
β
Pi
GAP
GTP
7
?
Ran
+
Ran
GTP
GDP
α
+
β
8
Recycle factors
Figure 46-3. Schematic representation of the proposed role of Ran in the import of cargo
carrying an NLS signal. (1) The targeting complex forms when the NLS receptor (α, an importin)
binds NLS cargo and the docking factor (β). (2) Docking occurs at filamentous sites that pro-
trude from the NPC. Ran-GDP docks independently. (3) Transfer to the translocation channel is
triggered when a RanGEF converts Ran-GDP to Ran-GTP. (4) The NPC catalyzes translocation of
the targeting complex. (5) Ran-GTP is recycled to Ran-GDP by docked RanGAP. (6) Ran-GTP dis-
rupts the targeting complex by binding to a site on β that overlaps with a binding site. (7) NLS
cargo dissociates from α, and Ran-GTP may dissociate from β. (8) α and β factors are recycled to
the cytoplasm. Inset: The Ran translocation switch is off in the cytoplasm and on in the nucleus.
Ran-GTP promotes NLS- and NES-directed translocation. However, cytoplasmic Ran is enriched
in Ran-GDP (OFF) by an active RanGAP, and nuclear pools are enriched in Ran-GTP (ON) by an
active GEF. RanBP1 promotes the contrary activities of these two factors. Direct linkage of nu-
clear and cytoplasmic pools of Ran occurs through the NPC by an unknown shuttling mecha-
nism. Pi, inorganic phosphate; NLS, nuclear localization signal; NPC, nuclear pore complex; GEF,
guanine nucleotide exchange factor; GAP, guanine-activating protein; NES, nuclear export sig-
nal; BP, binding protein. (Reprinted, with permission, from Goldfarb DS: Whose finger is on the
switch? Science 1997;276:1814.)
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
503
Proteins similar to importins, referred to as ex-
the synthesis of bile acids, and a marked reduction of
portins, are involved in export of many macromole-
plasmalogens. The condition is believed to be due to
cules from the nucleus. Cargo molecules for export
mutations in genes encoding certain proteins—so
carry nuclear export signals (NESs). Ran proteins are
called peroxins—involved in various steps of peroxi-
involved in this process also, and it is now established
some biogenesis (such as the import of proteins de-
that the processes of import and export share a number
scribed above), or in genes encoding certain peroxiso-
of common features.
mal enzymes themselves. Two closely related conditions
are neonatal adrenoleukodystrophy and infantile
Refsum disease. Zellweger syndrome and these two
MOST CASES OF ZELLWEGER SYNDROME
conditions represent a spectrum of overlapping fea-
ARE DUE TO MUTATIONS IN GENES
tures, with Zellweger syndrome being the most severe
(many proteins affected) and infantile Refsum disease
INVOLVED IN THE BIOGENESIS
the least severe (only one or a few proteins affected).
OF PEROXISOMES
Table 46-2 lists some features of these and related con-
The peroxisome is an important organelle involved in
ditions.
aspects of the metabolism of many molecules, including
fatty acids and other lipids (eg, plasmalogens, choles-
THE SIGNAL HYPOTHESIS EXPLAINS
terol, bile acids), purines, amino acids, and hydrogen
HOW POLYRIBOSOMES BIND TO THE
peroxide. The peroxisome is bounded by a single mem-
ENDOPLASMIC RETICULUM
brane and contains more than 50 enzymes; catalase and
urate oxidase are marker enzymes for this organelle. Its
As indicated above, the rough ER branch is the second
proteins are synthesized on cytosolic polyribosomes and
of the two branches involved in the synthesis and sort-
fold prior to import. The pathways of import of a num-
ing of proteins. In this branch, proteins are synthesized
ber of its proteins and enzymes have been studied, some
on membrane-bound polyribosomes and translocated
being matrix components and others membrane com-
into the lumen of the rough ER prior to further sorting
ponents. At least two peroxisomal-matrix targeting
(Figure 46-2).
sequences (PTSs) have been discovered. One, PTS1, is
The signal hypothesis was proposed by Blobel and
a tripeptide (ie, Ser-Lys-Leu [SKL], but variations of
Sabatini partly to explain the distinction between free
this sequence have been detected) located at the car-
and membrane-bound polyribosomes. They found that
boxyl terminal of a number of matrix proteins, includ-
proteins synthesized on membrane-bound polyribo-
ing catalase. Another, PTS2, consisting of about 26-36
somes contained a peptide extension (signal peptide)
amino acids, has been found in at least four matrix pro-
teins (eg, thiolase) and, unlike PTS1, is cleaved after
Table 46-2. Disorders due to peroxisomal
entry into the matrix. Proteins containing PTS1 se-
quences form complexes with a soluble receptor protein
abnormalities.1
(PTS1R) and proteins containing PTS2 sequences
complex with another, PTS2R. The resulting com-
MIM Number2
plexes then interact with a membrane receptor, Pex14p.
Zellweger syndrome
214100
Proteins involved in further transport of proteins into
Neonatal adrenoleukodystrophy
202370
the matrix are also present. Most peroxisomal mem-
Infantile Refsum disease
266510
brane proteins have been found to contain neither of
Hyperpipecolic acidemia
239400
the above two targeting sequences, but apparently con-
Rhizomelic chondrodysplasia punctata
215100
tain others. The import system can handle intact
Adrenoleukodystrophy
300100
oligomers (eg, tetrameric catalase). Import of matrix
Pseudo-neonatal adrenoleukodystrophy
264470
proteins requires ATP, whereas import of membrane
Pseudo-Zellweger syndrome
261510
proteins does not.
Hyperoxaluria type 1
259900
Interest in import of proteins into peroxisomes has
Acatalasemia
115500
been stimulated by studies on Zellweger syndrome.
Glutaryl-CoA oxidase deficiency
231690
This condition is apparent at birth and is characterized
1Reproduced, with permission, from Seashore MR, Wappner RS:
by profound neurologic impairment, victims often
Genetics in Primary Care & Clinical Medicine. Appleton & Lange,
dying within a year. The number of peroxisomes can
1996.
vary from being almost normal to being virtually absent
2MIM = Mendelian Inheritance in Man. Each number specifies a ref-
in some patients. Biochemical findings include an accu-
erence in which information regarding each of the above condi-
mulation of very long chain fatty acids, abnormalities of
tions can be found.
504
/
CHAPTER 46
at their amino terminals which mediated their attach-
and the β subunit spans the membrane. When the SRP-
ment to the membranes of the ER. As noted above,
signal peptide complex interacts with the receptor, the
proteins whose entire synthesis occurs on free polyribo-
exchange of GDP for GTP is stimulated. This form of
somes lack this signal peptide. An important aspect of
the receptor (with GTP bound) has a high affinity for
the signal hypothesis was that it suggested—as turns
the SRP and thus releases the signal peptide, which binds
out to be the case—that all ribosomes have the same
to the translocation machinery (translocon) also present
structure and that the distinction between membrane-
in the ER membrane. The α subunit then hydrolyzes its
bound and free ribosomes depends solely on the for-
bound GTP, restoring GDP and completing a GTP-
mer’s carrying proteins that have signal peptides. Much
GDP cycle. The unidirectionality of this cycle helps drive
evidence has confirmed the original hypothesis. Because
the interaction of the polyribosome and its signal peptide
many membrane proteins are synthesized on mem-
with the ER membrane in the forward direction.
brane-bound polyribosomes, the signal hypothesis plays
The translocon consists of a number of membrane
an important role in concepts of membrane assembly.
proteins that form a protein-conducting channel in the
Some characteristics of signal peptides are summarized
ER membrane through which the newly synthesized
in Table 46-3.
protein may pass. The channel appears to be open only
Figure 46-4 illustrates the principal features in rela-
when a signal peptide is present, preserving conductance
tion to the passage of a secreted protein through the
across the ER membrane when it closes. The conduc-
membrane of the ER. It incorporates features from the
tance of the channel has been measured experimentally.
original signal hypothesis and from subsequent work.
Specific functions of a number of components of the
The mRNA for such a protein encodes an amino termi-
translocon have been identified or suggested. TRAM
nal signal peptide (also variously called a leader se-
(translocating chain-associated membrane) protein may
quence, a transient insertion signal, a signal sequence,
bind the signal sequence as it initially interacts with the
or a presequence). The signal hypothesis proposed that
translocon and the Sec61p complex (consisting of three
the protein is inserted into the ER membrane at the
proteins) binds the heavy subunit of the ribosome.
same time as its mRNA is being translated on polyribo-
The insertion of the signal peptide into the conduct-
somes, so-called cotranslational insertion. As the sig-
ing channel, while the other end of the parent protein is
nal peptide emerges from the large subunit of the ribo-
still attached to ribosomes, is termed
“cotranslational
some, it is recognized by a signal recognition particle
insertion.” The process of elongation of the remaining
(SRP) that blocks further translation after about 70
portion of the protein probably facilitates passage of the
amino acids have been polymerized (40 buried in the
nascent protein across the lipid bilayer as the ribosomes
large ribosomal subunit and 30 exposed). The block is
remain attached to the membrane of the ER. Thus, the
referred to as elongation arrest. The SRP contains six
rough (or ribosome-studded) ER is formed. It is impor-
proteins and has a 7S RNA associated with it that is
tant that the protein be kept in an unfolded state prior
closely related to the Alu family of highly repeated
to entering the conducting channel—otherwise, it may
DNA sequences (Chapter 36). The SRP-imposed block
not be able to gain access to the channel.
is not released until the SRP-signal peptide-polyribo-
Ribosomes remain attached to the ER during syn-
some complex has bound to the so-called docking pro-
thesis of signal peptide-containing proteins but are re-
tein (SRP-R, a receptor for the SRP) on the ER mem-
leased and dissociated into their two types of subunits
brane; the SRP thus guides the signal peptide to the
when the process is completed. The signal peptide is
SRP-R and prevents premature folding and expulsion
hydrolyzed by signal peptidase, located on the luminal
of the protein being synthesized into the cytosol.
side of the ER membrane (Figure 46-4), and then is
The SRP-R is an integral membrane protein com-
apparently rapidly degraded by proteases.
posed of α and β subunits. The α subunit binds GDP
Cytochrome P450 (Chapter 53), an integral ER
membrane protein, does not completely cross the mem-
brane. Instead, it resides in the membrane with its sig-
Table 46-3. Some properties of signal peptides.
nal peptide intact. Its passage through the membrane is
prevented by a sequence of amino acids called a halt- or
• Usually, but not always, located at the amino terminal
stop-transfer signal.
• Contain approximately 12-35 amino acids
Secretory proteins and proteins destined for mem-
• Methionine is usually the amino terminal amino acid
branes distal to the ER completely traverse the mem-
• Contain a central cluster of hydrophobic amino acids
brane bilayer and are discharged into the lumen of the
• Contain at least one positively charged amino acid near
ER. N-Glycan chains, if present, are added (Chapter
their amino terminal
47) as these proteins traverse the inner part of the ER
• Usually cleaved off at the carboxyl terminal end of an Ala
membrane—a process called “cotranslational glycosyla-
residue by signal peptidase
tion.” Subsequently, the proteins are found in the
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
505
3′
Signal codons
Signal peptide
SRP
Signal peptidase
Ribosome receptor
Signal receptor
Figure 46-4. Diagram of the signal hypothesis for the transport of secreted proteins across the
ER membrane. The ribosomes synthesizing a protein move along the messenger RNA specifying the
amino acid sequence of the protein. (The messenger is represented by the line between 5′ and 3′.)
The codon AUG marks the start of the message for the protein; the hatched lines that follow AUG
represent the codons for the signal sequence. As the protein grows out from the larger ribosomal
subunit, the signal sequence is exposed and bound by the signal recognition particle (SRP). Transla-
tion is blocked until the complex binds to the “docking protein,” also designated SRP-R (repre-
sented by the solid bar) on the ER membrane. There is also a receptor (open bar) for the ribosome
itself. The interaction of the ribosome and growing peptide chain with the ER membrane results in
the opening of a channel through which the protein is transported to the interior space of the ER.
During translocation, the signal sequence of most proteins is removed by an enzyme called the
“signal peptidase,” located at the luminal surface of the ER membrane. The completed protein is
eventually released by the ribosome, which then separates into its two components, the large and
small ribosomal subunits. The protein ends up inside the ER. See text for further details. (Slightly
modified and reproduced, with permission, from Marx JL: Newly made proteins zip through the cell. Sci-
ence 1980;207:164. Copyright © 1980 by the American Association for the Advancement of Science.)
lumen of the Golgi apparatus, where further changes in
PROTEINS FOLLOW SEVERAL ROUTES
glycan chains occur (Figure 47-9) prior to intracellular
TO BE INSERTED INTO OR ATTACHED
distribution or secretion. There is strong evidence that
TO THE MEMBRANES OF THE
the signal peptide is involved in the process of protein
ENDOPLASMIC RETICULUM
insertion into ER membranes. Mutant proteins, con-
taining altered signal peptides in which a hydrophobic
The routes that proteins follow to be inserted into the
amino acid is replaced by a hydrophilic one, are not in-
membranes of the ER include the following.
serted into ER membranes. Nonmembrane proteins
A. COTRANSLATIONAL INSERTION
(eg, α-globin) to which signal peptides have been at-
tached by genetic engineering can be inserted into the
Figure 46-5 shows a variety of ways in which proteins
lumen of the ER or even secreted.
are distributed in the plasma membrane. In particular,
There is considerable evidence that a second trans-
the amino terminals of certain proteins (eg, the LDL re-
poson in the ER membrane is involved in retrograde
ceptor) can be seen to be on the extracytoplasmic face,
transport of various molecules from the ER lumen to
whereas for other proteins (eg, the asialoglycoprotein re-
the cytosol. These molecules include unfolded or mis-
ceptor) the carboxyl terminals are on this face. To ex-
folded glycoproteins, glycopeptides, and oligosaccha-
plain these dispositions, one must consider the initial
rides. Some at least of these molecules are degraded in
biosynthetic events at the ER membrane. The LDL re-
proteasomes. Thus, there is two-way traffic across the
ceptor enters the ER membrane in a manner analogous
ER membrane.
to a secretory protein (Figure 46-4); it partly traverses
506
/
CHAPTER 46
N
N
N
N
N
EXTRACYTOPLASMIC
C
FACE
N
C C
N
C
Various transporters (eg, glucose)
C
C
C
C
N
Insulin and
N
Influenza neuraminidase
IGF-I receptors
G protein-coupled receptors
Asialoglycoprotein receptor
Transferrin receptor
HLA-DR invariant chain
LDL receptor
HLA-A heavy chain
Influenza hemagglutinin
Figure 46-5. Variations in the way in which proteins are inserted into membranes. This
schematic representation, which illustrates a number of possible orientations, shows the seg-
ments of the proteins within the membrane as α-helices and the other segments as lines. The
LDL receptor, which crosses the membrane once and has its amino terminal on the exterior, is
called a type I transmembrane protein. The asialoglycoprotein receptor, which also crosses the
membrane once but has its carboxyl terminal on the exterior, is called a type II transmembrane
protein. The various transporters indicated (eg, glucose) cross the membrane a number of times
and are called type III transmembrane proteins; they are also referred to as polytopic membrane
proteins. (N, amino terminal; C, carboxyl terminal.) (Adapted, with permission, from Wickner WT,
Lodish HF: Multiple mechanisms of protein insertion into and across membranes. Science
1985;230:400. Copyright © 1985 by the American Association for the Advancement of Science.)
the ER membrane, its signal peptide is cleaved, and its
cleaved insertion sequences and as halt-transfer signals,
amino terminal protrudes into the lumen. However, it is
respectively. Each pair of helical segments is inserted as a
retained in the membrane because it contains a highly
hairpin. Sequences that determine the structure of a
hydrophobic segment, the halt- or stop-transfer signal.
protein in a membrane are called topogenic sequences.
This sequence forms the single transmembrane segment
As explained in the legend to Figure 46-5, the above
of the protein and is its membrane-anchoring domain.
three proteins are examples of type I, type II, and type
The small patch of ER membrane in which the newly
III transmembrane proteins.
synthesized LDL receptor is located subsequently buds
off as a component of a transport vesicle, probably from
B. SYNTHESIS ON FREE POLYRIBOSOMES
the transitional elements of the ER (Figure 46-2). As
& SUBSEQUENT ATTACHMENT TO THE
described below in the discussion of asymmetry of pro-
ENDOPLASMIC RETICULUM MEMBRANE
teins and lipids in membrane assembly, the disposition
of the receptor in the ER membrane is preserved in the
An example is cytochrome b5, which enters the ER
vesicle, which eventually fuses with the plasma mem-
membrane spontaneously.
brane. In contrast, the asialoglycoprotein receptor pos-
sesses an internal insertion sequence, which inserts into
C. RETENTION AT THE LUMINAL ASPECT
the membrane but is not cleaved. This acts as an anchor,
OF THE ENDOPLASMIC RETICULUM
and its carboxyl terminal is extruded through the mem-
BY SPECIFIC AMINO ACID SEQUENCES
brane. The more complex disposition of the trans-
porters (eg, for glucose) can be explained by the fact
A number of proteins possess the amino acid sequence
that alternating transmembrane α-helices act as un-
KDEL (Lys-Asp-Glu-Leu) at their carboxyl terminal.
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
507
This sequence specifies that such proteins will be at-
denote transport steps that may be independent of tar-
tached to the inner aspect of the ER in a relatively loose
geting signals, whereas the vertical open arrows repre-
manner. The chaperone BiP (see below) is one such
sent steps that depend on specific signals. Thus, flow of
protein. Actually, KDEL-containing proteins first travel
certain proteins
(including membrane proteins) from
to the Golgi, interact there with a specific KDEL recep-
the ER to the plasma membrane (designated “bulk
tor protein, and then return in transport vesicles to the
flow,” as it is nonselective) probably occurs without any
ER, where they dissociate from the receptor.
targeting sequences being involved, ie, by default. On
the other hand, insertion of resident proteins into the
D. RETROGRADE TRANSPORT FROM
ER and Golgi membranes is dependent upon specific
THE GOLGI APPARATUS
signals
(eg, KDEL or halt-transfer sequences for the
Certain other non-KDEL-containing proteins destined
ER). Similarly, transport of many enzymes to lysosomes
for the membranes of the ER also pass to the Golgi and
is dependent upon the Man 6-P signal (Chapter 47),
then return, by retrograde vesicular transport, to the ER
and a signal may be involved for entry of proteins into
to be inserted therein (see below).
secretory granules. Table 46-4 summarizes informa-
The foregoing paragraphs demonstrate that a vari-
tion on sequences that are known to be involved in tar-
ety of routes are involved in assembly of the proteins of
geting various proteins to their correct intracellular sites.
the ER membranes; a similar situation probably holds
for other membranes
(eg, the mitochondrial mem-
CHAPERONES ARE PROTEINS
branes and the plasma membrane). Precise targeting se-
THAT PREVENT FAULTY FOLDING
quences have been identified in some instances (eg,
& UNPRODUCTIVE INTERACTIONS
KDEL sequences).
The topic of membrane biogenesis is discussed fur-
OF OTHER PROTEINS
ther later in this chapter.
Exit from the ER may be the rate-limiting step in the
secretory pathway. In this context, it has been found
PROTEINS MOVE THROUGH CELLULAR
that certain proteins play a role in the assembly or
proper folding of other proteins without themselves
COMPARTMENTS TO SPECIFIC
being components of the latter. Such proteins are called
DESTINATIONS
molecular chaperones; a number of important proper-
A scheme representing the possible flow of proteins
ties of these proteins are listed in Table 46-5, and the
along the ER → Golgi apparatus → plasma membrane
names of some of particular importance in the ER are
route is shown in Figure 46-6. The horizontal arrows
listed in Table 46-6. Basically, they stabilize unfolded
Lysosomes
cis
medial
trans
Cell
ER
Golgi
Golgi
Golgi
surface
Secretory
storage vesicles
Figure 46-6. Flow of membrane proteins from the endoplas-
mic reticulum (ER) to the cell surface. Horizontal arrows denote
steps that have been proposed to be signal independent and
thus represent bulk flow. The open vertical arrows in the boxes
denote retention of proteins that are resident in the membranes
of the organelle indicated. The open vertical arrows outside the
boxes indicate signal-mediated transport to lysosomes and secre-
tory storage granules. (Reproduced, with permission, from Pfeffer
SR, Rothman JE: Biosynthetic protein transport and sorting by the en-
doplasmic reticulum and Golgi. Annu Rev Biochem 1987;56:829.)
508
/
CHAPTER 46
Table 46-4. Some sequences or compounds that
Table 46-6. Some chaperones and enzymes
direct proteins to specific organelles.
involved in folding that are located in the rough
endoplasmic reticulum.
Targeting Sequence
or Compound
Organelle Targeted
• BiP (immunoglobulin heavy chain binding protein)
• GRP94 (glucose-regulated protein)
Signal peptide sequence
Membrane of ER
• Calnexin
Amino terminal
Luminal surface of ER
• Calreticulin
KDEL sequence
• PDI (protein disulfide isomerase)
(Lys-Asp-Glu-Leu)
• PPI (peptidyl prolyl cis-trans isomerase)
Amino terminal sequence
Mitochondrial matrix
(20-80 residues)
NLS1 (eg, Pro2-Lys2-Ala-
Nucleus
Several examples of chaperones were introduced
Lys-Val)
above when the sorting of mitochondrial proteins was
discussed. The immunoglobulin heavy chain binding
PTS1 (eg, Ser-Lys-Leu)
Peroxisome
protein (BiP) is located in the lumen of the ER. This
Mannose 6-phosphate
Lysosome
protein will bind abnormally folded immunoglobulin
1NLS, nuclear localization signal; PTS, peroxisomal-matrix target-
heavy chains and certain other proteins and prevent
ing sequence.
them from leaving the ER, in which they are degraded.
Another important chaperone is calnexin, a calcium-
binding protein located in the ER membrane. This pro-
tein binds a wide variety of proteins, including mixed
histocompatibility
(MHC) antigens and a variety of
or partially folded intermediates, allowing them time to
serum proteins. As mentioned in Chapter 47, calnexin
fold properly, and prevent inappropriate interactions,
binds the monoglycosylated species of glycoproteins
thus combating the formation of nonfunctional struc-
that occur during processing of glycoproteins, retaining
tures. Most chaperones exhibit ATPase activity and
them in the ER until the glycoprotein has folded prop-
bind ADP and ATP. This activity is important for their
erly. Calreticulin, which is also a calcium-binding pro-
effect on folding. The ADP-chaperone complex often
tein, has properties similar to those of calnexin; it is not
has a high affinity for the unfolded protein, which,
membrane-bound. Chaperones are not the only pro-
when bound, stimulates release of ADP with replace-
teins in the ER lumen that are concerned with proper
ment by ATP. The ATP-chaperone complex, in turn,
folding of proteins. Two enzymes are present that play
releases segments of the protein that have folded prop-
an active role in folding. Protein disulfide isomerase
erly, and the cycle involving ADP and ATP binding is
(PDI) promotes rapid reshuffling of disulfide bonds
repeated until the folded protein is released.
until the correct set is achieved. Peptidyl prolyl isom-
erase (PPI) accelerates folding of proline-containing
proteins by catalyzing the cis-trans isomerization of
X-Pro bonds, where X is any amino acid residue.
Table 46-5. Some properties of chaperone
proteins.
TRANSPORT VESICLES ARE KEY PLAYERS
IN INTRACELLULAR PROTEIN TRAFFIC
• Present in a wide range of species from bacteria to humans
• Many are so-called heat shock proteins (Hsp)
Most proteins that are synthesized on membrane-
• Some are inducible by conditions that cause unfolding of
bound polyribosomes and are destined for the Golgi
newly synthesized proteins (eg, elevated temperature and
apparatus or plasma membrane reach these sites inside
various chemicals)
transport vesicles. The precise mechanisms by which
• They bind to predominantly hydrophobic regions of un-
proteins synthesized in the rough ER are inserted into
folded and aggregated proteins
these vesicles are not known. Those involved in trans-
• They act in part as a quality control or editing mechanism
port from the ER to the Golgi apparatus and vice
for detecting misfolded or otherwise defective proteins
versa—and from the Golgi to the plasma membrane—
• Most chaperones show associated ATPase activity, with ATP
are mainly clathrin-free, unlike the coated vesicles in-
or ADP being involved in the protein-chaperone interaction
volved in endocytosis (see discussions of the LDL re-
• Found in various cellular compartments such as cytosol,
ceptor in Chapters 25 and 26). For the sake of clarity,
mitochondria, and the lumen of the endoplasmic reticulum
the non-clathrin-coated vesicles will be referred to in
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
509
this text as transport vesicles. There is evidence that
Table 46-7. Factors involved in the formation of
proteins destined for the membranes of the Golgi appa-
non-clathrin-coated vesicles and their transport.
ratus contain specific signal sequences. On the other
hand, most proteins destined for the plasma membrane
•
ARF: ADP-ribosylation factor, a GTPase
or for secretion do not appear to contain specific sig-
•
Coatomer: A family of at least seven coat proteins (α, β, γ, δ,
nals, reaching these destinations by default.
ε, β′, and ζ). Different transport vesicles have different com-
plements of coat proteins.
The Golgi Apparatus Is Involved in
•
SNAP: Soluble NSF attachment factor
•
SNARE: SNAP receptor
Glycosylation & Sorting of Proteins
•
v-SNARE: Vesicle SNARE
The Golgi apparatus plays two important roles in mem-
•
t-SNARE: Target SNARE
brane synthesis. First, it is involved in the processing
•
GTP-γ-S: A nonhydrolyzable analog of GTP, used to test the
of the oligosaccharide chains of membrane and other
involvement of GTP
N-linked glycoproteins and also contains enzymes in-
•
NEM: N-Ethylmaleimide, a chemical that alkylates sulfhy-
volved in O-glycosylation (see Chapter 47). Second, it
dryl groups
is involved in the sorting of various proteins prior to
•
NSF: NEM-sensitive factor, an ATPase
their delivery to their appropriate intracellular destina-
•
Rab proteins: A family of ras-related proteins first observed
in rat brain; they are GTPases and are active when GTP is
tions. All parts of the Golgi apparatus participate in the
found
first role, whereas the trans-Golgi is particularly in-
•
Sec1: A member of a family of proteins that attach to
volved in the second and is very rich in vesicles. Because
t-SNAREs and are displaced from them by Rab proteins,
of their central role in protein transport, considerable
thereby allowing v-SNARE-t-SNARE interactions to occur.
research has been conducted in recent years concerning
the formation and fate of transport vesicles.
A Model of Non-Clathrin-Coated Vesicles
Step 2: Membrane-associated ARF recruits the coat
proteins that comprise the coatomer shell from the
Involves SNAREs & Other Factors
cytosol, forming a coated bud.
Vesicles lie at the heart of intracellular transport of
Step 3: The bud pinches off in a process involving
many proteins. Recently, significant progress has been
acyl-CoA—and probably ATP—to complete the
made in understanding the events involved in vesicle
formation of the coated vesicle.
formation and transport. This has transpired because of
Step 4: Coat disassembly (involving dissociation of
the use of a number of approaches. These include es-
ARF and coatomer shell) follows hydrolysis of
tablishment of cell-free systems with which to study
bound GTP; uncoating is necessary for fusion to
vesicle formation. For instance, it is possible to observe,
occur.
by electron microscopy, budding of vesicles from Golgi
Step 5: Vesicle targeting is achieved via members of
preparations incubated with cytosol and ATP. The de-
a family of integral proteins, termed v-SNAREs,
velopment of genetic approaches for studying vesicles
that tag the vesicle during its budding. v-SNAREs
in yeast has also been crucial. The picture is complex,
pair with cognate t-SNAREs in the target membrane
with its own nomenclature (Table 46-7), and involves
to dock the vesicle.
a variety of cytosolic and membrane proteins, GTP,
ATP, and accessory factors.
It is presumed that steps 4 and 5 are closely coupled
Based largely on a proposal by Rothman and col-
and that step 4 may follow step 5, with ARF and the
leagues, anterograde vesicular transport can be consid-
coatomer shell rapidly dissociating after docking.
ered to occur in eight steps (Figure 46-7). The basic
concept is that each transport vesicle bears a unique ad-
Step 6: The general fusion machinery then assem-
dress marker consisting of one or more v-SNARE pro-
bles on the paired SNARE complex; it includes an
teins, while each target membrane bears one or more
ATPase (NSF; NEM-sensitive factor) and the SNAP
complementary t-SNARE proteins with which the
(soluble NSF attachment factor) proteins. SNAPs
former interact specifically.
bind to the SNARE (SNAP receptor) complex, en-
abling NSF to bind.
Step 1: Coat assembly is initiated when ARF is ac-
tivated by binding GTP, which is exchanged for
Step 7: Hydrolysis of ATP by NSF is essential for
GDP. This leads to the association of GTP-bound
fusion, a process that can be inhibited by NEM (N-
ARF with its putative receptor (hatched in Figure
ethylmaleimide). Certain other proteins and calcium
46-7) in the donor membrane.
are also required.
510
/
CHAPTER 46
Coated
3
vesicle
4
t-SNARE
Coated
GTP-γ-S
bud
5
Acyl-CoA
SNAPs
NSF
ATP
P
i
GDP
Budding
6
SNAPs
NSF
Ca2+
2
20S fusion
Coatomer
particle
v-SNARE
ATP
1
GTP
NEM
7
Fusion
GTP
BFA
GDP
GDP
GDP
ARF
Nocodazole
Donor
Target
membrane
membrane
8
(eg, ER)
(eg, CGN)
Figure 46-7. Model of the steps in a round of anterograde vesicular transport. The
cycle starts in the bottom left-hand side of the figure, where two molecules of ARF
are represented as small ovals containing GDP. The steps in the cycle are described in
the text. Most of the abbreviations used are explained in Table 46-7. The roles of Rab
and Sec1 proteins (see text) in the overall process are not dealt with in this figure.
(CGN, cis-Golgi network; BFA, Brefeldin A.) (Adapted from Rothman JE: Mechanisms of
intracellular protein transport. Nature 1994;372:55.) (Courtesy of E Degen.)
Step 8: Retrograde transport occurs to restart the
doubt remain to be discovered. COPI vesicles are in-
cycle. This last step may retrieve certain proteins
volved in bidirectional transport from the ER to the
or recycle v-SNAREs. Nocodazole, a microtubule-
Golgi and in the reverse direction, whereas COPII vesi-
disrupting agent, inhibits this step.
cles are involved mainly in transport in the former di-
rection. Clathrin-containing vesicles are involved in
transport from the trans-Golgi network to prelysosomes
Brefeldin A Inhibits the Coating Process
and from the plasma membrane to endosomes, respec-
tively. Regarding selection of cargo molecules by vesi-
The following points expand and clarify the above.
cles, this appears to be primarily a function of the coat
(a) To participate in step 1, ARF must first be modi-
proteins of vesicles. Cargo molecules may interact with
fied by addition of myristic acid (C14:0), employing
coat proteins either directly or via intermediary proteins
myristoyl-CoA as the acyl donor. Myristoylation is one
that attach to coat proteins, and they then become en-
of a number of enzyme-catalyzed posttranslational mod-
closed in their appropriate vesicles.
ifications, involving addition of certain lipids to specific
(c) The fungal metabolite brefeldin A prevents
residues of proteins, that facilitate the binding of pro-
GTP from binding to ARF in step 1 and thus inhibits
teins to the cytosolic surfaces of membranes or vesicles.
the entire coating process. In its presence, the Golgi ap-
Others are addition of palmitate, farnesyl, and geranyl-
paratus appears to disintegrate, and fragments are lost.
geranyl; the two latter molecules are polyisoprenoids
It may do this by inhibiting the guanine nucleotide ex-
containing 15 and 20 carbon atoms, respectively.
changer involved in step 1.
(b) At least three different types of coated vesicles
(d) GTP- -S (a nonhydrolyzable analog of GTP
have been distinguished: COPI, COPII, and clathrin-
often used in investigations of the role of GTP in bio-
coated vesicles; the first two are referred to here as
chemical processes) blocks disassembly of the coat from
transport vesicles. Many other types of vesicles no
coated vesicles, leading to a build-up of coated vesicles.
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
511
(e) A family of Ras-like proteins, called the Rab pro-
Asymmetry of Both Proteins & Lipids Is
tein family, are required in several steps of intracellular
Maintained During Membrane Assembly
protein transport, regulated secretion, and endocytosis.
Vesicles formed from membranes of the ER and Golgi
They are small monomeric GTPases that attach to the
apparatus, either naturally or pinched off by homoge-
cytosolic faces of membranes via geranylgeranyl chains.
nization, exhibit transverse asymmetries of both lipid
They attach in the GTP-bound state (not shown in
and protein. These asymmetries are maintained during
Figure 46-7) to the budding vesicle. Another family of
fusion of transport vesicles with the plasma membrane.
proteins (Sec1) binds to t-SNAREs and prevents inter-
The inside of the vesicles after fusion becomes the out-
action with them and their complementary v-SNAREs.
side of the plasma membrane, and the cytoplasmic side
When a vesicle interacts with its target membrane, Rab
of the vesicles remains the cytoplasmic side of the mem-
proteins displace Sec1 proteins and the v-SNARE-
brane (Figure 46-8). Since the transverse asymmetry of
t-SNARE interaction is free to occur. It appears that
the membranes already exists in the vesicles of the ER
the Rab and Sec1 families of proteins regulate the speed
well before they are fused to the plasma membrane, a
of vesicle formation, opposing each other. Rab proteins
major problem of membrane assembly becomes under-
have been likened to throttles and Sec1 proteins to
standing how the integral proteins are inserted into the
dampers on the overall process of vesicle formation.
lipid bilayer of the ER. This problem was addressed
(f) Studies using v- and t-SNARE proteins reconsti-
earlier in this chapter.
tuted into separate lipid bilayer vesicles have indicated
Phospholipids are the major class of lipid in mem-
that they form SNAREpins, ie, SNARE complexes that
branes. The enzymes responsible for the synthesis of
link two membranes (vesicles). SNAPs and NSF are re-
phospholipids reside in the cytoplasmic surface of the
quired for formation of SNAREpins, but once they
cisternae of the ER. As phospholipids are synthesized at
have formed they can apparently lead to spontaneous
that site, they probably self-assemble into thermody-
fusion of membranes at physiologic temperature, sug-
namically stable bimolecular layers, thereby expanding
gesting that they are the minimal machinery required
the membrane and perhaps promoting the detachment
for membrane fusion.
of so-called lipid vesicles from it. It has been proposed
(g) The fusion of synaptic vesicles with the plasma
that these vesicles travel to other sites, donating their
membrane of neurons involves a series of events similar
lipids to other membranes; however, little is known
to that described above. For example, one v-SNARE is
about this matter. As indicated above, cytosolic pro-
designated synaptobrevin and two t-SNAREs are des-
teins that take up phospholipids from one membrane
ignated syntaxin and SNAP 25 (synaptosome-associ-
and release them to another (ie, phospholipid exchange
ated protein of 25 kDa). Botulinum B toxin is one of
proteins) have been demonstrated; they probably play a
the most lethal toxins known and the most serious
role in contributing to the specific lipid composition of
cause of food poisoning. One component of this toxin
various membranes.
is a protease that appears to cleave only synaptobrevin,
thus inhibiting release of acetylcholine at the neuro-
muscular junction and possibly proving fatal, depend-
Lipids & Proteins Undergo Turnover at
ing on the dose taken.
Different Rates in Different Membranes
(h) Although the above model describes non-
clathrin-coated vesicles, it appears likely that many of
It has been shown that the half-lives of the lipids of the
the events described above apply, at least in principle,
ER membranes of rat liver are generally shorter than
to clathrin-coated vesicles.
those of its proteins, so that the turnover rates of
lipids and proteins are independent. Indeed, differ-
ent lipids have been found to have different half-lives.
Furthermore, the half-lives of the proteins of these
THE ASSEMBLY OF MEMBRANES
membranes vary quite widely, some exhibiting short
IS COMPLEX
(hours) and others long (days) half-lives. Thus, individ-
There are many cellular membranes, each with its own
ual lipids and proteins of the ER membranes appear to
specific features. No satisfactory scheme describing the
be inserted into it relatively independently; this is the
assembly of any one of these membranes is available.
case for many other membranes.
How various proteins are initially inserted into the
The biogenesis of membranes is thus a complex
membrane of the ER has been discussed above. The
process about which much remains to be learned. One
transport of proteins, including membrane proteins, to
indication of the complexity involved is to consider the
various parts of the cell inside vesicles has also been de-
number of posttranslational modifications that mem-
scribed. Some general points about membrane assembly
brane proteins may be subjected to prior to attaining
remain to be addressed.
their mature state. These include proteolysis, assembly
512
/
CHAPTER 46
Membrane protein
Exterior surface
Table 46-8. Major features of membrane
assembly.
• Lipids and proteins are inserted independently into mem-
branes.
• Individual membrane lipids and proteins turn over indepen-
C
dently and at different rates.
Plasma
membrane
• Topogenic sequences (eg, signal [amino terminal or inter-
Lumen
nal] and stop-transfer) are important in determining the in-
N
sertion and disposition of proteins in membranes.
N
Cytoplasm
Integral
• Membrane proteins inside transport vesicles bud off the en-
protein
doplasmic reticulum on their way to the Golgi; final sorting
of many membrane proteins occurs in the trans-Golgi net-
Vesicle
work.
membrane
C
• Specific sorting sequences guide proteins to particular
organelles such as lysosomes, peroxisomes, and mitochon-
dria.
into multimers, glycosylation, addition of a glycophos-
phatidylinositol (GPI) anchor, sulfation on tyrosine or
carbohydrate moieties, phosphorylation, acylation, and
N
prenylation—a list that is undoubtedly not complete.
N
Nevertheless, significant progress has been made; Table
C
46-8 summarizes some of the major features of mem-
brane assembly that have emerged to date.
Table 46-9. Some disorders due to mutations in
genes encoding proteins involved in intracellular
N
N
membrane transport.1
Disorder2
Protein Involved
C
Chédiak-Higashi syndrome,
Lysosomal trafficking regula-
C
214500
tor
Figure 46-8. Fusion of a vesicle with the plasma
Combined deficiency of factors
ERGIC-53, a mannose-
membrane preserves the orientation of any integral
V and VIII, 227300
binding lectin
proteins embedded in the vesicle bilayer. Initially, the
Hermansky-Pudlak syndrome,
AP-3 adaptor complex β3A
amino terminal of the protein faces the lumen, or inner
203300
subunit
cavity, of such a vesicle. After fusion, the amino termi-
I-cell disease, 252500
N-Acetylglucosamine
nal is on the exterior surface of the plasma membrane.
1-phosphotransferase
That the orientation of the protein has not been re-
versed can be perceived by noting that the other end
Oculocerebrorenal syndrome,
OCRL-1, an inositol poly-
of the molecule, the carboxyl terminal, is always im-
30900
phosphate 5-phosphatase
mersed in the cytoplasm. The lumen of a vesicle and
1Modified from Olkonnen VM, Ikonen E: Genetic defects of intra-
the outside of the cell are topologically equivalent. (Re-
cellular-membrane transport. N Eng J Med 2000;343:1095. Certain
drawn and modified, with permission, from Lodish HF,
related conditions not listed here are also described in this publi-
Rothman JE: The assembly of cell membranes. Sci Am
cation. I-cell disease is described in Chapter 47. The majority of
the disorders listed above affect lysosomal function; readers
[Jan] 1979;240:43.)
should consult a textbook of medicine for information on the
clinical manifestations of these conditions.
2The numbers after each disorder are the OMIM numbers.
INTRACELLULAR TRAFFIC & SORTING OF PROTEINS
/
513
Various Disorders Result From Mutations
and attachment of transport vesicles to a target mem-
in Genes Encoding Proteins Involved
brane is summarized.
in Intracellular Transport
• Membrane assembly is discussed and shown to be
complex. Asymmetry of both lipids and proteins is
Some of these are listed in Table 46-9; the majority af-
maintained during membrane assembly.
fect lysosomal function. A number of other mutations
• A number of disorders have been shown to be due to
affecting intracellular protein transport have been re-
mutations in genes encoding proteins involved in
ported but are not included here.
various aspects of protein traffic and sorting.
SUMMARY
REFERENCES
• Many proteins are targeted to their destinations by
Fuller GM, Shields DL: Molecular Basis of Medical Cell Biology.
signal sequences. A major sorting decision is made
McGraw-Hill, 1998.
when proteins are partitioned between cytosolic and
Gould SJ et al: The peroxisome biogenesis disorders. In: The Meta-
membrane-bound polyribosomes by virtue of the ab-
bolic and Molecular Bases of Inherited Disease, 8th ed. Scriver
sence or presence of a signal peptide.
CR et al (editors). McGraw-Hill, 2001.
• The pathways of protein import into mitochondria,
Graham JM, Higgins JA: Membrane Analysis. BIOS Scientific,
1997.
nuclei, peroxisomes, and the endoplasmic reticulum
are described.
Griffith J, Sansom C: The Transporter Facts Book. Academic Press,
1998.
• Many proteins synthesized on membrane-bound
Lodish H et al: Molecular Cell Biology, 4th ed. Freeman, 2000.
polyribosomes proceed to the Golgi apparatus and
(Chapter 17 contains comprehensive coverage of protein sort-
the plasma membrane in transport vesicles.
ing and organelle biogenesis.)
• A number of glycosylation reactions occur in com-
Olkkonen VM, Ikonen E: Genetic defects of intracellular-mem-
partments of the Golgi, and proteins are further
brane transport. N Engl J Med 2000;343:1095.
sorted in the trans-Golgi network.
Reithmeier RAF: Assembly of proteins into membranes. In: Bio-
chemistry of Lipids, Lipoproteins and Membranes. Vance DE,
• Most proteins destined for the plasma membrane
Vance JE (editors). Elsevier, 1996.
and for secretion appear to lack specific signals—a
Sabatini DD, Adesnik MB: The biogenesis of membranes and or-
default mechanism.
ganelles. In: The Metabolic and Molecular Bases of Inherited
• The role of chaperone proteins in the folding of pro-
Disease,
8th ed. Scriver CR et al (editors). McGraw-Hill,
teins is presented, and a model describing budding
2001.
Glycoproteins
47
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
are firmly established; others are still under investi-
gation.
Glycoproteins are proteins that contain oligosaccha-
ride
(glycan) chains covalently attached to their
OLIGOSACCHARIDE CHAINS ENCODE
polypeptide backbones. They are one class of glycocon-
jugate or complex carbohydrates—equivalent terms
BIOLOGIC INFORMATION
used to denote molecules containing one or more car-
An enormous number of glycosidic linkages can be gen-
bohydrate chains covalently linked to protein (to form
erated between sugars. For example, three different hex-
glycoproteins or proteoglycans) or lipid (to form glyco-
oses may be linked to each other to form over 1000 dif-
lipids). (Proteoglycans are discussed in Chapter 48 and
ferent trisaccharides. The conformations of the sugars in
glycolipids in Chapter 14). Almost all the plasma pro-
oligosaccharide chains vary depending on their linkages
teins of humans—except albumin—are glycoproteins.
and proximity to other molecules with which the oligo-
Many proteins of cellular membranes (Chapter 41)
saccharides may interact. It is now established that certain
contain substantial amounts of carbohydrate. A num-
oligosaccharide chains encode considerable biologic in-
ber of the blood group substances are glycoproteins,
formation and that this depends upon their constituent
whereas others are glycosphingolipids. Certain hor-
sugars, their sequences, and their linkages. For instance,
mones (eg, chorionic gonadotropin) are glycoproteins.
mannose 6-phosphate residues target newly synthesized
A major problem in cancer is metastasis, the phenome-
lysosomal enzymes to that organelle (see below).
non whereby cancer cells leave their tissue of origin (eg,
the breast), migrate through the bloodstream to some
distant site in the body (eg, the brain), and grow there
TECHNIQUES ARE AVAILABLE
in an unregulated manner, with catastrophic results for
FOR DETECTION, PURIFICATION,
the affected individual. Many cancer researchers think
& STRUCTURAL ANALYSIS
that alterations in the structures of glycoproteins and
OF GLYCOPROTEINS
other glycoconjugates on the surfaces of cancer cells are
important in the phenomenon of metastasis.
A variety of methods used in the detection, purifica-
tion, and structural analysis of glycoproteins are listed
in Table 47-3. The conventional methods used to pu-
GLYCOPROTEINS OCCUR WIDELY
rify proteins and enzymes are also applicable to the pu-
& PERFORM NUMEROUS FUNCTIONS
rification of glycoproteins. Once a glycoprotein has
Glycoproteins occur in most organisms, from bacteria
been purified, the use of mass spectrometry and high-
to humans. Many viruses also contain glycoproteins,
resolution NMR spectroscopy can often identify the
some of which have been much investigated, in part be-
structures of its glycan chains. Analysis of glycoproteins
cause they are very suitable for biosynthetic studies.
can be complicated by the fact that they often exist as
Numerous proteins with diverse functions are glycopro-
glycoforms; these are proteins with identical amino
teins (Table 47-1); their carbohydrate content ranges
acid sequences but somewhat different oligosaccharide
from 1% to over 85% by weight.
compositions. Although linkage details are not stressed
Many studies have been conducted in an attempt to
in this chapter, it is critical to appreciate that the precise
define the precise roles oligosaccharide chains play in
natures of the linkages between the sugars of glycopro-
the functions of glycoproteins. Table 47-2 summarizes
teins are of fundamental importance in determining the
results from such studies. Some of the functions listed
structures and functions of these molecules.
514
GLYCOPROTEINS
/
515
Table 47-1. Some functions served
Table 47-3. Some important methods used to
by glycoproteins.
study glycoproteins.
Function
Glycoproteins
Method
Use
Structural molecule
Collagens
Periodic acid-Schiff reagent
Detects glycoproteins as pink
bands after electrophoretic sep-
Lubricant and
Mucins
aration.
protective agent
Incubation of cultured cells
Leads to detection of a radio-
Transport molecule
Transferrin, ceruloplasmin
with glycoproteins as
active sugar after electropho-
Immunologic molecule
Immunoglobulins, histocompatibil-
radioactive bands
retic separation.
ity antigens
Treatment with appropriate
Resultant shifts in electropho-
Hormone
Chorionic gonadotropin, thyroid-
endo- or exoglycosidase
retic migration help distinguish
stimulating hormone (TSH)
or phospholipases
among proteins with N-glycan,
O-glycan, or GPI linkages and
Enzyme
Various, eg, alkaline phosphatase
also between high mannose
Cell attachment-
Various proteins involved in cell-cell
and complex N-glycans.
recognition site
(eg, sperm-oocyte), virus-cell,
Sepharose-lectin column
To purify glycoproteins or gly-
bacterium-cell, and hormone-cell
chromatography
copeptides that bind the par-
interactions
ticular lectin used.
Antifreeze
Certain plasma proteins of cold
Compositional analysis
Identifies sugars that the gly-
water fish
following acid hydrolysis
coprotein contains and their
Interact with specific
Lectins, selectins (cell adhesion
stoichiometry.
carbohydrates
lectins), antibodies
Mass spectrometry
Provides information on molec-
Receptor
Various proteins involved in hor-
ular mass, composition, se-
mone and drug action
quence, and sometimes branch-
ing of a glycan chain.
Affect folding of
Calnexin, calreticulin
certain proteins
NMR spectroscopy
To identify specific sugars, their
sequence, linkages, and the
anomeric nature of glycosidic
linkages.
Methylation (linkage)
To determine linkages between
analysis
sugars.
Amino acid or cDNA
Determination of amino acid
Table 47-2. Some functions of the
sequencing
sequence.
oligosaccharide chains of glycoproteins.1
• Modulate physicochemical properties, eg, solubility, vis-
cosity, charge, conformation, denaturation, and binding
sites for bacteria and viruses
EIGHT SUGARS PREDOMINATE
• Protect against proteolysis, from inside and outside of cell
IN HUMAN GLYCOPROTEINS
• Affect proteolytic processing of precursor proteins to
smaller products
About 200 monosaccharides are found in nature; how-
• Are involved in biologic activity, eg, of human chorionic
ever, only eight are commonly found in the oligosac-
gonadotropin (hCG)
charide chains of glycoproteins (Table 47-4). Most of
• Affect insertion into membranes, intracellular migration,
these sugars were described in Chapter 13. N-Acetyl-
sorting and secretion
neuraminic acid (NeuAc) is usually found at the ter-
• Affect embryonic development and differentiation
mini of oligosaccharide chains, attached to subterminal
• May affect sites of metastases selected by cancer cells
galactose
(Gal) or N-acetylgalactosamine
(GalNAc)
residues. The other sugars listed are generally found in
1Adapted from Schachter H: Biosynthetic controls that determine
the branching and heterogeneity of protein-bound oligosaccha-
more internal positions. Sulfate is often found in glyco-
rides. Biochem Cell Biol 1986;64:163.
proteins, usually attached to Gal, GalNAc, or GlcNAc.
516
/
CHAPTER 47
Table 47-4. The principal sugars found in human glycoproteins. Their structures are illustrated in
Chapter 13.
Nucleotide
Sugar
Type
Abbreviation
Sugar
Comments
Galactose
Hexose
Gal
UDP-Gal
Often found subterminal to NeuAc in N-linked gly-
coproteins. Also found in core trisaccharide of pro-
teoglycans.
Glucose
Hexose
Glc
UDP-Glc
Present during the biosynthesis of N-linked glyco-
proteins but not usually present in mature glyco-
proteins. Present in some clotting factors.
Mannose
Hexose
Man
GDP-Man
Common sugar in N-linked glycoproteins.
N-Acetylneur-
Sialic acid (nine
NeuAc
CMP-NeuAc
Often the terminal sugar in both N- and O-linked
aminic acid
C atoms)
glycoproteins. Other types of sialic acid are also
found, but NeuAc is the major species found in hu-
mans. Acetyl groups may also occur as O-acetyl
species as well as N-acetyl.
Fucose
Deoxyhexose
Fuc
GDP-Fuc
May be external in both N- and O-linked glycopro-
teins or internal, linked to the GlcNAc residue at-
tached to Asn in N-linked species. Can also occur
internally attached to the OH of Ser (eg, in t-PA and
certain clotting factors).
N-Acetylgalactosamine
Aminohexose
GalNAc
UDP-GalNAc
Present in both N- and O-linked glycoproteins.
N-Acetylglucosamine
Aminohexose
GlcNAc
UDP-GlcNAc
The sugar attached to the polypeptide chain via
Asn in N-linked glycoproteins; also found at other
sites in the oligosaccharides of these proteins.
Many nuclear proteins have GlcNAc attached to the
OH of Ser or Thr as a single sugar.
Xylose
Pentose
Xyl
UDP-Xyl
Xyl is attached to the OH of Ser in many proteogly-
cans. Xyl in turn is attached to two Gal residues,
forming a link trisaccharide. Xyl is also found in t-PA
and certain clotting factors.
NUCLEOTIDE SUGARS ACT
suitable acceptors provided appropriate transferases are
AS SUGAR DONORS IN MANY
available.
Most nucleotide sugars are formed in the cytosol,
BIOSYNTHETIC REACTIONS
generally from reactions involving the corresponding
The first nucleotide sugar to be reported was uridine
nucleoside triphosphate. CMP-sialic acids are formed
diphosphate glucose (UDP-Glc); its structure is shown
in the nucleus. Formation of uridine diphosphate galac-
in Figure 18-2. The common nucleotide sugars in-
tose (UDP-Gal) requires the following two reactions in
volved in the biosynthesis of glycoproteins are listed in
mammalian tissues:
Table 47-4; the reasons some contain UDP and others
UDP-Glc
guanosine diphosphate (GDP) or cytidine monophos-
PYROPHOS-
phate (CMP) are obscure. Many of the glycosylation re-
PHORYLASE
actions involved in the biosynthesis of glycoproteins
UTP + Glucose 1-phosphate
utilize these compounds (see below). The anhydro na-
UDP-Glc + Pyrophosphate
ture of the linkage between the phosphate group and
the sugars is of the high-energy, high-group-transfer-
UDP-Glc
potential type (Chapter 10). The sugars of these com-
EPIMERASE
pounds are thus “activated” and can be transferred to
UDP-Glc
UDP-Gal
GLYCOPROTEINS
/
517
Because many glycosylation reactions occur within
tide backbone (ie, at internal sites; Figure 47-5) and are
the lumen of the Golgi apparatus, carrier systems (per-
thus useful in releasing large oligosaccharide chains for
meases, transporters) are necessary to transport nu-
structural analyses. A glycoprotein can be treated with
cleotide sugars across the Golgi membrane. Systems
one or more of the above glycosidases to analyze the
transporting UDP-Gal, GDP-Man, and CMP-NeuAc
effects on its biologic behavior of removal of specific
into the cisternae of the Golgi apparatus have been de-
sugars.
scribed. They are antiport systems; ie, the influx of one
molecule of nucleotide sugar is balanced by the efflux
THE MAMMALIAN
of one molecule of the corresponding nucleotide (eg,
ASIALOGLYCOPROTEIN RECEPTOR
UMP, GMP, or CMP) formed from the nucleotide
IS INVOLVED IN CLEARANCE
sugars. This mechanism ensures an adequate concentra-
tion of each nucleotide sugar inside the Golgi appara-
OF CERTAIN GLYCOPROTEINS
tus. UMP is formed from UDP-Gal in the above
FROM PLASMA BY HEPATOCYTES
process as follows:
Experiments performed by Ashwell and his colleagues
in the early 1970s played an important role in focusing
GALACTOSYL-
TRANSFERASE
attention on the functional significance of the oligosac-
UDP-Gal + Protein
Protein Gal + UDP
charide chains of glycoproteins. They treated rabbit
ceruloplasmin (a plasma protein; see Chapter 50) with
neuraminidase in vitro. This procedure exposed subter-
NUCLEOSIDE
DIPHOSPHATE
minal Gal residues that were normally masked by ter-
PHOSPHATASE
minal NeuAc residues. Neuraminidase-treated radioac-
UDP
UMP + Pi
tive ceruloplasmin was found to disappear rapidly from
the circulation, in contrast to the slow clearance of the
untreated protein. Very significantly, when the Gal
EXO- & ENDOGLYCOSIDASES
residues exposed to treatment with neuraminidase were
FACILITATE STUDY
removed by treatment with a galactosidase, the clear-
OF GLYCOPROTEINS
ance rate of the protein returned to normal. Further
studies demonstrated that liver cells contain a mam-
A number of glycosidases of defined specificity have
malian asialoglycoprotein receptor that recognizes
proved useful in examining structural and functional
the Gal moiety of many desialylated plasma proteins
aspects of glycoproteins (Table 47-5). These enzymes
and leads to their endocytosis. This work indicated that
act at either external (exoglycosidases) or internal (en-
an individual sugar, such as Gal, could play an impor-
doglycosidases) positions of oligosaccharide chains. Ex-
tant role in governing at least one of the biologic prop-
amples of exoglycosidases are neuraminidases and
erties (ie, time of residence in the circulation) of certain
galactosidases; their sequential use removes terminal
glycoproteins. This greatly strengthened the concept
NeuAc and subterminal Gal residues from most glyco-
that oligosaccharide chains could contain biologic in-
proteins. Endoglycosidases F and H are examples of
formation.
the latter class; these enzymes cleave the oligosaccharide
chains at specific GlcNAc residues close to the polypep-
LECTINS CAN BE USED TO
PURIFY GLYCOPROTEINS
Table 47-5. Some glycosidases used to study the
& TO PROBE THEIR FUNCTIONS
structure and function of glycoproteins.1
Lectins are carbohydrate-binding proteins that aggluti-
nate cells or precipitate glycoconjugates; a number of
Enzymes
Type
lectins are themselves glycoproteins. Immunoglobulins
Neuraminidases
Exoglycosidase
that react with sugars are not considered lectins. Lectins
Galactosidases
Exo- or endoglycosidase
contain at least two sugar-binding sites; proteins with a
Endoglycosidase F
Endoglycosidase
single sugar-binding site will not agglutinate cells or
Endoglycosidase H
Endoglycosidase
precipitate glycoconjugates. The specificity of a lectin is
usually defined by the sugars that are best at inhibiting
1The enzymes are available from a variety of sources and are often
its ability to cause agglutination or precipitation. En-
specific for certain types of glycosidic linkages and also for their
zymes, toxins, and transport proteins can be classified
anomeric natures. The sites of action of endoglycosidases F and H
are shown in Figure 47-5. F acts on both high-mannose and com-
as lectins if they bind carbohydrate. Lectins were first
plex oligosaccharides, whereas H acts on the former.
discovered in plants and microbes, but many lectins of
518
/
CHAPTER 47
animal origin are now known. The mammalian asialo-
O-linked), involving the hydroxyl side chain of serine
glycoprotein receptor described above is an important
or threonine and a sugar such as N-acetylgalactosamine
example of an animal lectin. Some important lectins are
(GalNAc-Ser[Thr]); (2) those containing an N-glyco-
listed in Table 47-6. Much current research is centered
sidic linkage (ie, N-linked), involving the amide nitro-
on the roles of various animal lectins (eg, the selectins)
gen of asparagine and N-acetylglucosamine (GlcNAc-
in cell-cell interactions that occur in pathologic condi-
Asn); and (3) those linked to the carboxyl terminal
tions such as inflammation and cancer metastasis (see
amino acid of a protein via a phosphoryl-ethanolamine
below).
moiety joined to an oligosaccharide (glycan), which in
Numerous lectins have been purified and are com-
turn is linked via glucosamine to phosphatidylinositol
mercially available; three plant lectins that have been
(PI). This latter class is referred to as glycosylphos-
widely used experimentally are listed in Table 47-7.
phatidylinositol-anchored (GPI-anchored, or GPI-
Among many uses, lectins have been employed to pu-
linked) glycoproteins. Other minor classes of glycopro-
rify specific glycoproteins, as tools for probing the gly-
teins also exist.
coprotein profiles of cell surfaces, and as reagents for
The number of oligosaccharide chains attached to
generating mutant cells deficient in certain enzymes in-
one protein can vary from one to 30 or more, with the
volved in the biosynthesis of oligosaccharide chains.
sugar chains ranging from one or two residues in
length to much larger structures. Many proteins con-
tain more than one type of linkage; for instance, gly-
THERE ARE THREE MAJOR CLASSES
cophorin, an important red cell membrane glycopro-
OF GLYCOPROTEINS
tein
(Chapter 52), contains both O- and N-linked
Based on the nature of the linkage between their poly-
oligosaccharides.
peptide chains and their oligosaccharide chains, glyco-
proteins can be divided into three major classes (Figure
GLYCOPROTEINS CONTAIN SEVERAL
47-1): (1) those containing an O-glycosidic linkage (ie,
TYPES OF O-GLYCOSIDIC LINKAGES
At least four subclasses of O-glycosidic linkages are
found in human glycoproteins:
(1) The GalNAc-
Table 47-6. Some important lectins.
Ser(Thr) linkage shown in Figure 47-1 is the predomi-
nant linkage. Two typical oligosaccharide chains found
in members of this subclass are shown in Figure 47-2.
Lectins
Examples or Comments
Usually a Gal or a NeuAc residue is attached to the
Legume lectins
Concanavalin A, pea lectin
GalNAc, but many variations in the sugar compositions
Wheat germ
Widely used in studies of surfaces of nor-
and lengths of such oligosaccharide chains are found.
agglutinin
mal cells and cancer cells
This type of linkage is found in mucins (see below).
(2) Proteoglycans contain a Gal-Gal-Xyl-Ser trisac-
Ricin
Cytotoxic glycoprotein derived from seeds
charide (the so-called link trisaccharide). (3) Collagens
of the castor plant
contain a Gal-hydroxylysine (Hyl) linkage. (Subclasses
Bacterial toxins
Heat-labile enterotoxin of E coli and
[2] and
[3] are discussed further in Chapter
48.)
cholera toxin
(4) Many nuclear proteins (eg, certain transcription
factors) and cytosolic proteins contain side chains con-
Influenza virus
Responsible for host-cell attachment and
sisting of a single GlcNAc attached to a serine or threo-
hemagglutinin
membrane fusion
nine residue (GlcNAc-Ser[Thr]).
C-type lectins
Characterized by a Ca2+-dependent carbo-
hydrate recognition domain (CRD); in-
cludes the mammalian asialoglycoprotein
receptor, the selectins, and the mannose-
Table 47-7. Three plant lectins and the sugars
binding protein
with which they interact.1
S-type lectins
β-Galactoside-binding animal lectins with
roles in cell-cell and cell-matrix interac-
Lectin
Abbreviation
Sugars
tions
Concanavalin A
ConA
Man and Glc
P-type lectins
Mannose 6-P receptor
Soybean lectin
Gal and GalNAc
l-type lectins
Members of the immunoglobulin super-
Wheat germ agglutinin
WGA
Glc and NeuAc
family, eg, sialoadhesin mediating adhe-
1In most cases, lectins show specificity for the anomeric nature of
sion of macrophages to various cells
the glycosidic linkage (α or β); this is not indicated in the table.
GLYCOPROTEINS
/
519
A
CH2OH
C
NH2
O
C
OH
α
Protein
H
C
C
O
CH2
C
OH
H
Glycine
H
H
C
C
Ser
Ethanolamine
H H
N
P
C
O
Mannose
Ethanolamine
P
Mannose
CH3
Mannose
B
CH2OH
Glucosamine
C
O
Inositol
H
H
H
O
β
PI-PLC
P
Additional fatty acid
C
C
N
C
CH
2
C
OH
H
Plasma
HO
C
C
Asn
membrane
H H
N
C
O
CH3
Figure 47-1.
Depictions of (A) an O-linkage (N-acetylgalactosamine to serine); (B) an N-linkage (N-acetylglu-
cosamine to asparagine) and (C) a glycosylphosphatidylinositol (GPI) linkage. The GPI structure shown is that
linking acetylcholinesterase to the plasma membrane of the human red blood cell. The carboxyl terminal amino
acid is glycine joined in amide linkage via its COOH group to the NH2 group of phosphorylethanolamine, which
in turn is joined to a mannose residue. The core glycan contains three mannose and one glucosamine residues.
The glucosamine is linked to inositol, which is attached to phosphatidic acid. The site of action of PI-phospholi-
pase C (PI-PLC) is indicated. The structure of the core glycan is shown in the text. This particular GPI contains an
extra fatty acid attached to inositol and also an extra phosphorylethanolamine moiety attached to the middle of
the three mannose residues. Variations found among different GPI structures include the identity of the carboxyl
terminal amino acid, the molecules attached to the mannose residues, and the precise nature of the lipid moiety.
Mucins Have a High Content of O-Linked
Oligosaccharides & Exhibit Repeating
Amino Acid Sequences
A
α 2,6
NeuAc
GalNAc
Ser(Thr)
Mucins are glycoproteins with two major characteris-
tics: (1) a high content of O-linked oligosaccharides
(the carbohydrate content of mucins is generally more
B
than 50%); and (2) the presence of repeating amino
β 1,3
Gal
GalNAc
Ser(Thr)
acid sequences (tandem repeats) in the center of their
α 2,3
α 2,6
polypeptide backbones, to which the O-glycan chains
NeuAc
NeuAc
are attached in clusters (Figure 47-3). These sequences
are rich in serine, threonine, and proline. Although O-
Figure 47-2. Structures of two O-linked oligosac-
glycans predominate, mucins often contain a number
charides found in (A) submaxillary mucins and (B) fe-
of N-glycan chains. Both secretory and membrane-
tuin and in the sialoglycoprotein of the membrane of
bound mucins occur. The former are found in the
human red blood cells. (Modified and reproduced, with
mucus present in the secretions of the gastrointestinal,
permission, from Lennarz WJ: The Biochemistry of Glyco-
respiratory, and reproductive tracts. Mucus consists of
proteins and Proteoglycans. Plenum Press, 1980.)
about 94% water and 5% mucins, with the remainder
520
/
CHAPTER 47
O-glycan chain
epitopes have been used to stimulate an immune re-
N-glycan chain
sponse against cancer cells.
The genes encoding the polypeptide backbones of a
number of mucins derived from various tissues (eg,
pancreas, small intestine, trachea and bronchi, stomach,
and salivary glands) have been cloned and sequenced.
N
C
These studies have revealed new information about the
polypeptide backbones of mucins (size of tandem re-
peats, potential sites of N-glycosylation, etc) and ulti-
Tandem repeat sequence
mately should reveal aspects of their genetic control.
Some important properties of mucins are summarized
Figure 47-3. Schematic diagram of a mucin. O-gly-
in Table 47-8.
can chains are shown attached to the central region of
the extended polypeptide chain and N-glycan chains to
The Biosynthesis of O-Linked
the carboxyl terminal region. The narrow rectangles
Glycoproteins Uses Nucleotide Sugars
represent a series of tandem repeat amino acid se-
The polypeptide chains of O-linked and other glyco-
quences. Many mucins contain cysteine residues whose
proteins are encoded by mRNA species; because most
SH groups form interchain linkages; these are not
glycoproteins are membrane-bound or secreted, they
shown in the figure. (Adapted from Strous GJ, Dekker J:
are generally translated on membrane-bound polyribo-
Mucin-type glycoproteins. Crit Rev Biochem Mol Biol
somes (Chapter 38). Hundreds of different oligosaccha-
1992;27:57.)
ride chains of the O-glycosidic type exist. These glyco-
proteins are built up by the stepwise donation of
sugars from nucleotide sugars, such as UDP-GalNAc,
being a mixture of various cell molecules, electrolytes,
UDP-Gal, and CMP-NeuAc. The enzymes catalyzing
and remnants of cells. Secretory mucins generally have
this type of reaction are membrane-bound glycopro-
an oligomeric structure and thus often have a very high
tein glycosyltransferases. Generally, synthesis of one
molecular mass. The oligomers are composed of
specific type of linkage requires the activity of a corre-
monomers linked by disulfide bonds. Mucus exhibits a
spondingly specific transferase. The factors that deter-
high viscosity and often forms a gel. These qualities are
mine which specific serine and threonine residues are
functions of its content of mucins. The high content of
glycosylated have not been identified but are probably
O-glycans confers an extended structure on mucins.
found in the peptide structure surrounding the glycosy-
This is in part explained by steric interactions between
lation site. The enzymes assembling O-linked chains are
their GalNAc moieties and adjacent amino acids, re-
located in the Golgi apparatus, sequentially arranged in
sulting in a chain-stiffening effect so that the conforma-
an assembly line with terminal reactions occurring in
tions of mucins often become those of rigid rods. Inter-
the trans-Golgi compartments.
molecular noncovalent interactions between various
The major features of the biosynthesis of O-linked
sugars on neighboring glycan chains contribute to gel
glycoproteins are summarized in Table 47-9.
formation. The high content of NeuAc and sulfate
residues found in many mucins confers a negative
charge on them. With regard to function, mucins help
lubricate and form a protective physical barrier on
Table 47-8. Some properties of mucins.
epithelial surfaces. Membrane-bound mucins partici-
pate in various cell-cell interactions (eg, involving se-
• Found in secretions of the gastrointestinal, respiratory,
lectins; see below). The density of oligosaccharide
and reproductive tracts and also in membranes of various
chains makes it difficult for proteases to approach their
cells.
polypeptide backbones, so that mucins are often resis-
• Exhibit high content of O-glycan chains, usually containing
tant to their action. Mucins also tend to “mask” certain
NeuAc.
surface antigens. Many cancer cells form excessive
• Contain repeating amino acid sequences rich in serine, thre-
amounts of mucins; perhaps the mucins may mask cer-
onine, and proline.
tain surface antigens on such cells and thus protect the
• Extended structure contributes to their high visco-
elasticity.
cells from immune surveillance. Mucins also carry can-
• Form protective physical barrier on epithelial surfaces, are
cer-specific peptide and carbohydrate epitopes (an epi-
involved in cell-cell interactions, and may contain or mask
tope is a site on an antigen recognized by an antibody,
certain surface antigens.
also called an antigenic determinant). Some of these
GLYCOPROTEINS
/
521
Table 47-9. Summary of main features
penta-antennary structures may all be found. A bewil-
of O-glycosylation.
dering number of chains of the complex type exist, and
that indicated in Figure 47-4 is only one of many.
Other complex chains may terminate in Gal or Fuc.
• Involves a battery of membrane-bound glycoprotein glyco-
High-mannose oligosaccharides typically have two to
syltransferases acting in a stepwise manner; each trans-
six additional Man residues linked to the pentasaccha-
ferase is generally specific for a particular type of linkage.
ride core. Hybrid molecules contain features of both of
• The enzymes involved are located in various subcompart-
the two other classes.
ments of the Golgi apparatus.
• Each glycosylation reaction involves the appropriate
nucleotide-sugar.
The Biosynthesis of N-Linked
• Dolichol-P-P-oligosaccharide is not involved, nor are gly-
Glycoproteins Involves
cosidases; and the reactions are not inhibited by tuni-
Dolichol-P-P-Oligosaccharide
camycin.
• O-Glycosylation occurs posttranslationally at certain Ser and
Leloir and his colleagues described the occurrence of
Thr residues.
a dolichol-pyrophosphate-oligosaccharide (Dol-P-P-
oligosaccharide), which subsequent research showed
to play a key role in the biosynthesis of N-linked glyco-
proteins. The oligosaccharide chain of this compound
N-LINKED GLYCOPROTEINS CONTAIN
generally has the structure R-GlcNAc2Man9Glc3 (R =
AN Asn-GlcNAc LINKAGE
Dol-P-P). The sugars of this compound are first assem-
N-Linked glycoproteins are distinguished by the pres-
bled on the Dol-P-P backbone, and the oligosaccharide
ence of the Asn-GlcNAc linkage (Figure 47-1). It is the
chain is then transferred en bloc to suitable Asn resi-
major class of glycoproteins and has been much stud-
dues of acceptor apoglycoproteins during their synthe-
ied, since the most readily accessible glycoproteins (eg,
sis on membrane-bound polyribosomes. All N-glycans
plasma proteins) mainly belong to this group. It in-
have a common pentasaccharide core structure (Fig-
cludes both membrane-bound and circulating glyco-
ure 47-5).
proteins. The principal difference between this and the
To form high-mannose chains, only the Glc
previous class, apart from the nature of the amino acid
residues plus certain of the peripheral Man residues are
to which the oligosaccharide chain is attached (Asn ver-
removed. To form an oligosaccharide chain of the com-
sus Ser or Thr), concerns their biosynthesis.
plex type, the Glc residues and four of the Man
residues are removed by glycosidases in the endoplas-
mic reticulum and Golgi. The sugars characteristic of
Complex, Hybrid, & High-Mannose
complex chains (GlcNAc, Gal, NeuAc) are added by
Are the Three Major Classes
the action of individual glycosyltransferases located in
of N-Linked Oligosaccharides
the Golgi apparatus. The phenomenon whereby the
There are three major classes of N-linked oligosaccha-
glycan chains of N-linked glycoproteins are first par-
rides: complex, hybrid, and high-mannose (Figure
tially degraded and then in some cases rebuilt is referred
47-4). Each type shares a common pentasaccharide,
to as oligosaccharide processing. Hybrid chains are
Man3GlcNAc2—shown within the boxed area in Figure
formed by partial processing, forming complex chains
47-4 and depicted also in Figure 47-5—but they differ
on one arm and Man structures on the other arm.
in their outer branches. The presence of the common
Thus, the initial steps involved in the biosynthesis of
pentasaccharide is explained by the fact that all three
the N-linked glycoproteins differ markedly from those
classes share an initial common mechanism of biosyn-
involved in the biosynthesis of the O-linked glycopro-
thesis. Glycoproteins of the complex type generally
teins. The former involves Dol-P-P-oligosaccharide; the
contain terminal NeuAc residues and underlying Gal
latter, as described earlier, does not.
and GlcNAc residues, the latter often constituting the
The process of N-glycosylation can be broken down
disaccharide N-acetyllactosamine. Repeating N-acetyl-
into two stages: (1) assembly of Dol-P-P-oligosaccha-
lactosamine units—[Galβ1-3/4GlcNAcβ1-3]n (poly-
ride and transfer of the oligosaccharide; and (2) pro-
N-acetyllactosaminoglycans)—are often found on N-
cessing of the oligosaccharide chain.
linked glycan chains. I/i blood group substances belong
A. ASSEMBLY & TRANSFER OF
to this class. The majority of complex-type oligosaccha-
DOLICHOL-P-P-OLIGOSACCHARIDE
rides contain two, three, or four outer branches (Figure
47-4), but structures containing five branches have also
Polyisoprenol compounds exist in both bacteria and
been described. The oligosaccharide branches are often
eukaryotic cells. They participate in the synthesis of
referred to as antennae, so that bi-, tri-, tetra-, and
bacterial polysaccharides and in the biosynthesis of N-
522
/
CHAPTER 47
Sialic acid
Sialic acid
α2,3 or 2,6
α2,3 or 2,6
Gal
Gal
Gal
Man
Man
Man
β1,4
β1,4
β1,4
α1,2
α1,2
α1,2
GlcNAc
GlcNAc
GlcNAc
Man
Man
Man
Man
Man
β1,2
β1,2
β1,2
α1,3
α1,6
α1,2
α1,3
α1,6
Man
Man
Man
Man
Man
Man
α1,6
α1,3
α1,6
α1,3
α1,3
α1,6
β1,4
β1,4
±GlcNAc
Man
Man
GlcNAc
Man
β1,4
β1,4
β1,4
GlcNAc
GlcNAc
GlcNAc
β1,4
β1,4
β1,4
α1,6
±Fuc
GlcNAc
GlcNAc
GlcNAc
Asn
Asn
Asn
Complex
Hybrid
High-mannose
Figure 47-4. Structures of the major types of asparagine-linked oligosaccharides. The boxed area en-
closes the pentasaccharide core common to all N-linked glycoproteins. (Reproduced, with permission,
from Kornfeld R, Kornfeld S: Assembly of asparagine-linked oligosaccharides. Annu Rev Biochem
1985;54:631.)
linked glycoproteins and GPI anchors. The polyiso-
in the membranes of the endoplasmic reticulum from
prenol used in eukaryotic tissues is dolichol, which is,
Dol-P and UDP-GlcNAc in the following reaction,
next to rubber, the longest naturally occurring hydro-
catalyzed by GlcNAc-P transferase:
carbon made up of a single repeating unit. Dolichol is
composed of 17-20 repeating isoprenoid units (Figure
Dol - P + UDP - GlcNAc → Dol - P - P - GlcNAc + UMP
47-6).
Before it participates in the biosynthesis of Dol-P-P-
The above reaction—which is the first step in the as-
oligosaccharide, dolichol must first be phosphorylated
sembly of Dol-P-P-oligosaccharide—and the other later
to form dolichol phosphate (Dol-P) in a reaction cat-
reactions are summarized in Figure 47-7. The essential
alyzed by dolichol kinase and using ATP as the phos-
features of the subsequent steps in the assembly of Dol-
phate donor.
P-P-oligosaccharide are as follows:
Dolichol-P-P-GlcNAc (Dol-P-P-GlcNAc) is the
key lipid that acts as an acceptor for other sugars in the
(1) A second GlcNAc residue is added to the first,
assembly of Dol-P-P-oligosaccharide. It is synthesized
again using UDP-GlcNAc as the donor.
(2) Five Man residues are added, using GDP-man-
nose as the donor.
Endoglycosidase F
Man
(3) Four additional Man residues are next added,
α1,6
using Dol-P-Man as the donor. Dol-P-Man is
β1,4
β1,4
Man
GlcNAc
GlcNAc
Asn
formed by the following reaction:
α1,3
Man
Endoglycosidase H
Dol - P + GDP - Man → Dol - P - Man + GDP
Figure 47-5. Schematic diagram of the pentasac-
charide core common to all N-linked glycoproteins and
(4) Finally, the three peripheral glucose residues are
to which various outer chains of oligosaccharides may
donated by Dol-P-Glc, which is formed in a reac-
be attached. The sites of action of endoglycosidases F
tion analogous to that just presented except that
and H are also indicated.
Dol-P and UDP-Glc are the substrates.
GLYCOPROTEINS
/
523
Figure 47-6. The structure of dolichol.
H
CH3
CH3
The phosphate in dolichol phosphate is at-
tached to the primary alcohol group at the HO CH2
CH2
C
CH2
CH2
CH C
CH2
CH2
CH C
CH3
left-hand end of the molecule. The group
CH3
within the brackets is an isoprene unit (n =
n
17-20 isoprenoid units).
It should be noted that the first seven sugars (two
preference for the Dol-P-P-GlcNAc2Man9Glc3 struc-
GlcNAc and five Man residues) are donated by nu-
ture. Glycosylation occurs at the Asn residue of an Asn-
cleotide sugars, whereas the last seven sugars (four Man
X-Ser/Thr tripeptide sequence, where X is any amino
and three Glc residues) added are donated by dolichol-
acid except proline, aspartic acid, or glutamic acid. A
P-sugars. The net result is assembly of the compound
tripeptide site contained within a β turn is favored.
illustrated in Figure 47-8 and referred to in shorthand
Only about one-third of the Asn residues that are po-
as Dol-P-P-GlcNAc2Man9Glc3.
tential acceptor sites are actually glycosylated, suggest-
The oligosaccharide linked to dolichol-P-P is trans-
ing that factors other than the tripeptide are also im-
ferred en bloc to form an N-glycosidic bond with one
portant. The acceptor proteins are of both the secretory
or more specific Asn residues of an acceptor protein
and integral membrane class. Cytosolic proteins are
emerging from the luminal surface of the membrane of
rarely glycosylated. The transfer reaction and subse-
the endoplasmic reticulum. The reaction is catalyzed by
quent processes in the glycosylation of N-linked glyco-
oligosaccharide:protein transferase, a membrane-
proteins, along with their subcellular locations, are de-
associated enzyme complex. The transferase will recog-
picted in Figure
47-9. The other product of the
nize and transfer any substrate with the general struc-
oligosaccharide:protein transferase reaction is dolichol-
ture Dol-P-P-(GlcNAc)2-R, but it has a strong
P-P, which is subsequently converted to dolichol-P by a
UDP-GIcNAc
Dol-P
Tunicamycin
UMP
M M
GIcNAc P P Dol
M
UDP-GIcNAc
M M
M
(GIcNAc)
2
P P Dol
UDP
G G G
M M M
P Dol
GIcNAc GIcNAc P P Dol
GDP-M
M P
Dol and G P Dol
(M)6
(GIcNAc)
2
P P Dol
GDP
P Dol
M GIcNAc GIcNAc P P Dol
M
M P
Dol
M
(GIcNAc)
2
P P Dol
(GDP-M)4
(GDP)4
M M M
Figure 47-7. Pathway of biosynthesis of dolichol-P-P-oligosaccharide. The specific linkages formed are indi-
cated in Figure 47-8. Note that the first five internal mannose residues are donated by GDP-mannose, whereas
the more external mannose residues and the glucose residues are donated by dolichol-P-mannose and dolichol-
P-glucose. (UDP, uridine diphosphate; Dol, dolichol; P, phosphate; UMP, uridine monophosphate; GDP, guanosine
diphosphate; M, mannose; G, glucose.)
524
/
CHAPTER 47
α1,2
Man
Man
α1,6
Man
α1,6
α1,3
α1,2
Man
Man
β1,4
β1,4
α
Man
GlcNAc
GlcNAc P P Dolichol
α1,2
α1,3
α1,3
α1,2
α1,2
α1,3
Glc
Glc
Glc
Man
Man
Man
Figure 47-8. Structure of dolichol-P-P-oligosaccharide. (Reproduced, with permission, from Lennarz WJ:
The Biochemistry of Glycoproteins and Proteoglycans. Plenum Press, 1980.)
phosphatase. The dolichol-P can serve again as an ac-
PHOSPHO-
ceptor for the synthesis of another molecule of Dol-P-
DIESTERASE
P-oligosaccharide.
GlcNAc-1-P-6-Man Protein
P-6-Man Protein + GlcNAc
B. PROCESSING OF THE OLIGOSACCHARIDE CHAIN
1. Early phase—The various reactions involved are in-
Man 6-P receptors, located in the Golgi apparatus,
dicated in Figure
47-9. The oligosaccharide:protein
bind the Man 6-P residue of these enzymes and direct
transferase catalyzes reaction 1 (see above). Reactions 2
them to the lysosomes. Fibroblasts from patients with
and 3 involve the removal of the terminal Glc residue
I-cell disease (see below) are severely deficient in the
by glucosidase I and of the next two Glc residues by
activity of the GlcNAc phosphotransferase.
glucosidase II, respectively. In the case of high-
2. Late phase—To assemble a typical complex
mannose glycoproteins, the process may stop here, or
oligosaccharide chain, additional sugars must be added
up to four Man residues may also be removed. How-
to the structure formed in reaction 7. Hence, in reac-
ever, to form complex chains, additional steps are nec-
tion 8, a second GlcNAc is added to the peripheral
essary, as follows. Four external Man residues are re-
Man residue of the other arm of the bi-antennary struc-
moved in reactions 4 and 5 by at least two different
ture shown in Figure 47-9; the enzyme catalyzing this
mannosidases. In reaction 6, a GlcNAc residue is added
step is GlcNAc transferase II. Reactions 9, 10, and 11
to the Man residue of the Manα1-3 arm by GlcNAc
involve the addition of Fuc, Gal, and NeuAc residues at
transferase I. The action of this latter enzyme permits
the sites indicated, in reactions catalyzed by fucosyl,
the occurrence of reaction 7, a reaction catalyzed by yet
galactosyl, and sialyl transferases, respectively. The as-
another mannosidase (Golgi α-mannosidase II) and
sembly of poly-N-acetyllactosamine chains requires ad-
which results in a reduction of the Man residues to the
ditional GlcNAc transferases.
core number of three (Figure 47-5).
An important additional pathway is indicated in re-
The Endoplasmic Reticulum
actions I and II of Figure 47-9. This involves enzymes
& Golgi Apparatus Are the
destined for lysosomes. Such enzymes are targeted to
Major Sites of Glycosylation
the lysosomes by a specific chemical marker. In reaction
I, a residue of GlcNAc-1-P is added to carbon 6 of one
As indicated in Figure 47-9, the endoplasmic reticulum
or more specific Man residues of these enzymes. The
and the Golgi apparatus are the major sites involved in
reaction is catalyzed by a GlcNAc phosphotransferase,
glycosylation processes. The assembly of Dol-P-P-
which uses UDP-GlcNAc as the donor and generates
oligosaccharide occurs on both the cytoplasmic and lu-
UMP as the other product:
minal surfaces of the ER membranes. Addition of the
oligosaccharide to protein occurs in the rough endo-
GlcNAc
PHOSPHO-
plasmic reticulum during or after translation. Removal
TRANSFERASE
of the Glc and some of the peripheral Man residues also
UDP-GlcNAc + Man Protein
occurs in the endoplasmic reticulum. The Golgi appa-
GlcNAc-1-P-6-Man Protein + UMP
ratus is composed of cis, medial, and trans cisternae;
these can be separated by appropriate centrifugation
In reaction II, the GlcNAc is removed by the action of
procedures. Vesicles containing glycoproteins appear to
a phosphodiesterase, leaving the Man residues phos-
bud off in the endoplasmic reticulum and are trans-
phorylated in the 6 position:
ported to the cis Golgi. Various studies have shown
GLYCOPROTEINS
/
525
ROUGH ENDOPLASMIC RETICULUM
1
2
3
4
P
P
Dol
GOLGI APPARATUS
P-
-P
II
-P-
-P-
I
5
cis
UDP-
6
7
8
9
medial
UDP-
UDP-
GDP-
10
11
Exit
trans
UDP-
CMP-
Figure 47-9. Schematic pathway of oligosaccharide processing. The reactions are cat-
alyzed by the following enzymes: 1 , oligosaccharide:protein transferase; 2 , α-glucosidase I;
3 , α-glucosidase II;
4 , endoplasmic reticulum α1,2-mannosidase; l , N-acetylglu-
cosaminylphosphotransferase; ll , N-acetylglucosamine-1-phosphodiester α-N-acetylglu-
cosaminidase; 5 , Golgi apparatus α-mannosidase I; 6 , N-acetylglucosaminyltransferase I;
7 , Golgi apparatus α-mannosidase II; 8 , N-acetylglucosaminyltransferase II; 9 , fucosyltrans-
ferase; 10, galactosyltransferase; 11 , sialyltransferase. The thick arrows indicate various nu-
cleotide sugars involved in the oveall scheme. (Solid square, N-acetylglucosamine; open cir-
cle, mannose; solid triangle, glucose; open triangle, fucose; solid circle, galactose; solid
diamond, sialic acid.) (Reproduced, with permission, from Kornfeld R, Kornfeld S: Assembly of
asparagine-linked oligosaccharides. Annu Rev Biochem 1985;54:631.)
that the enzymes involved in glycoprotein processing
the fucosyl, galactosyl, and sialyl transferases (catalyzing
show differential locations in the cisternae of the Golgi.
reactions 9, 10, and 11) are located mainly in the trans
As indicated in Figure 47-9, Golgi α-mannosidase I
Golgi. The major features of the biosynthesis of N-
(catalyzing reaction
5) is located mainly in the cis
linked glycoproteins are summarized in Table 47-10
Golgi, whereas GlcNAc transferase I (catalyzing reac-
and should be contrasted with those previously listed
tion 6) appears to be located in the medial Golgi, and
(Table 47-9) for O-linked glycoproteins.
526
/
CHAPTER 47
Table 47-10. Summary of main features of
ing in the lumen of the ER. The soluble protein cal-
N-glycosylation.
reticulin appears to play a similar function.
Several Factors Regulate the Glycosylation
• The oligosaccharide Glc3Man9(GIcNAc)2 is transferred from
dolichol-P-P-oligosaccharide in a reaction catalyzed by
of Glycoproteins
oligosaccharide:protein transferase, which is inhibited by
It is evident that glycosylation of glycoproteins is a
tunicamycin.
complex process involving a large number of enzymes.
• Transfer occurs to specific Asn residues in the sequence
One index of its complexity is that more than ten dis-
Asn-X-Ser/Thr, where X is any residue except Pro, Asp, or
tinct GlcNAc transferases involved in glycoprotein
Glu.
biosynthesis have been reported, and many others are
• Transfer can occur cotranslationally in the endoplasmic
theoretically possible. Multiple species of the other gly-
reticulum.
• The protein-bound oligosaccharide is then partially
cosyltransferases (eg, sialyltransferases) also exist. Con-
processed by glucosidases and mannosidases; if no addi-
trolling factors of the first stage of N-linked glycopro-
tional sugars are added, this results in a high-mannose
tein biosynthesis
(ie, oligosaccharide assembly and
chain.
transfer) include (1) the presence of suitable acceptor
• If processing occurs down to the core heptasaccharide
sites in proteins, (2) the tissue level of Dol-P, and (3) the
(Man5[GlcNAc]2), complex chains are synthesized by the ad-
activity of the oligosaccharide:protein transferase.
dition of GlcNAc, removal of two Man, and the stepwise ad-
Some factors known to be involved in the regulation
dition of individual sugars in reactions catalyzed by specific
of oligosaccharide processing are listed in Table
transferases (eg, GlcNAc, Gal, NeuAc transferases) that em-
47-11. Two of the points listed merit further com-
ploy appropriate nucleotide sugars.
ment. First, species variations among processing en-
zymes have assumed importance in relation to produc-
tion of glycoproteins of therapeutic use by means of
recombinant DNA technology. For instance, recombi-
nant erythropoietin (epoetin alfa; EPO) is sometimes
Some Glycan Intermediates
administered to patients with certain types of chronic
Formed During N-Glycosylation
anemia in order to stimulate erythropoiesis. The half-
Have Specific Functions
life of EPO in plasma is influenced by the nature of its
The following are a number of specific functions of N-
glycosylation pattern, with certain patterns being asso-
glycan chains that have been established or are under
ciated with a short half-life, appreciably limiting its pe-
investigation. (1) The involvement of the mannose 6-P
riod of therapeutic effectiveness. It is thus important to
signal in targeting of certain lysosomal enzymes is clear
harvest EPO from host cells that confer a pattern of
(see above and discussion of I-cell disease, below). (2) It
glycosylation consistent with a normal half-life in
is likely that the large N-glycan chains present on newly
plasma. Second, there is great interest in analysis of the
synthesized glycoproteins may assist in keeping these
activities of glycoprotein-processing enzymes in various
proteins in a soluble state inside the lumen of the endo-
types of cancer cells. These cells have often been found
plasmic reticulum. (3) One species of N-glycan chains
to synthesize different oligosaccharide chains (eg, they
has been shown to play a role in the folding and reten-
often exhibit greater branching) from those made in
tion of certain glycoproteins in the lumen of the endo-
control cells. This could be due to cancer cells contain-
plasmic reticulum. Calnexin is a protein present in the
ing different patterns of glycosyltransferases from those
endoplasmic reticulum membrane that acts as a “chap-
exhibited by corresponding normal cells, due to speci-
erone” (Chapter 46). It has been found that calnexin
fic gene activation or repression. The differences in
will bind specifically to a number of glycoproteins (eg,
oligosaccharide chains could affect adhesive interactions
the influenza virus hemagglutinin [HA]) that possess the
between cancer cells and their normal parent tissue
monoglycosylated core structure. This species is the
cells, contributing to metastasis. If a correlation could
product of reaction 2 shown in Figure 47-9 but from
be found between the activity of particular processing
which the terminal glucose residue has been removed,
enzymes and the metastatic properties of cancer cells,
leaving only the innermost glucose attached. The re-
this could be important as it might permit synthesis of
lease of fully folded HA from calnexin requires the en-
drugs to inhibit these enzymes and, secondarily, metas-
zymatic removal of this last glucosyl residue by α-glu-
tasis.
cosidase II. In this way, calnexin retains certain partly
The genes encoding many glycosyltransferases have
folded (or misfolded) glycoproteins and releases them
already been cloned, and others are under study.
when proper folding has occurred; it is thus an impor-
Cloning has revealed new information on both protein
tant component of the quality control systems operat-
and gene structures. The latter should also cast light on
GLYCOPROTEINS
/
527
Table 47-11. Some factors affecting the activities
Table 47-12. Three inhibitors of enzymes
of glycoprotein processing enzymes.
involved in the glycosylation of glycoproteins and
their sites of action.
Factor
Comment
Inhibitor
Site of Action
Cell type
Different cell types contain different pro-
files of processing enzymes.
Tunicamycin
Inhibits GlcNAc-P transferase, the enzyme
catalyzing addition of GlcNAc to dolichol-
Previous enzyme
Certain glycosyltransferases will only act
P, the first step in the biosynthesis of
on an oligosaccharide chain if it has al-
oligosaccharide-P-P-dolichol
ready been acted upon by another pro-
cessing enzyme.1
Deoxynojirimycin
Inhibitor of glucosidases I and II
Development
The cellular profile of processing enzymes
Swainsonine
Inhibitor of mannosidase II
may change during development if their
genes are turned on or off.
Intracellular
For instance, if an enzyme is destined for
location
insertion into the membrane of the ER
increase the susceptibility of these proteins to proteoly-
(eg, HMG-CoA reductase), it may never
sis. Inhibition of glycosylation does not appear to have
encounter Golgi-located processing
a consistent effect upon the secretion of glycoproteins
enzymes.
that are normally secreted. The inhibitors of glycopro-
Protein
Differences in conformation of different
tein processing listed in Table 47-12 do not affect the
conformation
proteins may facilitate or hinder access of
biosynthesis of O-linked glycoproteins. The extension
processing enzymes to identical oligosac-
of O-linked chains can be prevented by GalNAc-
charide chains.
benzyl. This compound competes with natural glyco-
Species
Same cells (eg, fibroblasts) from different
protein substrates and thus prevents chain growth be-
species may exhibit different patterns of
yond GalNAc.
processing enzymes.
Cancer
Cancer cells may exhibit processing en-
SOME PROTEINS ARE ANCHORED
zymes different from those of correspond-
TO THE PLASMA MEMBRANE
ing normal cells.
BY GLYCOSYLPHOSPHATIDYL-
1For example, prior action of GlcNAc transferase I is necessary for
INOSITOL STRUCTURES
the action of Golgi α-mannosidase II.
Glycosylphosphatidylinositol
(GPI)-linked glycopro-
teins comprise the third major class of glycoprotein.
the mechanisms involved in their transcriptional con-
The GPI structure (sometimes called a “sticky foot”)
trol, and gene knockout studies are being used to evalu-
involved in linkage of the enzyme acetylcholinesterase
ate the biologic importance of various glycosyltrans-
(ACh esterase) to the plasma membrane of the red
ferases.
blood cell is shown in Figure 47-1. GPI-linked pro-
teins are anchored to the outer leaflet of the plasma
membrane by the fatty acids of phosphatidylinositol
(PI). The PI is linked via a GlcNH2 moiety to a glycan
Tunicamycin Inhibits
chain that contains various sugars (eg, Man, GlcNH2).
N- but Not O-Glycosylation
In turn, the oligosaccharide chain is linked via phos-
A number of compounds are known to inhibit various
phorylethanolamine in an amide linkage to the car-
reactions involved in glycoprotein processing. Tuni-
boxyl terminal amino acid of the attached protein. The
camycin, deoxynojirimycin, and swainsonine are
core of most GPI structures contains one molecule of
three such agents. The reactions they inhibit are indi-
phosphorylethanolamine, three Man residues, one mol-
cated in Table 47-12. These agents can be used experi-
ecule of GlcNH2, and one molecule of phosphatidyl-
mentally to inhibit various stages of glycoprotein
inositol, as follows:
biosynthesis and to study the effects of specific alter-
ations upon the process. For instance, if cells are grown
Ethanolamine - phospho
6Manα1
→
→
in the presence of tunicamycin, no glycosylation of
2Manα1→
6Manα1→
GlcNα1→
their normally N-linked glycoproteins will occur. In
certain cases, lack of glycosylation has been shown to
6—
myo
- inositol -1- phospholipid
528
/
CHAPTER 47
Additional constituents are found in many GPI struc-
text (eg, transport molecules, immunologic molecules,
tures; for example, that shown in Figure 47-1 contains
and hormones). Here, their involvement in two specific
an extra phosphorylethanolamine attached to the mid-
processes—fertilization and inflammation—will be
dle of the three Man moieties of the glycan and an extra
briefly described. In addition, the bases of a number of
fatty acid attached to GlcNH2. The functional signifi-
diseases that are due to abnormalities in the synthesis
cance of these variations among structures is not under-
and degradation of glycoproteins will be summarized.
stood. This type of linkage was first detected by the use
of bacterial PI-specific phospholipase C
(PI-PLC),
Glycoproteins Are Important
which was found to release certain proteins from the
in Fertilization
plasma membrane of cells by splitting the bond indi-
cated in Figure 47-1. Examples of some proteins that
To reach the plasma membrane of an oocyte, a sperm
are anchored by this type of linkage are given in Table
has to traverse the zona pellucida (ZP), a thick, trans-
47-13. At least three possible functions of this type of
parent, noncellular envelope that surrounds the oocyte.
linkage have been suggested: (1) The GPI anchor may
The zona pellucida contains three glycoproteins of inter-
allow greatly enhanced mobility of a protein in the
est, ZP1-3. Of particular note is ZP3, an O-linked gly-
plasma membrane compared with that observed for a
coprotein that functions as a receptor for the sperm. A
protein that contains transmembrane sequences. This is
protein on the sperm surface, possibly galactosyl trans-
perhaps not surprising, as the GPI anchor is attached
ferase, interacts specifically with oligosaccharide chains of
only to the outer leaflet of the lipid bilayer, so that it is
ZP3; in at least certain species (eg, the mouse), this inter-
freer to diffuse than a protein anchored via both leaflets
action, by transmembrane signaling, induces the acroso-
of the bilayer. Increased mobility may be important in fa-
mal reaction, in which enzymes such as proteases and
cilitating rapid responses to appropriate stimuli. (2) Some
hyaluronidase and other contents of the acrosome of the
GPI anchors may connect with signal transduction
sperm are released. Liberation of these enzymes helps
pathways. (3) It has been shown that GPI structures can
the sperm to pass through the zona pellucida and reach
target certain proteins to apical domains of the plasma
the plasma membrane (PM) of the oocyte. In hamsters,
membrane of certain epithelial cells. The biosynthesis of
it has been shown that another glycoprotein, PH-30, is
GPI anchors is complex and begins in the endoplasmic
important in both the binding of the PM of the sperm to
reticulum. The GPI anchor is assembled independently
the PM of the oocyte and also in the subsequent fusion
by a series of enzyme-catalyzed reactions and then trans-
of the two membranes. These interactions enable the
ferred to the carboxyl terminal end of its acceptor pro-
sperm to enter and thus fertilize the oocyte. It may be
tein, accompanied by cleavage of the preexisting car-
possible to inhibit fertilization by developing drugs or
boxyl terminal hydrophobic peptide from that protein.
antibodies that interfere with the normal functions of
This process is sometimes called glypiation. An ac-
ZP3 and PH-30 and which would thus act as contracep-
quired defect in an early stage of the biosynthesis of the
tive agents.
GPI structure has been implicated in the causation of
paroxysmal nocturnal hemoglobinuria (see below).
Selectins Play Key Roles in Inflammation
& in Lymphocyte Homing
GLYCOPROTEINS ARE INVOLVED
Leukocytes play important roles in many inflammatory
IN MANY BIOLOGIC PROCESSES
and immunologic phenomena. The first steps in many
& IN MANY DISEASES
of these phenomena are interactions between circulat-
As listed in Table 47-1, glycoproteins have many dif-
ing leukocytes and endothelial cells prior to passage of
ferent functions; some have already been addressed in
the former out of the circulation. Work done to iden-
this chapter and others are described elsewhere in this
tify specific molecules on the surfaces of the cells in-
volved in such interactions has revealed that leukocytes
and endothelial cells contain on their surfaces specific
lectins, called selectins, that participate in their inter-
Table 47-13. Some GPI-linked proteins.
cellular adhesion. Features of the three major classes of
selectins are summarized in Table 47-14. Selectins are
• Acetylcholinesterase (red cell membrane)
single-chain Ca2+-binding transmembrane proteins that
• Alkaline phosphatase (intestinal, placental)
contain a number of domains (Figure 47-10). Their
• Decay-accelerating factor (red cell membrane)
amino terminal ends contain the lectin domain, which
•
5′-Nucleotidase (T lymphocytes, other cells)
is involved in binding to specific carbohydrate ligands.
• Thy-1 antigen (brain, T lymphocytes)
The adhesion of neutrophils to endothelial cells of
• Variable surface glycoprotein (Trypanosoma brucei)
postcapillary venules can be considered to occur in four
GLYCOPROTEINS
/
529
Table 47-14. Some molecules involved in leukocyte-endothelial cell interactions.1
Molecule
Cell
Ligands
Selectins
L-selectin
PMN, lymphs
CD34, Gly-CAM-12
Sialyl-Lewisx and others
P-selectin
EC, platelets
P-selectin glycoprotein ligand-1 (PSGL-1)
Sialyl-Lewisx and others
E-selectin
EC
Sialyl-Lewisx and others
Integrins
LFA-1
PMN, lymphs
ICAM-1, ICAM-2 (CD11a/CD18)
Mac-1
PMN
ICAM-1 and others (CD11b/CD18)
Immunoglobulin superfamily
ICAM-1
Lymphs, EC
LFA-1, Mac-1
ICAM-2
Lymphs, EC
LFA-1
PECAM-1
EC, PMN, lymphs
Various platelets
1Modified from Albelda SM, Smith CW, Ward PA: Adhesion molecules and inflammatory injury. FASEB J 1994;8:504.
2These are ligands for lymphocyte L-selectin; the ligands for neutrophil L-selectin have not been identified.
Key: PMN, polymorphonuclear leukocytes; EC, endothelial cell; lymphs, lymphocytes; CD, cluster of differentiation; ICAM, intercellular ad-
hesion molecule; LFA-1, lymphocyte function-associated antigen-1; PECAM-1, platelet endothelial cell adhesion cell molecule-1.
stages, as shown in Figure 47-11. The initial baseline
this stage, activation of the neutrophils by various
stage is succeeded by slowing or rolling of the neu-
chemical mediators (discussed below) occurs, resulting
trophils, mediated by selectins. Interactions between
in a change of shape of the neutrophils and firm adhe-
L-selectin on the neutrophil surface and CD34 and
sion of these cells to the endothelium. An additional set
GlyCAM-1 or other glycoproteins on the endothelial
of adhesion molecules is involved in firm adhesion,
surface are involved. These particular interactions are
namely, LFA-1 and Mac-1 on the neutrophils and
initially short-lived, and the overall binding is of rela-
ICAM-1 and ICAM-2 on endothelial cells. LFA-1 and
tively low affinity, permitting rolling. However, during
Mac-1 are CD11/CD18 integrins (see Chapter 52 for a
discussion of integrins), whereas ICAM-1 and ICAM-2
are members of the immunoglobulin superfamily. The
L-selectin
fourth stage is transmigration of the neutrophils across
the endothelial wall. For this to occur, the neutrophils
NH2
Lectin
EGF
1
2
COOH
insert pseudopods into the junctions between endothe-
lial cells, squeeze through these junctions, cross the
Figure 47-10. Schematic diagram of the structure
basement membrane, and then are free to migrate in
of human L-selectin. The extracellular portion contains
the extravascular space. Platelet-endothelial cell adhe-
an amino terminal domain homologous to C-type
sion molecule-1 (PECAM-1) has been found to be lo-
lectins and an adjacent epidermal growth factor-like
calized at the junctions of endothelial cells and thus
domain. These are followed by a variable number of
may have a role in transmigration. A variety of biomol-
complement regulatory-like modules (numbered cir-
ecules have been found to be involved in activation of
cles) and a transmembrane sequence (black diamond).
neutrophil and endothelial cells, including tumor
A short cytoplasmic sequence (open rectangle) is at the
necrosis factor α, various interleukins, platelet activat-
carboxyl terminal. The structures of P- and E-selectin
ing factor (PAF), leukotriene B4, and certain comple-
are similar to that shown except that they contain more
ment fragments. These compounds stimulate various
complement-regulatory modules. The numbers of
signaling pathways, resulting in changes in cell shape
amino acids in L-, P-, and E- selectins, as deduced from
and function, and some are also chemotactic. One im-
the cDNA sequences, are 385, 789, and 589, respec-
portant functional change is recruitment of selectins to
tively. (Reproduced, with permission, from Bevilacqua MP,
the cell surface, as in some cases selectins are stored in
Nelson RM: Selectins. J Clin Invest 1993;91:370.)
granules (eg, in endothelial cells and platelets).
530
/
CHAPTER 47
A
lished. Sulfated molecules, such as the sulfatides (Chap-
ter 14), may be ligands in certain instances. This basic
Baseline
knowledge is being used in attempts to synthesize com-
pounds that block selectin-ligand interactions and thus
may inhibit the inflammatory response. Approaches in-
clude administration of specific monoclonal antibodies
B
or of chemically synthesized analogs of sialyl-LewisX,
Rolling
both of which bind selectins. Cancer cells often exhibit
sialyl-LewisX and other selectin ligands on their sur-
faces. It is thought that these ligands play a role in the
invasion and metastasis of cancer cells.
C
Activation
and
Abnormalities in the Synthesis of
firm adhesion
Glycoproteins Underlie Certain Diseases
Table 47-15 lists a number of conditions in which ab-
D
normalities in the synthesis of glycoproteins are of im-
Transmigration
Table 47-15. Some diseases due to or involving
Figure 47-11. Schematic diagram of neutrophil-
abnormalities in the biosynthesis of
endothelial cell interactions. A: Baseline conditions:
glycoproteins.
Neutrophils do not adhere to the vessel wall. B: The first
event is the slowing or rolling of the neutrophils within
Disease
Abnormality
the vessel (venule) mediated by selectins. C: Activation
Cancer
Increased branching of cell surface
occurs, resulting in neutrophils firmly adhering to the
glycans or presentation of selectin li-
surfaces of endothelial cells and also assuming a flat-
gands may be important in metastasis.
tened shape. This requires interaction of activated
CD18 integrins on neutrophils with ICAM-1 on the en-
Congenital disorders
See Table 47-16.
dothelium. D: The neutrophils then migrate through
of glycosylation1
the junctions of endothelial cells into the interstitial tis-
HEMPAS2 (MIM
Abnormalities in certain enzymes (eg,
sue; this requires involvement of PECAM-1. Chemotaxis
224100)
mannosidase II and others) involved in
is also involved in this latter stage. (Reproduced, with
the biosynthesis of N-glycans, particu-
permission, from Albelda SM, Smith CW, Ward PA: Adhe-
larly affecting the red blood cell mem-
sion molecules and inflammatory injury. FASEB J
brane.
1994;8;504.)
Leukocyte adhesion
Probably mutations affecting a Golgi-
deficiency, type II
located GDP-fucose transporter, re-
(MIM 266265)
sulting in defective fucosylation.
The precise chemical nature of some of the ligands
Paroxysmal nocturnal
Acquired defect in biosynthesis of the
involved in selectin-ligand interactions has been deter-
hemoglobinuria
GPI3 structures of decay accelerating
mined. All three selectins bind sialylated and fucosy-
(MIM 311770)
factor (DAF) and CD59.
lated oligosaccharides, and in particular all three bind
I-cell disease
Deficiency of GlcNAc phosphotrans-
sialyl-LewisX (Figure 47-12), a structure present on
(MIM 252500)
ferase, resulting in abnormal targeting
both glycoproteins and glycolipids. Whether this com-
of certain lysosomal enzymes.
pound is the actual ligand involved in vivo is not estab-
1The MIM number for congenital disorder of glycosylation type Ia
is 212065.
2Hereditary erythroblastic multinuclearity with a positive acidified
NeuAcα2
3Galβ1
4GlcNAc
serum lysis test (congenital dyserythropoietic anemia type II). This
α 1-3
is a relatively mild form of anemia. It reflects at least in part the
Fuc
presence in the red cell membranes of various glycoproteins with
abnormal N-glycan chains, which contribute to the susceptibility
Figure 47-12. Schematic representation of the
to lysis.
structure of sialyl-LewisX.
3Glycosylphosphatidylinositol.
GLYCOPROTEINS
/
531
portance. As mentioned above, many cancer cells ex-
of somatic mutations in the PIG-A (for phosphatidyl-
hibit different profiles of oligosaccharide chains on
inositol glycan class A) gene of certain hematopoietic
their surfaces, some of which may contribute to metas-
cells. The product of this gene appears to be the en-
tasis. The congenital disorders of glycosylation
zyme that links glucosamine to phosphatidylinositol in
(CDG) are a group of disorders of considerable current
the GPI structure (Figure 47-1). Thus, proteins that
interest. The major features of these conditions are
are anchored by a GPI linkage are deficient in the red
summarized in Table 47-16. Leukocyte adhesion de-
cell membrane. Two proteins are of particular interest:
ficiency (LAD) II is a rare condition probably due to
decay accelerating factor (DAF) and another protein
mutations affecting the activity of a Golgi-located
designated CD59. They normally interact with certain
GDP-fucose transporter. It can be considered a congen-
components of the complement system (Chapter 50) to
ital disorder of glycosylation. The absence of fucosy-
prevent the hemolytic actions of the latter. However,
lated ligands for selectins leads to a marked decrease in
when they are deficient, the complement system can act
neutrophil rolling. Subjects suffer life-threatening, re-
on the red cell membrane to cause hemolysis. Paroxys-
current bacterial infections and also psychomotor and
mal nocturnal hemoglobinuria can be diagnosed rela-
mental retardation. The condition appears to respond
tively simply, as the red cells are much more sensitive to
to oral fucose. Hereditary erythroblastic multinuclear-
hemolysis in normal serum acidified to pH 6.2 (Ham’s
ity with a positive acidified lysis test (HEMPAS)—
test); the complement system is activated under these
congenital dyserythropoietic anemia type II—is an-
conditions, but normal cells are not affected. Figure
other disorder due to abnormalities in the processing of
47-13 summarizes the etiology of paroxysmal noctur-
N-glycans. Some cases have been claimed to be due to
nal hemoglobinuria.
defects in alpha-mannosidase II. I-cell disease is dis-
cussed further below. Paroxysmal nocturnal hemo-
I-Cell Disease Results From Faulty
globinuria is an acquired mild anemia characterized by
Targeting of Lysosomal Enzymes
the presence of hemoglobin in urine due to hemolysis
of red cells, particularly during sleep. This latter phe-
As indicated above, Man
6-P serves as a chemical
nomenon may reflect a slight drop in plasma pH dur-
marker to target certain lysosomal enzymes to that or-
ing sleep, which increases susceptibility to lysis by the
ganelle. Analysis of cultured fibroblasts derived from
complement system (Chapter 50). The basic defect in
patients with I-cell (inclusion cell) disease played a large
paroxysmal nocturnal hemoglobinuria is the acquisition
part in revealing the above role of Man 6-P. I-cell dis-
ease is an uncommon condition characterized by severe
progressive psychomotor retardation and a variety of
physical signs, with death often occurring in the first
Table 47-16. Major features of the congenital
decade. Cultured cells from patients with I-cell disease
were found to lack almost all of the normal lysosomal
disorders of glycosylation.
enzymes; the lysosomes thus accumulate many different
• Autosomal recessive disorders
• Multisystem disorders that have probably not been recog-
nized in the past
Acquired mutations in the PIG-A gene
• Generally affect the central nervous system, resulting in
of certain hematopoietic cells
psychomotor retardation and other features
• Type I disorders are due to mutations in genes encoding en-
zymes (eg, phosphomannomutase-2 [PMM-2], causing CDG
Defective synthesis of the GlcNH2-PI
Ia) involved in the synthesis of dolichol-P-P-oligo-
linkage of GPI anchors
saccharide
• Type II disorders are due to mutations in genes encoding
enzymes (eg, GlcNAc transferase-2, causing CDG IIa) in-
Decreased amounts in the red blood membrane of
volved in the processing of N-glycan chains
GPI-anchored proteins, with decay accelerating factor
• About 11 distinct disorders have been recognized
(DAF) and CD59 being of especial importance
• Isoelectric focusing of transferrin is a useful biochemical test
for assisting in the diagnosis of these conditions; trun-
Certain components of the complement system
cation of the oligosaccharide chains of this protein alters its
are not opposed by DAF and CD59, resulting
isolectric focusing pattern
in complement-mediated lysis of red cells
• Oral mannose has proved of benefit in the treatment of
CDG Ia
Figure 47-13. Scheme of causation of paroxysmal
Key: CDG, congenital disorder of glycosylation.
nocturnal hemoglobinuria (MIM 311770).
532
/
CHAPTER 47
types of undegraded molecules, forming inclusion bod-
Mutations in DNA
ies. Samples of plasma from patients with the disease
were observed to contain very high activities of lysoso-
Mutant GlcNAc phosphotransferase
mal enzymes; this suggested that the enzymes were
being synthesized but were failing to reach their proper
intracellular destination and were instead being se-
Lack of normal transfer of GlcNAc 1-P
creted. Cultured cells from patients with the disease
to specific mannose residues of certain enzymes
were noted to take up exogenously added lysosomal en-
destined for lysosomes
zymes obtained from normal subjects, indicating that
the cells contained a normal receptor on their surfaces
These enzymes consequently lack Man 6-P
for endocytic uptake of lysosomal enzymes. In addition,
and are secreted from cells (eg, into the plasma)
this finding suggested that lysosomal enzymes from pa-
rather than targeted to lysosomes
tients with I-cell disease might lack a recognition
marker. Further studies revealed that lysosomal en-
Lysosomes are thus deficient in certain hydrolases, do
zymes from normal individuals carried the Man 6-P
not function properly, and accumulate partly digested
recognition marker described above, which interacted
cellular material, manifesting as inclusion bodies
with a specific intracellular protein, the Man 6-P recep-
tor. Cultured cells from patients with I-cell disease were
Figure 47-14. Summary of the causation of I-cell
then found to be deficient in the activity of the cis
disease (MIM 252500).
Golgi-located GlcNAc phosphotransferase, explaining
how their lysosomal enzymes failed to acquire the Man
6-P marker. It is now known that there are two Man
6-P receptor proteins, one of high (275 kDa) and one
cally recognizes and interacts with lysosomal enzymes.
of low (46 kDa) molecular mass. These proteins are
It has been proposed that the defect in pseudo-Hurler
lectins, recognizing Man 6-P. The former is cation-
polydystrophy lies in the latter domain, and the reten-
independent and also binds IGF-II (hence it is named
tion of some catalytic activity results in a milder condi-
the Man 6-P-IGF-II receptor), whereas the latter is
tion.
cation-dependent in some species and does not bind
IGF-II. It appears that both receptors function in the
Genetic Deficiencies of Glycoprotein
intracellular sorting of lysosomal enzymes into clathrin-
Lysosomal Hydrolases Cause Diseases
coated vesicles, which occurs in the trans Golgi subse-
Such as α-Mannosidosis
quent to synthesis of Man 6-P in the cis Golgi. These
vesicles then leave the Golgi and fuse with a prelysoso-
Glycoproteins, like most other biomolecules, undergo
mal compartment. The low pH in this compartment
both synthesis and degradation (ie, turnover). Degrada-
causes the lysosomal enzymes to dissociate from their
tion of the oligosaccharide chains of glycoproteins in-
receptors and subsequently enter into lysosomes. The
volves a battery of lysosomal hydrolases, including
receptors are recycled and reused. Only the smaller re-
α-neuraminidase, β-galactosidase, β-hexosaminidase,
ceptor functions in the endocytosis of extracellular lyso-
α- and β-mannosidases, α-N-acetylgalactosaminidase,
somal enzymes, which is a minor pathway for lysosomal
α-fucosidase, endo-β-N-acetylglucosaminidase, and as-
location. Not all cells employ the Man 6-P receptor to
partylglucosaminidase. The sites of action of the last
target their lysosomal enzymes (eg, hepatocytes use a
two enzymes are indicated in the legend to Figure
different but undefined pathway); furthermore, not all
47-5. Genetically determined defects of the activities of
lysosomal enzymes are targeted by this mechanism.
these enzymes can occur, resulting in abnormal degra-
Thus, biochemical investigations of I-cell disease not
dation of glycoproteins. The accumulation in tissues of
only led to elucidation of its basis but also contributed
such abnormally degraded glycoproteins can lead to
significantly to knowledge of how newly synthesized
various diseases. Among the best-recognized of these
proteins are targeted to specific organelles, in this case
diseases are mannosidosis, fucosidosis, sialidosis, as-
the lysosome. Figure 47-14 summarizes the causation
partylglycosaminuria, and Schindler disease, due re-
of I-cell disease.
spectively to deficiencies of α-mannosidase, α-fucosi-
Pseudo-Hurler polydystrophy is another genetic
dase, α-neuraminidase, aspartylglucosaminidase, and
disease closely related to I-cell disease. It is a milder
α-N-acetyl-galactosaminidase. These diseases, which
condition, and patients may survive to adulthood.
are relatively uncommon, have a variety of manifesta-
Studies have revealed that the GlcNAc phosphotrans-
tions; some of their major features are listed in Table
ferase involved in I-cell disease has several domains, in-
47-17. The fact that patients affected by these disor-
cluding a catalytic domain and a domain that specifi-
ders all show signs referable to the central nervous sys-
GLYCOPROTEINS
/
533
Table 47-17. Major features of some diseases
It can thus bind the agalactosyl IgG molecules, which
(eg, α-mannosidosis, β-mannosidosis, fucosidosis,
subsequently activate the complement system, con-
tributing to chronic inflammation in the synovial mem-
sialidosis, aspartylglycosaminuria, and Schindler
branes of joints. This protein can also bind the above
disease) due to deficiencies of glycoprotein
sugars when they are present on the surfaces of certain
hydrolases.1
bacteria, fungi, and viruses, preparing these pathogens
for opsonization or for destruction by the complement
• Usually exhibit mental retardation or other neurologic ab-
system. This is an example of innate immunity, not in-
normalities, and in some disorders coarse features or vis-
volving immunoglobulins. Deficiency of this protein in
ceromegaly (or both)
young infants, due to mutation, renders them very sus-
• Variations in severity from mild to rapidly progressive
ceptible to recurrent infections.
• Autosomal recessive inheritance
• May show ethnic distribution (eg, aspartylglycosaminuria is
Other disorders in which glycoproteins have been
common in Finland)
implicated include hepatitis B and C, Creutzfeldt-
• Vacuolization of cells observed by microscopy in some
Jakob disease, and diarrheas due to a number of bacter-
disorders
ial enterotoxins. It is hoped that basic studies of glyco-
• Presence of abnormal degradation products (eg, oligo-
proteins and other glycoconjugates
(ie, the field of
saccharides that accumulate because of the enzyme
glycobiology) will lead to effective treatments for dis-
deficiency) in urine, detectable by TLC and characterizable
eases in which these molecules are involved. Already, at
by GLC-MS
least two disorders have been found to respond to oral
• Definitive diagnosis made by assay of appropriate enzyme,
supplements of sugars.
often using leukocytes
The fantastic progress made in relation to the
• Possibility of prenatal diagnosis by appropriate enzyme
human genome has stimulated intense interest in both
assays
genomics and proteomics. It is anticipated that the pace
• No definitive treatment at present
of research in glycomics—characterization of the entire
1MIM numbers: α-mannosidosis,
248500; β-mannosidosis,
complement of sugar chains found in cells (the gly-
248510; fucosidosis,
230000; sialidosis,
256550; aspartylgly-
come)—will also accelerate markedly. For a number of
cosaminuria, 208400; Schindler disease, 104170.
reasons, this field will prove more challenging than ei-
ther genomics or proteomics. These reasons include the
complexity of the structures of oligosaccharide chains
due to linkage variations—in contrast to the generally
tem reflects the importance of glycoproteins in the de-
uniform nature of the linkages between nucleotides and
velopment and normal function of that system.
between amino acids. There are also significant varia-
From the above, it should be apparent that glyco-
tions in oligosaccharide structures among cells and at
proteins are involved in a wide variety of biologic
different stages of development. In addition, no simple
processes and diseases. Glycoproteins play direct or in-
technique exists for amplifying oligosaccharides, com-
direct roles in a number of other diseases, as shown in
parable to the PCR reaction. Despite these and other
the following examples.
problems, it seems certain that research in this area will
uncover many new important biologic interactions that
(1) The influenza virus possesses a neuraminidase
are sugar-dependent and will provide targets for drug
that plays a key role in elution of newly synthesized
and other therapies.
progeny from infected cells. If this process is inhibited,
spread of the virus is markedly diminished. Inhibitors
of this enzyme are now available for use in treating pa-
SUMMARY
tients with influenza.
• Glycoproteins are widely distributed proteins—with
(2) HIV-1, thought by many to be the causative
diverse functions—that contain one or more cova-
agent of AIDS, attaches to cells via one of its surface
lently linked carbohydrate chains.
glycoproteins, gp120.
(3) Rheumatoid arthritis is associated with an al-
• The carbohydrate components of a glycoprotein
range from 1% to more than 85% of its weight and
teration in the glycosylation of circulating im-
munoglobulin-γ (IgG) molecules (Chapter 50), such
may be simple or very complex in structure.
that they lack galactose in their Fc regions and termi-
• At least certain of the oligosaccharide chains of glyco-
nate in GlcNAc. Mannose-binding protein (not to be
proteins encode biologic information; they are also
confused with the mannose-6-P receptor), a C-lectin
important to glycoproteins in modulating their solu-
synthesized by liver cells and secreted into the circula-
bility and viscosity, in protecting them against prote-
tion, binds mannose, GlcNAc, and certain other sugars.
olysis, and in their biologic actions.
534
/
CHAPTER 47
•
The structures of many oligosaccharide chains can be
• Developments in the new field of glycomics are likely
elucidated by gas-liquid chromatography, mass spec-
to provide much new information on the roles of
trometry, and high-resolution NMR spectrometry.
sugars in health and disease and also indicate targets
•
Glycosidases hydrolyze specific linkages in oligosac-
for drug and other types of therapies.
charides and are used to explore both the structures
and functions of glycoproteins.
•
Lectins are carbohydrate-binding proteins involved
in cell adhesion and other biologic processes.
REFERENCES
•
The major classes of glycoproteins are O-linked (in-
Brockhausen I, Kuhns W: Glycoproteins and Human Disease. Chap-
volving an OH of serine or threonine), N-linked (in-
man & Hall, 1997.
volving the N of the amide group of asparagine), and
Kornfeld R, Kornfeld S: Assembly of asparagine-linked oligosaccha-
glycosylphosphatidylinositol (GPI)-linked.
rides. Annu Rev Biochem 1985;54:631.
•
Mucins are a class of O-linked glycoproteins that are
Lehrman MA Oligosaccharide-based information in endoplasmic
distributed on the surfaces of epithelial cells of the
reticulum quality control and other biological systems. J Biol
Chem. 2001;276:8623.
respiratory, gastrointestinal, and reproductive tracts.
Perkel JM: Glycobiology goes to the ball. The Scientist 2002;
•
The Golgi apparatus plays a major role in glycosyla-
16:32.
tion reactions involved in the biosynthesis of glyco-
Roseman S: Reflections on glycobiology. J Biol Chem 2001;276:
proteins.
41527.
•
The oligosaccharide chains of O-linked glycoproteins
Schachter H: The clinical relevance of glycobiology. J Clin Invest
are synthesized by the stepwise addition of sugars do-
2001;108:1579.
nated by nucleotide sugars in reactions catalyzed by
Schwartz NB, Domowicz M: Chondrodysplasias due to proteogly-
individual specific glycoprotein glycosyltransferases.
can defects. Glycobiology 2002;12:57R.
Science 2001;21(5512):2263. (This issue contains a special section
•
In contrast, the biosynthesis of N-linked glycopro-
entitled Carbohydrates and Glycobiology. It contains articles
teins involves a specific dolichol-P-P-oligosaccharide
on the synthesis, structural determination, and functions of
and various glycosidases. Depending on the glycosi-
sugar-containing molecules and the roles of glycosylation in
dases and precursor proteins synthesized by a tissue,
the immune system).
it can synthesize complex, hybrid, or high-mannose
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
types of N-linked oligosaccharides.
herited Disease, 8th ed. McGraw-Hill, 2001.(Various chapters
in this text give in-depth coverage of topics such as I-cell dis-
•
Glycoproteins are implicated in many biologic
ease and disorders of glycoprotein degradation.)
processes. For instance, they have been found to play
Spiro RG: Protein glycosylation: nature, distribution, enzymatic
key roles in fertilization and inflammation.
formation, and disease implications of glycopeptide bonds.
•
A number of diseases involving abnormalities in the
Glycobiology 2002;12:43R.
synthesis and degradation of glycoproteins have been
Varki A et al (editors): Essentials of Glycobiology. Cold Spring Har-
recognized. Glycoproteins are also involved in many
bor Laboratory Press, 1999.
other diseases, including influenza, AIDS, and
Vestweber W, Blanks JE: Mechanisms that regulate the function of
rheumatoid arthritis.
the selectins and their ligands. Physiol Rev 1999;79:181.
The Extracellular Matrix
48
Robert K. Murray, MD, PhD, & Frederick W. Keeley, PhD
BIOMEDICAL IMPORTANCE
collagen-like domains in their structures; these proteins
are sometimes referred to as “noncollagen collagens.”
Most mammalian cells are located in tissues where they
Table 48-1 summarizes the types of collagens found
are surrounded by a complex extracellular matrix
in human tissues; the nomenclature used to designate
(ECM) often referred to as “connective tissue.” The
types of collagen and their genes is described in the
ECM contains three major classes of biomolecules: (1)
footnote.
the structural proteins, collagen, elastin, and fibrillin;
The 19 types of collagen mentioned above can be
(2) certain specialized proteins such as fibrillin, fi-
subdivided into a number of classes based primarily on
bronectin, and laminin; and (3) proteoglycans, whose
the structures they form (Table 48-2). In this chapter,
chemical natures are described below. The ECM has
we shall be primarily concerned with the fibril-forming
been found to be involved in many normal and patho-
collagens I and II, the major collagens of skin and bone
logic processes—eg, it plays important roles in develop-
and of cartilage, respectively. However, mention will be
ment, in inflammatory states, and in the spread of can-
made of some of the other collagens.
cer cells. Involvement of certain components of the
ECM has been documented in both rheumatoid arthri-
tis and osteoarthritis. Several diseases (eg, osteogenesis
COLLAGEN TYPE I IS COMPOSED
imperfecta and a number of types of the Ehlers-Danlos
OF A TRIPLE HELIX STRUCTURE
syndrome) are due to genetic disturbances of the syn-
& FORMS FIBRILS
thesis of collagen. Specific components of proteogly-
cans (the glycosaminoglycans; GAGs) are affected in
All collagen types have a triple helical structure. In
the group of genetic disorders known as the mu-
some collagens, the entire molecule is triple helical,
copolysaccharidoses. Changes occur in the ECM dur-
whereas in others the triple helix may involve only a
ing the aging process. This chapter describes the basic
fraction of the structure. Mature collagen type I, con-
biochemistry of the three major classes of biomolecules
taining approximately 1000 amino acids, belongs to the
found in the ECM and illustrates their biomedical sig-
former type; in it, each polypeptide subunit or alpha
nificance. Major biochemical features of two specialized
chain is twisted into a left-handed helix of three
forms of ECM—bone and cartilage—and of a number
residues per turn (Figure 48-1). Three of these alpha
of diseases involving them are also briefly considered.
chains are then wound into a right-handed superhelix,
forming a rod-like molecule 1.4 nm in diameter and
about 300 nm long. A striking characteristic of collagen
COLLAGEN IS THE MOST ABUNDANT
is the occurrence of glycine residues at every third posi-
PROTEIN IN THE ANIMAL WORLD
tion of the triple helical portion of the alpha chain.
Collagen, the major component of most connective tis-
This is necessary because glycine is the only amino acid
sues, constitutes approximately 25% of the protein of
small enough to be accommodated in the limited space
mammals. It provides an extracellular framework for all
available down the central core of the triple helix. This
metazoan animals and exists in virtually every animal
repeating structure, represented as (Gly-X-Y)n, is an ab-
tissue. At least 19 distinct types of collagen made up of
solute requirement for the formation of the triple helix.
30 distinct polypeptide chains (each encoded by a sepa-
While X and Y can be any other amino acids, about
rate gene) have been identified in human tissues. Al-
100 of the X positions are proline and about 100 of the
though several of these are present only in small pro-
Y positions are hydroxyproline. Proline and hydroxy-
portions, they may play important roles in determining
proline confer rigidity on the collagen molecule. Hy-
the physical properties of specific tissues. In addition, a
droxyproline is formed by the posttranslational hy-
number of proteins (eg, the C1q component of the
droxylation of peptide-bound proline residues catalyzed
complement system, pulmonary surfactant proteins
by the enzyme prolyl hydroxylase, whose cofactors are
SP-A and SP-D) that are not classified as collagens have
ascorbic acid (vitamin C) and α-ketoglutarate. Lysines
535
536
/
CHAPTER 48
Table 48-1. Types of collagen and their genes.1,2
Table 48-2. Classification of collagens, based
primarily on the structures that they form.1
Type
Genes
Tissue
Class
Type
I
COL1A1, COL1A2
Most connective tissues,
including bone
Fibril-forming
I, II, III, V, and XI
II
COL2A1
Cartilage, vitreous humor
Network-like
IV, VIII, X
III
COL3A1
Extensible connective tissues
FACITs2
IX, XII, XIV, XVI, XIX
such as skin, lung, and the
Beaded filaments
VI
vascular system
Anchoring fibrils
VII
IV
COL4A1-COL4A6
Basement membranes
Transmembrane domain
XIII, XVII
V
COL5A1-COL5A3
Minor component in tissues
containing collagen I
Others
XV, XVIII
VI
COL6A1-COL6A3
Most connective tissues
1Based on Prockop DJ, Kivirrikko KI: Collagens: molecular biology,
diseases, and potentials for therapy. Annu Rev Biochem
VII
COL7A1
Anchoring fibrils
1995;64:403.
VIII
COL8A1-COL8A2
Endothelium, other tissues
2FACITs
= fibril-associated
collagens
with
interrupted triple
helices.
IX
COL9A1-COL9A3
Tissues containing collagen II
X
COL10A1
Hypertrophic cartilage
XI
COL11A1, COL11A2,
Tissues containing collagen II
COL2A1
XII
COL12A1
Tissues containing collagen I
XIII
COL13A1
Many tissues
67 nm
XIV
COL14A1
Tissues containing collagen I
Fibril
XV
COL15A1
Many tissues
XVI
COL16A1
Many tissues
XVII
COL17A1
Skin hemidesmosomes
300 nm
XVIII
COL18A1
Many tissues (eg, liver, kidney)
XIX
COL19A1
Rhabdomyosarcoma cells
Molecule
1Adapted slightly from Prockop DJ, Kivirrikko KI: Collagens: mole-
cular biology, diseases, and potentials for therapy. Annu Rev
Biochem 1995;64:403.
2The types of collagen are designated by Roman numerals. Con-
stituent procollagen chains, called proα chains, are numbered
Triple helix
1.4 nm
using Arabic numerals, followed by the collagen type in paren-
theses. For instance, type I procollagen is assembled from two
proα1(I) and one proα2(I) chain. It is thus a heterotrimer, whereas
type 2 procollagen is assembled from three proα1(II) chains and
Alpha chain
is thus a homotrimer. The collagen genes are named according to
the collagen type, written in Arabic numerals for the gene sym-
Amino acid
bol, followed by an A and the number of the proα chain that they
- Gly - X - Y - Gly - X - Y - Gly - X - Y -
sequence
encode. Thus, the COL1A1 and COL1A2 genes encode the α1 and
α2 chains of type I collagen, respectively.
Figure 48-1. Molecular features of collagen struc-
ture from primary sequence up to the fibril. (Slightly
modified and reproduced, with permission, from Eyre DR:
Collagen: Molecular diversity in the body’s protein scaf-
fold. Science 1980;207:1315. Copyright © 1980 by the
American Association for the Advancement of Science.)
THE EXTRACELLULAR MATRIX
/
537
in the Y position may also be posttranslationally modi-
Table 48-3. Order and location of processing of
fied to hydroxylysine through the action of lysyl hy-
the fibrillar collagen precursor.
droxylase, an enzyme with similar cofactors. Some of
these hydroxylysines may be further modified by the
Intracellular
addition of galactose or galactosyl-glucose through an
1. Cleavage of signal peptide
O-glycosidic linkage, a glycosylation site that is
2. Hydroxylation of prolyl residues and some lysyl
unique to collagen.
residues; glycosylation of some hydroxylysyl residues
Collagen types that form long rod-like fibers in tis-
3. Formation of intrachain and interchain S-S bonds in ex-
sues are assembled by lateral association of these triple
tension peptides
helical units into a “quarter staggered” alignment such
4. Formation of triple helix
that each is displaced longitudinally from its neighbor
Extracellular
by slightly less than one-quarter of its length (Figure
1. Cleavage of amino and carboxyl terminal propeptides
48-1, upper part). This arrangement is responsible for
2. Assembly of collagen fibers in quarter-staggered align-
the banded appearance of these fibers in connective tis-
ment
sues. Collagen fibers are further stabilized by the forma-
3. Oxidative deamination of ε-amino groups of lysyl and
tion of covalent cross-links, both within and between
hydroxylysyl residues to aldehydes
the triple helical units. These cross-links form through
4. Formation of intra- and interchain cross-links via Schiff
the action of lysyl oxidase, a copper-dependent en-
bases and aldol condensation products
zyme that oxidatively deaminates the ε-amino groups of
certain lysine and hydroxylysine residues, yielding reac-
tive aldehydes. Such aldehydes can form aldol conden-
sation products with other lysine- or hydroxylysine-
polypeptide extensions (extension peptides) of 20-35
derived aldehydes or form Schiff bases with the
kDa at both its amino and carboxyl terminal ends, nei-
ε-amino groups of unoxidized lysines or hydroxy-
ther of which is present in mature collagen. Both exten-
lysines. These reactions, after further chemical re-
sion peptides contain cysteine residues. While the
arrangements, result in the stable covalent cross-links
amino terminal propeptide forms only intrachain disul-
that are important for the tensile strength of the fibers.
fide bonds, the carboxyl terminal propeptides form
Histidine may also be involved in certain cross-links.
both intrachain and interchain disulfide bonds. Forma-
Several collagen types do not form fibrils in tissues
tion of these disulfide bonds assists in the registration of
(Table 48-2). They are characterized by interruptions
the three collagen molecules to form the triple helix,
of the triple helix with stretches of protein lacking Gly-
winding from the carboxyl terminal end. After forma-
X-Y repeat sequences. These non-Gly-X-Y sequences
tion of the triple helix, no further hydroxylation of pro-
result in areas of globular structure interspersed in the
line or lysine or glycosylation of hydroxylysines can
triple helical structure.
take place. Self-assembly is a cardinal principle in the
Type IV collagen, the best-characterized example of
biosynthesis of collagen.
a collagen with discontinuous triple helices, is an im-
Following secretion from the cell by way of the
portant component of basement membranes, where it
Golgi apparatus, extracellular enzymes called procolla-
forms a mesh-like network.
gen aminoproteinase and procollagen carboxypro-
teinase remove the extension peptides at the amino and
carboxyl terminal ends, respectively. Cleavage of these
Collagen Undergoes Extensive
propeptides may occur within crypts or folds in the cell
Posttranslational Modifications
membrane. Once the propeptides are removed, the
Newly synthesized collagen undergoes extensive post-
triple helical collagen molecules, containing approxi-
translational modification before becoming part of a
mately 1000 amino acids per chain, spontaneously as-
mature extracellular collagen fiber (Table 48-3). Like
semble into collagen fibers. These are further stabilized
most secreted proteins, collagen is synthesized on ribo-
by the formation of inter- and intrachain cross-links
somes in a precursor form, preprocollagen, which con-
through the action of lysyl oxidase, as described previ-
tains a leader or signal sequence that directs the
ously.
polypeptide chain into the lumen of the endoplasmic
The same cells that secrete collagen also secrete fi-
reticulum. As it enters the endoplasmic reticulum, this
bronectin, a large glycoprotein present on cell surfaces,
leader sequence is enzymatically removed. Hydroxyla-
in the extracellular matrix, and in blood (see below). Fi-
tion of proline and lysine residues and glycosylation of
bronectin binds to aggregating precollagen fibers and
hydroxylysines in the procollagen molecule also take
alters the kinetics of fiber formation in the pericellular
place at this site. The procollagen molecule contains
matrix. Associated with fibronectin and procollagen in
538
/
CHAPTER 48
this matrix are the proteoglycans heparan sulfate and
Table 48-4. Diseases caused by mutations in
chondroitin sulfate (see below). In fact, type IX colla-
collagen genes or by deficiencies in the activities
gen, a minor collagen type from cartilage, contains at-
of posttranslational enzymes involved in the
tached proteoglycan chains. Such interactions may
biosynthesis of collagen.1
serve to regulate the formation of collagen fibers and to
determine their orientation in tissues.
Gene or Enzyme
Disease2
Once formed, collagen is relatively metabolically sta-
ble. However, its breakdown is increased during starva-
COL1A1, COL1A2
Osteogenesis imperfecta, type 13 (MIM
tion and various inflammatory states. Excessive produc-
1566200)
tion of collagen occurs in a number of conditions, eg,
Osteoporosis4 (MIM 166710)
hepatic cirrhosis.
Ehlers-Danlos syndrome type VII auto-
somal dominant (130060)
A Number of Genetic Diseases Result From
COL2A1
Severe chondrodysplasias
Abnormalities in the Synthesis of Collagen
Osteoarthritis4 (MIM 120140)
COL3A1
Ehlers-Danlos syndrome type IV (MIM
About 30 genes encode collagen, and its pathway of
130050)
biosynthesis is complex, involving at least eight en-
zyme-catalyzed posttranslational steps. Thus, it is not
COL4A3-COL4A6
Alport syndrome (including both auto-
surprising that a number of diseases (Table 48-4) are
somal and X-linked forms) (MIM 104200)
due to mutations in collagen genes or in genes en-
COL7A1
Epidermolysis bullosa, dystrophic (MIM
coding some of the enzymes involved in these post-
131750)
translational modifications. The diseases affecting bone
(eg, osteogenesis imperfecta) and cartilage
(eg, the
COL10A1
Schmid metaphysial chondrodysplasia
chondrodysplasias) will be discussed later in this chap-
(MIM 156500)
ter.
Lysyl hydroxylase
Ehlers-Danlos syndrome type VI (MIM
Ehlers-Danlos syndrome comprises a group of in-
225400)
herited disorders whose principal clinical features are
Procollagen
Ehlers-Danlos syndrome type VII auto-
hyperextensibility of the skin, abnormal tissue fragility,
N-proteinase
somal recessive (MIM 225410)
and increased joint mobility. The clinical picture is
variable, reflecting underlying extensive genetic hetero-
Lysyl hydroxylase
Menkes disease5 (MIM 309400)
geneity. At least 10 types have been recognized, most
1Adapted from Prockop DJ, Kivirrikko KI: Collagens: molecular bi-
but not all of which reflect a variety of lesions in the
ology, diseases, and potentials for therapy. Annu Rev Biochem
synthesis of collagen. Type IV is the most serious be-
1995;64:403.
cause of its tendency for spontaneous rupture of arteries
2Genetic linkage to collagen genes has been shown for a few
or the bowel, reflecting abnormalities in type III colla-
other conditions not listed here.
gen. Patients with type VI, due to a deficiency of lysyl
3At least four types of osteogenesis imperfecta are recognized;
the great majority of mutations in all types are in the COL1A1 and
hydroxylase, exhibit marked joint hypermobility and a
COL1A2 genes.
tendency to ocular rupture. A deficiency of procollagen
4At present applies to only a relatively small number of such
N-proteinase, causing formation of abnormal thin, ir-
patients.
regular collagen fibrils, results in type VIIC, manifested
5Secondary to a deficiency of copper (Chapter 50).
by marked joint hypermobility and soft skin.
Alport syndrome is the designation applied to a
number of genetic disorders (both X-linked and autoso-
mal) affecting the structure of type IV collagen fibers,
due to mutations in COL7A1, affecting the structure of
the major collagen found in the basement membranes
type VII collagen. This collagen forms delicate fibrils
of the renal glomeruli
(see discussion of laminin,
that anchor the basal lamina to collagen fibrils in the
below). Mutations in several genes encoding type IV
dermis. These anchoring fibrils have been shown to be
collagen fibers have been demonstrated. The presenting
markedly reduced in this form of the disease, probably
sign is hematuria, and patients may eventually develop
resulting in the blistering. Epidermolysis bullosa sim-
end-stage renal disease. Electron microscopy reveals
plex, another variant, is due to mutations in keratin 5
characteristic abnormalities of the structure of the base-
(Chapter 49).
ment membrane and lamina densa.
Scurvy affects the structure of collagen. However, it
In epidermolysis bullosa, the skin breaks and blis-
is due to a deficiency of ascorbic acid (Chapter 45) and
ters as a result of minor trauma. The dystrophic form is
is not a genetic disease. Its major signs are bleeding
THE EXTRACELLULAR MATRIX
/
539
gums, subcutaneous hemorrhages, and poor wound
Table 48-5. Major differences between collagen
healing. These signs reflect impaired synthesis of colla-
and elastin.
gen due to deficiencies of prolyl and lysyl hydroxylases,
both of which require ascorbic acid as a cofactor.
Collagen
Elastin
1. Many different genetic
One genetic type
ELASTIN CONFERS EXTENSIBILITY
types
& RECOIL ON LUNG, BLOOD
2. Triple helix
No triple helix; random coil
VESSELS, & LIGAMENTS
conformations permitting
stretching
Elastin is a connective tissue protein that is responsible
3.
(Gly-X-Y)n repeating
No (Gly-X-Y)n repeating
for properties of extensibility and elastic recoil in tis-
structure
structure
sues. Although not as widespread as collagen, elastin is
4. Presence of hydroxylysine
No hydroxylysine
present in large amounts, particularly in tissues that re-
5. Carbohydrate-containing
No carbohydrate
quire these physical properties, eg, lung, large arterial
6. Intramolecular aldol
Intramolecular desmosine
blood vessels, and some elastic ligaments. Smaller quan-
cross-links
cross-links
tities of elastin are also found in skin, ear cartilage, and
7. Presence of extension
No extension peptides present
several other tissues. In contrast to collagen, there ap-
peptides during bio-
during biosynthesis
pears to be only one genetic type of elastin, although
synthesis
variants arise by alternative splicing (Chapter 37) of the
hnRNA for elastin. Elastin is synthesized as a soluble
monomer of 70 kDa called tropoelastin. Some of the
prolines of tropoelastin are hydroxylated to hydroxy-
MARFAN SYNDROME IS DUE TO
proline by prolyl hydroxylase, though hydroxylysine
MUTATIONS IN THE GENE FOR FIBRILLIN,
and glycosylated hydroxylysine are not present. Unlike
A PROTEIN PRESENT IN MICROFIBRILS
collagen, tropoelastin is not synthesized in a pro- form
with extension peptides. Furthermore, elastin does not
Marfan syndrome is a relatively prevalent inherited dis-
contain repeat Gly-X-Y sequences, triple helical struc-
ease affecting connective tissue; it is inherited as an au-
ture, or carbohydrate moieties.
tosomal dominant trait. It affects the eyes (eg, causing
After secretion from the cell, certain lysyl residues of
dislocation of the lens, known as ectopia lentis), the
tropoelastin are oxidatively deaminated to aldehydes by
skeletal system (most patients are tall and exhibit long
lysyl oxidase, the same enzyme involved in this process
digits
[arachnodactyly] and hyperextensibility of the
in collagen. However, the major cross-links formed in
joints), and the cardiovascular system
(eg, causing
elastin are the desmosines, which result from the con-
weakness of the aortic media, leading to dilation of the
densation of three of these lysine-derived aldehydes with
ascending aorta). Abraham Lincoln may have had this
an unmodified lysine to form a tetrafunctional cross-
condition. Most cases are caused by mutations in the
link unique to elastin. Once cross-linked in its mature,
gene (on chromosome 15) for fibrillin; missense muta-
extracellular form, elastin is highly insoluble and ex-
tions have been detected in several patients with Mar-
tremely stable and has a very low turnover rate. Elastin
fan syndrome.
exhibits a variety of random coil conformations that per-
Fibrillin is a large glycoprotein (about 350 kDa)
mit the protein to stretch and subsequently recoil during
that is a structural component of microfibrils, 10- to
the performance of its physiologic functions.
12-nm fibers found in many tissues. Fibrillin is secreted
Table 48-5 summarizes the main differences be-
(subsequent to a proteolytic cleavage) into the extracel-
tween collagen and elastin.
lular matrix by fibroblasts and becomes incorporated
Deletions in the elastin gene (located at 7q11.23)
into the insoluble microfibrils, which appear to provide
have been found in approximately 90% of subjects with
a scaffold for deposition of elastin. Of special relevance
Williams syndrome, a developmental disorder affect-
to Marfan syndrome, fibrillin is found in the zonular
ing connective tissue and the central nervous system.
fibers of the lens, in the periosteum, and associated
The mutations, by affecting synthesis of elastin, proba-
with elastin fibers in the aorta (and elsewhere); these lo-
bly play a causative role in the supravalvular aortic
cations respectively explain the ectopia lentis, arach-
stenosis often found in this condition. A number of
nodactyly, and cardiovascular problems found in the
skin diseases (eg, scleroderma) are associated with accu-
syndrome. Other proteins (eg, emelin and two mi-
mulation of elastin. Fragmentation or, alternatively, a
crofibril-associated proteins) are also present in these
decrease of elastin is found in conditions such as pul-
microfibrils, and it appears likely that abnormalities of
monary emphysema, cutis laxa, and aging of the skin.
them may cause other connective tissue disorders. An-
540
/
CHAPTER 48
other gene for fibrillin exists on chromosome 5; muta-
ECM, with a typical cell (eg, fibroblast) present in the
tions in this gene are linked to causation of congenital
matrix.
contractural arachnodactyly but not to Marfan syn-
The fibronectin receptor interacts indirectly with
drome. The probable sequence of events leading to
actin microfilaments (Chapter 49) present in the cy-
Marfan syndrome is summarized in Figure 48-2.
tosol (Figure 48-5). A number of proteins, collectively
known as attachment proteins, are involved; these in-
clude talin, vinculin, an actin-filament capping protein,
FIBRONECTIN IS AN IMPORTANT
and α-actinin. Talin interacts with the receptor and
GLYCOPROTEIN INVOLVED IN CELL
vinculin, whereas the latter two interact with actin. The
ADHESION & MIGRATION
interaction of fibronectin with its receptor provides one
Fibronectin is a major glycoprotein of the extracellular
route whereby the exterior of the cell can communicate
matrix, also found in a soluble form in plasma. It con-
with the interior and thus affect cell behavior. Via the
sists of two identical subunits, each of about 230 kDa,
interaction with its cell receptor, fibronectin plays an
joined by two disulfide bridges near their carboxyl ter-
important role in the adhesion of cells to the ECM. It is
minals. The gene encoding fibronectin is very large,
also involved in cell migration by providing a binding
containing some 50 exons; the RNA produced by its
site for cells and thus helping them to steer their way
transcription is subject to considerable alternative splic-
through the ECM. The amount of fibronectin around
ing, and as many as 20 different mRNAs have been de-
many transformed cells is sharply reduced, partly ex-
tected in various tissues. Fibronectin contains three
plaining their faulty interaction with the ECM.
types of repeating motifs (I, II, and III), which are orga-
nized into functional domains (at least seven); func-
tions of these domains include binding heparin (see
LAMININ IS A MAJOR PROTEIN
below) and fibrin, collagen, DNA, and cell surfaces
COMPONENT OF RENAL GLOMERULAR
(Figure
48-3). The amino acid sequence of the fi-
& OTHER BASAL LAMINAS
bronectin receptor of fibroblasts has been derived, and
the protein is a member of the transmembrane integrin
Basal laminas are specialized areas of the ECM that sur-
round epithelial and some other cells (eg, muscle cells);
class of proteins (Chapter 51). The integrins are het-
erodimers, containing various types of α and β
here we discuss only the laminas found in the renal
glomerulus. In that structure, the basal lamina is con-
polypeptide chains. Fibronectin contains an Arg-Gly-
Asp (RGD) sequence that binds to the receptor. The
tributed by two separate sheets of cells (one endothelial
and one epithelial), each disposed on opposite sides of
RGD sequence is shared by a number of other proteins
present in the ECM that bind to integrins present in
the lamina; these three layers make up the glomerular
membrane. The primary components of the basal lam-
cell surfaces. Synthetic peptides containing the RGD
sequence inhibit the binding of fibronectin to cell sur-
ina are three proteins—laminin, entactin, and type IV
collagen—and the GAG heparin or heparan sulfate.
faces. Figure 48-4 illustrates the interaction of collagen,
fibronectin, and laminin, all major proteins of the
These components are synthesized by the underlying
cells.
Laminin (about 850 kDa, 70 nm long) consists of
three distinct elongated polypeptide chains (A, B1, and
Mutations in gene (on chromosome 15)
B2) linked together to form an elongated cruciform
for fibrillin, a large glycoprotein present in
shape. It has binding sites for type IV collagen, heparin,
elastin-associated microfibrils
and integrins on cell surfaces. The collagen interacts
with laminin (rather than directly with the cell surface),
Abnormalities of the structure of fibrillin
which in turn interacts with integrins or other laminin
receptor proteins, thus anchoring the lamina to the
cells. Entactin, also known as “nidogen,” is a glycopro-
Structures of the suspensory ligament of the eye,
tein containing an RGD sequence; it binds to laminin
the periosteum, and the media of the aorta affected
and is a major cell attachment factor. The relatively
thick basal lamina of the renal glomerulus has an im-
Ectopia lentis, arachnodactyly,
portant role in glomerular filtration, regulating the
and dilation of the ascending aorta
passage of large molecules (most plasma proteins) across
the glomerulus into the renal tubule. The glomerular
Figure 48-2. Probable sequence of events in the
membrane allows small molecules, such as inulin (5.2
causation of the major signs exhibited by patients with
kDa), to pass through as easily as water. On the other
Marfan syndrome (MIM 154700).
hand, only a small amount of the protein albumin (69
THE EXTRACELLULAR MATRIX
/
541
RGD
Heparin A
Collagen
DNA
Cell A
Heparin B
Cell B
Fibrin B
Fibrin A
Figure 48-3. Schematic representation of fibronectin. Seven functional
domains of fibronectin are represented; two different types of domain for
heparin, cell-binding, and fibrin are shown. The domains are composed of
various combinations of three structural motifs (I, II, and III), not depicted
in the figure. Also not shown is the fact that fibronectin is a dimer joined
by disulfide bridges near the carboxyl terminals of the monomers. The ap-
proximate location of the RGD sequence of fibronectin, which interacts
with a variety of fibronectin integrin receptors on cell surfaces, is indicated
by the arrow. (Redrawn after Yamada KM: Adhesive recognition sequences.
J Biol Chem 1991;266:12809.)
kDa), the major plasma protein, passes through the
normal glomerulus. This is explained by two sets of
facts: (1) The pores in the glomerular membrane are
large enough to allow molecules up to about 8 nm to
pass through. (2) Albumin is smaller than this pore size,
but it is prevented from passing through easily by the
Collagen
negative charges of heparan sulfate and of certain sialic
acid-containing glycoproteins present in the lamina.
These negative charges repel albumin and most plasma
proteins, which are negatively charged at the pH of
Heparin
Fibronectin
blood. The normal structure of the glomerulus may be
severely damaged in certain types of glomerulonephri-
OUTSIDE
S-S
tis
(eg, caused by antibodies directed against various
S-S
components of the glomerular membrane). This alters
the pores and the amounts and dispositions of the nega-
tively charged macromolecules referred to above, and
relatively massive amounts of albumin (and of certain
Integrin
receptor
Plasma membrane
Collagen
Fibronectin
Talin
Vinculin
INSIDE
Capping protein
α-Actin
Actin
Laminin
Figure 48-5.
Schematic representation of fibro-
Figure 48-4. Schematic representation of a cell in-
nectin interacting with an integrin fibronectin receptor
teracting through various integrin receptors with colla-
situated in the exterior of the plasma membrane of a
gen, fibronectin, and laminin present in the ECM. (Spe-
cell of the ECM and of various attachment proteins in-
cific subunits are not indicated.) (Redrawn after Yamada
teracting indirectly or directly with an actin microfila-
KM: Adhesive recognition sequences. J Biol Chem
ment in the cytosol. For simplicity, the attachment pro-
1991;266:12809.)
teins are represented as a complex.
542
/
CHAPTER 48
other plasma proteins) can pass through into the urine,
resulting in severe albuminuria.
PROTEOGLYCANS
& GLYCOSAMINOGLYCANS
The Glycosaminoglycans Found
in Proteoglycans Are Built Up
of Repeating Disaccharides
Proteoglycans are proteins that contain covalently
linked glycosaminoglycans. A number of them have
been characterized and given names such as syndecan,
betaglycan, serglycin, perlecan, aggrecan, versican,
decorin, biglycan, and fibromodulin. They vary in tis-
sue distribution, nature of the core protein, attached
glycosaminoglycans, and function. The proteins bound
covalently to glycosaminoglycans are called “core pro-
teins”; they have proved difficult to isolate and charac-
terize, but the use of recombinant DNA technology is
beginning to yield important information about their
structures. The amount of carbohydrate in a proteogly-
can is usually much greater than is found in a glycopro-
tein and may comprise up to 95% of its weight. Figures
48-6 and 48-7 show the general structure of one par-
ticular proteoglycan, aggrecan, the major type found in
cartilage. It is very large (about 2 × 103 kDa), with its
overall structure resembling that of a bottle brush. It
contains a long strand of hyaluronic acid (one type of
Figure 48-6.
Dark field electron micrograph of a
GAG) to which link proteins are attached noncova-
proteoglycan aggregate in which the proteoglycan
lently. In turn, these latter interact noncovalently with
subunits and filamentous backbone are particularly
core protein molecules from which chains of other
well extended. (Reproduced, with permission, from
GAGs (keratan sulfate and chondroitin sulfate in this
Rosenberg L, Hellman W, Kleinschmidt AK: Electron micro-
case) project. More details on this macromolecule are
scopic studies of proteoglycan aggregates from bovine
given when cartilage is discussed below.
articular cartilage. J Biol Chem 1975;250:1877.)
There are at least seven glycosaminoglycans
(GAGs): hyaluronic acid, chondroitin sulfate, keratan
sulfates I and II, heparin, heparan sulfate, and der-
matan sulfate. A GAG is an unbranched polysaccharide
biologic roles; and they are involved in a number of dis-
made up of repeating disaccharides, one component of
ease processes—so that interest in them is increasing
which is always an amino sugar
(hence the name
rapidly.
GAG), either D-glucosamine or D-galactosamine. The
other component of the repeating disaccharide (except
in the case of keratan sulfate) is a uronic acid, either
Biosynthesis of Glycosaminoglycans
L-glucuronic acid (GlcUA) or its 5′-epimer, L-iduronic
Involves Attachment to Core Proteins,
acid (IdUA). With the exception of hyaluronic acid, all
Chain Elongation, & Chain Termination
the GAGs contain sulfate groups, either as O-esters or
A. ATTACHMENT TO CORE PROTEINS
as N-sulfate
(in heparin and heparan sulfate).
Hyaluronic acid affords another exception because
The linkage between GAGs and their core proteins is
there is no clear evidence that it is attached covalently
generally one of three types.
to protein, as the definition of a proteoglycan given
above specifies. Both GAGs and proteoglycans have
1. An O-glycosidic bond between xylose (Xyl) and
proved difficult to work with, partly because of their
Ser, a bond that is unique to proteoglycans. This link-
complexity. However, they are major components of
age is formed by transfer of a Xyl residue to Ser from
the ground substance; they have a number of important
UDP-xylose. Two residues of Gal are then added to the
THE EXTRACELLULAR MATRIX
/
543
as in the case of certain types of linkages found in gly-
coproteins. The enzyme systems involved in chain elon-
Hyaluronic acid
gation are capable of high-fidelity reproduction of com-
Link protein
plex GAGs.
Keratan sulfate
C. CHAIN TERMINATION
Chondroitin sulfate
This appears to result from (1) sulfation, particularly at
certain positions of the sugars, and (2) the progression
Core protein
of the growing GAG chain away from the membrane
site where catalysis occurs.
D. FURTHER MODIFICATIONS
After formation of the GAG chain, numerous chemical
Subunits
modifications occur, such as the introduction of sulfate
groups onto GalNAc and other moieties and the
epimerization of GlcUA to IdUA residues. The enzymes
catalyzing sulfation are designated sulfotransferases and
use 3′-phosphoadenosine-5′-phosphosulfate (PAPS; ac-
tive sulfate) as the sulfate donor. These Golgi-located
enzymes are highly specific, and distinct enzymes cat-
alyze sulfation at different positions (eg, carbons 2, 3, 4,
and 6) on the acceptor sugars. An epimerase catalyzes
conversions of glucuronyl to iduronyl residues.
Figure 48-7.
Schematic representation of the pro-
The Various Glycosaminoglycans Exhibit
teoglycan aggrecan. (Reproduced, with permission, from
Differences in Structure & Have
Lennarz WJ:The Biochemistry of Glycoproteins and Proteo-
Characteristic Distributions
glycans. Plenum Press, 1980.)
The seven GAGs named above differ from each other in
a number of the following properties: amino sugar com-
position, uronic acid composition, linkages between
these components, chain length of the disaccharides, the
Xyl residue, forming a link trisaccharide, Gal-Gal-Xyl-
presence or absence of sulfate groups and their positions
Ser. Further chain growth of the GAG occurs on the
of attachment to the constituent sugars, the nature of
terminal Gal.
the core proteins to which they are attached, the nature
2. An O-glycosidic bond forms between GalNAc
of the linkage to core protein, their tissue and subcellu-
(N-acetylgalactosamine) and Ser
(Thr)
(Figure
47-
lar distribution, and their biologic functions.
1[a]), present in keratan sulfate II. This bond is formed
The structures (Figure 48-8) and the distributions
by donation to Ser (or Thr) of a GalNAc residue, em-
of each of the GAGs will now be briefly discussed. The
ploying UDP-GalNAc as its donor.
major features of the seven GAGs are summarized in
3. An N-glycosylamine bond between GlcNAc
Table 48-6.
(N-acetylglucosamine) and the amide nitrogen of Asn,
as found in N-linked glycoproteins (Figure 47-1[b]).
A. HYALURONIC ACID
Its synthesis is believed to involve dolichol-P-P-
Hyaluronic acid consists of an unbranched chain of re-
oligosaccharide.
peating disaccharide units containing GlcUA and Glc-
The synthesis of the core proteins occurs in the en-
NAc. Hyaluronic acid is present in bacteria and is
doplasmic reticulum, and formation of at least some
widely distributed among various animals and tissues,
of the above linkages also occurs there. Most of the later
including synovial fluid, the vitreous body of the eye,
steps in the biosynthesis of GAG chains and their sub-
cartilage, and loose connective tissues.
sequent modifications occur in the Golgi apparatus.
B. CHONDROITIN SULFATES (CHONDROITIN
B. CHAIN ELONGATION
4-SULFATE & CHONDROITIN 6-SULFATE)
Appropriate nucleotide sugars and highly specific
Proteoglycans linked to chondroitin sulfate by the Xyl-
Golgi-located glycosyltransferases are employed to syn-
Ser O-glycosidic bond are prominent components of
thesize the oligosaccharide chains of GAGs. The “one
cartilage
(see below). The repeating disaccharide is
enzyme, one linkage” relationship appears to hold here,
similar to that found in hyaluronic acid, containing
544
/
CHAPTER 48
β1,4
β1,3
β1,4
β1,3
β1,4
Hyaluronic acid:
GlcUA
GlcNAc
GlcUA
GlcNAc
β1,4
β1,3
β1,4
β1,3
β1,3
β1,4
β
Chondroitin sulfates:
GlcUA
GalNAc
GlcUA
Gal
Gal
Xyl
Ser
4- or 6-Sulfate
β
GlcNAc
Asn (keratan sulfate I)
Keratan sulfates
β1,4
β1,3
β1,4
β1,3
I and II:
GlcNAc
Gal
GlcNAc
Gal
α
6-Sulfate
6-Sulfate
GalNAc
Thr (Ser) (keratan sulfate II)
Gal-NeuAc
6-Sulfate
Heparin and
α1,4
α1,4
α1,4
β1,4
α1,4
β1,3
β1,3
β1,4
β
heparan sulfate:
IdUA
GlcN
GlcUA
GlcNAc
GlcUA
Gal
Gal
Xyl
Ser
2-Sulfate
SO3- or Ac
β1,4
α1,3
β1,4
β1,3
β1,4
β1,3
β1,3
β1,4
β
Dermatan sulfate:
IdUA
GalNAc
GlcUA
GalNAc
GlcUA
Gal
Gal
Xyl
Ser
2-Sulfate
4-Sulfate
Figure 48-8. Summary of structures of glycosaminoglycans and their attachments to core proteins. (GlcUA,
D-glucuronic acid; IdUA, L-iduronic acid; GlcN, D-glucosamine; GalN, D-galactosamine; Ac, acetyl; Gal, D-galac-
tose; Xyl, D-xylose; Ser, L-serine; Thr, L-threonine; Asn, L-asparagine; Man, D-mannose; NeuAc, N-acetylneu-
raminic acid.) The summary structures are qualitative representations only and do not reflect, for example, the
uronic acid composition of hybrid glycosaminoglycans such as heparin and dermatan sulfate, which contain
both L-iduronic and D-glucuronic acid. Neither should it be assumed that the indicated substituents are always
present, eg, whereas most iduronic acid residues in heparin carry a 2′-sulfate group, a much smaller proportion
of these residues are sulfated in dermatan sulfate. The presence of link trisaccharides (Gal-Gal-Xyl) in the chon-
droitin sulfates, heparin, and heparan and dermatan sulfates is shown. (Slightly modified and reproduced, with
permission, from Lennarz WJ: The Biochemistry of Glycoproteins and Proteoglycans. Plenum Press, 1980.)
Table 48-6. Major properties of the glycosaminoglycans.
GAG
Sugars
Sulfate1
Linkage of Protein
Location
HA
GIcNAc, GlcUA
Nil
No firm evidence
Synovial fluid, vitreous humor, loose connective tissue
CS
GaINAc, GlcUA
GalNAc
Xyl-Ser; associated with
HA via link proteins
Cartilage, bone, cornea
KS I
GlcNAc, Gal
GlcNAc
GlcNAc-Asn
Cornea
KS II
GlcNAc, Gal
Same as KS I
GalNAc-Thr
Loose connective tissue
Heparin
GlcN, IdUA
GlcN
Ser
Mast cells
GlcN
IdUA
Heparan sulfate
GlcN, GlcUA
GlcN
Xyl-Ser
Skin fibroblasts, aortic wall
Dermatan
GalNAc, IdUA,
GaINAc
Xyl-Ser
Wide distribution
sulfate
(GlcUA)
IdUa
1The sulfate is attached to various positions of the sugars indicated (see Figure 48-7).
THE EXTRACELLULAR MATRIX
/
545
GlcUA but with GalNAc replacing GlcNAc. The
cept that in place of a GlcUA in β-1,3 linkage to
GalNAc is substituted with sulfate at either its 4′ or its
GalNAc it contains an IdUA in an α-1,3 linkage to
6′ position, with approximately one sulfate being pre-
GalNAc. Formation of the IdUA occurs, as in heparin
sent per disaccharide unit.
and heparan sulfate, by 5′-epimerization of GlcUA. Be-
cause this is regulated by the degree of sulfation and be-
C. KERATAN SULFATES I & II
cause sulfation is incomplete, dermatan sulfate contains
As shown in Figure 48-8, the keratan sulfates consist of
both IdUA-GalNAc and GlcUA-GalNAc disaccha-
repeating Gal-GlcNAc disaccharide units containing
rides.
sulfate attached to the 6′ position of GlcNAc or occa-
sionally of Gal. Type I is abundant in cornea, and type
II is found along with chondroitin sulfate attached to
hyaluronic acid in loose connective tissue. Types I and
Deficiencies of Enzymes That Degrade
II have different attachments to protein (Figure 48-8).
Glycosaminoglycans Result in
Mucopolysaccharidoses
D. HEPARIN
The repeating disaccharide contains glucosamine
Both exo- and endoglycosidases degrade GAGs. Like
most other biomolecules, GAGs are subject to
(GlcN) and either of the two uronic acids
(Figure
48-9). Most of the amino groups of the GlcN residues
turnover, being both synthesized and degraded. In
adult tissues, GAGs generally exhibit relatively slow
are N-sulfated, but a few are acetylated. The GlcN also
carries a C6 sulfate ester.
turnover, their half-lives being days to weeks.
Understanding of the degradative pathways for
Approximately 90% of the uronic acid residues are
IdUA. Initially, all of the uronic acids are GlcUA, but a
GAGs, as in the case of glycoproteins (Chapter 47) and
glycosphingolipids (Chapter 24), has been greatly aided
5′-epimerase converts approximately
90% of the
GlcUA residues to IdUA after the polysaccharide chain
by elucidation of the specific enzyme deficiencies that
occur in certain inborn errors of metabolism. When
is formed. The protein molecule of the heparin proteo-
glycan is unique, consisting exclusively of serine and
GAGs are involved, these inborn errors are called mu-
copolysaccharidoses (Table 48-7).
glycine residues. Approximately two-thirds of the serine
residues contain GAG chains, usually of 5-15 kDa but
Degradation of GAGs is carried out by a battery of
lysosomal hydrolases. These include certain endogly-
occasionally much larger. Heparin is found in the gran-
ules of mast cells and also in liver, lung, and skin.
cosidases, various exoglycosidases, and sulfatases, gener-
ally acting in sequence to degrade the various GAGs. A
E. HEPARAN SULFATE
number of them are indicated in Table 48-7.
This molecule is present on many cell surfaces as a
The mucopolysaccharidoses share a common
proteoglycan and is extracellular. It contains GlcN with
mechanism of causation, as illustrated in Figure 48-10.
fewer N-sulfates than heparin, and, unlike heparin, its
They are inherited in an autosomal recessive manner,
predominant uronic acid is GlcUA.
with Hurler and Hunter syndromes being perhaps the
most widely studied. None are common. In some cases,
F. DERMATAN SULFATE
a family history of a mucopolysaccharidosis is obtained.
This substance is widely distributed in animal tissues.
Specific laboratory investigations of help in their diag-
Its structure is similar to that of chondroitin sulfate, ex-
nosis are urine testing for the presence of increased
-
-
-
-
CH2OSO3-
CH2OSO
3
CH2OSO3
CO
2
CH2OSO3
O
O
O
O
O
O
O
-
CO2-
CO
2
O
O
O
O
OH
OH
OH
OH
OH
OH
OH
O
O
O
O
HNSO3
–
OSO3
–
HNSO3
–
OH
HNSO
-
OH
HNAc
3
GlcN
IdUA
GlcN
IdUA
GlcN
GlcUA
GlcNAc
Figure 48-9. Structure of heparin. The polymer section illustrates structural features typical of heparin;
however, the sequence of variously substituted repeating disaccharide units has been arbitrarily selected. In
addition, non-O-sulfated or 3-O-sulfated glucosamine residues may also occur. (Modified, redrawn, and repro-
duced, with permission, from Lindahl U et al: Structure and biosynthesis of heparin-like polysaccharides. Fed Proc
1977;36:19.)
546
/
CHAPTER 48
Table 48-7. Biochemical defects and diagnostic tests in mucopolysaccharidoses (MPS) and
mucolipidoses (ML).1
Alternative
Urinary
Name
Designation2,3
Enzymatic Defect
Metabolites
Mucopolysaccharidoses
Hurler, Scheie,
MPS I
α-L-Iduronidase
Dermatan sulfate, heparan sulfate
Hurler-Scheie
(MIM 252800)
Hunter (MIM 309900)
MPS II
Iduronate sulfatase
Dermatan sulfate, heparan sulfate
Sanfilippo A
MPS IIIA
Heparan sulfate N-sulfatase
Heparan sulfate
(MIM 252900)
(sulfamidase)
Sanfilippo B
MPS IIIB
α-N-Acetylglucosaminidase
Heparan sulfate
(MIM 252920)
Sanfilippo C
MPS IIIC
Acetyltransferase
Heparan sulfate
(MIM 252930)
Sanfilippo D
MPS IIID
N-Acetylglucosamine
Heparan sulfate
(MIM 252940)
6-sulfatase
Morquio A
MPS IVA
Galactosamine 6-sulfatase
Keratan sulfate, chondroitin 6-sulfate
(MIM 253000)
Morquio B
MPS IVB
β-Galactosidase
Keratan sulfate
(MIM 253010)
Maroteaux-Lamy
MPS VI
N-Acetylgalactosamine 4-
Dermatan sulfate
(MIM 253200)
sulfatase (arylsulfatase B)
Sly (MIM 253220)
MPS VII
β-Glucuronidase
Dermatan sulfate, heparan sulfate, chondroitin
4-sulfate, chondroitin 6-sulfate
Mucolipidoses
Sialidosis
M LI
Sialidase (neuraminidase)
Glycoprotein fragments
(MIM 256550)
I-cell disease
ML II
UDP-N-acetylglucosamine:
Glycoprotein fragments
(MIM 252500)
glycoprotein N-acetylglu-
cosamininylphosphotrans-
ferase. (Acid hydrolases
thus lack phosphoman-
nosyl residues.)
Pseudo-Hurler
ML III
As for ML II but deficiency
Glycoprotein fragments
polydystrophy
is incomplete
(MIM 252600)
1Modified and reproduced, with permission, from DiNatale P, Neufeld EF: The biochemical diagnosis of mucopolysaccharidoses,
mucolipidoses and related disorders. In: Perspectives in Inherited Metabolic Diseases, vol 2. Barr B et al (editors). Editiones Ermes
(Milan), 1979.
2Fibroblasts, leukocytes, tissues, amniotic fluid cells, or serum can be used for the assay of many of the above enzymes. Patients
with these disorders exhibit a variety of clinical findings that may include cloudy corneas, mental retardation, stiff joints, cardiac ab-
normalities, hepatosplenomegaly, and short stature, depending on the specific disease and its severity.
3The term MPS V is no longer used. The existence of MPS VIII (suspected glucosamine 6-sulfatase deficiency: MIM 253230) has not
been confirmed. At least one case of hyaluronidase deficiency (MPS IX; MIM 601492) has been reported.
amounts of GAGs and assays of suspected enzymes in
The term “mucolipidosis” was introduced to de-
white cells, fibroblasts, or sometimes in serum. In cer-
note diseases that combined features common to both
tain cases, a tissue biopsy is performed and the GAG
mucopolysaccharidoses and sphingolipidoses (Chapter
that has accumulated can be determined by elec-
24). Three mucolipidoses are listed in Table 48-7. In
trophoresis. DNA tests are increasingly available. Pre-
sialidosis (mucolipidosis I, ML-I), various oligosaccha-
natal diagnosis can be made using amniotic cells or
rides derived from glycoproteins and certain ganglio-
chorionic villus biopsy.
sides can accumulate in tissues. I-cell disease (ML-II)
THE EXTRACELLULAR MATRIX
/
547
TGF-β, modulating their effects on cells. In addition,
Mutation(s) in a gene encoding a lysosomal hydrolase
some of them interact with certain adhesive proteins
involved in the degradation of one or more GAGs
such as fibronectin and laminin (see above), also found
in the matrix. The GAGs present in the proteoglycans
Defective lysosomal hydrolase
are polyanions and hence bind polycations and cations
such as Na+ and K+. This latter ability attracts water by
Accumulation of substrate in various tissues, including
osmotic pressure into the extracellular matrix and con-
liver, spleen, bone, skin, and central nervous system
tributes to its turgor. GAGs also gel at relatively low
concentrations. Because of the long extended nature of
Figure 48-10. Simplified scheme of causation of a
the polysaccharide chains of GAGs and their ability to
mucopolysaccharidosis, such as Hurler syndrome (MIM
gel, the proteoglycans can act as sieves, restricting the
252800), in which the affected enzyme is α-L-iduroni-
passage of large macromolecules into the ECM but al-
dase. Marked accumulation of the GAGs in the tissues
lowing relatively free diffusion of small molecules.
mentioned in the figure could cause hepatomegaly,
Again, because of their extended structures and the
splenomegaly, disturbances of growth, coarse facies,
huge macromolecular aggregates they often form, they
occupy a large volume of the matrix relative to proteins.
and mental retardation, respectively.
A. SOME FUNCTIONS OF SPECIFIC
GAGS & PROTEOGLYCANS
Hyaluronic acid is especially high in concentration in
and pseudo-Hurler polydystrophy (ML-III) are de-
embryonic tissues and is thought to play an important
scribed in Chapter 47. The term “mucolipidosis” is re-
role in permitting cell migration during morphogenesis
tained because it is still in relatively widespread clinical
and wound repair. Its ability to attract water into the
usage, but it is not appropriate for these two latter dis-
extracellular matrix and thereby “loosen it up” may be
eases since the mechanism of their causation involves
important in this regard. The high concentrations of
mislocation of certain lysosomal enzymes. Genetic de-
hyaluronic acid and chondroitin sulfates present in car-
fects of the catabolism of the oligosaccharide chains of
tilage contribute to its compressibility (see below).
glycoproteins (eg, mannosidosis, fucosidosis) are also
Chondroitin sulfates are located at sites of calcifica-
described in Chapter 47. Most of these defects are char-
tion in endochondral bone and are also found in carti-
acterized by increased excretion of various fragments of
lage. They are also located inside certain neurons and
glycoproteins in the urine, which accumulate because
may provide an endoskeletal structure, helping to
of the metabolic block, as in the case of the mucolipi-
maintain their shape.
doses.
Both keratan sulfate I and dermatan sulfate are
Hyaluronidase is one important enzyme involved
present in the cornea. They lie between collagen fibrils
in the catabolism of both hyaluronic acid and chondro-
and play a critical role in corneal transparency. Changes
itin sulfate. It is a widely distributed endoglycosidase
in proteoglycan composition found in corneal scars dis-
that cleaves hexosaminidic linkages. From hyaluronic
appear when the cornea heals. The presence of der-
acid, the enzyme will generate a tetrasaccharide with
matan sulfate in the sclera may also play a role in main-
the structure
(GlcUA-β-1,3-GlcNAc-β-1,4)2, which
taining the overall shape of the eye. Keratan sulfate I is
can be degraded further by a β-glucuronidase and β-N-
also present in cartilage.
acetylhexosaminidase. Surprisingly, only one case of an
Heparin is an important anticoagulant. It binds
apparent genetic deficiency of this enzyme appears to
with factors IX and XI, but its most important interac-
have been reported.
tion is with plasma antithrombin III (discussed in
Chapter 51). Heparin can also bind specifically to
Proteoglycans Have Numerous Functions
lipoprotein lipase present in capillary walls, causing a
As indicated above, proteoglycans are remarkably com-
release of this enzyme into the circulation.
plex molecules and are found in every tissue of the
Certain proteoglycans (eg, heparan sulfate) are as-
body, mainly in the ECM or “ground substance.”
sociated with the plasma membrane of cells, with their
There they are associated with each other and also with
core proteins actually spanning that membrane. In it
the other major structural components of the matrix,
they may act as receptors and may also participate in
collagen and elastin, in quite specific manners. Some
the mediation of cell growth and cell-cell communica-
proteoglycans bind to collagen and others to elastin.
tion. The attachment of cells to their substratum in cul-
These interactions are important in determining the
ture is mediated at least in part by heparan sulfate. This
structural organization of the matrix. Some proteogly-
proteoglycan is also found in the basement membrane
cans (eg, decorin) can also bind growth factors such as
of the kidney along with type IV collagen and laminin
548
/
CHAPTER 48
(see above), where it plays a major role in determining
In various types of arthritis, proteoglycans may act
the charge selectiveness of glomerular filtration.
as autoantigens, thus contributing to the pathologic
Proteoglycans are also found in intracellular loca-
features of these conditions. The amount of chon-
tions such as the nucleus; their function in this or-
droitin sulfate in cartilage diminishes with age, whereas
ganelle has not been elucidated. They are present in
the amounts of keratan sulfate and hyaluronic acid in-
some storage or secretory granules, such as the chromaf-
crease. These changes may contribute to the develop-
fin granules of the adrenal medulla. It has been postu-
ment of osteoarthritis. Changes in the amounts of cer-
lated that they play a role in release of the contents of
such granules. The various functions of GAGs are sum-
marized in Table 48-8.
B. ASSOCIATIONS WITH MAJOR DISEASES
Table 48-9. The principal proteins found
& WITH AGING
in bone.1
Hyaluronic acid may be important in permitting
tumor cells to migrate through the ECM. Tumor cells
Proteins
Comments
can induce fibroblasts to synthesize greatly increased
Collagens
amounts of this GAG, thereby perhaps facilitating their
Collagen type I
Approximately 90% of total bone
own spread. Some tumor cells have less heparan sulfate
protein. Composed of two α1(I)
at their surfaces, and this may play a role in the lack of
and one α2(I) chains.
adhesiveness that these cells display.
Collagen type V
Minor component.
The intima of the arterial wall contains hyaluronic
acid and chondroitin sulfate, dermatan sulfate, and he-
Noncollagen proteins
paran sulfate proteoglycans. Of these proteoglycans,
Plasma proteins
Mixture of various plasma proteins.
dermatan sulfate binds plasma low-density lipopro-
Proteoglycans2
teins. In addition, dermatan sulfate appears to be the
CS-PG I (biglycan)
Contains two GAG chains; found in
major GAG synthesized by arterial smooth muscle
other tissues.
cells. Because it is these cells that proliferate in athero-
CS-PG II (decorin)
Contains one GAG chain; found in
sclerotic lesions in arteries, dermatan sulfate may play
other tissues.
an important role in development of the atheroscle-
rotic plaque.
CS-PG III
Bone-specific.
Bone SPARC3 protein
Not bone-specific.
(osteonectin)
Table 48-8. Some functions of
Osteocalcin (bone Gla
Contains γ-carboxyglutamate
glycosaminoglycans and proteoglycans.1
protein)
residues that bind to hydroxyap-
atite. Bone-specific.
• Act as structural components of the ECM
Osteopontin
Not bone-specific. Glycosylated
• Have specific interactions with collagen, elastin, fibronectin,
and phosphorylated.
laminin, and other proteins such as growth factors
• As polyanions, bind polycations and cations
Bone sialoprotein
Bone-specific. Heavily glycosylated,
• Contribute to the characteristic turgor of various tissues
and sulfated on tyrosine.
• Act as sieves in the ECM
Bone morphogenetic
A family (eight or more) of secreted
• Facilitate cell migration (HA)
proteins (BMPs)
proteins with a variety of actions
• Have role in compressibility of cartilage in weight-bearing
on bone; many induce ectopic
(HA, CS)
bone growth.
• Play role in corneal transparency (KS I and DS)
• Have structural role in sclera (DS)
1Various functions have been ascribed to the noncollagen
• Act as anticoagulant (heparin)
proteins, including roles in mineralization; however, most of
them are still speculative. It is considered unlikely that the
• Are components of plasma membranes, where they may
noncollagen proteins that are not bone-specific play a key
act as receptors and participate in cell adhesion and cell-cell
role in mineralization. A number of other proteins are also
interactions (eg, HS)
present in bone, including a tyrosine-rich acidic matrix pro-
• Determine charge-selectiveness of renal glomerulus (HS)
tein (TRAMP), some growth factors (eg, TGFβ), and enzymes
• Are components of synaptic and other vesicles (eg, HS)
involved in collagen synthesis (eg, lysyl oxidase).
1ECM, extracellular matrix; HA, hyaluronic acid; CS, chondroitin
2CS-PG, chondroitin sulfate-proteoglycan; these are similar to
sulfate; KS I, keratan sulfate I; DS, dermatan sulfate; HS, heparan
the dermatan sulfate PGs (DS-PGs) of cartilage (Table 48-11).
sulfate.
3SPARC, secreted protein acidic and rich in cysteine.
THE EXTRACELLULAR MATRIX
/
549
Osteoclast
Mesenchyme
Newly formed matrix (osteoid)
Osteoblast
Osteocyte
Bone matrix
Figure 48-11. Schematic illustration of the major cells present in membranous
bone. Osteoblasts (lighter color) are synthesizing type I collagen, which forms a matrix
that traps cells. As this occurs, osteoblasts gradually differentiate to become osteo-
cytes. (Reproduced, with permission, from Junqueira LC, Carneiro J: Basic Histology: Text &
Atlas, 10th ed. McGraw-Hill, 2003.)
tain GAGs in the skin are also observed with aging and
bone (Chapter 45). Hydroxyapatite confers on bone
help to account for the characteristic changes noted in
the strength and resilience required by its physiologic
this organ in the elderly.
roles.
An exciting new phase in proteoglycan research is
Bone is a dynamic structure that undergoes continu-
opening up with the findings that mutations that affect
ing cycles of remodeling, consisting of resorption fol-
individual proteoglycans or the enzymes needed for
lowed by deposition of new bone tissue. This remodel-
their synthesis alter the regulation of specific signaling
ing permits bone to adapt to both physical
(eg,
pathways in drosophila and Caenorhabditis elegans, thus
increases in weight-bearing) and hormonal signals.
affecting development; it already seems likely that simi-
The major cell types involved in bone resorption
lar effects exist in mice and humans.
and deposition are osteoclasts and osteoblasts (Figure
48-11). The former are associated with resorption and
the latter with deposition of bone. Osteocytes are de-
scended from osteoblasts; they also appear to be in-
BONE IS A MINERALIZED
volved in maintenance of bone matrix but will not be
CONNECTIVE TISSUE
discussed further here.
Bone contains both organic and inorganic material.
Osteoclasts are multinucleated cells derived from
The organic matter is mainly protein. The principal
pluripotent hematopoietic stem cells. Osteoclasts pos-
proteins of bone are listed in Table 48-9; type I colla-
sess an apical membrane domain, exhibiting a ruffled
gen is the major protein, comprising 90-95% of the
border that plays a key role in bone resorption (Figure
organic material. Type V collagen is also present in
48-12). A proton-translocating ATPase expels protons
small amounts, as are a number of noncollagen pro-
across the ruffled border into the resorption area, which
teins, some of which are relatively specific to bone.
is the microenvironment of low pH shown in the fig-
The inorganic or mineral component is mainly crys-
ure. This lowers the local pH to 4.0 or less, thus in-
talline
hydroxyapatite—Ca10(PO4)6(OH)2—along
creasing the solubility of hydroxyapatite and allowing
with sodium, magnesium, carbonate, and fluoride; ap-
demineralization to occur. Lysosomal acid proteases are
proximately 99% of the body’s calcium is contained in
released that digest the now accessible matrix proteins.
550
/
CHAPTER 48
Blood capillary
Nucleus
Osteoclast
Golgi
Nucleus
Lysosomes
-
CO2 + H2O
H+
+ HCO3
Section of
circumferential
clear zone
Ruffled
border
Microenvironment of low pH
Bone matrix
and lysosomal enzymes
Figure 48-12. Schematic illustration of some aspects of the role of the osteoclast in
bone resorption. Lysosomal enzymes and hydrogen ions are released into the confined
microenvironment created by the attachment between bone matrix and the peripheral
clear zone of the osteoclast. The acidification of this confined space facilitates the dis-
solution of calcium phosphate from bone and is the optimal pH for the activity of lyso-
somal hydrolases. Bone matrix is thus removed, and the products of bone resorption
are taken up into the cytoplasm of the osteoclast, probably digested further, and trans-
ferred into capillaries. The chemical equation shown in the figure refers to the action of
carbonic anhydrase II, described in the text. (Reproduced, with permission, from Jun-
queira LC, Carneiro J: Basic Histology: Text & Atlas, 10th ed. McGraw-Hill, 2003.)
Osteoblasts—mononuclear cells derived from pluripo-
Recent interest has focused on acidic phosphoproteins,
tent mesenchymal precursors—synthesize most of the
such as bone sialoprotein, acting as sites of nucleation.
proteins found in bone (Table 48-9) as well as various
These proteins contain motifs (eg, poly-Asp and poly-
growth factors and cytokines. They are responsible for
Glu stretches) that bind calcium and may provide an
the deposition of new bone matrix (osteoid) and its
initial scaffold for mineralization. Some macromole-
subsequent mineralization. Osteoblasts control miner-
cules, such as certain proteoglycans and glycoproteins,
alization by regulating the passage of calcium and phos-
can also act as inhibitors of nucleation.
phate ions across their surface membranes. The latter
It is estimated that approximately 4% of compact
contain alkaline phosphatase, which is used to generate
bone is renewed annually in the typical healthy adult,
phosphate ions from organic phosphates. The mecha-
whereas approximately 20% of trabecular bone is re-
nisms involved in mineralization are not fully under-
placed.
stood, but several factors have been implicated. Alkaline
Many factors are involved in the regulation of bone
phosphatase contributes to mineralization but in itself
metabolism, only a few of which will be mentioned
is not sufficient. Small vesicles (matrix vesicles) contain-
here. Some stimulate osteoblasts (eg, parathyroid hor-
ing calcium and phosphate have been described at sites
mone and 1,25-dihydroxycholecalciferol) and others
of mineralization, but their role is not clear. Type I col-
inhibit them (eg, corticosteroids). Parathyroid hormone
lagen appears to be necessary, with mineralization being
and 1,25-dihydroxycholecalciferol also stimulate osteo-
first evident in the gaps between successive molecules.
clasts, whereas calcitonin and estrogens inhibit them.
THE EXTRACELLULAR MATRIX
/
551
Table 48-10. Some metabolic and genetic
MANY METABOLIC & GENETIC
diseases affecting bone and cartilage.
DISORDERS INVOLVE BONE
A number of the more important examples of meta-
Disease
Comments
bolic and genetic disorders that affect bone are listed in
Dwarfism
Often due to a deficiency of growth
Table 48-10.
hormone, but has many other causes.
Osteogenesis imperfecta (brittle bones) is charac-
terized by abnormal fragility of bones. The scleras are
Rickets
Due to a deficiency of vitamin D
often abnormally thin and translucent and may appear
during childhood.
blue owing to a deficiency of connective tissue. Four
Osteomalacia
Due to a deficiency of vitamin D
types of this condition (mild, extensive, severe, and
during adulthood.
variable) have been recognized, of which the extensive
Hyperparathyroidism
Excess parathormone causes bone
type occurring in the newborn is the most ominous.
resorption.
Affected infants may be born with multiple fractures
and not survive. Over 90% of patients with osteogene-
Osteogenesis
Due to a variety of mutations in the
sis imperfecta have mutations in the COL1A1
and
imperfecta (eg,
COL1A1 and COL1A2 genes affecting
COL1A2
genes, encoding proα1(I) and proα2(I)
MIM 166200)
the synthesis and structure of type I
chains, respectively. Over 100 mutations in these two
collagen.
genes have been documented and include partial gene
Osteoporosis
Commonly postmenopausal or in
deletions and duplications. Other mutations affect
other cases is more gradual and re-
RNA splicing, and the most frequent type results in the
lated to age; a small number of cases
replacement of glycine by another bulkier amino acid,
are due to mutations in the COL1A1
affecting formation of the triple helix. In general, these
and COL1A2 genes and possibly in the
mutations result in decreased expression of collagen or
vitamin D receptor gene (MIM 166710)
Osteoarthritis
A small number of cases are due to
mutations in the COL1A genes.
Table 48-11. The principal proteins found
Several chondro-
Due to mutations in COL2A1 genes.
in cartilage.
dysplasias
Pfeiffer syndrome1
Mutations in the gene encoding fi-
Proteins
Comments
(MIM 100600)
broblast growth receptor 1 (FGFR1).
Collagen proteins
Jackson-Weiss
Mutations in the gene encoding
Collagen type II
90-98% of total articular cartilage
(MIM 123150)
FGFR2.
collagen. Composed of three
and Crouzon
α1(II) chains.
(MIM 123500)
Collagens V, VI, IX,
Type IX cross-links to type II colla-
syndromes1
X, XI
gen. Type XI may help control di-
Achondroplasia
Mutations in the gene encoding
ameter of type II fibrils.
(MIM 100800)
FGFR3.
Noncollagen proteins
and thanatophoric
Proteoglycans
dysplasia2
Aggrecan
The major proteoglycan of cartilage.
(MIM 187600)
Large non-
Found in some types of cartilage.
1The Pfeiffer, Jackson-Weiss, and Crouzon syndromes are cran-
aggregating
iosynostosis syndromes; craniosynostosis is a term signifying pre-
proteoglycan
mature fusion of sutures in the skull.
DS-PG I (biglycan)1
Similar to CS-PG I of bone.
2Thanatophoric (Gk thanatos “death” + phoros “bearing”) dyspla-
DS-PG II (decorin)
Similar to CS-PG II of bone.
sia is the most common neonatal lethal skeletal dysplasia, dis-
Chondronectin
May play role in binding type II colla-
playing features similar to those of homozygous achondroplasia.
gen to surface of cartilage.
Anchorin C II
May bind type II collagen to surface
of chondrocyte.
1The core proteins of DS-PG I and DS-PG II are homologous to
those of CS-PG I and CS-PG II found in bone (Table 48-9). A possi-
ble explanation is that osteoblasts lack the epimerase required to
convert glucuronic acid to iduronic acid, the latter of which is
found in dermatan sulfate.
552
/
CHAPTER 48
in structurally abnormal proα chains that assemble into
ions are pumped across their ruffled borders
(see
abnormal fibrils, weakening the overall structure of
above). Thus, if CA II is deficient in activity in osteo-
bone. When one abnormal chain is present, it may in-
clasts, normal bone resorption does not occur, and os-
teract with two normal chains, but folding may be pre-
teopetrosis results. The mechanism of the cerebral calci-
vented, resulting in enzymatic degradation of all of the
fication is not clear, whereas the renal tubular acidosis
chains. This is called “procollagen suicide” and is an ex-
reflects deficient activity of CA II in the renal tubules.
ample of a dominant negative mutation, a result often
Osteoporosis is a generalized progressive reduction
seen when a protein consists of multiple different sub-
in bone tissue mass per unit volume causing skeletal
units.
weakness. The ratio of mineral to organic elements is
Osteopetrosis (marble bone disease), characterized
unchanged in the remaining normal bone. Fractures of
by increased bone density, is due to inability to resorb
various bones, such as the head of the femur, occur very
bone. One form occurs along with renal tubular acido-
easily and represent a huge burden to both the affected
sis and cerebral calcification. It is due to mutations in
patients and to the health care budget of society.
the gene (located on chromosome 8q22) encoding car-
Among other factors, estrogens and interleukins-1 and
bonic anhydrase II (CA II), one of four isozymes of car-
-6 appear to be intimately involved in the causation of
bonic anhydrase present in human tissues. The reaction
osteoporosis.
catalyzed by carbonic anhydrase is shown below:
THE MAJOR COMPONENTS OF
67 nm
CARTILAGE ARE TYPE II COLLAGEN
Fibril
& CERTAIN PROTEOGLYCANS
The principal proteins of hyaline cartilage (the major
Reaction II is spontaneous. In osteoclasts involved in
type of cartilage) are listed in Table 48-11. Type II colla-
bone resorption, CA II apparently provides protons to
gen is the principal protein (Figure 48-13), and a num-
neutralize the OH− ions left inside the cell when H+
ber of other minor types of collagen are also present. In
Hyaluronic acid
Type II collagen fibril
Hyaluronic acid
Link protein
Chondroitin sulfate
Proteoglycan
Core protein
Collagen (type II)
Figure 48-13. Schematic representation of the molecular organization in cartilage
matrix. Link proteins noncovalently bind the core protein (lighter color) of proteogly-
cans to the linear hyaluronic acid molecules (darker color). The chondroitin sulfate side
chains of the proteoglycan electrostatically bind to the collagen fibrils, forming a
cross-linked matrix. The oval outlines the area enlarged in the lower part of the figure.
(Reproduced, with permission, from Junqueira LC, Carneiro J: Basic Histology: Text & Atlas,
10th ed. McGraw-Hill, 2003.)
THE EXTRACELLULAR MATRIX
/
553
addition to these components, elastic cartilage contains
degrade collagen and the other proteins found in carti-
elastin and fibroelastic cartilage contains type I collagen.
lage. Interleukin-1 (IL-1) and tumor necrosis factor α
Cartilage contains a number of proteoglycans, which
(TNFα) appear to stimulate the production of such
play an important role in its compressibility. Aggrecan
proteases, whereas transforming growth factor β
(about 2 × 103 kDa) is the major proteoglycan. As shown
(TGFβ) and insulin-like growth factor 1 (IGF-I) gener-
in Figure 48-14, it has a very complex structure, con-
ally exert an anabolic influence on cartilage.
taining several GAGs (hyaluronic acid, chondroitin sul-
fate, and keratan sulfate) and both link and core proteins.
THE MOLECULAR BASES OF THE
The core protein contains three domains: A, B, and C.
The hyaluronic acid binds noncovalently to domain A of
CHONDRODYSPLASIAS INCLUDE
the core protein as well as to the link protein, which sta-
MUTATIONS IN GENES ENCODING
bilizes the hyaluronate-core protein interactions. The
TYPE II COLLAGEN & FIBROBLAST
keratan sulfate chains are located in domain B, whereas
GROWTH FACTOR RECEPTORS
the chondroitin sulfate chains are located in domain C;
both of these types of GAGs are bound covalently to the
Chondrodysplasias are a mixed group of hereditary dis-
core protein. The core protein also contains both O- and
orders affecting cartilage. They are manifested by short-
N-linked oligosaccharide chains.
limbed dwarfism and numerous skeletal deformities. A
The other proteoglycans found in cartilage have
number of them are due to a variety of mutations in the
simpler structures than aggrecan.
COL2A1 gene, leading to abnormal forms of type II
Chondronectin is involved in the attachment of
collagen. One example is Stickler syndrome, mani-
type II collagen to chondrocytes.
fested by degeneration of joint cartilage and of the vit-
Cartilage is an avascular tissue and obtains most of
reous body of the eye.
its nutrients from synovial fluid. It exhibits slow but
The best-known of the chondrodysplasias is achon-
continuous turnover. Various proteases (eg, collage-
droplasia, the commonest cause of short-limbed
nases and stromalysin) synthesized by chondrocytes can
dwarfism. Affected individuals have short limbs, nor-
Domain A
Domain B
Domain C
Hyaluronate-
binding
region
Core
N-linked
protein
oligosaccharide
Link
protein
Keratan
Chondroitin
O-linked
Hyaluronic acid
sulfate
sulfate
oligosaccharide
Figure 48-14. Schematic diagram of the aggrecan from bovine nasal cartilage. A
strand of hyaluronic acid is shown on the left. The core protein (about 210 kDa) has
three major domains. Domain A, at its amino terminal end, interacts with approxi-
mately five repeating disaccharides in hyaluronate. The link protein interacts with
both hyaluronate and domain A, stabilizing their interactions. Approximately 30 ker-
atan sulfate chains are attached, via GalNAc-Ser linkages, to domain B. Domain C
contains about 100 chondroitin sulfate chains attached via Gal-Gal-Xyl-Ser linkages
and about 40 O-linked oligosaccharide chains. One or more N-linked glycan chains
are also found near the carboxyl terminal of the core protein. (Reproduced, with per-
mission, from Moran LA et al: Biochemistry, 2nd ed. Neil Patterson Publishers, 1994.)
554
/
CHAPTER 48
mal trunk size, macrocephaly, and a variety of other
SUMMARY
skeletal abnormalities. The condition is often inherited
•
The major components of the ECM are the struc-
as an autosomal dominant trait, but many cases are due
tural proteins collagen, elastin, and fibrillin; a num-
to new mutations. The molecular basis of achondropla-
ber of specialized proteins
(eg, fibronectin and
sia is outlined in Figure 48-15. Achondroplasia is not a
laminin); and various proteoglycans.
collagen disorder but is due to mutations in the gene
encoding fibroblast growth factor receptor
3
•
Collagen is the most abundant protein in the animal
(FGFR3). Fibroblast growth factors are a family of at
kingdom; approximately 19 types have been isolated.
least nine proteins that affect the growth and differenti-
All collagens contain greater or lesser stretches of
ation of cells of mesenchymal and neuroectodermal ori-
triple helix and the repeating structure (Gly-X-Y)n.
gin. Their receptors are transmembrane proteins and
•
The biosynthesis of collagen is complex, featuring
form a subgroup of the family of receptor tyrosine ki-
many posttranslational events, including hydroxyla-
nases. FGFR3 is one member of this subgroup and me-
tion of proline and lysine.
diates the actions of FGF3 on cartilage. In almost all
•
Diseases associated with impaired synthesis of colla-
cases of achondroplasia that have been investigated, the
gen include scurvy, osteogenesis imperfecta, Ehlers-
mutations were found to involve nucleotide 1138 and
Danlos syndrome (many types), and Menkes disease.
resulted in substitution of arginine for glycine (residue
•
Elastin confers extensibility and elastic recoil on tis-
number 380) in the transmembrane domain of the pro-
sues. Elastin lacks hydroxylysine, Gly-X-Y sequences,
tein, rendering it inactive. No such mutation was found
triple helical structure, and sugars but contains
in unaffected individuals. As indicated in Table 48-10,
desmosine and isodesmosine cross-links not found in
other skeletal dysplasias (including certain craniosynos-
collagen.
tosis syndromes) are also due to mutations in genes en-
•
Fibrillin is located in microfibrils. Mutations in the
coding FGF receptors. Another type of skeletal dyspla-
gene for fibrillin cause Marfan syndrome.
sia (diastrophic dysplasia) has been found to be due to
•
The glycosaminoglycans (GAGs) are made up of re-
mutation in a sulfate transporter. Thus, thanks to re-
combinant DNA technology, a new era in understand-
peating disaccharides containing a uronic acid (glu-
curonic or iduronic) or hexose (galactose) and a hex-
ing of skeletal dysplasias has begun.
osamine (galactosamine or glucosamine). Sulfate is
also frequently present.
•
The major GAGs are hyaluronic acid, chondroitin
4- and 6-sulfates, keratan sulfates I and II, heparin,
Mutations of nucleotide 1138 in the gene
heparan sulfate, and dermatan sulfate.
encoding FGFR3 on chromosome 4
•
The GAGs are synthesized by the sequential actions
of a battery of specific enzymes (glycosyltransferases,
Replacement in FGFR3 of Gly (codon 380) by Arg
epimerases, sulfotransferases, etc) and are degraded
by the sequential action of lysosomal hydrolases. Ge-
Defective function of FGFR3
netic deficiencies of the latter result in mucopolysac-
charidoses (eg, Hurler syndrome).
Abnormal development and growth of cartilage
•
GAGs occur in tissues bound to various proteins
leading to short-limbed dwarfism and other features
(linker proteins and core proteins), constituting pro-
teoglycans. These structures are often of very high
Figure 48-15. Simplified scheme of the causation of
molecular weight and serve many functions in tis-
achondroplasia (MIM 100800). In most cases studied so
sues.
far, the mutation has been a G to A transition at nu-
•
Many components of the ECM bind to proteins of
cleotide 1138. In a few cases, the mutation was a G to C
the cell surface named integrins; this constitutes one
transversion at the same nucleotide. This particular nu-
pathway by which the exteriors of cells can commu-
cleotide is a real “hot spot” for mutation. Both muta-
nicate with their interiors.
tions result in replacement of a Gly residue by an Arg
•
Bone and cartilage are specialized forms of the ECM.
residue in the transmembrane segment of the receptor.
Collagen I and hydroxyapatite are the major con-
A few cases involving replacement of Gly by Cys at
stituents of bone. Collagen II and certain proteogly-
codon 375 have also been reported.
cans are major constituents of cartilage.
THE EXTRACELLULAR MATRIX
/
555
• The molecular causes of a number of heritable dis-
Prockop DJ, Kivirikko KI: Collagens: molecular biology, diseases,
and potential therapy. Annu Rev Biochem 1995;64:403.
eases of bone (eg, osteogenesis imperfecta) and of car-
Pyeritz RE: Ehlers-Danlos syndrome. N Engl J Med 2000;342:730.
tilage (eg, the chondrodystrophies) are being revealed
by the application of recombinant DNA technology.
Sage E: Regulation of interactions between cells and extracellular
matrix: a command performance on several stages. J Clin In-
vest 2001;107:781. (This article introduces a series of six arti-
REFERENCES
cles on cell-matrix interaction. The topics covered are cell
adhesion and de-adhesion, thrombospondins, syndecans,
Bandtlow CE, Zimmermann DR: Proteoglycans in the developing
SPARC, osteopontin, and Ehlers-Danlos syndrome. All of
brain: new conceptual insights for old proteins. Physiol Rev
2000;80:1267.
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
Bikle DD: Biochemical markers in the assessment of bone diseases.
herited Disease, 8th ed. McGraw-Hill, 2001 (This compre-
Am J Med 1997;103:427.
hensive four-volume text contains chapters on disorders of
Burke D et al: Fibroblast growth factor receptors: lessons from the
collagen biosynthesis and structure, Marfan syndrome, the
genes. Trends Biochem Sci 1998;23:59.
mucopolysaccharidoses, achondroplasia, Alport syndrome,
Compston JE: Sex steroids and bone. Physiol Rev 2001;81:419.
and craniosynostosis syndromes.)
Fuller GM, Shields D: Molecular Basis of Medical Cell Biology. Ap-
Selleck SB: Genetic dissection of proteoglycan function in
pleton & Lange, 1998.
Drosophila and C. elegans. Semin Cell Dev Biol 2001;12:127.
Herman T, Horvitz HR: Three proteins involved in Caenorhabditis
elegans vulval invagination are similar to components of a gly-
cosylation pathway. Proc Natl Acad Sci U S A 1999;96:974.
Muscle & the Cytoskeleton
49
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
shall discuss aspects of the three types of muscle found
in vertebrates: skeletal, cardiac, and smooth. Both
Proteins play an important role in movement at both
skeletal and cardiac muscle appear striated upon micro-
the organ (eg, skeletal muscle, heart, and gut) and cellu-
scopic observation; smooth muscle is nonstriated. Al-
lar levels. In this chapter, the roles of specific proteins
though skeletal muscle is under voluntary nervous con-
and certain other key molecules (eg, Ca2+) in muscular
trol, the control of both cardiac and smooth muscle is
contraction are described. A brief coverage of cyto-
involuntary.
skeletal proteins is also presented.
Knowledge of the molecular bases of a number of
The Sarcoplasm of Muscle Cells
conditions that affect muscle has advanced greatly in re-
cent years. Understanding of the molecular basis of
Contains ATP, Phosphocreatine,
Duchenne-type muscular dystrophy was greatly en-
& Glycolytic Enzymes
hanced when it was found that it was due to mutations
Striated muscle is composed of multinucleated muscle
in the gene encoding dystrophin. Significant progress
fiber cells surrounded by an electrically excitable plasma
has also been made in understanding the molecular
membrane, the sarcolemma. An individual muscle
basis of malignant hyperthermia, a serious complica-
fiber cell, which may extend the entire length of the
tion for some patients undergoing certain types of anes-
muscle, contains a bundle of many myofibrils arranged
thesia. Heart failure is a very common medical condi-
in parallel, embedded in intracellular fluid termed sar-
tion, with a variety of causes; its rational therapy
coplasm. Within this fluid is contained glycogen, the
requires understanding of the biochemistry of heart
high-energy compounds ATP and phosphocreatine,
muscle. One group of conditions that cause heart fail-
and the enzymes of glycolysis.
ure are the cardiomyopathies, some of which are ge-
netically determined. Nitric oxide
(NO) has been
The Sarcomere Is the Functional
found to be a major regulator of smooth muscle tone.
Many widely used vasodilators—such as nitroglycerin,
Unit of Muscle
used in the treatment of angina pectoris—act by in-
An overall view of voluntary muscle at several levels of
creasing the formation of NO. Muscle, partly because
organization is presented in Figure 49-1.
of its mass, plays major roles in the overall metabolism
When the myofibril is examined by electron mi-
of the body.
croscopy, alternating dark and light bands (anisotropic
bands, meaning birefringent in polarized light; and
isotropic bands, meaning not altered by polarized light)
MUSCLE TRANSDUCES CHEMICAL
can be observed. These bands are thus referred to as A
and I bands, respectively. The central region of the A
ENERGY INTO MECHANICAL ENERGY
band (the H band) appears less dense than the rest of
Muscle is the major biochemical transducer (machine)
the band. The I band is bisected by a very dense and
that converts potential (chemical) energy into kinetic
narrow Z line (Figure 49-2).
(mechanical) energy. Muscle, the largest single tissue in
The sarcomere is defined as the region between two
the human body, makes up somewhat less than 25% of
Z lines (Figures 49-1 and 49-2) and is repeated along
body mass at birth, more than 40% in the young adult,
the axis of a fibril at distances of 1500-2300 nm de-
and somewhat less than 30% in the aged adult. We
pending upon the state of contraction.
556
MUSCLE & THE CYTOSKELETON
/
557
A
Muscle
B
Muscle fasciculus
C
20-100 µm
Muscle fiber
H
Z
A
I
band line
band band
D
1-2 µm
Myofibril
Z - Sarcomere - Z
Figure 49-1. The structure of voluntary muscle. The sarcomere is the region between the
Z lines. (Drawing by Sylvia Colard Keene. Reproduced, with permission, from Bloom W, Fawcett
DW: A Textbook of Histology, 10th ed. Saunders, 1975.)
The striated appearance of voluntary and cardiac
section), and each thick filament is surrounded sym-
muscle in light microscopic studies results from their
metrically by six thin filaments.
high degree of organization, in which most muscle fiber
The thick and thin filaments interact via cross-
cells are aligned so that their sarcomeres are in parallel
bridges that emerge at intervals of 14 nm along the
register (Figure 49-1).
thick filaments. As depicted in Figure 49-2, the cross-
bridges (drawn as arrowheads at each end of the myosin
filaments, but not shown extending fully across to the
Thick Filaments Contain Myosin;
thin filaments) have opposite polarities at the two ends
Thin Filaments Contain Actin,
of the thick filaments. The two poles of the thick fila-
Tropomyosin, & Troponin
ments are separated by a 150-nm segment (the M band,
When myofibrils are examined by electron microscopy,
not labeled in the figure) that is free of projections.
it appears that each one is constructed of two types of
longitudinal filaments. One type, the thick filament,
confined to the A band, contains chiefly the protein
The Sliding Filament Cross-Bridge
myosin. These filaments are about 16 nm in diameter
Model Is the Foundation on Which
and arranged in cross-section as a hexagonal array (Fig-
Current Thinking About Muscle
ure 49-2, center; right-hand cross-section).
Contraction Is Built
The thin filament (about 7 nm in diameter) lies in
the I band and extends into the A band but not into its
This model was proposed independently in the 1950s
H zone (Figure 49-2). Thin filaments contain the pro-
by Henry Huxley and Andrew Huxley and their col-
teins actin, tropomyosin, and troponin (Figure 49-3).
leagues. It was largely based on careful morphologic ob-
In the A band, the thin filaments are arranged around
servations on resting, extended, and contracting mus-
the thick (myosin) filament as a secondary hexagonal
cle. Basically, when muscle contracts, there is no change
array. Each thin filament lies symmetrically between
in the lengths of the thick and thin filaments, but the
three thick filaments (Figure 49-2, center; mid cross-
H zones and the I bands shorten (see legend to Fig-
558
/
CHAPTER 49
H band
A. Extended
I band
A band
Z line
2300 nm
α-Actinin
Actin filaments
6-nm diameter
Myosin filaments
16-nm diameter
Cross section:
B. Contracted
Thin
6-nm diameter
filament
Thick
16-nm diameter
filament
1500 nm
Figure 49-2. Arrangement of filaments in striated muscle. A: Extended. The positions of the
I, A, and H bands in the extended state are shown. The thin filaments partly overlap the ends of the
thick filaments, and the thin filaments are shown anchored in the Z lines (often called Z disks). In
the lower part of Figure 49-2A, “arrowheads,” pointing in opposite directions, are shown emanat-
ing from the myosin (thick) filaments. Four actin (thin) filaments are shown attached to two Z lines
via α-actinin. The central region of the three myosin filaments, free of arrowheads, is called the
M band (not labeled). Cross-sections through the M bands, through an area where myosin and
actin filaments overlap and through an area in which solely actin filaments are present, are shown.
B: Contracted. The actin filaments are seen to have slipped along the sides of the myosin fibers to-
ward each other. The lengths of the thick filaments (indicated by the A bands) and the thin fila-
ments (distance between Z lines and the adjacent edges of the H bands) have not changed. How-
ever, the lengths of the sarcomeres have been reduced (from 2300 nm to 1500 nm), and the
lengths of the H and I bands are also reduced because of the overlap between the thick and thin
filaments. These morphologic observations provided part of the basis for the sliding filament
model of muscle contraction.
MUSCLE & THE CYTOSKELETON
/
559
G-actin
F-actin
6-7 nm
Tropomyosin
Troponin
TpC
38.5 nm
TpI
TpT
35.5 nm
The assembled thin filament
Figure 49-3. Schematic representation of the thin filament, showing the spatial configuration of its three
major protein components: actin, myosin, and tropomyosin. The upper panel shows individual molecules of
G-actin. The middle panel shows actin monomers assembled into F-actin. Individual molecules of tropomyosin
(two strands wound around one another) and of troponin (made up of its three subunits) are also shown. The
lower panel shows the assembled thin filament, consisting of F-actin, tropomyosin, and the three subunits of
troponin (TpC, TpI, and TpT).
ure 49-2). Thus, the arrays of interdigitating filaments
ACTIN & MYOSIN ARE THE MAJOR
must slide past one another during contraction. Cross-
PROTEINS OF MUSCLE
bridges that link thick and thin filaments at certain
stages in the contraction cycle generate and sustain the
The mass of a muscle is made up of 75% water and
tension. The tension developed during muscle contrac-
more than 20% protein. The two major proteins are
tion is proportionate to the filament overlap and to the
actin and myosin.
number of cross-bridges. Each cross-bridge head is con-
Monomeric G-actin (43 kDa; G, globular) makes
nected to the thick filament via a flexible fibrous seg-
up 25% of muscle protein by weight. At physiologic
ment that can bend outward from the thick filament.
ionic strength and in the presence of Mg2+, G-actin
This flexible segment facilitates contact of the head
polymerizes noncovalently to form an insoluble double
with the thin filament when necessary but is also suffi-
helical filament called F-actin
(Figure
49-3). The
ciently pliant to be accommodated in the interfilament
F-actin fiber is 6-7 nm thick and has a pitch or repeat-
spacing.
ing structure every 35.5 nm.
560
/
CHAPTER 49
Myosins constitute a family of proteins, with at
light chain. Skeletal muscle myosin binds actin to form
least 15 members having been identified. The myosin
actomyosin (actin-myosin), and its intrinsic ATPase ac-
discussed in this chapter is myosin-II, and when myosin
tivity is markedly enhanced in this complex. Isoforms
is referred to in this text, it is this species that is meant
of myosin exist whose amounts can vary in different
unless otherwise indicated. Myosin-I is a monomeric
anatomic, physiologic, and pathologic situations.
species that binds to cell membranes. It may serve as a
The structures of actin and of the head of myosin
linkage between microfilaments and the cell membrane
have been determined by x-ray crystallography; these
in certain locations.
studies have confirmed a number of earlier findings
Myosin contributes
55% of muscle protein by
concerning their structures and have also given rise to
weight and forms the thick filaments. It is an asymmet-
much new information.
ric hexamer with a molecular mass of approximately
460 kDa. Myosin has a fibrous tail consisting of two in-
Limited Digestion of Myosin With
tertwined helices. Each helix has a globular head por-
Proteases Has Helped to Elucidate
tion attached at one end (Figure 49-4). The hexamer
Its Structure & Function
consists of one pair of heavy (H) chains each of ap-
proximately 200 kDA molecular mass, and two pairs of
When myosin is digested with trypsin, two myosin
light (L) chains each with a molecular mass of approxi-
fragments (meromyosins) are generated. Light mero-
mately 20 kDa. The L chains differ, one being called
myosin (LMM) consists of aggregated, insoluble α-he-
the essential light chain and the other the regulatory
lical fibers from the tail of myosin (Figure 49-4). LMM
L
L
L
L
L
L
G
G
G
G
HMM S-1
G
G
9 nm
L
L
L
L
PAPAIN
L
L
HMM
TRYPSIN
HMM S-2
134 nm
LMM
85 nm
Figure 49-4. Diagram of a myosin molecule showing the two intertwined α-helices (fibrous portion), the
globular region or head (G), the light chains (L), and the effects of proteolytic cleavage by trypsin and papain.
The globular region (myosin head) contains an actin-binding site and an L chain-binding site and also attaches
to the remainder of the myosin molecule.
MUSCLE & THE CYTOSKELETON
/
561
exhibits no ATPase activity and does not bind to
in the power stroke, which drives movement of actin
F-actin.
filaments past myosin filaments. The energy for the
Heavy meromyosin (HMM; molecular mass about
power stroke is ultimately supplied by ATP, which is
340 kDa) is a soluble protein that has both a fibrous
hydrolyzed to ADP and Pi. However, the power stroke
portion and a globular portion (Figure 49-4). It ex-
itself occurs as a result of conformational changes in
hibits ATPase activity and binds to F-actin. Digestion
the myosin head when ADP leaves it.
of HMM with papain generates two subfragments, S-1
The major biochemical events occurring during one
and S-2. The S-2 fragment is fibrous in character, has
cycle of muscle contraction and relaxation can be repre-
no ATPase activity, and does not bind to F-actin.
sented in the five steps shown in Figure 49-6:
S-1 (molecular mass approximately 115 kDa) does
exhibit ATPase activity, binds L chains, and in the ab-
(1) In the relaxation phase of muscle contraction,
sence of ATP will bind to and decorate actin with “ar-
the S-1 head of myosin hydrolyzes ATP to ADP and Pi,
rowheads” (Figure 49-5). Both S-1 and HMM exhibit
but these products remain bound. The resultant ADP-
ATPase activity, which is accelerated 100- to 200-fold by
Pi-myosin complex has been energized and is in a so-
complexing with F-actin. As discussed below, F-actin
called high-energy conformation.
greatly enhances the rate at which myosin ATPase re-
(2) When contraction of muscle is stimulated (via
leases its products, ADP and Pi. Thus, although F-actin
events involving Ca2+, troponin, tropomyosin, and
does not affect the hydrolysis step per se, its ability to
actin, which are described below), actin becomes acces-
promote release of the products produced by the ATPase
sible and the S-1 head of myosin finds it, binds it, and
activity greatly accelerates the overall rate of catalysis.
forms the actin-myosin-ADP-Pi complex indicated.
(3) Formation of this complex promotes the re-
CHANGES IN THE CONFORMATION
lease of Pi, which initiates the power stroke. This is fol-
lowed by release of ADP and is accompanied by a large
OF THE HEAD OF MYOSIN DRIVE
conformational change in the head of myosin in rela-
MUSCLE CONTRACTION
tion to its tail (Figure 49-7), pulling actin about 10 nm
How can hydrolysis of ATP produce macroscopic
toward the center of the sarcomere. This is the power
movement? Muscle contraction essentially consists of
stroke. The myosin is now in a so-called low-energy
the cyclic attachment and detachment of the S-1 head of
state, indicated as actin-myosin.
myosin to the F-actin filaments. This process can also be
(4) Another molecule of ATP binds to the S-1 head,
referred to as the making and breaking of cross-bridges.
forming an actin-myosin-ATP complex.
The attachment of actin to myosin is followed by con-
(5) Myosin-ATP has a low affinity for actin, and
formational changes which are of particular importance
actin is thus released. This last step is a key compo-
in the S-1 head and are dependent upon which nu-
nent of relaxation and is dependent upon the binding
cleotide is present (ADP or ATP). These changes result
of ATP to the actin-myosin complex.
Actin
ATP-Myosin
H2O
5
1
Actin-Myosin
ATP
ADP-Pi-Myosin
ATP
4
Actin
Actin-Myosin
2
3
ADP
+
Actin-Myosin
Pi
ADP-Pi
Figure 49-6. The hydrolysis of ATP drives the cyclic
Figure 49-5. The decoration of actin filaments with
association and dissociation of actin and myosin in five
the S-1 fragments of myosin to form “arrowheads.”
reactions described in the text. (Modified from Stryer L:
(Courtesy of JA Spudich.)
Biochemistry, 2nd ed. Freeman, 1981.)
562
/
CHAPTER 49
ADP. The hinge regions of myosin (referred to as flexi-
ble points at each end of S-2 in the legend to Figure
1
49-7) permit the large range of movement of S-1 and
also allow S-1 to find actin filaments.
If intracellular levels of ATP drop (eg, after death),
Thick filament
ATP is not available to bind the S-1 head (step 4
LMM
above), actin does not dissociate, and relaxation (step 5)
2
S-2
S-1
does not occur. This is the explanation for rigor mor-
tis, the stiffening of the body that occurs after death.
Calculations have indicated that the efficiency of
Thin filament
contraction is about 50%; that of the internal combus-
tion engine is less than 20%.
3
Tropomyosin & the Troponin Complex
Present in Thin Filaments Perform Key
Figure 49-7. Representation of the active cross-
Functions in Striated Muscle
bridges between thick and thin filaments. This diagram
In striated muscle, there are two other proteins that are
was adapted by AF Huxley from HE Huxley: The
minor in terms of their mass but important in terms of
mechanism of muscular contraction. Science
their function. Tropomyosin is a fibrous molecule that
1969;164:1356. The latter proposed that the force in-
consists of two chains, alpha and beta, that attach to
volved in muscular contraction originates in a tendency
F-actin in the groove between its filaments (Figure 49-3).
for the myosin head (S-1) to rotate relative to the thin
Tropomyosin is present in all muscular and muscle-like
filament and is transmitted to the thick filament by the
structures. The troponin complex is unique to striated
S-2 portion of the myosin molecule acting as an inex-
muscle and consists of three polypeptides. Troponin T
tensible link. Flexible points at each end of S-2 permit
(TpT) binds to tropomyosin as well as to the other two
S-1 to rotate and allow for variations in the separation
troponin components. Troponin I (TpI) inhibits the
between filaments. The present figure is based on HE
F-actin-myosin interaction and also binds to the other
Huxley’s proposal but also incorporates elastic (the coils
components of troponin. Troponin C (TpC) is a cal-
in the S-2 portion) and stepwise-shortening elements
cium-binding polypeptide that is structurally and func-
(depicted here as four sites of interaction between the
tionally analogous to calmodulin, an important cal-
S-1 portion and the thin filament). (See Huxley AF, Sim-
cium-binding protein widely distributed in nature.
mons RM: Proposed mechanism of force generation in
Four molecules of calcium ion are bound per molecule
striated muscle. Nature [Lond] 1971;233:533.) The
of troponin C or calmodulin, and both molecules have
strengths of binding of the attached sites are higher in
a molecular mass of 17 kDa.
position 2 than in position 1 and higher in position 3
than position 2. The myosin head can be detached from
Ca2+ Plays a Central Role in Regulation
position 3 with the utilization of a molecule of ATP; this
of Muscle Contraction
is the predominant process during shortening. The
The contraction of muscles from all sources occurs by
myosin head is seen to vary in its position from about
the general mechanism described above. Muscles from
90° to about 45°, as indicated in the text. (S-1, myosin
different organisms and from different cells and tissues
head; S-2, portion of the myosin molecule; LMM, light
within the same organism may have different molecular
meromyosin) (see legend to Figure 49-4). (Reproduced
mechanisms responsible for the regulation of their con-
from Huxley AF: Muscular contraction. J Physiol 1974;
traction and relaxation. In all systems, Ca2+ plays a key
243:1. By kind permission of the author and the Journal of
regulatory role. There are two general mechanisms of
Physiology.)
regulation of muscle contraction: actin-based and
myosin-based. The former operates in skeletal and car-
diac muscle, the latter in smooth muscle.
Another cycle then commences with the hydrolysis
of ATP (step 1 of Figure 49-6), re-forming the high-
Actin-Based Regulation Occurs
energy conformation.
in Striated Muscle
Thus, hydrolysis of ATP is used to drive the cycle,
with the actual power stroke being the conformational
Actin-based regulation of muscle occurs in vertebrate
change in the S-1 head that occurs upon the release of
skeletal and cardiac muscles, both striated. In the gen-
MUSCLE & THE CYTOSKELETON
/
563
eral mechanism described above
(Figure
49-6), the
T tubule
only potentially limiting factor in the cycle of muscle
Sarcolemma
contraction might be ATP. The skeletal muscle system
Dihydropyridine
is inhibited at rest; this inhibition is relieved to activate
receptor
contraction. The inhibitor of striated muscle is the tro-
Ca2+ release
ponin system, which is bound to tropomyosin and
Calsequestrin
channel
F-actin in the thin filament (Figure 49-3). In striated
muscle, there is no control of contraction unless the
Ca2+
tropomyosin-troponin systems are present along with
Ca2+
the actin and myosin filaments. As described above,
Cister na
Ca2+
tropomyosin lies along the groove of F-actin, and
Ca2+
Cister na
the three components of troponin—TpT, TpI, and
TpC—are bound to the F-actin-tropomyosin complex.
Ca2+ ATPase
TpI prevents binding of the myosin head to its F-actin
Calsequestrin
Ca2+
attachment site either by altering the conformation of
F-actin via the tropomyosin molecules or by simply
Ca2+
rolling tropomyosin into a position that directly blocks
the sites on F-actin to which the myosin heads attach.
Either way prevents activation of the myosin ATPase
Sarcomere
that is mediated by binding of the myosin head to
F-actin. Hence, the TpI system blocks the contraction
Figure 49-8. Diagram of the relationships among
cycle at step 2 of Figure 49-6. This accounts for the in-
the sarcolemma (plasma membrane), a T tubule, and
hibited state of relaxed striated muscle.
two cisternae of the sarcoplasmic reticulum of skeletal
muscle (not to scale). The T tubule extends inward from
The Sarcoplasmic Reticulum
the sarcolemma. A wave of depolarization, initiated by
Regulates Intracellular Levels
a nerve impulse, is transmitted from the sarcolemma
in Skeletal Muscle
of Ca2+
down the T tubule. It is then conveyed to the Ca2+ re-
In the sarcoplasm of resting muscle, the concentration
lease channel (ryanodine receptor), perhaps by interac-
of Ca2+ is 10−8 to 10−7 mol/L. The resting state is
tion between it and the dihydropyridine receptor (slow
achieved because Ca2+ is pumped into the sarcoplasmic
Ca2+ voltage channel), which are shown in close prox-
reticulum through the action of an active transport sys-
imity. Release of Ca2+ from the Ca2+ release channel into
tem, called the Ca2+ ATPase (Figure 49-8), initiating
the cytosol initiates contraction. Subsequently, Ca2+ is
relaxation. The sarcoplasmic reticulum is a network of
pumped back into the cisternae of the sarcoplasmic
fine membranous sacs. Inside the sarcoplasmic reticu-
reticulum by the Ca2+ ATPase (Ca2+ pump) and stored
lum, Ca2+ is bound to a specific Ca2+-binding protein
there, in part bound to calsequestrin.
designated calsequestrin. The sarcomere is surrounded
by an excitable membrane (the T tubule system) com-
posed of transverse (T) channels closely associated with
the sarcoplasmic reticulum.
receptor, RYR1 and RYR2, the former being present in
When the sarcolemma is excited by a nerve impulse,
skeletal muscle and the latter in heart muscle and brain.
the signal is transmitted into the T tubule system and a
Ryanodine is a plant alkaloid that binds to RYR1 and
Ca2+ release channel in the nearby sarcoplasmic reticu-
RYR2 specifically and modulates their activities. The
lum opens, releasing Ca2+ from the sarcoplasmic reticu-
Ca2+ release channel is a homotetramer made up of four
lum into the sarcoplasm. The concentration of Ca2+ in
subunits of kDa 565. It has transmembrane sequences
the sarcoplasm rises rapidly to 10−5 mol/L. The Ca2+-
at its carboxyl terminal, and these probably form the
binding sites on TpC in the thin filament are quickly
Ca2+ channel. The remainder of the protein protrudes
occupied by Ca2+. The TpC-4Ca2+ interacts with TpI
into the cytosol, bridging the gap between the sar-
and TpT to alter their interaction with tropomyosin.
coplasmic reticulum and the transverse tubular mem-
Accordingly, tropomyosin moves out of the way or al-
brane. The channel is ligand-gated, Ca2+ and ATP
ters the conformation of F-actin so that the myosin
working synergistically in vitro, although how it oper-
head-ADP-Pi (Figure 49-6) can interact with F-actin to
ates in vivo is not clear. A possible sequence of events
start the contraction cycle.
leading to opening of the channel is shown in Figure
The Ca2+ release channel is also known as the ryan-
49-9. The channel lies very close to the dihydropyri-
odine receptor (RYR). There are two isoforms of this
dine receptor (DHPR; a voltage-gated slow K type
564
/
CHAPTER 49
Depolarization of nerve
Table 49-1 summarizes the overall events in con-
traction and relaxation of skeletal muscle.
Depolarization of skeletal muscle
Mutations in the Gene Encoding the Ca2+
Release Channel Are One Cause of Human
Depolarization of the transverse tubular membrane
Malignant Hyperthermia
Some genetically predisposed patients experience a se-
Charge movement of the slow Ca2+ voltage
vere reaction, designated malignant hyperthermia, on
channel (DHPR) of the transverse tubular membrane
exposure to certain anesthetics (eg, halothane) and de-
polarizing skeletal muscle relaxants
(eg, succinyl-
Opening of the Ca2+ release channel (RYR1)
choline). The reaction consists primarily of rigidity of
skeletal muscles, hypermetabolism, and high fever. A
Figure 49-9. Possible chain of events leading to
high cytosolic concentration of Ca2+ in skeletal mus-
opening of the Ca2+ release channel. As indicated in the
cle is a major factor in its causation. Unless malignant
text, the Ca2+ voltage channel and the Ca2+ release
hyperthermia is recognized and treated immediately,
channel have been shown to interact with each other in
patients may die acutely of ventricular fibrillation or
vitro via specific regions in their polypeptide chains.
survive to succumb subsequently from other serious
(DHPR, dihydropyridine receptor; RYR1, ryanodine re-
complications. Appropriate treatment is to stop the
ceptor 1.)
anesthetic and administer the drug dantrolene intra-
venously. Dantrolene is a skeletal muscle relaxant that
acts to inhibit release of Ca2+ from the sarcoplasmic
reticulum into the cytosol, thus preventing the increase
Ca2+ channel) of the transverse tubule system (Figure
of cytosolic Ca2+ found in malignant hyperthermia.
49-8). Experiments in vitro employing an affinity col-
umn chromatography approach have indicated that a
37-amino-acid stretch in RYR1 interacts with one spe-
cific loop of DHPR.
Table 49-1. Sequence of events in contraction
Relaxation occurs when sarcoplasmic Ca2+ falls
and relaxation of skeletal muscle.1
below 10−7 mol/L owing to its resequestration into the
sarcoplasmic reticulum by Ca2+ ATPase. TpC.4Ca2+
Steps in contraction
thus loses its Ca2+. Consequently, troponin, via interac-
(1) Discharge of motor neuron
tion with tropomyosin, inhibits further myosin head
(2) Release of transmitter (acetylcholine) at motor end-
and F-actin interaction, and in the presence of ATP the
plate
myosin head detaches from the F-actin.
(3) Binding of acetylcholine to nicotinic acetylcholine re-
Thus, Ca2+ controls skeletal muscle contraction and
ceptors
relaxation by an allosteric mechanism mediated by
(4) Increased Na+ and K+ conductance in endplate mem-
TpC, TpI, TpT, tropomyosin, and F-actin.
brane
A decrease in the concentration of ATP in the sar-
(5) Generation of endplate potential
coplasm (eg, by excessive usage during the cycle of con-
(6) Generation of action potential in muscle fibers
traction-relaxation or by diminished formation, such as
(7) Inward spread of depolarization along T tubules
might occur in ischemia) has two major effects: (1) The
(8) Release of Ca2+ from terminal cisterns of sarcoplasmic
Ca2+ ATPase (Ca2+ pump) in the sarcoplasmic reticu-
reticulum and diffusion to thick and thin filaments
lum ceases to maintain the low concentration of Ca2+
(9) Binding of Ca2+ to troponin C, uncovering myosin
in the sarcoplasm. Thus, the interaction of the myosin
binding sites of actin
heads with F-actin is promoted. (2) The ATP-depen-
(10) Formation of cross-linkages between actin and
dent detachment of myosin heads from F-actin cannot
myosin and sliding of thin on thick filaments, produc-
ing shortening
occur, and rigidity (contracture) sets in. The condition
Steps in relaxation
of rigor mortis, following death, is an extension of
(1) Ca2+ pumped back into sarcoplasmic reticulum
these events.
(2) Release of Ca2+ from troponin
Muscle contraction is a delicate dynamic balance of
(3) Cessation of interaction between actin and myosin
the attachment and detachment of myosin heads to
F-actin, subject to fine regulation via the nervous
1Reproduced, with permission, from Ganong WF: Review of Med-
system.
ical Physiology, 21st ed. McGraw-Hill, 2003.
MUSCLE & THE CYTOSKELETON
/
565
Malignant hyperthermia also occurs in swine. Sus-
Mutations in the RYR1 gene
ceptible animals homozygous for malignant hyperther-
mia respond to stress with a fatal reaction (porcine
Altered Ca2+ release channel protein (RYR1)
stress syndrome) similar to that exhibited by humans.
(eg, substitution of Cys for Arg615 )
If the reaction occurs prior to slaughter, it affects the
quality of the pork adversely, resulting in an inferior
product. Both events can result in considerable eco-
Mutated channel opens more easily and stays open
nomic losses for the swine industry.
longer, thus flooding the cytosol with Ca2+
The finding of a high level of cytosolic Ca2+ in mus-
cle in malignant hyperthermia suggested that the con-
High intracellular levels of Ca2+ stimulate sustained
dition might be caused by abnormalities of the Ca2+
muscle contraction (rigidity); high Ca2+ also stimulates
ATPase or of the Ca2+ release channel. No abnormali-
breakdown of glycogen, glycolysis, and aerobic
ties were detected in the former, but sequencing of
metabolism (resulting in excessive production of heat)
cDNAs for the latter protein proved insightful, particu-
larly in swine. All cDNAs from swine with malignant
Figure 49-10. Simplified scheme of the causation of
hyperthermia so far examined have shown a substitu-
malignant hyperthermia (MIM 145600). At least 17 dif-
tion of T for C1843, resulting in the substitution of
ferent point mutations have been detected in the RYR1
release channel. The muta-
Cys for Arg615 in the Ca2+
gene, some of which are associated with central core
tion affects the function of the channel in that it opens
disease (MIM 117000). It is estimated that at least 50%
more easily and remains open longer; the net result is
of families with members who have malignant hyper-
massive release of Ca2+ into the cytosol, ultimately caus-
thermia are linked to the RYR1 gene. Some individuals
ing sustained muscle contraction.
with mutations in the gene encoding DHPR have also
The picture is more complex in humans, since ma-
been detected; it is possible that mutations in other
lignant hyperthermia exhibits genetic heterogeneity.
genes for proteins involved in certain aspects of muscle
Members of a number of families who suffer from ma-
metabolism will also be found.
lignant hyperthermia have not shown genetic linkage
to the RYR1 gene. Some humans susceptible to malig-
nant hyperthermia have been found to exhibit the
same mutation found in swine, and others have a vari-
ety of point mutations at different loci in the RYR1
MUTATIONS IN THE GENE ENCODING
gene. Certain families with malignant hypertension
DYSTROPHIN CAUSE DUCHENNE
have been found to have mutations affecting the
MUSCULAR DYSTROPHY
DHPR. Figure 49-10 summarizes the probable chain
of events in malignant hyperthermia. The major
A number of additional proteins play various roles in
promise of these findings is that, once additional mu-
the structure and function of muscle. They include titin
tations are detected, it will be possible to screen, using
(the largest protein known), nebulin, α-actinin, desmin,
suitable DNA probes, for individuals at risk of devel-
dystrophin, and calcineurin. Some properties of these
oping malignant hyperthermia during anesthesia. Cur-
proteins are summarized in Table 49-2.
rent screening tests (eg, the in vitro caffeine-halothane
Dystrophin is of special interest. Mutations in the
test) are relatively unreliable. Affected individuals
gene encoding this protein have been shown to be the
could then be given alternative anesthetics, which
cause of Duchenne muscular dystrophy and the milder
would not endanger their lives. It should also be possi-
Becker muscular dystrophy (see Figure 49-11). They
ble, if desired, to eliminate malignant hyperthermia
are also implicated in some cases of dilated cardiomy-
from swine populations using suitable breeding prac-
opathy (see below). The gene encoding dystrophin is
tices.
the largest gene known (≈ 2300 kb) and is situated on
Another condition due to mutations in the RYR1
the X chromosome, accounting for the maternal inheri-
gene is central core disease. This is a rare myopathy
tance pattern of Duchenne and Becker muscular dys-
presenting in infancy with hypotonia and proximal
trophies. As shown in Figure 49-12, dystrophin forms
muscle weakness. Electron microscopy reveals an ab-
part of a large complex of proteins that attach to or in-
sence of mitochondria in the center of many type I (see
teract with the plasmalemma. Dystrophin links the
below) muscle fibers. Damage to mitochondria induced
actin cytoskeleton to the ECM and appears to be
by high intracellular levels of Ca2+ secondary to abnor-
needed for assembly of the synaptic junction. Impair-
mal functioning of RYR1 appears to be responsible for
ment of these processes by formation of defective dys-
the morphologic findings.
trophin is presumably critical in the causation of
566
/
CHAPTER 49
Table 49-2. Some other important proteins
Duchenne muscular dystrophy. Mutations in the genes
of muscle.
encoding some of the components of the sarcoglycan
complex shown in Figure 49-12 are responsible for
limb-girdle and certain other congenital forms of mus-
Protein
Location
Comment or Function
cular dystrophy.
Titin
Reaches from the Z
Largest protein in body.
line to the M line
Role in relaxation of
muscle.
CARDIAC MUSCLE RESEMBLES SKELETAL
MUSCLE IN MANY RESPECTS
Nebulin
From Z line along
May regulate assembly
length of actin
and length of actin
The general picture of muscle contraction in the heart
filaments
filaments.
resembles that of skeletal muscle. Cardiac muscle, like
α-Actinin
Anchors actin to Z
Stabilizes actin
skeletal muscle, is striated and uses the actin-myosin-
lines
filaments.
tropomyosin-troponin system described above. Unlike
skeletal muscle, cardiac muscle exhibits intrinsic rhyth-
Desmin
Lies alongside actin
Attaches to plasma
micity, and individual myocytes communicate with
filaments
membrane (plasma-
each other because of its syncytial nature. The T tubu-
lemma).
lar system is more developed in cardiac muscle,
Dystrophin
Attached to plasma-
Deficient in Duchenne
whereas the sarcoplasmic reticulum is less extensive
lemma
muscular dystrophy.
and consequently the intracellular supply of Ca2+ for
Mutations of its gene
contraction is less. Cardiac muscle thus relies on extra-
can also cause dilated
cellular Ca2+ for contraction; if isolated cardiac muscle
cardiomyopathy.
is deprived of Ca2+, it ceases to beat within approxi-
Calcineurin
Cytosol
A calmodulin-regulated
mately 1 minute, whereas skeletal muscle can continue
protein phosphatase.
to contract without an extracellular source of Ca2+.
May play important
Cyclic AMP plays a more prominent role in cardiac
roles in cardiac hyper-
than in skeletal muscle. It modulates intracellular levels
trophy and in regulating
of Ca2+ through the activation of protein kinases; these
amounts of slow and
enzymes phosphorylate various transport proteins in
fast twitch muscles.
the sarcolemma and sarcoplasmic reticulum and also in
Myosin-
Arranged trans-
Binds myosin and titin.
the troponin-tropomyosin regulatory complex, affect-
binding
versely in sarcomere
Plays a role in main-
ing intracellular levels of Ca2+ or responses to it. There
protein C
A-bands
taining the structural
is a rough correlation between the phosphorylation of
integrity of the sarco-
TpI and the increased contraction of cardiac muscle in-
mere.
duced by catecholamines. This may account for the in-
otropic effects (increased contractility) of β-adrenergic
compounds on the heart. Some differences among
skeletal, cardiac, and smooth muscle are summarized in
Table 49-3.
Deletion of part of the structural gene for dystrophin,
located on the X chromosome
Ca2+ Enters Myocytes via Ca2+ Channels
& Leaves via the Na+-Ca2+ Exchanger
Diminished synthesis of the mRNA for dystrophin
& the Ca2+ ATPase
As stated above, extracellular Ca2+ plays an important
Low levels or absence of dystrophin
role in contraction of cardiac muscle but not in skeletal
muscle. This means that Ca2+ both enters and leaves
Muscle contraction/relaxation affected;
myocytes in a regulated manner. We shall briefly con-
precise mechanisms not elucidated
sider three transmembrane proteins that play roles in
this process.
Progressive, usually fatal muscular weakness
A. Ca2+ CHANNELS
Figure 49-11. Summary of the causation of
Ca2+ enters myocytes via these channels, which allow
Duchenne muscular dystrophy (MIM 310200).
entry only of Ca2+ ions. The major portal of entry is the
MUSCLE & THE CYTOSKELETON
/
567
Figure 49-12. Organization of dystrophin and other proteins in relation to the plasma membrane of muscle cells.
Dystrophin is part of a large oligomeric complex associated with several other protein complexes. The dystroglycan
complex consists of α-dystroglycan, which associates with the basal lamina protein merosin, and β-dystroglycan,
which binds α-dystroglycan and dystrophin. Syntrophin binds to the carboxyl terminal of dystrophin. The sarcogly-
can complex consists of four transmembrane proteins: α-, β-, γ-, and δ-sarcoglycan. The function of the sarcoglycan
complex and the nature of the interactions within the complex and between it and the other complexes are not
clear. The sarcoglycan complex is formed only in striated muscle, and its subunits preferentially associate with each
other, suggesting that the complex may function as a single unit. Mutations in the gene encoding dystrophin cause
Duchenne and Becker muscular dystrophy; mutations in the genes encoding the various sarcoglycans have been
shown to be responsible for limb-girdle dystrophies (eg, MIM 601173). (Reproduced, with permission, from Duggan DJ
et al: Mutations in the sarcoglycan genes in patients with myopathy. N Engl J Med 1997;336:618.)
L-type (long-duration current, large conductance) or
(CICR). It is estimated that approximately 10% of the
slow Ca2+ channel, which is voltage-gated, opening
Ca2+ involved in contraction enters the cytosol from
during depolarization induced by spread of the cardiac
the extracellular fluid and 90% from the sarcoplasmic
action potential and closing when the action potential
reticulum. However, the former 10% is important, as
declines. These channels are equivalent to the dihy-
the rate of increase of Ca2+ in the myoplasm is impor-
dropyridine receptors of skeletal muscle (Figure 49-8).
tant, and entry via the Ca2+ channels contributes appre-
Slow Ca2+ channels are regulated by cAMP-dependent
ciably to this.
protein kinases
(stimulatory) and cGMP-protein ki-
nases (inhibitory) and are blocked by so-called calcium
channel blockers (eg, verapamil). Fast (or T, transient)
B. Ca2+-Na+ EXCHANGER
Ca2+ channels are also present in the plasmalemma,
This is the principal route of exit of Ca2+ from myo-
though in much lower numbers; they probably con-
cytes. In resting myocytes, it helps to maintain a low
tribute to the early phase of increase of myoplasmic
level of free intracellular Ca2+ by exchanging one Ca2+
Ca2+.
for three Na+. The energy for the uphill movement of
The resultant increase of Ca2+ in the myoplasm acts
Ca2+ out of the cell comes from the downhill move-
on the Ca2+ release channel of the sarcoplasmic reticu-
ment of Na+ into the cell from the plasma. This ex-
lum to open it. This is called Ca2+-induced Ca2+ release
change contributes to relaxation but may run in the re-
568
/
CHAPTER 49
Table 49-3. Some differences between skeletal, cardiac, and smooth muscle.
Skeletal Muscle
Cardiac Muscle
Smooth Muscle
1.
Striated.
1.
Striated.
1. Nonstriated.
2.
No syncytium.
2.
Syncytial.
2. Syncytial.
3.
Small T tubules.
3.
Large T tubules.
3. Generally rudimentary T tubules.
4.
Sarcoplasmic reticulum well-
4.
Sarcoplasmic reticulum present and
4. Sarcoplasmic reticulum often rudimen-
developed and Ca2+ pump acts
Ca2+ pump acts relatively rapidly.
tary and Ca2+ pump acts slowly.
rapidly.
5.
Plasmalemma lacks many hormone
5.
Plasmalemma contains a variety of
5. Plasmalemma contains a variety of
receptors.
receptors (eg, α- and β-adrenergic).
receptors (eg, α- and β-adrenergic).
6.
Nerve impulse initiates contraction.
6.
Has intrinsic rhythmicity.
6. Contraction initiated by nerve impulses,
hormones, etc.
7.
Extracellular fluid Ca2+ not important
7.
Extracellular fluid Ca2+ important
7. Extracellular fluid Ca2+ important for
for contraction.
for contraction.
contraction.
8.
Troponin system present.
8.
Troponin system present.
8. Lacks troponin system; uses regulatory
head of myosin.
9.
Caldesmon not involved.
9.
Caldesmon not involved.
9. Caldesmon is important regulatory
protein.
10.
Very rapid cycling of the
10.
Relatively rapid cycling of the cross-
10. Slow cycling of the cross-bridges per-
cross-bridges.
bridges.
mits slow prolonged contraction
and less utilization of ATP.
important in skeletal muscle. Mutations in genes en-
verse direction during excitation. Because of the Ca2+-
coding ion channels have been shown to be responsible
Na+ exchanger, anything that causes intracellular Na+
for a number of relatively rare conditions affecting mus-
(Na+i) to rise will secondarily cause Ca2+i to rise, caus-
cle. These and other diseases due to mutations of ion
ing more forceful contraction. This is referred to as a
channels have been termed channelopathies; some are
positive inotropic effect. One example is when the drug
listed in Table 49-5.
digitalis is used to treat heart failure. Digitalis inhibits
the sarcolemmal Na+-K+ ATPase, diminishing exit of
Na+ and thus increasing Na+i. This in turn causes Ca2+
to increase, via the Ca2+-Na+ exchanger. The increased
Table 49-4. Major types of ion channels found
Ca2+i results in increased force of cardiac contraction, of
in cells.
benefit in heart failure.
C. Ca2+ ATPASE
Type
Comment
This Ca2+ pump, situated in the sarcolemma, also con-
External
Open in response to a specific extracellular
tributes to Ca2+ exit but is believed to play a relatively
ligand-gated
molecule, eg, acetylcholine.
minor role as compared with the Ca2+-Na+ exchanger.
It should be noted that there are a variety of ion
Internal
Open or close in response to a specific intra-
channels (Chapter 41) in most cells, for Na+, K+, Ca2+,
ligand-gated
cellular molecule, eg, a cyclic nucleotide.
etc. Many of them have been cloned in recent years and
Voltage-gated
Open in response to a change in membrane
their dispositions in their respective membranes worked
potential, eg, Na+, K+, and Ca2+ channels in
out (number of times each one crosses its membrane,
heart.
location of the actual ion transport site in the protein,
Mechanically
Open in response to change in mechanical
etc). They can be classified as indicated in Table 49-4.
gated
pressure.
Cardiac muscle is rich in ion channels, and they are also
MUSCLE & THE CYTOSKELETON
/
569
Table 49-5. Some disorders (channelopathies)
Table 49-6. Biochemical causes of inherited
due to mutations in genes encoding polypeptide
cardiomyopathies.1,2
constituents of ion channels.1
Proteins or Process
Ion Channel and Major
Cause
Affected
Disorder2
Organs Involved
Inborn errors of fatty acid
Carnitine entry into cells and
Central core disease
Ca2+ release channel (RYR1)
oxidation
mitochondria
(MIM 117000)
Skeletal muscle
Certain enzymes of fatty acid
oxidation
Cystic fibrosis
CFTR (Cl− channel)
(MIM 219700)
Lungs, pancreas
Disorders of mitochondrial
Proteins encoded by mito-
oxidative phosphorylation
chondrial genes
Hyperkalemic periodic
Sodium channel
Proteins encoded by nuclear
paralysis (MIM 170500)
Skeletal muscle
genes
Hypokalemic periodic
Slow Ca2+ voltage channel (DHPR)
Abnormalities of myocardial
β-Myosin heavy chains, tropo-
paralysis (MIM 114208)
Skeletal muscle
contractile and structural
nin, tropomyosin, dys-
Malignant hyperthermia
Ca2+ release channel (RYR1)
proteins
trophin
(MIM 180901)
Skeletal muscle
1Based on Kelly DP, Strauss AW: Inherited cardiomyopathies.
N Engl J Med 1994;330:913.
Myotonia congenita
Chloride channel
2Mutations (eg, point mutations, or in some cases deletions) in
(MIM 160800)
Skeletal muscle
the genes (nuclear or mitochondrial) encoding various proteins,
1Data in part from Ackerman NJ, Clapham DE: Ion channels—
enzymes, or tRNA molecules are the fundamental causes of the
basic science and clinical disease. N Engl J Med 1997;336:1575.
inherited cardiomyopathies. Some conditions are mild, whereas
2Other channelopathies include the long QT syndrome (MIM
others are severe and may be part of a syndrome affecting other
192500); pseudoaldosteronism (Liddle syndrome, MIM 177200);
tissues.
persistent hyperinsulinemic hypoglycemia of infancy
(MIM
601820); hereditary X-linked recessive type II nephrolithiasis of in-
fancy (Dent syndrome, MIM 300009); and generalized myotonia,
recessive (Becker disease, MIM 255700). The term “myotonia” sig-
Mutations in the Cardiac
-Myosin Heavy
nifies any condition in which muscles do not relax after contrac-
tion.
Chain Gene Are One Cause of Familial
Hypertrophic Cardiomyopathy
Familial hypertrophic cardiomyopathy is one of the
Inherited Cardiomyopathies Are Due
most frequent hereditary cardiac diseases. Patients ex-
hibit hypertrophy—often massive—of one or both ven-
to Disorders of Cardiac Energy
tricles, starting early in life, and not related to any ex-
Metabolism or to Abnormal
trinsic cause such as hypertension. Most cases are
Myocardial Proteins
transmitted in an autosomal dominant manner; the rest
An inherited cardiomyopathy is any structural or func-
are sporadic. Until recently, its cause was obscure. How-
tional abnormality of the ventricular myocardium due
ever, this situation changed when studies of one affected
to an inherited cause. There are nonheritable types of
family showed that a missense mutation (ie, substitu-
cardiomyopathy, but these will not be described here.
tion of one amino acid by another) in the β-myosin
As shown in Table 49-6, the causes of inherited car-
heavy chain gene was responsible for the condition.
diomyopathies fall into two broad classes: (1) disorders
Subsequent studies have shown a number of missense
of cardiac energy metabolism, mainly reflecting muta-
mutations in this gene, all coding for highly conserved
tions in genes encoding enzymes or proteins involved in
residues. Some individuals have shown other mutations,
fatty acid oxidation (a major source of energy for the
such as formation of an α/β-myosin heavy chain hybrid
myocardium) and oxidative phosphorylation; and
gene. Patients with familial hypertrophic cardiomyopa-
(2) mutations in genes encoding proteins involved in or
thy can show great variation in clinical picture. This in
affecting myocardial contraction, such as myosin,
part reflects genetic heterogeneity; ie, mutation in a
tropomyosin, the troponins, and cardiac myosin-
number of other genes (eg, those encoding cardiac
binding protein C. Mutations in the genes encoding
actin, tropomyosin, cardiac troponins I and T, essential
these latter proteins cause familial hypertrophic car-
and regulatory myosin light chains, and cardiac myosin-
diomyopathy, which will now be discussed.
binding protein C) may also cause familial hypertrophic
570
/
CHAPTER 49
cardiomyopathy. In addition, mutations at different
in the regulation of a number of genes in these cells.
sites in the gene for β-myosin heavy chain may affect the
Current research is not only elucidating the molecular
function of the protein to a greater or lesser extent. The
causes of the cardiomyopathies but is also disclosing
missense mutations are clustered in the head and head-
mutations that cause cardiac developmental disorders
rod regions of myosin heavy chain. One hypothesis is
(eg, septal defects) and arrhythmias (eg, due to muta-
that the mutant polypeptides (“poison polypeptides”)
tions affecting ion channels).
cause formation of abnormal myofibrils, eventually re-
sulting in compensatory hypertrophy. Some mutations
Ca2+ Also Regulates Contraction
alter the charge of the amino acid (eg, substitution of
of Smooth Muscle
arginine for glutamine), presumably affecting the con-
formation of the protein more markedly and thus affect-
While all muscles contain actin, myosin, and tropo-
ing its function. Patients with these mutations have a
myosin, only vertebrate striated muscles contain the
significantly shorter life expectancy than patients in
troponin system. Thus, the mechanisms that regulate
whom the mutation produced no alteration in charge.
contraction must differ in various contractile systems.
Thus, definition of the precise mutations involved in the
Smooth muscles have molecular structures similar to
genesis of FHC may prove to be of important prognos-
those in striated muscle, but the sarcomeres are not
tic value; it can be accomplished by appropriate use of
aligned so as to generate the striated appearance.
the polymerase chain reaction on genomic DNA ob-
Smooth muscles contain α-actinin and tropomyosin
tained from one sample of blood lymphocytes. Figure
molecules, as do skeletal muscles. They do not have the
49-13 is a simplified scheme of the events causing fa-
troponin system, and the light chains of smooth muscle
milial hypertrophic cardiomyopathy.
myosin molecules differ from those of striated muscle
Another type of cardiomyopathy is termed dilated
myosin. Regulation of smooth muscle contraction is
cardiomyopathy. Mutations in the genes encoding dys-
myosin-based, unlike striated muscle, which is actin-
trophin, muscle LIM protein (so called because it was
based. However, like striated muscle, smooth muscle
found to contain a cysteine-rich domain originally de-
contraction is regulated by Ca2+.
tected in three proteins: Lin-II, Isl-1, and Mec-3), and
the cyclic response-element binding protein (CREB)
Phosphorylation of Myosin Light Chains
have been implicated in the causation of this condition.
Initiates Contraction of Smooth Muscle
The first two proteins help organize the contractile ap-
paratus of cardiac muscle cells, and CREB is involved
When smooth muscle myosin is bound to F-actin in the
absence of other muscle proteins such as tropomyosin,
there is no detectable ATPase activity. This absence of
activity is quite unlike the situation described for stri-
Predominantly missense mutations in the β-myosin
ated muscle myosin and F-actin, which has abundant
heavy chain gene on chromosome 14
ATPase activity. Smooth muscle myosin contains light
chains that prevent the binding of the myosin head to
Mutant polypeptide chains (“poison polypeptides”)
F-actin; they must be phosphorylated before they allow
that lead to formation of defective myofibrils
F-actin to activate myosin ATPase. The ATPase activity
then attained hydrolyzes ATP about tenfold more
slowly than the corresponding activity in skeletal mus-
Compensatory hypertrophy of one
cle. The phosphate on the myosin light chains may form
or both cardiac ventricles
a chelate with the Ca2+ bound to the tropomyosin-TpC-
actin complex, leading to an increased rate of formation
Cardiomegaly and various cardiac signs and
of cross-bridges between the myosin heads and actin.
symptoms, including sudden death
The phosphorylation of light chains initiates the attach-
ment-detachment contraction cycle of smooth muscle.
Simplified scheme of the causation of
Figure 49-13.
familial hypertrophic cardiomyopathy (MIM 192600)
Myosin Light Chain Kinase Is Activated
due to mutations in the gene encoding β-myosin heavy
by Calmodulin-4Ca2+ & Then
chain. Mutations in genes encoding other proteins,
Phosphorylates the Light Chains
such as the troponins, tropomyosin, and cardiac
myosin-binding protein C can also cause this condition.
Smooth muscle sarcoplasm contains a myosin light
Mutations in genes encoding yet other proteins (eg,
chain kinase that is calcium-dependent. The Ca2+ acti-
dystrophin) are involved in the causation of dilated
vation of myosin light chain kinase requires binding of
cardiomyopathy.
calmodulin-4Ca2+ to its kinase subunit (Figure 49-14).
MUSCLE & THE CYTOSKELETON
/
571
Calmodulin
Table 49-7 summarizes and compares the regula-
Myosin kinase
tion of actin-myosin interactions (activation of myosin
(inactive)
+
10-5 mol/L Ca2+
10-7 mol/L Ca2
ATPase) in striated and smooth muscles.
The myosin light chain kinase is not directly af-
Ca2+ • calmodulin
fected or activated by cAMP. However, cAMP-acti-
vated protein kinase can phosphorylate the myosin
light chain kinase (not the light chains themselves). The
phosphorylated myosin light chain kinase exhibits a sig-
nificantly lower affinity for calmodulin-Ca2+ and thus is
ATP
Ca2+ • CALMODULIN-MYOSIN
less sensitive to activation. Accordingly, an increase in
KINASE (ACTIVE)
cAMP dampens the contraction response of smooth
muscle to a given elevation of sarcoplasmic Ca2+. This
L-myosin (inhibits
ADP
molecular mechanism can explain the relaxing effect of
myosin-actin interaction)
β-adrenergic stimulation on smooth muscle.
Another protein that appears to play a Ca2+-depen-
dent role in the regulation of smooth muscle contrac-
pL-myosin (does not
tion is caldesmon (87 kDa). This protein is ubiquitous
inhibit myosin-actin interaction)
in smooth muscle and is also found in nonmuscle tis-
sue. At low concentrations of Ca2+, it binds to tro-
-
pomyosin and actin. This prevents interaction of actin
H2PO
4
with myosin, keeping muscle in a relaxed state. At
higher concentrations of Ca2+, Ca2+-calmodulin binds
PHOSPHATASE
caldesmon, releasing it from actin. The latter is then
free to bind to myosin, and contraction can occur.
Figure 49-14. Regulation of smooth muscle con-
Caldesmon is also subject to phosphorylation-dephos-
traction by Ca2+. pL-myosin is the phosphorylated light
phorylation; when phosphorylated, it cannot bind
chain of myosin; L-myosin is the dephosphorylated
actin, again freeing the latter to interact with myosin.
light chain. (Adapted from Adelstein RS, Eisenberg R: Reg-
Caldesmon may also participate in organizing the struc-
ulation and kinetics of actin-myosin ATP interaction. Annu
ture of the contractile apparatus in smooth muscle.
Rev Biochem 1980;49:921.)
Many of its effects have been demonstrated in vitro,
and its physiologic significance is still under investiga-
tion.
As noted in Table 49-3, slow cycling of the cross-
The calmodulin-4Ca2+-activated light chain kinase
bridges permits slow prolonged contraction of smooth
phosphorylates the light chains, which then ceases to in-
muscle (eg, in viscera and blood vessels) with less uti-
hibit the myosin-F-actin interaction. The contraction
lization of ATP compared with striated muscle. The
cycle then begins.
ability of smooth muscle to maintain force at reduced
velocities of contraction is referred to as the latch state;
this is an important feature of smooth muscle, and its
Smooth Muscle Relaxes When
precise molecular bases are under study.
the Concentration of Ca2+ Falls
Below 10−7 Molar
Nitric Oxide Relaxes the Smooth Muscle
Relaxation of smooth muscle occurs when sarcoplasmic
of Blood Vessels & Also Has Many Other
Ca2+ falls below 10−7 mol/L. The Ca2+ dissociates from
Important Biologic Functions
calmodulin, which in turn dissociates from the myosin
light chain kinase, inactivating the kinase. No new
Acetylcholine is a vasodilator that acts by causing relax-
phosphates are attached to the p-light chain, and light
ation of the smooth muscle of blood vessels. However,
chain protein phosphatase, which is continually active
it does not act directly on smooth muscle. A key obser-
and calcium-independent, removes the existing phos-
vation was that if endothelial cells were stripped away
phates from the light chains. Dephosphorylated myosin
from underlying smooth muscle cells, acetylcholine no
p-light chain then inhibits the binding of myosin heads
longer exerted its vasodilator effect. This finding indi-
to F-actin and the ATPase activity. The myosin head
cated that vasodilators such as acetylcholine initially in-
detaches from the F-actin in the presence of ATP, but
teract with the endothelial cells of small blood vessels
it cannot reattach because of the presence of dephos-
via receptors. The receptors are coupled to the phos-
phorylated p-light chain; hence, relaxation occurs.
phoinositide cycle, leading to the intracellular release of
572
/
CHAPTER 49
Table 49-7. Actin-myosin interactions in striated and smooth muscle.
Smooth Muscle
Striated Muscle
(and Nonmuscle Cells)
Proteins of muscle filaments
Actin
Actin
Myosin
Myosin1
Tropomyosin
Tropomyosin
Troponin (Tpl, TpT, TpC)
Spontaneous interaction of F-actin and
Yes
No
myosin alone (spontaneous activation
of myosin ATPase by F-actin
Inhibitor of F-actin-myosin interaction (in-
Troponin system (Tpl)
Unphosphorylated myosin light chain
hibitor of F-actin-dependent activation
of ATPase)
Contraction activated by
Ca2+
Ca2+
Direct effect of Ca2+
4Ca2+ bind to TpC
4Ca2+ bind to calmodulin
Effect of protein-bound Ca2+
TpC ⋅ 4Ca2+ antagonizes Tpl inhibition
Calmodulin ⋅ 4Ca2+ activates myosin light
of F-actin-myosin interaction (allows
chain kinase that phosphorylates myosin
F-actin activation of ATPase)
p-light chain. The phosphorylated p-light
chain no longer inhibits F-actin-myosin
interaction (allows F-actin activation of
ATPase).
1Light chains of myosin are different in striated and smooth muscles.
Ca2+ through the action of inositol trisphosphate. In
zyme. NO synthase is a very complex enzyme, employ-
turn, the elevation of Ca2+ leads to the liberation of en-
ing five redox cofactors: NADPH, FAD, FMN, heme,
dothelium-derived relaxing factor
(EDRF), which
and tetrahydrobiopterin. NO can also be formed from
diffuses into the adjacent smooth muscle. There, it re-
nitrite, derived from vasodilators such as glyceryl trini-
acts with the heme moiety of a soluble guanylyl cyclase,
trate during their metabolism. NO has a very short
resulting in activation of the latter, with a consequent
half-life (approximately 3-4 seconds) in tissues because
elevation of intracellular levels of cGMP
(Figure
it reacts with oxygen and superoxide. The product of
49-15). This in turn stimulates the activities of certain
the reaction with superoxide is peroxynitrite (ONOO−),
cGMP-dependent protein kinases, which probably
which decomposes to form the highly reactive OH•
phosphorylate specific muscle proteins, causing relax-
radical. NO is inhibited by hemoglobin and other
ation; however, the details are still being clarified. The
heme proteins, which bind it tightly. Chemical in-
important coronary artery vasodilator nitroglycerin,
hibitors of NO synthase are now available that can
widely used to relieve angina pectoris, acts to increase
markedly decrease formation of NO. Administration of
intracellular release of EDRF and thus of cGMP.
such inhibitors to animals and humans leads to vaso-
Quite unexpectedly, EDRF was found to be the gas
constriction and a marked elevation of blood pressure,
nitric oxide (NO). NO is formed by the action of the
indicating that NO is of major importance in the main-
enzyme NO synthase, which is cytosolic. The endothe-
tenance of blood pressure in vivo. Another important
lial and neuronal forms of NO synthase are activated by
cardiovascular effect is that by increasing synthesis of
Ca2+ (Table 49-8). The substrate is arginine, and the
cGMP, it acts as an inhibitor of platelet aggregation
products are citrulline and NO:
(Chapter 51).
Since the discovery of the role of NO as a vasodila-
NO SYNTHASE
tor, there has been intense experimental interest in this
Arginine
Citrulline + NO
substance. It has turned out to have a variety of physio-
logic roles, involving virtually every tissue of the body
NO synthase catalyzes a five-electron oxidation of
(Table 49-9). Three major isoforms of NO synthase
an amidine nitrogen of arginine. L-Hydroxyarginine is
have been identified, each of which has been cloned,
an intermediate that remains tightly bound to the en-
and the chromosomal locations of their genes in hu-
MUSCLE & THE CYTOSKELETON
/
573
Glyceryl
Acetylcholine
phosphate, and (4) from two molecules of ADP in a re-
trinitrate
action catalyzed by adenylyl kinase (Figure 49-16). The
amount of ATP in skeletal muscle is only sufficient to
R
ENDOTHELIAL
provide energy for contraction for a few seconds, so
CELL
that ATP must be constantly renewed from one or
Arginine
more of the above sources, depending upon metabolic
conditions. As discussed below, there are at least two
↑Ca2+
+
NO synthase
distinct types of fibers in skeletal muscle, one predomi-
nantly active in aerobic conditions and the other in
NO + Citrulline
anaerobic conditions; not unexpectedly, they use each
of the above sources of energy to different extents.
Skeletal Muscle Contains Large
GTP
Supplies of Glycogen
Guanylyl
The sarcoplasm of skeletal muscle contains large stores
Nitrate
Nitrite
NO
+
cyclase
of glycogen, located in granules close to the I bands.
cGMP
cGMP
The release of glucose from glycogen is dependent on a
protein
specific muscle glycogen phosphorylase (Chapter 18),
kinases
which can be activated by Ca2+, epinephrine, and AMP.
To generate glucose 6-phosphate for glycolysis in skele-
+
tal muscle, glycogen phosphorylase b must be activated
to phosphorylase a via phosphorylation by phosphory-
Relaxation
SMOOTH MUSCLE CELL
lase b kinase (Chapter 18). Ca2+ promotes the activa-
tion of phosphorylase b kinase, also by phosphoryla-
Figure 49-15. Diagram showing formation in an en-
tion. Thus, Ca2+ both initiates muscle contraction and
dothelial cell of nitric oxide (NO) from arginine in a re-
activates a pathway to provide necessary energy. The
action catalyzed by NO synthase. Interaction of an ago-
hormone epinephrine also activates glycogenolysis in
nist (eg, acetylcholine) with a receptor (R) probably
muscle. AMP, produced by breakdown of ADP during
leads to intracellular release of Ca2+ via inositol trisphos-
muscular exercise, can also activate phosphorylase b
phate generated by the phosphoinositide pathway, re-
without causing phosphorylation. Muscle glycogen
sulting in activation of NO synthase. The NO subse-
phosphorylase b is inactive in McArdle disease, one of
quently diffuses into adjacent smooth muscle, where it
the glycogen storage diseases (Chapter 18).
leads to activation of guanylyl cyclase, formation of
cGMP, stimulation of cGMP-protein kinases, and subse-
Under Aerobic Conditions, Muscle
quent relaxation. The vasodilator nitroglycerin is shown
Generates ATP Mainly by Oxidative
entering the smooth muscle cell, where its metabolism
Phosphorylation
also leads to formation of NO.
Synthesis of ATP via oxidative phosphorylation re-
quires a supply of oxygen. Muscles that have a high de-
mand for oxygen as a result of sustained contraction
mans have been determined. Gene knockout experi-
(eg, to maintain posture) store it attached to the heme
ments have been performed on each of the three iso-
moiety of myoglobin. Because of the heme moiety,
forms and have helped establish some of the postulated
muscles containing myoglobin are red, whereas muscles
functions of NO.
with little or no myoglobin are white. Glucose, derived
To summarize, research in the past decade has
from the blood glucose or from endogenous glycogen,
shown that NO plays an important role in many physi-
and fatty acids derived from the triacylglycerols of adi-
ologic and pathologic processes.
pose tissue are the principal substrates used for aerobic
metabolism in muscle.
SEVERAL MECHANISMS REPLENISH
STORES OF ATP IN MUSCLE
Creatine Phosphate Constitutes a
Major Energy Reserve in Muscle
The ATP required as the constant energy source for the
contraction-relaxation cycle of muscle can be generated
Creatine phosphate prevents the rapid depletion of
(1) by glycolysis, using blood glucose or muscle glyco-
ATP by providing a readily available high-energy phos-
gen, (2) by oxidative phosphorylation, (3) from creatine
phate that can be used to regenerate ATP from ADP.
574
/
CHAPTER 49
Table 49-8. Summary of the nomenclature of the NO synthases and of the effects of knockout of their
genes in mice.1
Result of Gene
Subtype
Name2
Comments
Knockout in Mice3
1
nNOS
Activity depends on elevated Ca2+. First
Pyloric stenosis, resistant to vascular stroke, aggressive
identified in neurons. Calmodulin-activated.
sexual behavior (males).
2
iNOS4
Independent of elevated Ca2+.
More susceptible to certain types of infection.
Prominent in macrophages.
3
eNOS
Activity depends on elevated Ca2+.
Elevated mean blood pressure.
First identified in endothelial cells.
1Adapted from Snyder SH: No endothelial NO. Nature 1995;377:196.
2n, neuronal; i, inducible; e, endothelial.
3Gene knockouts were performed by homologous recombination in mice. The enzymes are characterized as neuronal, inducible
(macrophage), and endothelial because these were the sites in which they were first identified. However, all three enzymes have been
found in other sites, and the neuronal enzyme is also inducible. Each gene has been cloned, and its chromosomal location in humans has
been determined.
4iNOS is Ca2+ -independent but binds calmodulin very tightly.
Creatine phosphate is formed from ATP and creatine
idative) and type II
(fast twitch, glycolytic)
(Table
(Figure 49-16) at times when the muscle is relaxed and
49-10). The type I fibers are red because they contain
demands for ATP are not so great. The enzyme catalyz-
myoglobin and mitochondria; their metabolism is aero-
ing the phosphorylation of creatine is creatine kinase
bic, and they maintain relatively sustained contractions.
(CK), a muscle-specific enzyme with clinical utility in
The type II fibers, lacking myoglobin and containing
the detection of acute or chronic diseases of muscle.
few mitochondria, are white: they derive their energy
from anaerobic glycolysis and exhibit relatively short du-
rations of contraction. The proportion of these two
SKELETAL MUSCLE CONTAINS SLOW
types of fibers varies among the muscles of the body, de-
(RED) & FAST (WHITE) TWITCH FIBERS
pending on function (eg, whether or not a muscle is in-
Different types of fibers have been detected in skeletal
volved in sustained contraction, such as maintaining
muscle. One classification subdivides them into type I
posture). The proportion also varies with training; for
(slow twitch), type IIA (fast twitch-oxidative), and type
example, the number of type I fibers in certain leg mus-
IIB (fast twitch-glycolytic). For the sake of simplicity,
cles increases in athletes training for marathons, whereas
we shall consider only two types: type I (slow twitch, ox-
the number of type II fibers increases in sprinters.
A Sprinter Uses Creatine Phosphate
& Anaerobic Glycolysis to Make ATP,
Table 49-9. Some physiologic functions and
Whereas a Marathon Runner Uses
pathologic involvements of nitric oxide (NO).
Oxidative Phosphorylation
• Vasodilator, important in regulation of blood pressure
In view of the two types of fibers in skeletal muscle and
• Involved in penile erection; sildenafil citrate (Viagra) affects
of the various energy sources described above, it is of
this process by inhibiting a cGMP phosphodiesterase
interest to compare their involvement in a sprint (eg,
• Neurotransmitter in the brain and peripheral autonomic
100 meters) and in the marathon (42.2 km; just over
nervous system
26 miles) (Table 49-11).
• Role in long-term potentiation
The major sources of energy in the 100-m sprint
• Role in neurotoxicity
are creatine phosphate (first 4-5 seconds) and then
• Low level of NO involved in causation of pylorospasm in in-
anaerobic glycolysis, using muscle glycogen as the
fantile hypertrophic pyloric stenosis
source of glucose. The two main sites of metabolic con-
• May have role in relaxation of skeletal muscle
trol are at glycogen phosphorylase and at PFK-1. The
• May constitute part of a primitive immune system
former is activated by Ca2+ (released from the sarcoplas-
• Inhibits adhesion, activation, and aggregation of platelets
mic reticulum during contraction), epinephrine, and
MUSCLE & THE CYTOSKELETON
/
575
Creatine phosphate
Muscle glycogen
CREATINE
PHOSPHOKINASE
ADP
MUSCLE
PHOSPHORYLASE
Creatine
Glucose 6-P
GLYCOLYSIS
Muscle
ATP
contraction
MYOSIN
OXIDATIVE
ATPase
PHOSPHORYLATION
ADP + Pi
AMP
ADP
ADENYLYL
KINASE
Figure 49-16. The multiple sources of ATP in muscle.
AMP. PFK-1 is activated by AMP, Pi, and NH3. Attest-
ergy during a marathon for 4 minutes, 18 minutes, 70
ing to the efficiency of these processes, the flux through
minutes, and approximately
4000 minutes, respec-
glycolysis can increase as much as 1000-fold during a
tively. However, the rate of oxidation of fatty acids by
sprint.
muscle is slower than that of glucose, so that oxidation
In contrast, in the marathon, aerobic metabolism is
of glucose and of fatty acids are both major sources of
the principal source of ATP. The major fuel sources are
energy in the marathon.
blood glucose and free fatty acids, largely derived from
A number of procedures have been used by athletes
the breakdown of triacylglycerols in adipose tissue,
to counteract muscle fatigue and inadequate strength.
stimulated by epinephrine. Hepatic glycogen is de-
These include carbohydrate loading, soda (sodium bi-
graded to maintain the level of blood glucose. Muscle
glycogen is also a fuel source, but it is degraded much
more gradually than in a sprint. It has been calculated
that the amounts of glucose in the blood, of glycogen in
Table 49-11. Types of muscle fibers and major
the liver, of glycogen in muscle, and of triacylglycerol in
fuel sources used by a sprinter and by a marathon
adipose tissue are sufficient to supply muscle with en-
runner.
Sprinter (100 m)
Marathon Runner
Table 49-10. Characteristics of type I and type II
Type II (glycolytic) fibers are
Type I (oxidative) fibers are
fibers of skeletal muscle.
used predominantly.
used predominantly.
Creatine phosphate is the
ATP is the major energy
Type I
Type II
major energy source dur-
source throughout.
Slow Twitch
Fast Twitch
ing the first 4-5 seconds.
Myosin ATPase
Low
High
Glucose derived from muscle
Blood glucose and free fatty
Energy utilization
Low
High
glycogen and metabolized
acids are the major fuel
Mitochondria
Many
Few
by anaerobic glycolysis is
sources.
Color
Red
White
the major fuel source.
Myoglobin
Yes
No
Contraction rate
Slow
Fast
Muscle glycogen is rapidly
Muscle glycogen is slowly
Duration
Prolonged
Short
depleted.
depleted.
576
/
CHAPTER 49
carbonate) loading, blood doping (administration of
Table 49-12. Summary of major features of
red blood cells), and ingestion of creatine and an-
the biochemistry of skeletal muscle related to
drostenedione. Their rationales and efficacies will not
its metabolism.1
be discussed here.
•
Skeletal muscle functions under both aerobic (resting) and
SKELETAL MUSCLE CONSTITUTES
anaerobic (eg, sprinting) conditions, so both aerobic and
THE MAJOR RESERVE OF
anaerobic glycolysis operate, depending on conditions.
PROTEIN IN THE BODY
•
Skeletal muscle contains myoglobin as a reservoir of oxy-
gen.
In humans, skeletal muscle protein is the major nonfat
•
Skeletal muscle contains different types of fibers primarily
source of stored energy. This explains the very large
suited to anaerobic (fast twitch fibers) or aerobic (slow
losses of muscle mass, particularly in adults, resulting
twitch fibers) conditions.
from prolonged caloric undernutrition.
•
Actin, myosin, tropomyosin, troponin complex (TpT, Tpl,
The study of tissue protein breakdown in vivo is dif-
and TpC), ATP, and Ca2+ are key constituents in relation to
ficult, because amino acids released during intracellular
contraction.
breakdown of proteins can be extensively reutilized for
•
The Ca2+ ATPase, the Ca2+ release channel, and calse-
protein synthesis within the cell, or the amino acids
questrin are proteins involved in various aspects of Ca2+ me-
may be transported to other organs where they enter
tabolism in muscle.
•
Insulin acts on skeletal muscle to increase uptake of glu-
anabolic pathways. However, actin and myosin are
cose.
methylated by a posttranslational reaction, forming
•
In the fed state, most glucose is used to synthesize glyco-
3-methylhistidine. During intracellular breakdown of
gen, which acts as a store of glucose for use in exercise;
actin and myosin, 3-methylhistidine is released and ex-
“preloading” with glucose is used by some long-distance
creted into the urine. The urinary output of the methy-
athletes to build up stores of glycogen.
lated amino acid provides a reliable index of the rate of
•
Epinephrine stimulates glycogenolysis in skeletal muscle,
myofibrillar protein breakdown in the musculature of
whereas glucagon does not because of absence of its re-
human subjects.
ceptors.
Various features of muscle metabolism, most of
•
Skeletal muscle cannot contribute directly to blood glucose
which are dealt with in other chapters of this text, are
because it does not contain glucose-6-phosphatase.
summarized in Table 49-12.
•
Lactate produced by anaerobic metabolism in skeletal mus-
cle passes to liver, which uses it to synthesize glucose,
THE CYTOSKELETON PERFORMS
which can then return to muscle (the Cori cycle).
•
Skeletal muscle contains phosphocreatine, which acts as an
MULTIPLE CELLULAR FUNCTIONS
energy store for short-term (seconds) demands.
Nonmuscle cells perform mechanical work, including
•
Free fatty acids in plasma are a major source of energy, par-
self-propulsion, morphogenesis, cleavage, endocytosis,
ticularly under marathon conditions and in prolonged star-
exocytosis, intracellular transport, and changing cell
vation.
shape. These cellular functions are carried out by an ex-
•
Skeletal muscle can utilize ketone bodies during starvation.
tensive intracellular network of filamentous structures
•
Skeletal muscle is the principal site of metabolism of
branched-chain amino acids, which are used as an energy
constituting the cytoskeleton. The cell cytoplasm is
source.
not a sac of fluid, as once thought. Essentially all eu-
•
Proteolysis of muscle during starvation supplies amino
karyotic cells contain three types of filamentous struc-
acids for gluconeogenesis.
tures: actin filaments
(7-9.5 nm in diameter; also
•
Major amino acids emanating from muscle are alanine (des-
known as microfilaments), microtubules (25 nm), and
tined mainly for gluconeogenesis in liver and forming part
intermediate filaments (10-12 nm). Each type of fila-
of the glucose-alanine cycle) and glutamine (destined
ment can be distinguished biochemically and by the
mainly for the gut and kidneys).
electron microscope.
1This table brings together material from various chapters in this
book.
Nonmuscle Cells Contain Actin
That Forms Microfilaments
G-actin is present in most if not all cells of the body.
With appropriate concentrations of magnesium and
potassium chloride, it spontaneously polymerizes to
form double helical F-actin filaments like those seen in
muscle. There are at least two types of actin in nonmus-
MUSCLE & THE CYTOSKELETON
/
577
cle cells: β-actin and γ-actin. Both types can coexist in
dynamin, and myosins are referred to as molecular
the same cell and probably even copolymerize in the
motors.
same filament. In the cytoplasm, F-actin forms micro-
An absence of dynein in cilia and flagella results in
filaments of 7-9.5 nm that frequently exist as bundles
immotile cilia and flagella, leading to male sterility and
of a tangled-appearing meshwork. These bundles are
chronic respiratory infection, a condition known as
prominent just underlying the plasma membrane of
Kartagener syndrome.
many cells and are there referred to as stress fibers. The
Certain drugs bind to microtubules and thus inter-
stress fibers disappear as cell motility increases or upon
fere with their assembly or disassembly. These include
malignant transformation of cells by chemicals or onco-
colchicine (used for treatment of acute gouty arthritis),
genic viruses.
vinblastine (a vinca alkaloid used for treating certain
Although not organized as in muscle, actin filaments
types of cancer), paclitaxel
(Taxol) (effective against
in nonmuscle cells interact with myosin to cause cellu-
ovarian cancer), and griseofulvin (an antifungal agent).
lar movements.
Intermediate Filaments Differ From
Microfilaments & Microtubules
Microtubules Contain
- &
-Tubulins
An intracellular fibrous system exists of filaments with
Microtubules, an integral component of the cellular cy-
an axial periodicity of 21 nm and a diameter of 8-10
toskeleton, consist of cytoplasmic tubes 25 nm in diam-
nm that is intermediate between that of microfilaments
eter and often of extreme length. Microtubules are nec-
(6 nm) and microtubules (23 nm). Four classes of inter-
essary for the formation and function of the mitotic
mediate filaments are found, as indicated in Table
spindle and thus are present in all eukaryotic cells.
49-13. They are all elongated, fibrous molecules, with
They are also involved in the intracellular movement of
a central rod domain, an amino terminal head, and a
endocytic and exocytic vesicles and form the major
carboxyl terminal tail. They form a structure like a
structural components of cilia and flagella. Micro-
rope, and the mature filaments are composed of
tubules are a major component of axons and dendrites,
tetramers packed together in a helical manner. They are
in which they maintain structure and participate in the
important structural components of cells, and most are
axoplasmic flow of material along these neuronal
relatively stable components of the cytoskeleton, not
processes.
undergoing rapid assembly and disassembly and not
Microtubules are cylinders of
13 longitudinally
arranged protofilaments, each consisting of dimers of
α-tubulin and β-tubulin, closely related proteins of ap-
proximately
50 kDa molecular mass. The tubulin
dimers assemble into protofilaments and subsequently
Table 49-13. Classes of intermediate filaments of
into sheets and then cylinders. A microtubule-organiz-
eukaryotic cells and their distributions.
ing center, located around a pair of centrioles, nucleates
the growth of new microtubules. A third species of
Molecular
tubulin, γ-tubulin, appears to play an important role in
Proteins
Mass
Distributions
this assembly. GTP is required for assembly. A variety
of proteins are associated with microtubules (micro-
Keratins
tubule-associated proteins [MAPs], one of which is tau)
Type I (acidic)
40-60 kDa
Epithelial cells, hair,
Type II (basic)
50-70 kDa
nails
and play important roles in microtubule assembly and
stabilization. Microtubules are in a state of dynamic
Vimentin-like
instability, constantly assembling and disassembling.
Vimentin
54 kDa
Various mesenchymal
They exhibit polarity (plus and minus ends); this is im-
cells
portant in their growth from centrioles and in their
Desmin
53 kDa
Muscle
ability to direct intracellular movement. For instance,
Glial fibrillary acid
50 kDa
Glial cells
in axonal transport, the protein kinesin, with a
protein
myosin-like ATPase activity, uses hydrolysis of ATP to
Peripherin
66 kDa
Neurons
move vesicles down the axon toward the positive end of
Neurofilaments
the microtubular formation. Flow of materials in the
Low (L), medium (M),
60-130 kDa
Neurons
opposite direction, toward the negative end, is powered
and high (H)1
by cytosolic dynein, another protein with ATPase ac-
Lamins
tivity. Similarly, axonemal dyneins power ciliary and
A, B, and C
65-75 kDa
Nuclear lamina
flagellar movement. Another protein, dynamin, uses
GTP and is involved in endocytosis. Kinesins, dyneins,
1Refers to their molecular masses.
578
/
CHAPTER 49
disappearing during mitosis, as do actin and many mi-
cle, the sarcoplasmic reticulum regulates distribution
crotubular filaments. An important exception to this is
of Ca2+ to the sarcomeres, whereas inflow of Ca2+ via
provided by the lamins, which, subsequent to phosphor-
Ca2+ channels in the sarcolemma is of major impor-
ylation, disassemble at mitosis and reappear when it ter-
tance in cardiac and smooth muscle.
minates.
•
Many cases of malignant hyperthermia in humans
Keratins form a large family, with about 30 mem-
are due to mutations in the gene encoding the Ca2+
bers being distinguished. As indicated in Table 49-13,
release channel.
two major types of keratins are found; all individual
•
A number of differences exist between skeletal and
keratins are heterodimers made up of one member of
cardiac muscle; in particular, the latter contains a va-
each class.
riety of receptors on its surface.
Vimentins are widely distributed in mesodermal
•
Some cases of familial hypertrophic cardiomyopathy
cells, and desmin, glial fibrillary acidic protein, and pe-
are due to missense mutations in the gene coding for
ripherin are related to them. All members of the vi-
β-myosin heavy chain.
mentin-like family can copolymerize with each other.
•
Smooth muscle, unlike skeletal and cardiac muscle,
Intermediate filaments are very prominent in nerve
cells; neurofilaments are classified as low, medium, and
does not contain the troponin system; instead, phos-
phorylation of myosin light chains initiates contrac-
high on the basis of their molecular masses. Lamins
form a meshwork in apposition to the inner nuclear
tion.
membrane. The distribution of intermediate filaments
•
Nitric oxide is a regulator of vascular smooth muscle;
in normal and abnormal (eg, cancer) cells can be stud-
blockage of its formation from arginine causes an
ied by the use of immunofluorescent techniques, using
acute elevation of blood pressure, indicating that reg-
antibodies of appropriate specificities. These antibodies
ulation of blood pressure is one of its many func-
to specific intermediate filaments can also be of use to
tions.
pathologists in helping to decide the origin of certain
•
Duchenne-type muscular dystrophy is due to muta-
dedifferentiated malignant tumors. These tumors may
tions in the gene, located on the X chromosome, en-
still retain the type of intermediate filaments found in
coding the protein dystrophin.
their cell of origin.
•
Two major types of muscle fibers are found in hu-
A number of skin diseases, mainly characterized by
mans: white (anaerobic) and red (aerobic). The for-
blistering, have been found to be due to mutations in
mer are particularly used in sprints and the latter in
genes encoding various keratins. Three of these disor-
prolonged aerobic exercise. During a sprint, muscle
ders are epidermolysis bullosa simplex, epidermolytic
uses creatine phosphate and glycolysis as energy
hyperkeratosis, and epidermolytic palmoplantar kerato-
sources; in the marathon, oxidation of fatty acids is
derma. The blistering probably reflects a diminished ca-
of major importance during the later phases.
pacity of various layers of the skin to resist mechanical
•
Nonmuscle cells perform various types of mechanical
stresses due to abnormalities in microfilament structure.
work carried out by the structures constituting the
cytoskeleton. These structures include actin filaments
SUMMARY
(microfilaments), microtubules (composed primarily
of α- tubulin and β-tubulin), and intermediate fila-
• The myofibrils of skeletal muscle contain thick and
ments. The latter include keratins, vimentin-like pro-
thin filaments. The thick filaments contain myosin.
teins, neurofilaments, and lamins.
The thin filaments contain actin, tropomyosin, and
the troponin complex (troponins T, I, and C).
• The sliding filament cross-bridge model is the foun-
dation of current thinking about muscle contraction.
REFERENCES
The basis of this model is that the interdigitating fila-
Ackerman MJ, Clapham DE: Ion channels—basic science and clin-
ments slide past one another during contraction and
ical disease. N Engl J Med 1997;336:1575.
cross-bridges between myosin and actin generate and
Andreoli TE: Ion transport disorders: introductory comments. Am
sustain the tension.
J Med 1998;104:85. (First of a series of articles on ion trans-
• The hydrolysis of ATP is used to drive movement of
port disorders published between January and August, 1998.
the filaments. ATP binds to myosin heads and is hy-
Topics covered were structure and function of ion channels,
arrhythmias and antiarrhythmic drugs, Liddle syndrome,
drolyzed to ADP and Pi by the ATPase activity of the
cholera, malignant hyperthermia, cystic fibrosis, the periodic
actomyosin complex.
paralyses and Bartter syndrome, and Gittelman syndrome.)
• Ca2+ plays a key role in the initiation of muscle con-
Fuller GM, Shields D: Molecular Basis of Medical Cell Biology. Ap-
traction by binding to troponin C. In skeletal mus-
pleton & Lange, 1998.
MUSCLE & THE CYTOSKELETON
/
579
Geeves MA, Holmes KC: Structural mechanism of muscle contrac-
Mayer B, Hemmens B: Biosynthesis and action of nitric oxide in
tion. Annu Rev Biochem 1999;68:728.
mammalian cells. Trends Biochem Sci 1998;22:477.
Hille B: Ion Channels of Excitable Membranes. Sinauer, 2001.
Scriver CR et al (editors): The Metabolic and Molecular Bases of In-
Howard J: Mechanics of Motor Proteins and the Cytoskeleton. Sin-
herited Disease, 8th ed. McGraw-Hill, 2001. (This compre-
auer, 2001.
hensive four-volume text contains coverage of malignant hy-
perthermia
[Chapter
9], channelopathies
[Chapter
204],
Lodish H et al (editors): Molecular Cell Biology, 4th ed. Freeman,
hypertrophic cardiomyopathy [Chapter 213], the muscular
2000. (Chapters 18 and 19 of this text contain comprehen-
dystrophies [Chapter 216], and disorders of intermediate fila-
sive descriptions of cell motility and cell shape.)
ments and their associated proteins [Chapter 221].)
Loke J, MacLennan DH: Malignant hyperthermia and central core
disease: disorders of Ca2+ release channels. Am J Med
1998;104:470.
Plasma Proteins & Immunoglobulins
50
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
solvents or electrolytes (or both) to remove different
protein fractions in accordance with their solubility
The fundamental role of blood in the maintenance of
characteristics. This is the basis of the so-called salting-
homeostasis and the ease with which blood can be ob-
out methods, which find some usage in the determina-
tained have meant that the study of its constituents has
tion of protein fractions in the clinical laboratory.
been of central importance in the development of bio-
Thus, one can separate the proteins of the plasma into
chemistry and clinical biochemistry. The basic proper-
three major groups—fibrinogen, albumin, and globu-
ties of a number of plasma proteins, including the
lins—by the use of varying concentrations of sodium
immunoglobulins
(antibodies), are described in this
or ammonium sulfate.
chapter. Changes in the amounts of various plasma pro-
The most common method of analyzing plasma
teins and immunoglobulins occur in many diseases and
proteins is by electrophoresis. There are many types of
can be monitored by electrophoresis or other suitable
electrophoresis, each using a different supporting
procedures. As indicated in an earlier chapter, alterations
medium. In clinical laboratories, cellulose acetate is
of the activities of certain enzymes found in plasma are
widely used as a supporting medium. Its use permits
of diagnostic use in a number of pathologic conditions.
resolution, after staining, of plasma proteins into five
bands, designated albumin, α1, α2, β, and γ fractions,
THE BLOOD HAS MANY FUNCTIONS
respectively (Figure 50-2). The stained strip of cellu-
lose acetate (or other supporting medium) is called an
The functions of blood—except for specific cellular
electrophoretogram. The amounts of these five bands
ones such as oxygen transport and cell-mediated im-
can be conveniently quantified by use of densitomet-
munologic defense—are carried out by plasma and its
ric scanning machines. Characteristic changes in the
constituents (Table 50-1).
amounts of one or more of these five bands are found
Plasma consists of water, electrolytes, metabolites,
in many diseases.
nutrients, proteins, and hormones. The water and elec-
trolyte composition of plasma is practically the same as
that of all extracellular fluids. Laboratory determina-
The Concentration of Protein in Plasma Is
tions of levels of Na+, K+, Ca2+, Cl−, HCO3−, PaCO2,
Important in Determining the Distribution
and of blood pH are important in the management of
of Fluid Between Blood & Tissues
many patients.
In arterioles, the hydrostatic pressure is about 37 mm
PLASMA CONTAINS A COMPLEX
Hg, with an interstitial (tissue) pressure of 1 mm Hg
opposing it. The osmotic pressure (oncotic pressure)
MIXTURE OF PROTEINS
exerted by the plasma proteins is approximately 25 mm
The concentration of total protein in human plasma is
Hg. Thus, a net outward force of about 11 mm Hg
approximately 7.0-7.5 g/dL and comprises the major
drives fluid out into the interstitial spaces. In venules,
part of the solids of the plasma. The proteins of the
the hydrostatic pressure is about 17 mm Hg, with the
plasma are actually a complex mixture that includes not
oncotic and interstitial pressures as described above;
only simple proteins but also conjugated proteins such
thus, a net force of about 9 mm Hg attracts water back
as glycoproteins and various types of lipoproteins.
into the circulation. The above pressures are often re-
Thousands of antibodies are present in human plasma,
ferred to as the Starling forces. If the concentration of
though the amount of any one antibody is usually quite
plasma proteins is markedly diminished (eg, due to se-
low under normal circumstances. The relative dimen-
vere protein malnutrition), fluid is not attracted back
sions and molecular masses of some of the most impor-
into the intravascular compartment and accumulates in
tant plasma proteins are shown in Figure 50-1.
the extravascular tissue spaces, a condition known as
The separation of individual proteins from a com-
edema. Edema has many causes; protein deficiency is
plex mixture is frequently accomplished by the use of
one of them.
580
PLASMA PROTEINS & IMMUNOGLOBULINS
/
581
Table 50-1. Major functions of blood.
Plasma Proteins Have Been
Studied Extensively
(1)
Respiration—transport of oxygen from the lungs to the
Because of the relative ease with which they can be ob-
tissues and of CO2 from the tissues to the lungs
tained, plasma proteins have been studied extensively in
(2)
Nutrition—transport of absorbed food materials
both humans and animals. Considerable information is
(3)
Excretion—transport of metabolic waste to the kidneys,
available about the biosynthesis, turnover, structure,
lungs, skin, and intestines for removal
and functions of the major plasma proteins. Alterations
(4)
Maintenance of the normal acid-base balance in the
of their amounts and of their metabolism in many dis-
body
ease states have also been investigated. In recent years,
(5)
Regulation of water balance through the effects of
many of the genes for plasma proteins have been cloned
blood on the exchange of water between the circulating
fluid and the tissue fluid
and their structures determined.
(6)
Regulation of body temperature by the distribution of
The preparation of antibodies specific for the indi-
body heat
vidual plasma proteins has greatly facilitated their
(7)
Defense against infection by the white blood cells and
study, allowing the precipitation and isolation of pure
circulating antibodies
proteins from the complex mixture present in tissues or
(8)
Transport of hormones and regulation of metabolism
plasma. In addition, the use of isotopes has made pos-
(9)
Transport of metabolites
sible the determination of their pathways of biosynthe-
(10)
Coagulation
sis and of their turnover rates in plasma.
The following generalizations have emerged from
studies of plasma proteins.
A. MOST PLASMA PROTEINS ARE
SYNTHESIZED IN THE LIVER
This has been established by experiments at the whole-
Scale
animal level (eg, hepatectomy) and by use of the iso-
lated perfused liver preparation, of liver slices, of liver
homogenates, and of in vitro translation systems using
10 nm
Na+
CI-
Glucose
preparations of mRNA extracted from liver. However,
the γ-globulins are synthesized in plasma cells and cer-
tain plasma proteins are synthesized in other sites, such
as endothelial cells.
Albumin
Hemoglobin
69,000
64,500
B. PLASMA PROTEINS ARE GENERALLY SYNTHESIZED
ON MEMBRANE-BOUND POLYRIBOSOMES
They then traverse the major secretory route in the cell
β1-Globulin
γ-Globulin
90,000
156,000
(rough endoplasmic membrane → smooth endoplasmic
membrane → Golgi apparatus → secretory vesicles) prior
to entering the plasma. Thus, most plasma proteins are
synthesized as preproteins and initially contain amino
terminal signal peptides (Chapter 46). They are usually
subjected to various posttranslational modifications (pro-
-Lipoprotein
α1
teolysis, glycosylation, phosphorylation, etc) as they travel
200,000
through the cell. Transit times through the hepatocyte
β1-Lipoprotein
1,300,000
from the site of synthesis to the plasma vary from 30 min-
utes to several hours or more for individual proteins.
C. MOST PLASMA PROTEINS ARE GLYCOPROTEINS
Fibrinogen
340,000
Accordingly, they generally contain either N- or O-
linked oligosaccharide chains, or both (Chapter 47). Al-
Figure 50-1. Relative dimensions and approximate
bumin is the major exception; it does not contain sugar
molecular masses of protein molecules in the blood
residues. The oligosaccharide chains have various func-
(Oncley).
tions
(Table 47-2). Removal of terminal sialic acid
582
/
CHAPTER 50
A
C
+
-
Albumin α1
α2
β
γ
B
D
+
-
Albumin α1
α2
β
γ
Figure 50-2. Technique of cellulose acetate zone electrophoresis. A: A small amount of serum or other
fluid is applied to a cellulose acetate strip. B: Electrophoresis of sample in electrolyte buffer is performed.
C: Separated protein bands are visualized in characteristic positions after being stained. D: Densitometer
scanning from cellulose acetate strip converts bands to characteristic peaks of albumin, α1-globulin, α2-glob-
ulin, β-globulin, and γ-globulin. (Reproduced, with permission, from Parslow TG et al [editors]: Medical Immunol-
ogy, 10th ed. McGraw-Hill, 2001.)
residues from certain plasma proteins (eg, ceruloplas-
nondenaturing conditions. This isotope unites covalently
min) by exposure to neuraminidase can markedly
with tyrosine residues in the protein. The labeled protein
shorten their half-lives in plasma (Chapter 47).
is freed of unbound 131I and its specific activity (disinte-
grations per minute per milligram of protein) deter-
D. MANY PLASMA PROTEINS EXHIBIT POLYMORPHISM
mined. A known amount of the radioactive protein is
A polymorphism is a mendelian or monogenic trait that
then injected into a normal adult subject, and samples of
exists in the population in at least two phenotypes, nei-
blood are taken at various time intervals for determina-
ther of which is rare (ie, neither of which occurs with
tions of radioactivity. The values for radioactivity are
frequency of less than 1-2%). The ABO blood group
plotted against time, and the half-life of the protein (the
substances (Chapter 52) are the best-known examples
time for the radioactivity to decline from its peak value
of human polymorphisms. Human plasma proteins
to one-half of its peak value) can be calculated from the
that exhibit polymorphism include α1-antitrypsin, hap-
resulting graph, discounting the times for the injected
toglobin, transferrin, ceruloplasmin, and immunoglob-
protein to equilibrate (mix) in the blood and in the ex-
ulins. The polymorphic forms of these proteins can be
travascular spaces. The half-lives obtained for albumin
distinguished by different procedures (eg, various types
and haptoglobin in normal healthy adults are approxi-
of electrophoresis or isoelectric focusing), in which each
mately 20 and 5 days, respectively. In certain diseases,
form may show a characteristic migration. Analyses of
the half-life of a protein may be markedly altered. For in-
these human polymorphisms have proved to be of ge-
stance, in some gastrointestinal diseases such as regional
netic, anthropologic, and clinical interest.
ileitis
(Crohn disease), considerable amounts of plasma
proteins, including albumin, may be lost into the bowel
E. EACH PLASMA PROTEIN HAS A CHARACTERISTIC
through the inflamed intestinal mucosa. Patients with
HALF-LIFE IN THE CIRCULATION
this condition have a protein-losing gastroenteropathy,
The half-life of a plasma protein can be determined by
and the half-life of injected iodinated albumin in these
labeling the isolated pure protein with 131I under mild,
subjects may be reduced to as little as 1 day.
PLASMA PROTEINS & IMMUNOGLOBULINS
/
583
Table 50-2. Some functions of plasma proteins.
F. THE LEVELS OF CERTAIN PROTEINS IN PLASMA
INCREASE DURING ACUTE INFLAMMATORY STATES OR
SECONDARY TO CERTAIN TYPES OF TISSUE DAMAGE
Function
Plasma Proteins
These proteins are called “acute phase proteins” (or re-
Antiproteases
Antichymotrypsin
actants) and include C-reactive protein (CRP, so-named
α1-Antitrypsin (α1-antiproteinase)
because it reacts with the C polysaccharide of pneumo-
α2-Macroglobulin
cocci), α1-antitrypsin, haptoglobin, α1-acid glycopro-
Antithrombin
tein, and fibrinogen. The elevations of the levels of these
Blood clotting
Various coagulation factors, fibrinogen
proteins vary from as little as 50% to as much as 1000-
fold in the case of CRP. Their levels are also usually ele-
Enzymes
Function in blood, eg, coagulation
vated during chronic inflammatory states and in pa-
factors, cholinesterase
tients with cancer. These proteins are believed to play a
Leakage from cells or tissues, eg, amino-
role in the body’s response to inflammation. For exam-
transferases
ple, C-reactive protein can stimulate the classic comple-
Hormones
Erythropoietin1
ment pathway, and α1-antitrypsin can neutralize certain
proteases released during the acute inflammatory state.
Immune defense
Immunoglobulins, complement proteins,
β2-microglobulin
CRP is used as a marker of tissue injury, infection, and
inflammation, and there is considerable interest in its
Involvement in
Acute phase response proteins (eg,
use as a predictor of certain types of cardiovascular con-
inflammatory
C-reactive protein, α1-acid glyco-
ditions secondary to atherosclerosis. Interleukin-1
responses
protein [orosomucoid])
(IL-1), a polypeptide released from mononuclear phago-
Oncofetal
α1-Fetoprotein (AFP)
cytic cells, is the principal—but not the sole—stimula-
tor of the synthesis of the majority of acute phase reac-
Transport or
Albumin (various ligands, including bili-
tants by hepatocytes. Additional molecules such as IL-6
binding
rubin, free fatty acids, ions [Ca2+],
are involved, and they as well as IL-1 appear to work at
proteins
metals [eg, Cu2+, Zn2+], metheme,
the level of gene transcription.
steroids, other hormones, and a vari-
ety of drugs
Table 50-2 summarizes the functions of many of
Ceruloplasmin (contains Cu2+; albumin
the plasma proteins. The remainder of the material in
probably more important in physio-
this chapter presents basic information regarding se-
logic transport of Cu2+)
lected plasma proteins: albumin, haptoglobin, transfer-
Corticosteroid-binding globulin (trans-
rin, ceruloplasmin, α1-antitrypsin, α2-macroglobulin,
cortin) (binds cortisol)
the immunoglobulins, and the complement system.
Haptoglobin (binds extracorpuscular
The lipoproteins are discussed in Chapter 25.
hemoglobin)
Lipoproteins (chylomicrons, VLDL, LDL,
Albumin Is the Major Protein
HDL)
in Human Plasma
Hemopexin (binds heme)
Retinol-binding protein (binds retinol)
Albumin (69 kDa) is the major protein of human
Sex hormone-binding globulin (binds
plasma (3.4-4.7 g/dL) and makes up approximately
testosterone, estradiol)
60% of the total plasma protein. About 40% of albu-
Thyroid-binding globulin (binds T4, T3)
min is present in the plasma, and the other 60% is pre-
Transferrin (transport iron)
sent in the extracellular space. The liver produces about
Transthyretin (formerly prealbumin;
12 g of albumin per day, representing about 25% of
binds T4 and forms a complex with
total hepatic protein synthesis and half its secreted pro-
retinol-binding protein)
tein. Albumin is initially synthesized as a prepropro-
1Various other protein hormones circulate in the blood but are
tein. Its signal peptide is removed as it passes into the
not usually designated as plasma proteins. Similarly, ferritin is also
cisternae of the rough endoplasmic reticulum, and a
found in plasma in small amounts, but it too is not usually charac-
hexapeptide at the resulting amino terminal is subse-
terized as a plasma protein.
quently cleaved off farther along the secretory pathway.
The synthesis of albumin is depressed in a variety of
diseases, particularly those of the liver. The plasma of
patients with liver disease often shows a decrease in the
ratio of albumin to globulins
(decreased albumin-
globulin ratio). The synthesis of albumin decreases rela-
584
/
CHAPTER 50
tively early in conditions of protein malnutrition, such
of the kidney, enters the tubules, and tends to precipi-
as kwashiorkor.
tate therein (as can happen after a massive incompatible
Mature human albumin consists of one polypeptide
blood transfusion, when the capacity of haptoglobin to
chain of 585 amino acids and contains 17 disulfide
bind hemoglobin is grossly exceeded) (Figure 50-3).
bonds. By the use of proteases, albumin can be subdi-
However, the Hb-Hp complex is too large to pass
vided into three domains, which have different func-
through the glomerulus. The function of Hp thus ap-
tions. Albumin has an ellipsoidal shape, which means
pears to be to prevent loss of free hemoglobin into the
that it does not increase the viscosity of the plasma as
kidney. This conserves the valuable iron present in he-
much as an elongated molecule such as fibrinogen does.
moglobin, which would otherwise be lost to the body.
Because of its relatively low molecular mass (about 69
Human haptoglobin exists in three polymorphic
kDa) and high concentration, albumin is thought to be
forms, known as Hp 1-1, Hp 2-1, and Hp 2-2. Hp 1-1
responsible for 75-80% of the osmotic pressure of
migrates in starch gel electrophoresis as a single band,
human plasma. Electrophoretic studies have shown that
whereas Hp 2-1 and Hp 2-2 exhibit much more com-
the plasma of certain humans lacks albumin. These
plex band patterns. Two genes, designated Hp1 and Hp2,
subjects are said to exhibit analbuminemia. One cause
direct these three phenotypes, with Hp 2-1 being the
of this condition is a mutation that affects splicing.
heterozygous phenotype. It has been suggested that the
Subjects with analbuminemia show only moderate
haptoglobin polymorphism may be associated with
edema, despite the fact that albumin is the major deter-
the prevalence of many inflammatory diseases.
minant of plasma osmotic pressure. It is thought that
The levels of haptoglobin in human plasma vary and
the amounts of the other plasma proteins increase and
are of some diagnostic use. Low levels of haptoglobin are
compensate for the lack of albumin.
found in patients with hemolytic anemias. This is ex-
Another important function of albumin is its ability
plained by the fact that whereas the half-life of haptoglo-
to bind various ligands. These include free fatty acids
bin is approximately 5 days, the half-life of the Hb-Hp
(FFA), calcium, certain steroid hormones, bilirubin,
complex is about 90 minutes, the complex being rapidly
and some of the plasma tryptophan. In addition, albu-
removed from plasma by hepatocytes. Thus, when hap-
min appears to play an important role in transport of
toglobin is bound to hemoglobin, it is cleared from the
copper in the human body (see below). A variety of
plasma about 80 times faster than normally. Accord-
drugs, including sulfonamides, penicillin G, dicumarol,
ingly, the level of haptoglobin falls rapidly in situations
and aspirin, are bound to albumin; this finding has im-
where hemoglobin is constantly being released from red
portant pharmacologic implications.
blood cells, such as occurs in hemolytic anemias. Hapto-
Preparations of human albumin have been widely
globin is an acute phase protein, and its plasma level is
used in the treatment of hemorrhagic shock and of
elevated in a variety of inflammatory states.
burns. However, this treatment is under review because
Certain other plasma proteins bind heme but not
some recent studies have suggested that administration of
hemoglobin. Hemopexin is a β1-globulin that binds
albumin in these conditions may increase mortality rates.
free heme. Albumin will bind some metheme (ferric
heme) to form methemalbumin, which then transfers
the metheme to hemopexin.
Haptoglobin Binds Extracorpuscular
Hemoglobin, Preventing Free Hemoglobin
Absorption of Iron From the Small
From Entering the Kidney
Intestine Is Tightly Regulated
Haptoglobin (Hp) is a plasma glycoprotein that binds
Transferrin (Tf) is a plasma protein that plays a central
extracorpuscular hemoglobin (Hb) in a tight noncova-
role in transporting iron around the body to sites where
lent complex (Hb-Hp). The amount of haptoglobin in
human plasma ranges from 40 mg to 180 mg of hemo-
globin-binding capacity per deciliter. Approximately
10% of the hemoglobin that is degraded each day is re-
A.
Hb → Kidney → Excreted in urine or precipitates in tubules;
leased into the circulation and is thus extracorpuscular.
(MW 65,000)
iron is lost to body
The other 90% is present in old, damaged red blood
B.
Hb
+ Hp → Hb : Hp complex → Kidney
cells, which are degraded by cells of the histiocytic sys-
(MW 65,000)
(MW 90,000)
(MW 155,000)
tem. The molecular mass of hemoglobin is approxi-
mately 65 kDa, whereas the molecular mass of the sim-
Catabolized by liver cells;
plest polymorphic form of haptoglobin (Hp 1-1) found
iron is conserved and reused
in humans is approximately 90 kDa. Thus, the Hb-Hp
complex has a molecular mass of approximately 155
Figure 50-3. Different fates of free hemoglobin and
kDa. Free hemoglobin passes through the glomerulus
of the hemoglobin-haptoglobin complex.
PLASMA PROTEINS & IMMUNOGLOBULINS
/
585
it is needed. Before we discuss it further, certain aspects
Table 50-3. Distribution of iron in a 70-kg
of iron metabolism will be reviewed.
adult male.1
Iron is important in the human body because of its
occurrence in many hemoproteins such as hemoglobin,
Transferrin
3-4 mg
myoglobin, and the cytochromes. It is ingested in the
Hemoglobin in red blood cells
2500 mg
diet either as heme or nonheme iron (Figure 50-4); as
In myoglobin and various enzymes
300 mg
shown, these different forms involve separate pathways.
In stores (ferritin and hemosiderin)
1000 mg
Absorption of iron in the proximal duodenum is tightly
Absorption
1 mg/d
regulated, as there is no physiologic pathway for its ex-
Losses
1 mg/d
cretion from the body. Under normal circumstances,
1In an adult female of similar weight, the amount in stores would
the body guards its content of iron zealously, so that a
generally be less (100-400 mg) and the losses would be greater
healthy adult male loses only about 1 mg/d, which is re-
(1.5-2 mg/d).
placed by absorption. Adult females are more prone to
states of iron deficiency because some may lose excessive
blood during menstruation. The amounts of iron in var-
ious body compartments are shown in Table 50-3.
brane into the plasma, where it is carried by transferrin
Enterocytes in the proximal duodenum are responsi-
(see below). Passage across the basolateral membrane
ble for absorption of iron. Incoming iron in the Fe3+
appears to be carried out by another protein, possibly
state is reduced to Fe2+ by a ferrireductase present on
iron regulatory protein 1 (IREG1). This protein may
the surface of enterocytes. Vitamin C in food also favors
interact with the copper-containing protein hephaestin,
reduction of ferric iron to ferrous iron. The transfer of
a protein similar to ceruloplasmin (see below). Hepha-
iron from the apical surfaces of enterocytes into their in-
estin is thought to have a ferroxidase activity, which is
teriors is performed by a proton-coupled divalent metal
important in the release of iron from cells. Thus, Fe2+ is
transporter
(DMT1). This protein is not specific for
converted back to Fe3+, the form in which it is trans-
iron, as it can transport a wide variety of divalent cations.
ported in the plasma by transferrin.
Once inside an enterocyte, iron can either be stored
Overall regulation of iron absorption is complex
as ferritin or transferred across the basolateral mem-
and not well understood mechanistically. It occurs at
Brush
border
Intestinal
Enterocyte
Blood
lumen
Heme
HT
Heme
HO
HP
Fe3+
Fe2+
Fe2+
Fe2+
reductase
FP
Fe2+
DMT1
Fe2+
Fe3+-ferritin
Fe3+
Shed
Fe3+−TF
Figure 50-4. Absorption of iron. Fe3+ is converted to Fe2+ by ferric reductase,
and Fe2+ is transported into the enterocyte by the apical membrane iron trans-
porter DMT1. Heme is transported into the enterocyte by a separate heme
transporter (HT), and heme oxidase (HO) releases Fe2+ from the heme. Some of
the intracellular Fe2+ is converted to Fe3+ and bound by ferritin. The remainder
binds to the basolateral Fe2+ transporter (FP) and is transported into the blood-
stream, aided by hephaestin (HP). In plasma, Fe3+ is bound to the iron transport
protein transferrin (TF). (Reproduced, with permission, from Ganong WF: Review of
Medical Physiology, 21st ed. McGraw-Hill, 2003.)
586
/
CHAPTER 50
the level of the enterocyte, where further absorption of
the protein is normally only one-third saturated with
iron is blocked if a sufficient amount has been taken up
iron. In iron deficiency anemia, the protein is even less
(so-called dietary regulation exerted by
“mucosal
saturated with iron, whereas in conditions of storage of
block”). It also appears to be responsive to the overall
excess iron in the body (eg, hemochromatosis) the satu-
requirement of erythropoiesis for iron (erythropoietic
ration with iron is much greater than one-third.
regulation). Absorption is excessive in hereditary he-
mochromatosis (see below).
Ferritin Stores Iron in Cells
Ferritin is another protein that is important in the me-
Transferrin Shuttles Iron to Sites
tabolism of iron. Under normal conditions, it stores
Where It Is Needed
iron that can be called upon for use as conditions re-
Transferrin (Tf ) is a β1-globulin with a molecular mass
quire. In conditions of excess iron (eg, hemochromato-
of approximately 76 kDa. It is a glycoprotein and is
sis), body stores of iron are greatly increased and much
synthesized in the liver. About 20 polymorphic forms
more ferritin is present in the tissues, such as the liver
of transferrin have been found. It plays a central role in
and spleen. Ferritin contains approximately 23% iron,
the body’s metabolism of iron because it transports iron
and apoferritin (the protein moiety free of iron) has a
(2 mol of Fe3+ per mole of Tf) in the circulation to sites
molecular mass of approximately 440 kDa. Ferritin is
where iron is required, eg, from the gut to the bone
composed of 24 subunits of 18.5 kDa, which surround
marrow and other organs. Approximately 200 billion
in a micellar form some 3000-4500 ferric atoms. Nor-
red blood cells (about 20 mL) are catabolized per day,
mally, there is a little ferritin in human plasma. How-
releasing about 25 mg of iron into the body—most of
ever, in patients with excess iron, the amount of ferritin
which will be transported by transferrin.
in plasma is markedly elevated. The amount of ferritin
There are receptors (TfRs) on the surfaces of many
in plasma can be conveniently measured by a sensitive
cells for transferrin. It binds to these receptors and is in-
and specific radioimmunoassay and serves as an index
ternalized by receptor-mediated endocytosis (compare
of body iron stores.
the fate of LDL; Chapter 25). The acid pH inside the
Synthesis of the transferrin receptor (TfR) and that
lysosome causes the iron to dissociate from the protein.
of ferritin are reciprocally linked to cellular iron con-
The dissociated iron leaves the endosome via DMT1 to
tent. Specific untranslated sequences of the mRNAs for
enter the cytoplasm. Unlike the protein component of
both proteins (named iron response elements) interact
LDL, apoTf is not degraded within the lysosome. In-
with a cytosolic protein sensitive to variations in levels
stead, it remains associated with its receptor, returns to
of cellular iron (iron-responsive element-binding pro-
the plasma membrane, dissociates from its receptor,
tein). When iron levels are high, cells use stored ferritin
reenters the plasma, picks up more iron, and again de-
mRNA to synthesize ferritin, and the TfR mRNA is de-
livers the iron to needy cells.
graded. In contrast, when iron levels are low, the TfR
Abnormalities of the glycosylation of transferrin
mRNA is stabilized and increased synthesis of receptors
occur in the congenital disorders of glycosylation
occurs, while ferritin mRNA is apparently stored in an
(Chapter 47) and in chronic alcohol abuse. Their detec-
inactive form. This is an important example of control
tion by, for example, isoelectric focusing is used to help
of expression of proteins at the translational level.
diagnose these conditions.
Hemosiderin is a somewhat ill-defined molecule; it
appears to be a partly degraded form of ferritin but still
containing iron. It can be detected by histologic stains
Iron Deficiency Anemia
(eg, Prussian blue) for iron, and its presence is deter-
Is Extremely Prevalent
mined histologically when excessive storage of iron
Attention to iron metabolism is particularly impor-
occurs.
tant in women for the reason mentioned above. Addi-
tionally, in pregnancy, allowances must be made for
Hereditary Hemochromatosis Is Due
the growing fetus. Older people with poor dietary
to Mutations in the HFE Gene
habits (“tea and toasters”) may develop iron deficiency.
Iron deficiency anemia due to inadequate intake, inade-
Hereditary (primary) hemochromatosis is a very preva-
quate utilization, or excessive loss of iron is one of the
lent autosomal recessive disorder in certain parts of the
most prevalent conditions seen in medical practice.
world (eg, Scotland, Ireland, and North America). It is
The concentration of transferrin in plasma is approx-
characterized by excessive storage of iron in tissues, lead-
imately 300 mg/dL. This amount of transferrin can
ing to tissue damage. Total body iron ranges between
bind 300 µg of iron per deciliter, so that this represents
2.5 g and 3.5 g in normal adults; in primary hemochro-
the total iron-binding capacity of plasma. However,
matosis it usually exceeds 15 g. The accumulated iron
PLASMA PROTEINS & IMMUNOGLOBULINS
/
587
damages organs and tissues such as the liver, pancreatic
Mutations in HFE, located on chromosome 6p21.3,
islets, and heart, perhaps in part due to effects on free
leading to abnormalities in the structure
radical production (Chapter 52). Melanin and various
of its protein product
amounts of iron accumulate in the skin, accounting for
the slate-gray color often seen. The precise cause of
Loss of regulation of absorption of iron
melanin accumulation is not clear. The frequent coexis-
in the small intestine
tence of diabetes mellitus (due to islet damage) and the
skin pigmentation led to use of the term bronze dia-
betes for this condition. In 1995, Feder and colleagues
Accumulation of iron in various tissues, but particularly
isolated a gene, now known as HFE, located on chromo-
liver, pancreatic islets, skin, and heart muscle
some 6 close to the major histocompatibility complex
genes. The encoded protein (HFE) was found to be re-
Iron directly or indirectly causes damage to the
lated to MHC class 1 antigens. Initially, two different
above tissues, resulting in hepatic cirrhosis, diabetes
missense mutations were found in HFE in individuals
mellitus, skin pigmentation, and cardiac problems
with hereditary hemochromatosis. The more frequent
mutation was one that changed cysteinyl residue 282 to
Figure 50-5. Tentative scheme of the main events
a tyrosyl residue (CY282Y), disrupting the structure of
in causation of primary hemochromatosis (MIM
the protein. The other mutation changed histidyl resi-
235200). The two principal mutations are CY282Y and
due 63 to an aspartyl residue (H63D). Some patients
H63D (see text). Mutations in genes other than HFE are
with hereditary hemochromatosis have neither muta-
also involved in some cases.
tion, perhaps due to other mutations in HFE or because
one or more other genes may be involved in its causa-
tion. Genetic screening for this condition has been eval-
uated but is not presently recommended. However, test-
carries 90% of the copper present in plasma. Each mol-
ing for HFE mutations in individuals with elevated
ecule of ceruloplasmin binds six atoms of copper very
serum iron concentrations may be useful.
tightly, so that the copper is not readily exchangeable.
HFE has been shown to be located in cells in the
Albumin carries the other 10% of the plasma copper
crypts of the small intestine, the site of iron absorption.
but binds the metal less tightly than does ceruloplas-
There is evidence that it associates with β2-microglobu-
min. Albumin thus donates its copper to tissues more
lin, an association that may be necessary for its stability,
readily than ceruloplasmin and appears to be more im-
intracellular processing, and cell surface expression. The
portant than ceruloplasmin in copper transport in the
complex interacts with the transferrin receptor (TfR);
human body. Ceruloplasmin exhibits a copper-depen-
how this leads to excessive storage of iron when HFE is
dent oxidase activity, but its physiologic significance
altered by mutation is under close study. The mouse
has not been clarified. The amount of ceruloplasmin in
homolog of HFE has been knocked out, resulting in a
plasma is decreased in liver disease. In particular, low
potentially useful animal model of hemochromatosis.
levels of ceruloplasmin are found in Wilson disease
A scheme of the likely main events in the causation of
(hepatolenticular degeneration), a disease due to abnor-
hereditary hemochromatosis is set forth in Figure 50-5.
mal metabolism of copper. In order to clarify the de-
Secondary hemochromatosis can occur after re-
scription of Wilson disease, we shall first consider the
peated transfusions (eg, for treatment of sickle cell ane-
metabolism of copper in the human body and then
mia), excessive oral intake of iron (eg, by African Bantu
Menkes disease, another condition involving abnormal
peoples who consume alcoholic beverages fermented in
copper metabolism.
containers made of iron), or a number of other condi-
tions.
Table 50-4 summarizes laboratory tests useful in the
assessment of patients with abnormalities of iron me-
Table 50-4. Laboratory tests for assessing
tabolism.
patients with disorders of iron metabolism.
• Red blood cell count and estimation of hemoglobin
Ceruloplasmin Binds Copper, & Low Levels
• Determinations of plasma iron, total iron-binding capacity
of This Plasma Protein Are Associated
(TIBC), and % transferrin saturation
With Wilson Disease
• Determination of ferritin in plasma by radioimmunoassay
• Prussian blue stain of tissue sections
Ceruloplasmin (about 160 kDa) is an α2-globulin. It
• Determination of amount of iron (µg/g) in a tissue biopsy
has a blue color because of its high copper content and
588
/
CHAPTER 50
Copper Is a Cofactor for Certain Enzymes
only male infants, involves the nervous system, connec-
tive tissue, and vasculature, and is usually fatal in in-
Copper is an essential trace element. It is required in
fancy. In 1993, it was reported that the basis of Menkes
the diet because it is the metal cofactor for a variety of
disease was mutations in the gene for a copper-binding
enzymes (see Table 50-5). Copper accepts and donates
P-type ATPase. Interestingly, the enzyme showed struc-
electrons and is involved in reactions involving dismu-
tural similarity to certain metal-binding proteins in mi-
tation, hydroxylation, and oxygenation. However, ex-
croorganisms. This ATPase is thought to be responsible
cess copper can cause problems because it can oxidize
for directing the efflux of copper from cells. When al-
proteins and lipids, bind to nucleic acids, and enhance
tered by mutation, copper is not mobilized normally
the production of free radicals. It is thus important to
from the intestine, in which it accumulates, as it does in
have mechanisms that will maintain the amount of
a variety of other cells and tissues, from which it cannot
copper in the body within normal limits. The body of
exit. Despite the accumulation of copper, the activities
the normal adult contains about 100 mg of copper, lo-
of many copper-dependent enzymes are decreased, per-
cated mostly in bone, liver, kidney, and muscle. The
haps because of a defect of its incorporation into the
daily intake of copper is about 2-4 mg, with about
apoenzymes. Normal liver expresses very little of the
50% being absorbed in the stomach and upper small
ATPase, which explains the absence of hepatic involve-
intestine and the remainder excreted in the feces. Cop-
ment in Menkes disease. This work led to the sugges-
per is carried to the liver bound to albumin, taken up
tion that liver might contain a different copper-binding
by liver cells, and part of it is excreted in the bile. Cop-
ATPase, which could be involved in the causation of
per also leaves the liver attached to ceruloplasmin,
Wilson disease. As described below, this turned out to
which is synthesized in that organ.
be the case.
The Tissue Levels of Copper & of Certain
Wilson Disease Is Also Due to Mutations
Other Metals Are Regulated in
in a Gene Encoding a Copper-Binding
Part by Metallothioneins
P-Type ATPase
Metallothioneins are a group of small proteins (about
Wilson disease is a genetic disease in which copper fails
6.5 kDa), found in the cytosol of cells, particularly of
to be excreted in the bile and accumulates in liver,
liver, kidney, and intestine. They have a high content of
brain, kidney, and red blood cells. It can be regarded as
cysteine and can bind copper, zinc, cadmium, and mer-
an inability to maintain a near-zero copper balance, re-
cury. The SH groups of cysteine are involved in binding
sulting in copper toxicosis. The increase of copper in
the metals. Acute intake (eg, by injection) of copper and
liver cells appears to inhibit the coupling of copper to
of certain other metals increases the amount (induction)
apoceruloplasmin and leads to low levels of ceruloplas-
of these proteins in tissues, as does administration of
min in plasma. As the amount of copper accumulates,
certain hormones or cytokines. These proteins may
patients may develop a hemolytic anemia, chronic liver
function to store the above metals in a nontoxic form
disease (cirrhosis, hepatitis), and a neurologic syndrome
and are involved in their overall metabolism in the
owing to accumulation of copper in the basal ganglia
body. Sequestration of copper also diminishes the
and other centers. A frequent clinical finding is the
amount of this metal available to generate free radicals.
Kayser-Fleischer ring. This is a green or golden pig-
ment ring around the cornea due to deposition of cop-
Menkes Disease Is Due to Mutations
per in Descemet’s membrane. The major laboratory
in the Gene Encoding a Copper-
tests of copper metabolism are listed in Table 50-6. If
Binding P-Type ATPase
Wilson disease is suspected, a liver biopsy should be
Menkes disease (“kinky” or “steely” hair disease) is a
performed; a value for liver copper of over 250 µg per
disorder of copper metabolism. It is X-linked, affects
gram dry weight along with a plasma level of cerulo-
plasmin of under 20 mg/dL is diagnostic.
The cause of Wilson disease was also revealed in
1993, when it was reported that a variety of mutations
Table 50-5. Some important enzymes that
in a gene encoding a copper-binding P-type ATPase
contain copper.
were responsible. The gene is estimated to encode a
protein of 1411 amino acids, which is highly homolo-
• Amine oxidase
gous to the product of the gene affected in Menkes dis-
• Copper-dependent superoxide dismutase
ease. In a manner not yet fully explained, a nonfunc-
• Cytochrome oxidase
tional ATPase causes defective excretion of copper into
• Tyrosinase
the bile, a reduction of incorporation of copper into
PLASMA PROTEINS & IMMUNOGLOBULINS
/
589
Table 50-6. Major laboratory tests used in the
other proteases by forming complexes with them. At
investigation of diseases of copper metabolism.1
least 75 polymorphic forms occur, many of which can
be separated by electrophoresis. The major genotype is
MM, and its phenotypic product is PiM. There are two
Normal Adult
areas of clinical interest concerning α1-antitrypsin. A de-
Test
Range
ficiency of this protein has a role in certain cases (ap-
Serum copper
10-22 µmol/L
proximately 5%) of emphysema. This occurs mainly in
subjects with the ZZ genotype, who synthesize PiZ, and
Ceruloplasmin
200-600 mg/L
also in PiSZ heterozygotes, both of whom secrete con-
Urinary copper
< 1 µmol/24 h
siderably less protein than PiMM individuals. Consider-
Liver copper
20-50 µg/g dry weight
ably less of this protein is secreted as compared with
PiM. When the amount of α1-antitrypsin is deficient
1Based on Gaw A et al: Clinical Biochemistry. Churchill Livingstone,
and polymorphonuclear white blood cells increase in the
1995.
lung (eg, during pneumonia), the affected individual
lacks a countercheck to proteolytic damage of the lung
by proteases such as elastase (Figure 50-6). It is of con-
apoceruloplasmin, and the accumulation of copper in
siderable interest that a particular methionine (residue
liver and subsequently in other organs such as brain.
358) of α1-antitrypsin is involved in its binding to pro-
Treatment for Wilson disease consists of a diet low
teases. Smoking oxidizes this methionine to methionine
in copper along with lifelong administration of penicil-
sulfoxide and thus inactivates it. As a result, affected
lamine, which chelates copper, is excreted in the urine,
molecules of α1-antitrypsin no longer neutralize
and thus depletes the body of the excess of this mineral.
proteases. This is particularly devastating in patients
Another condition involving ceruloplasmin is aceru-
(eg, PiZZ phenotype) who already have low levels of
loplasminemia. In this genetic disorder, levels of cerulo-
α1-antitrypsin. The further diminution in α1-antitrypsin
plasmin are very low and consequently its ferroxidase ac-
brought about by smoking results in increased proteolytic
tivity is markedly deficient. This leads to failure of release
destruction of lung tissue, accelerating the development
of iron from cells, and iron accumulates in certain brain
of emphysema. Intravenous administration of α1-anti-
cells, hepatocytes, and pancreatic islet cells. Affected indi-
trypsin (augmentation therapy) has been used as an ad-
viduals show severe neurologic signs and have diabetes
junct in the treatment of patients with emphysema due
mellitus. Use of a chelating agent or administration of
to α1-antitrypsin deficiency. Attempts are being made,
plasma or ceruloplasmin concentrate may be beneficial.
using the techniques of protein engineering, to replace
methionine 358 by another residue that would not be
subject to oxidation. The resulting “mutant” α1-anti-
Deficiency of
trypsin would thus afford protection against proteases
1-Antiproteinase
for a much longer period of time than would native
(
1-Antitrypsin) Is Associated
α1-antitrypsin. Attempts are also being made to develop
With Emphysema & One Type
gene therapy for this condition. One approach is to use
of Liver Disease
a modified adenovirus (a pathogen of the respiratory
α1-Antiproteinase (about 52 kDa) was formerly called
tract) into which the gene for α1-antitrypsin has been
α1-antitrypsin, and this name is retained here. It is a
inserted. The virus would then be introduced into the
single-chain protein of 394 amino acids, contains three
respiratory tract (eg, by an aerosol). The hope is that
oligosaccharide chains, and is the major component
pulmonary epithelial cells would express the gene and
(> 90%) of the α1 fraction of human plasma. It is syn-
secrete α1-antitrypsin locally. Experiments in animals
thesized by hepatocytes and macrophages and is the
have indicated the feasibility of this approach.
principal serine protease inhibitor (serpin, or Pi) of
Deficiency of α1-antitrypsin is also implicated in
human plasma. It inhibits trypsin, elastase, and certain
one type of liver disease (α1-antitrypsin deficiency liver
A. Active elastase + α1-AT → Inactive elastase: α1-AT complex → No proteolysis of lung → No tissue damage
B. Active elastase + or no α1-AT → Active elastase → Proteolysis of lung → Tissue damage
Figure 50-6. Scheme illustrating (A) normal inactivation of elastase by α1-antitrypsin and
(B) situation in which the amount of α1-antitrypsin is substantially reduced, resulting in pro-
teolysis by elastase and leading to tissue damage.
590
/
CHAPTER 50
disease). In this condition, molecules of the ZZ pheno-
comprises 8-10% of the total plasma protein in hu-
type accumulate and aggregate in the cisternae of the
mans. Approximately 10% of the zinc in plasma is
endoplasmic reticulum of hepatocytes. Aggregation is
transported by α2-macroglobulin, the remainder being
due to formation of polymers of mutant α1-antitrypsin,
transported by albumin. The protein is synthesized by a
the polymers forming via a strong interaction between a
variety of cell types, including monocytes, hepatocytes,
specific loop in one molecule and a prominent β-
and astrocytes. It is the major member of a group of
pleated sheet in another (loop-sheet polymerization).
plasma proteins that include complement proteins C3
By mechanisms that are not understood, hepatitis re-
and C4. These proteins contain a unique internal cyclic
sults with consequent cirrhosis (accumulation of mas-
thiol ester bond (formed between a cysteine and a glut-
sive amounts of collagen, resulting in fibrosis). It is pos-
amine residue) and for this reason have been designated
sible that administration of a synthetic peptide
as the thiol ester plasma protein family.
resembling the loop sequence could inhibit loop-sheet
α2-Macroglobulin binds many proteinases and is
polymerization. Diseases such as α1-antitrypsin defi-
thus an important panproteinase inhibitor. The α2-
ciency, in which cellular pathology is primarily caused
macroglobulin-proteinase complexes are rapidly cleared
by the presence of aggregates of aberrant forms of indi-
from the plasma by a receptor located on many cell
vidual proteins, have been named conformational dis-
types. In addition, α2-macroglobulin binds many cy-
eases. Most appear to be due to the formation by con-
tokines (platelet-derived growth factor, transforming
formationally unstable proteins of β sheets, which in
growth factor-β, etc) and appears to be involved in tar-
turn leads to formation of aggregates. Other members
geting them toward particular tissues or cells. Once
of this group of conditions include Alzheimer disease,
taken up by cells, the cytokines can dissociate from α2-
Parkinson disease, and Huntington disease.
macroglobulin and subsequently exert a variety of ef-
At present, severe α1-antitrypsin deficiency liver dis-
fects on cell growth and function. The binding of pro-
ease can be successfully treated by liver transplantation.
teinases and cytokines by α2-macroglobulin involves
In the future, introduction of the gene for normal α1-
different mechanisms that will not be considered here.
antitrypsin into hepatocytes may become possible, but
this would not stop production of the PiZ protein. Fig-
Amyloidosis Occurs by the Deposition
ure 50-7 is a scheme of the causation of this disease.
of Fragments of Various Plasma
Proteins in Tissues
2-Macroglobulin Neutralizes Many
Amyloidosis is the accumulation of various insoluble
Proteases & Targets Certain
fibrillar proteins between the cells of tissues to an extent
Cytokines to Tissues
that affects function. The fibrils generally represent
α2-Macroglobulin is a large plasma glycoprotein (720
proteolytic fragments of various plasma proteins and
kDa) made up of four identical subunits of 180 kDa. It
possess a β-pleated sheet structure. The term “amyloi-
dosis” is a misnomer, as it was originally thought that
the fibrils were starch-like in nature. Among the most
common precursor proteins are immunoglobulin light
chains (see below), amyloid-associated protein derived
GAG to AAG mutation in exon 5 of gene for α1-AT
from serum amyloid-associated protein (a plasma glyco-
on chromosome 14
protein), and transthyretin (Table 50-2). The precursor
proteins in plasma are generally either increased in
amount (eg, immunoglobulin light chains in multiple
Results in Glu342 to Lys342 substitution in α1-AT,
myeloma or β2-microglobulin in patients being main-
causing formation of PiZZ
tained on chronic dialysis) or mutant forms (eg, of
transthyretin in familial amyloidotic neuropathies).
PiZZ accumulates in cisternae
The precise factors that determine the deposition of
of endoplasmic reticulum and aggregates
proteolytic fragments in tissues await elucidation.
via loop-sheet polymerization
Other proteins have been found in amyloid fibrils, such
as calcitonin and amyloid β protein (not derived from a
Leads to hepatitis (mechanism unknown)
plasma protein) in Alzheimer disease; a total of about
and cirrhosis in ~10% of ZZ homozygotes
15 different proteins have been found. All fibrils have a
P component associated with them, which is derived
Figure 50-7. Scheme of causation of α1-antitrypsin-
from serum amyloid P component, a plasma protein
deficiency liver disease. The mutation shown causes
closely related to C-reactive protein. Tissue sections
formation of PiZZ (MIM 107400). (α1-AT, α1-antitrypsin.)
containing amyloid fibrils interact with Congo red stain
PLASMA PROTEINS & IMMUNOGLOBULINS
/
591
and display striking green birefringence when viewed
munoglobulins other than direct binding of antigens.
by polarizing microscopy. Deposition of amyloid oc-
Because there are two Fab regions, IgG molecules bind
curs in patients with a variety of disorders; treatment of
two molecules of antigen and are termed divalent. The
the underlying disorder should be provided if possible.
site on the antigen to which an antibody binds is
termed an antigenic determinant, or epitope. The
area in which papain cleaves the immunoglobulin mol-
PLASMA IMMUNOGLOBULINS PLAY
ecule—ie, the region between the CH1 and CH2 do-
A MAJOR ROLE IN THE BODY’S
mains—is referred to as the “hinge region.” The hinge
DEFENSE MECHANISMS
region confers flexibility and allows both Fab arms to
The immune system of the body consists of two major
move independently, thus helping them to bind to
components: B lymphocytes and T lymphocytes. The
antigenic sites that may be variable distances apart (eg,
B lymphocytes are mainly derived from bone marrow
on bacterial surfaces). Fc and hinge regions differ in the
cells in higher animals and from the bursa of Fabricius
different classes of antibodies, but the overall model of
in birds. The T lymphocytes are of thymic origin. The
antibody structure for each class is similar to that
B cells are responsible for the synthesis of circulating,
shown in Figure 50-8 for IgG.
humoral antibodies, also known as immunoglobulins.
The T cells are involved in a variety of important cell-
All Light Chains Are Either Kappa
mediated immunologic processes such as graft rejec-
or Lambda in Type
tion, hypersensitivity reactions, and defense against ma-
lignant cells and many viruses. This section considers
There are two general types of light chains, kappa (κ)
only the plasma immunoglobulins, which are synthe-
and lambda (λ), which can be distinguished on the
sized mainly in plasma cells. These are specialized cells
basis of structural differences in their CL regions. A
of B cell lineage that synthesize and secrete immuno-
given immunoglobulin molecule always contains two κ
globulins into the plasma in response to exposure to a
or two λ light chains—never a mixture of κ and λ. In
variety of antigens.
humans, the κ chains are more frequent than λ chains
in immunoglobulin molecules.
All Immunoglobulins Contain a Minimum
of Two Light & Two Heavy Chains
The Five Types of Heavy Chain Determine
Immunoglobulin Class
Immunoglobulins contain a minimum of two iden-
tical light (L) chains (23 kDa) and two identical heavy
Five classes of H chain have been found in humans
(H) chains (53-75 kDa), held together as a tetramer
(Table 50-7), distinguished by differences in their CH
(L2H2) by disulfide bonds The structure of IgG is
regions. They are designated γ, α, µ δ, and ε. The µ
shown in Figure 50-8; it is Y-shaped, with binding of
and ε chains each have four CH domains rather than
antigen occurring at both tips of the Y. Each chain can
the usual three. The type of H chain determines the
be divided conceptually into specific domains, or re-
class of immunoglobulin and thus its effector function.
gions, that have structural and functional significance.
There are thus five immunoglobulin classes: IgG, IgA,
The half of the light (L) chain toward the carboxyl ter-
IgM, IgD, and IgE. The biologic functions of these
minal is referred to as the constant region (CL), while
five classes are summarized in Table 50-8.
the amino terminal half is the variable region of the
light chain
(VL). Approximately one-quarter of the
No Two Variable Regions Are Identical
heavy (H) chain at the amino terminals is referred to as
its variable region (VH), and the other three-quarters of
The variable regions of immunoglobulin molecules
the heavy chain are referred to as the constant regions
consist of the VL and VH domains and are quite hetero-
(CH1, CH2, CH3) of that H chain. The portion of the
geneous. In fact, no two variable regions from different
immunoglobulin molecule that binds the specific anti-
humans have been found to have identical amino acid
gen is formed by the amino terminal portions (variable
sequences. However, amino acid analyses have shown
regions) of both the H and L chains—ie, the VH and
that the variable regions are comprised of relatively in-
VL domains. The domains of the protein chains consist
variable regions and other hypervariable regions (Figure
of two sheets of antiparallel distinct stretches of amino
50-9). L chains have three hypervariable regions (in
acids that bind antigen.
VL) and H chains have four (in VH). These hypervari-
As depicted in Figure 50-8, digestion of an im-
able regions comprise the antigen-binding site (located
munoglobulin by the enzyme papain produces two
at the tips of the Y shown in Figure 50-8) and dictate
antigen-binding fragments (Fab) and one crystallizable
the amazing specificity of antibodies. For this reason,
fragment (Fc), which is responsible for functions of im-
hypervariable regions are also termed complementar-
592
/
CHAPTER 50
+H3N
VL
Fab
+H3N
CL
Hinge region
VH
FC
CH2
CH3
CH1
S
S
S S
COO—
S S
H chain
S
S
H chain
COO—
S S
S S
Pepsin
Cleavage sites
Papain
+H3N
Fab
+H3N
Figure 50-8. Structure of IgG. The molecule consists of two light (L) chains and
two heavy (H) chains. Each light chain consists of a variable (VL) and a constant (CL)
region. Each heavy chain consists of a variable region (VH) and a constant region
that is divided into three domains (CH1, CH2, and CH3). The CH2 domain contains
the complement-binding site and the CH3 domain contains a site that attaches to
receptors on neutrophils and macrophages. The antigen-binding site is formed by
the hypervariable regions of both the light and heavy chains, which are located in
the variable regions of these chains (see Figure 50-9). The light and heavy chains
are linked by disulfide bonds, and the heavy chains are also linked to each other
by disulfide bonds. (Reproduced, with permission, from Parslow TG et al [editors]:
Medical Immunology, 10th ed. McGraw-Hill, 2001.)
ity-determining regions (CDRs). About five to ten
specificities, a feature that contributes to the tremen-
amino acids in each hypervariable region (CDR) con-
dous diversity of antibody molecules and is termed
tribute to the antigen-binding site. CDRs are located
combinatorial diversity. Large antigens interact with
on small loops of the variable domains, the surrounding
all of the CDRs of an antibody, whereas small ligands
polypeptide regions between the hypervariable regions
may interact with only one or a few CDRs that form a
being termed framework regions. CDRs from both
pocket or groove in the antibody molecule. The essence
VH and VL domains, brought together by folding of the
of antigen-antibody interactions is mutual complemen-
polypeptide chains in which they are contained, form a
tarity between the surfaces of CDRs and epitopes. The
single hypervariable surface comprising the antigen-
interactions between antibodies and antigens involve
binding site. Various combinations of H and L chain
noncovalent forces and bonds (electrostatic and van der
CDRs can give rise to many antibodies of different
Waals forces and hydrogen and hydrophobic bonds).
PLASMA PROTEINS & IMMUNOGLOBULINS
/
593
Table 50-7. Properties of human immunoglobulins.1
Property
IgG
IgA
IgM
IgD
IgE
Percentage of total immunoglo-
75
15
9
0.2
0.004
bulin in serum (approximate)
Serum concentration
1000
200
120
3
0.05
(mg/dL) (approximate)
Sedimentation coefficient
7S
7S or 11S2
19S
7S
8S
Molecular weight
150
170 or
900
180
190
(× 1000)
4002
Structure
Monomer
Monomer or dimer
Monomer or dimer
Monomer
Monomer
H-chain symbol
γ
α
µ
δ
ε
Complement fixation
+
−
+
−
−
Transplacental passage
+
−
−
?
−
Mediation of allergic responses
−
−
−
−
+
Found in secretions
−
+
−
−
−
Opsonization
+
−
−3
−
−
Antigen receptor on B cell
−
−
+
?
−
Polymeric form contains J chain
−
+
+
−
−
1Reproduced, with permission, from Levinson W, Jawetz E: Medical Microbiology and Immunology, 7th ed. McGraw-Hill,
2002.
2The 11S form is found in secretions (eg, saliva, milk, tears) and fluids of the respiratory, intestinal, and genital tracts.
3IgM opsonizes indirectly by activating complement. This produces C3b, which is an opsonin.
The Constant Regions Determine
(VL) gene, a joining region (J) gene (bearing no rela-
Class-Specific Effector Functions
tionship to the J chain of IgA or IgM), and a constant
region (CL) gene. Each heavy chain is the product of at
The constant regions of the immunoglobulin molecules,
least four different genes: a variable region (VH) gene, a
particularly the CH2 and CH3 (and CH4 of IgM and
diversity region (D) gene, a joining region (J) gene, and
IgE), which constitute the Fc fragment, are responsible
a constant region (CH) gene. Thus, the “one gene, one
for the class-specific effector functions of the different
protein” concept is not valid. The molecular mecha-
immunoglobulin molecules (Table 50-7, bottom part),
nisms responsible for the generation of the single im-
eg, complement fixation or transplacental passage.
munoglobulin chains from multiple structural genes are
Some immunoglobulins such as immune IgG exist
discussed in Chapters 36 and 39.
only in the basic tetrameric structure, while others such
as IgA and IgM can exist as higher order polymers of
two, three (IgA), or five (IgM) tetrameric units (Figure
Antibody Diversity Depends
50-10).
on Gene Rearrangements
The L chains and H chains are synthesized as sepa-
rate molecules and are subsequently assembled within
Each person is capable of generating antibodies directed
the B cell or plasma cell into mature immunoglobulin
against perhaps 1 million different antigens. The gener-
molecules, all of which are glycoproteins.
ation of such immense antibody diversity depends
upon a number of factors including the existence of
multiple gene segments (V, C, J, and D segments),
Both Light & Heavy Chains Are Products
their recombinations (see Chapters 36 and 39), the
of Multiple Genes
combinations of different L and H chains, a high fre-
Each immunoglobulin light chain is the product of at
quency of somatic mutations in immunoglobulin genes,
least three separate structural genes: a variable region
and junctional diversity. The latter reflects the addi-
594
/
CHAPTER 50
Table 50-8. Major functions of
Light chain
1
hypervariable
immunoglobulins.
regions
Immunoglobulin
Major Functions
IgG
Main antibody in the secondary re-
sponse. Opsonizes bacteria, making
Interchain
them easier to phagocytose. Fixes com-
disulfide
plement, which enhances bacterial
bonds
killing. Neutralizes bacterial toxins and
viruses. Crosses the placenta.
Heavy chain
hypervariable
IgA
Secretory IgA prevents attachment of
regions
bacteria and viruses to mucous mem-
Intrachain
branes. Does not fix complement.
disulfide
bonds
IgM
Produced in the primary response to an
antigen. Fixes complement. Does not
cross the placenta. Antigen receptor on
the surface of B cells.
IgD
Uncertain. Found on the surface of
many B cells as well as in serum.
IgE
Mediates immediate hypersensitivity by
causing release of mediators from mast
cells and basophils upon exposure to
Figure 50-9.
Schematic model of an IgG molecule
antigen (allergen). Defends against
showing approximate positions of the hypervariable re-
worm infections by causing release of
gions in heavy and light chains. The antigen-binding
enzymes from eosinophils. Does not fix
site is formed by these hypervariable regions. The hy-
complement. Main host defense against
pervariable regions are also called complementarity-
helminthic infections.
determining regions (CDRs). (Modified and reproduced,
1Reproduced, with permission, from Levinson W, Jawetz E: Med-
with permission, from Parslow TG et al [editors]: Medical
ical Microbiology and Immunology, 7th ed. McGraw-Hill, 2002.
Immunology, 10th ed. McGraw-Hill, 2001.)
tion or deletion of a random number of nucleotides
generate an IgG molecule with antigen specificity iden-
when certain gene segments are joined together, and in-
tical to that of the original IgM molecule. The same
troduces an additional degree of diversity. Thus, the
light chain can also combine with an α heavy chain,
above factors ensure that a vast number of antibodies
again containing the identical VH region, to form an IgA
can be synthesized from several hundred gene segments.
molecule with identical antigen specificity. These three
classes (IgM, IgG, and IgA) of immunoglobulin mole-
Class (Isotype) Switching Occurs
cules against the same antigen have identical variable do-
mains of both their light (VL ) chains and heavy (VH)
During Immune Responses
chains and are said to share an idiotype. (Idiotypes are
In most humoral immune responses, antibodies with
the antigenic determinants formed by the specific amino
identical specificity but of different classes are generated
acids in the hypervariable regions.) The different classes
in a specific chronologic order in response to the im-
of these three immunoglobulins (called isotypes) are
munogen (immunizing antigen). For instance, antibod-
thus determined by their different CH regions, which are
ies of the IgM class normally precede molecules of the
combined with the same antigen-specific VH regions.
IgG class. The switch from one class to another is desig-
nated “class or isotype switching,” and its molecular
Both Over- & Underproduction
basis has been investigated extensively. A single type of
of Immunoglobulins May Result
immunoglobulin light chain can combine with an anti-
in Disease States
gen-specific µ chain to generate a specific IgM molecule.
Subsequently, the same antigen-specific light chain
Disorders of immunoglobulins include increased pro-
combines with a γ chain with an identical VH region to
duction of specific classes of immunoglobulins or even
PLASMA PROTEINS & IMMUNOGLOBULINS
/
595
L
L
H
H
Monomer
Dimer
A. Serum lgA
H
J chain
H
L
L
L
H
B. Secretory lgA
(dimer)
J chain
H
Secretory
L
component
L
H
J chain
Figure 50-10. Schematic repre-
sentation of serum IgA, secretory
IgA, and IgM. Both IgA and IgM have C. lgM
(pentamer)
a J chain, but only secretory IgA has
H
a secretory component. Polypeptide
L
chains are represented by thick lines;
disulfide bonds linking different
polypeptide chains are represented
by thin lines. (Reproduced, with per-
mission, from Parslow TG et al [edi-
tors]: Medical Immunology, 10th ed.
McGraw-Hill, 2001.)
specific immunoglobulin molecules, the latter by clonal
Hybridomas Provide Long-Term Sources
tumors of plasma cells called myelomas. Multiple
of Highly Useful Monoclonal Antibodies
myeloma is a neoplastic condition; electrophoresis of
serum or urine will usually reveal a large increase of one
When an antigen is injected into an animal, the result-
particular immunoglobulin or one particular light chain
ing antibodies are polyclonal, being synthesized by a
(the latter termed a Bence Jones protein). Decreased
mixture of B cells. Polyclonal antibodies are directed
production may be restricted to a single class of im-
against a number of different sites (epitopes or determi-
munoglobulin molecules (eg, IgA or IgG) or may in-
nants) on the antigen and thus are not monospecific.
volve underproduction of all classes of immunoglobu-
However, by means of a method developed by Kohler
lins (IgA, IgD, IgE, IgG, and IgM). A severe reduction
and Milstein, large amounts of a single monoclonal an-
in synthesis of an immunoglobulin class due to a ge-
tibody specific for one epitope can be obtained.
netic abnormality can result in a serious immunodefi-
The method involves cell fusion, and the resulting
ciency disease—eg, agammaglobulinemia, in which
permanent cell line is called a hybridoma. Typically, B
production of IgG is markedly affected—because of
cells are obtained from the spleen of a mouse (or other
impairment of the body’s defense against microorgan-
suitable animal) previously injected with an antigen or
isms.
mixture of antigens (eg, foreign cells). The B cells are
596
/
CHAPTER 50
mixed with mouse myeloma cells and exposed to poly-
tery of monoclonal antibodies can be obtained, many of
ethylene glycol, which causes cell fusion. A summary of
which are specific for individual components of the im-
the principles involved in generating hybridoma cells is
munogenic mixture. The hybridoma cells can be frozen
given in Figure 50-11. Under the conditions used, only
and stored and subsequently thawed when more of the
the hybridoma cells multiply in cell culture. This in-
antibody is required; this ensures its long-term supply.
volves plating the hybrid cells into hypoxanthine-
The hybridoma cells can also be grown in the abdomen
aminopterin-thymidine (HAT)-containing medium at
of mice, providing relatively large supplies of anti-
a concentration such that each dish contains approxi-
bodies.
mately one cell. Thus, a clone of hybridoma cells mul-
Because of their specificity, monoclonal antibodies
tiplies in each dish. The culture medium is harvested
have become useful reagents in many areas of biology
and screened for antibodies that react with the original
and medicine. For example, they can be used to mea-
antigen or antigens. If the immunogen is a mixture of
sure the amounts of many individual proteins
(eg,
many antigens (eg, a cell membrane preparation), an
plasma proteins), to determine the nature of infectious
individual culture dish will contain a clone of hy-
agents (eg, types of bacteria), and to subclassify both
bridoma cells synthesizing a monoclonal antibody to
normal (eg, lymphocytes) and tumor cells (eg, leukemic
one specific antigenic determinant of the mixture. By
cells). In addition, they are being used to direct thera-
harvesting the media from many culture dishes, a bat-
peutic agents to tumor cells and also to accelerate re-
moval of drugs from the circulation when they reach
toxic levels (eg, digoxin).
Myeloma cell
B cell
The Complement System Comprises About
20 Plasma Proteins & Is Involved in Cell
Lysis, Inflammation, & Other Processes
Plasma contains approximately 20 proteins that are
Fused in presence of PEG
members of the complement system. This system was
discovered when it was observed that addition of fresh
Hybridoma cell
serum containing antibodies directed to a bacterium
caused its lysis. Unlike antibodies, the factor was labile
Grown in presence of HAT medium
when heated at 56 °C. Subsequent work has resolved
Hybridoma multiplies; myeloma and B cells die
the proteins of the system and how they function; most
have been cloned and sequenced. The major protein
components are designated C1-9, with C9 associated
Hybridoma cell
with the C5-8 complex
(together constituting the
Figure 50-11. Scheme of production of a hy-
membrane attack complex) being involved in generat-
bridoma cell. The myeloma cells are immortalized, do
ing a lipid-soluble pore in the cell membrane that
not produce antibody, and are HGPRT- (rendering the
causes osmotic lysis.
The details of this system are relatively complex, and
salvage pathway of purine synthesis [Chapter 34] inac-
a textbook of immunology should be consulted. The
tive). The B cells are not immortalized, each produces a
basic concept is that the normally inactive proteins of
specific antibody, and they are HGPRT+. Polyethylene
the system, when triggered by a stimulus, become acti-
glycol (PEG) stimulates cell fusion. The resulting hy-
vated by proteolysis and interact in a specific sequence
bridoma cells are immortalized (via the parental
with one or more of the other proteins of the system.
myeloma cells), produce antibody, and are HGPRT+
This results in cell lysis and generation of peptide or
(both latter properties gained from the parental B cells).
polypeptide fragments that are involved in various as-
The B cells will die in the medium because they are not
pects of inflammation (chemotaxis, phagocytosis, etc).
immortalized. In the presence of HAT, the myeloma
The system has other functions, such as clearance of
cells will also die, since the aminopterin in HAT sup-
antigen-antibody complexes from the circulation. Acti-
presses purine synthesis by the de novo pathway by in-
vation of the complement system is triggered by one of
hibiting reutilization of tetrahydrofolate (Chapter 34).
two routes, called the classic and the alternative path-
However, the hybridoma cells will survive, grow (be-
ways. The first involves interaction of C1 with antigen-
cause they are HGPRT+), and—if cloned—produce
antibody complexes, and the second (not involving an-
monoclonal antibody. (HAT, hypoxanthine,
tibody) involves direct interaction of bacterial cell
aminopterin, and thymidine; HGPRT, hypoxanthine-
surfaces or polysaccharides with a component desig-
guanine phosphoribosyl transferase.)
nated C3b.
PLASMA PROTEINS & IMMUNOGLOBULINS
/
597
The complement system resembles blood coagula-
trophils. Genetic deficiency of this protein is a cause
tion (Chapter 51) in that it involves both conversion of
of emphysema and can also lead to liver disease.
inactive precursors to active products by proteases and a
• α2-Macroglobulin is a major plasma protein that
cascade with amplification.
neutralizes many proteases and targets certain cy-
tokines to specific organs.
SUMMARY
• Immunoglobulins play a key role in the defense
mechanisms of the body, as do proteins of the com-
•
Plasma contains many proteins with a variety of
plement system. Some of the principal features of
functions. Most are synthesized in the liver and are
these proteins are described.
glycosylated.
•
Albumin, which is not glycosylated, is the major pro-
tein and is the principal determinant of intravascular
osmotic pressure; it also binds many ligands, such as
REFERENCES
drugs and bilirubin.
•
Haptoglobin binds extracorpuscular hemoglobin,
Andrews NC: Disorders of iron metabolism. N Engl J Med
1999;341:1986.
prevents its loss into the kidney and urine, and hence
Carrell RW, Lomas DA: Alpha1-antitrypsin deficiency—a model
preserves its iron for reutilization.
for conformational diseases. N Engl J Med 2002;346:45.
•
Transferrin binds iron, transporting it to sites where
Gabay C, Kushner I: Acute-phase proteins and other systemic re-
it is required. Ferritin provides an intracellular store
sponses to inflammation. New Engl J Med 1999;340:448.
of iron. Iron deficiency anemia is a very prevalent
Harris ED: Cellular copper transport and metabolism. Annu Rev
disorder. Hereditary hemochromatosis has been
Nutr 2000;20:291.
shown to be due to mutations in HFE, a gene encod-
Langlois MR, Delanghe JR: Biological and clinical significance of
ing the protein HFE, which appears to play an im-
haptoglobin polymorphism in humans. Clin Chem 1996;
portant role in absorption of iron.
2:1589.
•
Ceruloplasmin contains substantial amounts of cop-
Levinson W, Jawetz E: Medical Microbiology and Immunology, 6th
ed. Appleton & Lange, 2000.
per, but albumin appears to be more important with
Parslow TG et al (editors): Medical Immunology, 10th ed. Appleton
regard to its transport. Both Wilson disease and
& Lange, 2001.
Menkes disease, which reflect abnormalities of copper
Pepys MB, Berger A: The renaissance of C reactive protein. BMJ
metabolism, have been found to be due to mutations
2001;322:4.
in genes encoding copper-binding P-type ATPases.
Waheed A et al: Regulation of transferrin-mediated iron uptake by
•
α1-Antitrypsin is the major serine protease inhibitor
HFE, the protein defective in hereditary hemochromatosis.
of plasma, in particular inhibiting the elastase of neu-
Proc Natl Acad U S A 2002;99:3117.
Hemostasis & Thrombosis
51
Margaret L. Rand, PhD, & Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
There Are Three Types of Thrombi
Basic aspects of the proteins of the blood coagulation
Three types of thrombi or clots are distinguished. All
system and of fibrinolysis are described in this chapter.
three contain fibrin in various proportions.
Some fundamental aspects of platelet biology are also
(1) The white thrombus is composed of platelets and
presented. Hemorrhagic and thrombotic states can
fibrin and is relatively poor in erythrocytes. It
cause serious medical emergencies, and thromboses in
forms at the site of an injury or abnormal vessel
the coronary and cerebral arteries are major causes of
wall, particularly in areas where blood flow is
death in many parts of the world. Rational manage-
rapid (arteries).
ment of these conditions requires a clear understanding
of the bases of blood clotting and fibrinolysis.
(2) The red thrombus consists primarily of red cells
and fibrin. It morphologically resembles the clot
formed in a test tube and may form in vivo in
HEMOSTASIS & THROMBOSIS HAVE
areas of retarded blood flow or stasis (eg, veins)
THREE COMMON PHASES
with or without vascular injury, or it may form at
a site of injury or in an abnormal vessel in con-
Hemostasis is the cessation of bleeding from a cut or
junction with an initiating platelet plug.
severed vessel, whereas thrombosis occurs when the en-
(3) A third type is a disseminated fibrin deposit in
dothelium lining blood vessels is damaged or removed
very small blood vessels or capillaries.
(eg, upon rupture of an atherosclerotic plaque). These
processes encompass blood clotting (coagulation) and
We shall first describe the coagulation pathway lead-
involve blood vessels, platelet aggregation, and plasma
ing to the formation of fibrin. Then we shall briefly de-
proteins that cause formation or dissolution of platelet
scribe some aspects of the involvement of platelets and
aggregates.
blood vessel walls in the overall process. This separation
In hemostasis, there is initial vasoconstriction of the
of clotting factors and platelets is artificial, since both
injured vessel, causing diminished blood flow distal to
play intimate and often mutually interdependent roles
the injury. Then hemostasis and thrombosis share three
in hemostasis and thrombosis, but it facilitates descrip-
phases:
tion of the overall processes involved.
(1) Formation of a loose and temporary platelet ag-
gregate at the site of injury. Platelets bind to col-
Both Intrinsic & Extrinsic Pathways Result
lagen at the site of vessel wall injury and are acti-
in the Formation of Fibrin
vated by thrombin (the mechanism of activation
Two pathways lead to fibrin clot formation: the intrin-
of platelets is described below), formed in the co-
sic and the extrinsic pathways. These pathways are not
agulation cascade at the same site, or by ADP re-
independent, as previously thought. However, this arti-
leased from other activated platelets. Upon acti-
ficial distinction is retained in the following text to fa-
vation, platelets change shape and, in the
cilitate their description.
presence of fibrinogen, aggregate to form the he-
Initiation of the fibrin clot in response to tissue in-
mostatic plug (in hemostasis) or thrombus (in
jury is carried out by the extrinsic pathway. How the
thrombosis).
intrinsic pathway is activated in vivo is unclear, but it
(2) Formation of a fibrin mesh that binds to the
involves a negatively charged surface. The intrinsic and
platelet aggregate, forming a more stable hemosta-
extrinsic pathways converge in a final common path-
tic plug or thrombus.
way involving the activation of prothrombin to throm-
(3) Partial or complete dissolution of the hemostatic
bin and the thrombin-catalyzed cleavage of fibrinogen
plug or thrombus by plasmin.
to form the fibrin clot. The intrinsic, extrinsic, and
final common pathways are complex and involve many
different proteins (Figure 51-1 and Table 51-1). In
598
HEMOSTASIS & THROMBOSIS
/
599
Intrinsic pathway
PK
HK
XII
XIIa
HK
Ca2+
Extrinsic pathway
XI
XIa
VII
Ca2+
IX
IXa
VIIa/Tissue factor
2+
Ca
VIII
VIIIa
PL
X
Xa
X
Figure 51-1. The pathways of
blood coagulation. The intrinsic
Ca2+
V
Va
and extrinsic pathways are indi-
PL
cated. The events depicted below
factor Xa are designated the final
common pathway, culminating in
Prothrombin
Thrombin
the formation of cross-linked fibrin.
New observations (dotted arrow)
include the finding that complexes
of tissue factor and factor VIIa acti-
Fibrinogen
XIII
vate not only factor X (in the classic
extrinsic pathway) but also factor
IX in the intrinsic pathway. In ad-
Classic coagulation
cascade
dition, thrombin and factor Xa
Fibrin monomer
XIIIa
Positive feedback
feedback-activate at the two sites
(hypothesized)
indicated (dashed arrows). (PK,
Extrinsic-to-intrinsic
prekallikrein; HK, HMW kininogen;
activation
Fibrin polymer
PL, phospholipids.) (Reproduced,
with permission, from Roberts HR,
Lozier JN: New perspectives on the
coagulation cascade. Hosp Pract [Off
Cross-linked
Ed] 1992 Jan;27:97.)
fibrin polymer
600
/
CHAPTER 51
Table 51-1. Numerical system for nomenclature
Table 51-2. The functions of the proteins
of blood clotting factors. The numbers indicate
involved in blood coagulation.
the order in which the factors have been
discovered and bear no relationship to the order
Zymogens of serine proteases
in which they act.
Factor XII
Binds to negatively charged surface at site of
vessel wall injury; activated by high-MW
kininogen and kallikrein.
Factor
Common Name
Factor XI
Activated by factor XIIa.
I
Fibrinogen
These factors are usually referred
Factor IX
Activated by factor Xla in presence of Ca2+.
II
Prothrombin
to by their common names.
Factor VII
Activated thrombin in presence of Ca2+.
III
Tissue factor
These factors are usually not re-
Factor X
Activated on surface of activated platelets by
IV
Ca2+
ferred to as coagulation factors.
tenase complex (Ca2+, factors VIIIa and IXa)
V
Proaccelerin, labile factor, accelerator (Ac-)
and by factor VIIa in presence of tissue fac-
globulin
tor and Ca2+.
VII1
Proconvertin, serum prothrombin conversion
Factor II
Activated on surface of activated platelets by
accelerator (SPCA), cothromboplastin
prothrombinase complex (Ca2+, factors Va
VIII
Antihemophilic factor A, antihemophilic globulin
and Xa).
(AHG)
[Factors II, VII, IX, and X are Gla-containing
IX
Antihemophilic factor B, Christmas factor, plasma
zymogens.] (Gla = γ-carboxyglutamate.)
thromboplastin component (PTC)
Cofactors
X
Stuart-Prower factor
Factor VIII
Activated by thrombin; factor VIIIa is a co-
XI
Plasma thromboplastin antecedent (PTA)
factor in the activation of factor X by
XII
Hageman factor
factor IXa.
XIII
Fibrin stabilizing factor (FSF), fibrinoligase
Factor V
Activated by thrombin; factor Va is a co-
1There is no factor VI.
factor in the activation of prothrombin by
factor Xa.
Tissue factor
A glycoprotein expressed on the surface of
(factor III)
injured or stimulated endothelial cells to
general, as shown in Table 51-2, these proteins can be
act as a cofactor for factor VIIa.
classified into five types: (1) zymogens of serine-depen-
dent proteases, which become activated during the
Fibrinogen
process of coagulation; (2) cofactors;
(3) fibrinogen;
Factor I
Cleaved by thrombin to form fibrin clot.
(4) a transglutaminase, which stabilizes the fibrin clot;
Thiol-dependent transglutaminase
and (5) regulatory and other proteins.
Factor XIII
Activated by thrombin in presence of Ca2+;
stabilizes fibrin clot by covalent cross-
The Intrinsic Pathway Leads
linking.
to Activation of Factor X
Regulatory and other proteins
Protein C
Activated to protein Ca by thrombin bound
The intrinsic pathway (Figure 51-1) involves factors
to thrombomodulin; then degrades fac-
XII, XI, IX, VIII, and X as well as prekallikrein, high-
tors VIIIa and Va.
molecular-weight (HMW) kininogen, Ca2+, and plate-
Protein S
Acts as a cofactor of protein C; both proteins
let phospholipids. It results in the production of fac-
contain Gla (γ-carboxyglutamate)
tor Xa (by convention, activated clotting factors are
residues.
referred to by use of the suffix a).
Thrombo-
Protein on the surface of endothelial
This pathway commences with the “contact phase”
modulin
cells; binds thrombin, which then acti-
in which prekallikrein, HMW kininogen, factor XII,
vates protein C.
and factor XI are exposed to a negatively charged acti-
vating surface. In vivo, the proteins probably assemble
XIa and also releases bradykinin (a nonapeptide with
on endothelial cell membranes, whereas glass or kaolin
potent vasodilator action) from HMW kininogen.
can be used for in vitro tests of the intrinsic pathway.
Factor XIa in the presence of Ca2+ activates factor IX
When the components of the contact phase assemble
(55 kDa, a zymogen containing vitamin K-dependent
on the activating surface, factor XII is activated to fac-
γ-carboxyglutamate [Gla] residues; see Chapter 45), to
tor XIIa upon proteolysis by kallikrein. This factor
the serine protease, factor IXa. This in turn cleaves an
XIIa, generated by kallikrein, attacks prekallikrein to
Arg-Ile bond in factor X (56 kDa) to produce the two-
generate more kallikrein, setting up a reciprocal activa-
chain serine protease, factor Xa. This latter reaction re-
tion. Factor XIIa, once formed, activates factor XI to
quires the assembly of components, called the tenase
HEMOSTASIS & THROMBOSIS
/
601
cleave plasminogen and kallikrein can activate single-
complex, on the surface of activated platelets: Ca2+ and
chain urokinase.
factor VIIIa, as well as factors IXa and X. It should be
Tissue factor pathway inhibitor (TFPI) is a major
noted that in all reactions involving the Gla-containing
physiologic inhibitor of coagulation. It is a protein that
zymogens (factors II, VII, IX, and X), the Gla residues
circulates in the blood associated with lipoproteins.
in the amino terminal regions of the molecules serve as
TFPI directly inhibits factor Xa by binding to the en-
high-affinity binding sites for Ca2+. For assembly of the
zyme near its active site. This factor Xa-TFPI complex
tenase complex, the platelets must first be activated to
then inhibits the factor VIIa-tissue factor complex.
expose the acidic
(anionic) phospholipids, phos-
phatidylserine and phosphatidylinositol, that are
normally on the internal side of the plasma membrane
The Final Common Pathway of Blood
of resting, nonactivated platelets. Factor VIII
(330
Clotting Involves Activation of
kDa), a glycoprotein, is not a protease precursor but a
Prothrombin to Thrombin
cofactor that serves as a receptor for factors IXa and X
on the platelet surface. Factor VIII is activated by
In the final common pathway, factor Xa, produced by
minute quantities of thrombin to form factor VIIIa,
either the intrinsic or the extrinsic pathway, activates
which is in turn inactivated upon further cleavage by
prothrombin (factor II) to thrombin
(factor IIa),
thrombin.
which then converts fibrinogen to fibrin (Figure 51-1).
The activation of prothrombin, like that of factor X,
occurs on the surface of activated platelets and requires
The Extrinsic Pathway Also Leads
the assembly of a prothrombinase complex, consisting
to Activation of Factor X But
of platelet anionic phospholipids, Ca2+, factor Va, fac-
by a Different Mechanism
tor Xa, and prothrombin.
Factor Xa occurs at the site where the intrinsic and ex-
Factor V (330 kDa), a glycoprotein with homology
trinsic pathways converge (Figure 51-1) and lead into
to factor VIII and ceruloplasmin, is synthesized in the
the final common pathway of blood coagulation. The
liver, spleen, and kidney and is found in platelets as
extrinsic pathway involves tissue factor, factors VII and
well as in plasma. It functions as a cofactor in a manner
X, and Ca2+ and results in the production of factor Xa.
similar to that of factor VIII in the tenase complex.
It is initiated at the site of tissue injury with the expo-
When activated to factor Va by traces of thrombin, it
sure of tissue factor (Figure 51-1) on subendothelial
binds to specific receptors on the platelet membrane
cells. Tissue factor interacts with and activates factor
(Figure 51-2) and forms a complex with factor Xa and
VII (53 kDa), a circulating Gla-containing glycoprotein
prothrombin. It is subsequently inactivated by further
synthesized in the liver. Tissue factor acts as a cofactor
action of thrombin, thereby providing a means of limit-
for factor VIIa, enhancing its enzymatic activity to acti-
ing the activation of prothrombin to thrombin. Pro-
vate factor X. The association of tissue factor and factor
thrombin (72 kDa; Figure 51-3) is a single-chain gly-
VIIa is called tissue factor complex. Factor VIIa
coprotein synthesized in the liver. The amino terminal
cleaves the same Arg-Ile bond in factor X that is cleaved
region of prothrombin (1 in Figure 51-3) contains ten
by the tenase complex of the intrinsic pathway. Activa-
Gla residues, and the serine-dependent active protease
tion of factor X provides an important link between the
site (indicated by the arrowhead) is in the carboxyl ter-
intrinsic and extrinsic pathways.
minal region of the molecule. Upon binding to the
Another important interaction between the extrinsic
complex of factors Va and Xa on the platelet mem-
and intrinsic pathways is that complexes of tissue factor
brane, prothrombin is cleaved by factor Xa at two sites
and factor VIIa also activate factor IX in the intrinsic
(Figure 51-2) to generate the active, two-chain throm-
pathway. Indeed, the formation of complexes be-
bin molecule, which is then released from the platelet
tween tissue factor and factor VIIa is now consid-
surface. The A and B chains of thrombin are held to-
ered to be the key process involved in initiation of
gether by a disulfide bond.
blood coagulation in vivo. The physiologic signifi-
cance of the initial steps of the intrinsic pathway, in
Conversion of Fibrinogen to Fibrin
which factor XII, prekallikrein, and HMW kininogen
Is Catalyzed by Thrombin
are involved, has been called into question because pa-
tients with a hereditary deficiency of these components
Fibrinogen (factor I, 340 kDa; see Figures 51-1 and
do not exhibit bleeding problems. Similarly, patients
51-4 and Tables 51-1 and 51-2) is a soluble plasma
with a deficiency of factor XI may not have bleeding
glycoprotein that consists of three nonidentical pairs of
problems. The intrinsic pathway may actually be more
polypeptide chains
(Aα,Bβγ)2
covalently linked by
important in fibrinolysis (see below) than in coagula-
disulfide bonds. The Bβ and γ chains contain as-
tion, since kallikrein, factor XIIa, and factor XIa can
paragine-linked complex oligosaccharides. All three
602
/
CHAPTER 51
Prethrombin
Figure 51-2. Diagrammatic representation
(not to scale) of the binding of factors Va, Xa,
F-1.2
Ca2+, and prothrombin to the plasma membrane
+
S-S
COO-
of the activated platelet. The sites of cleavage of
NH
3
prothrombin by factor Xa are indicated by two
Ca2+ Ca2+
Platelet
Va
Xa
arrows. The part of prothrombin destined to
plasma
form thrombin is labeled prethrombin. The Ca2+
membrane
Indicates negative charges
is bound to anionic phospholipids of the plasma
to which Ca2+ binds.
membrane of the activated platelet.
chains are synthesized in the liver; the three structural
ture (α, β, γ)2. Since FPA and FPB contain only 16 and
genes involved are on the same chromosome, and their
14 residues, respectively, the fibrin molecule retains
expression is coordinately regulated in humans. The
98% of the residues present in fibrinogen. The removal
amino terminal regions of the six chains are held in
of the fibrinopeptides exposes binding sites that allow
close proximity by a number of disulfide bonds, while
the molecules of fibrin monomers to aggregate sponta-
the carboxyl terminal regions are spread apart, giving
neously in a regularly staggered array, forming an insol-
rise to a highly asymmetric, elongated molecule (Figure
uble fibrin clot. It is the formation of this insoluble fib-
51-4). The A and B portions of the Aα and Bβ chains,
rin polymer that traps platelets, red cells, and other
designated fibrinopeptides A (FPA) and B (FPB), re-
components to form the white or red thrombi. This
spectively, at the amino terminal ends of the chains,
initial fibrin clot is rather weak, held together only by
bear excess negative charges as a result of the presence
the noncovalent association of fibrin monomers.
of aspartate and glutamate residues, as well as an un-
In addition to converting fibrinogen to fibrin,
usual tyrosine O-sulfate in FPB. These negative charges
thrombin also converts factor XIII to factor XIIIa. This
contribute to the solubility of fibrinogen in plasma and
factor is a highly specific transglutaminase that cova-
also serve to prevent aggregation by causing electrosta-
lently cross-links fibrin molecules by forming peptide
tic repulsion between fibrinogen molecules.
bonds between the amide groups of glutamine and the
Thrombin (34 kDa), a serine protease formed by
ε-amino groups of lysine residues
(Figure
51-5B),
the prothrombinase complex, hydrolyzes the four Arg-
yielding a more stable fibrin clot with increased resis-
Gly bonds between the fibrinopeptides and the α and β
tance to proteolysis.
portions of the Aα and Bβ chains of fibrinogen (Figure
51-5A). The release of the fibrinopeptides by thrombin
generates fibrin monomer, which has the subunit struc-
Levels of Circulating Thrombin Must Be
Carefully Controlled or Clots May Form
Xa
Once active thrombin is formed in the course of hemo-
S - S
Gla
n
stasis or thrombosis, its concentration must be carefully
controlled to prevent further fibrin formation or
1
2
A
B
platelet activation. This is achieved in two ways.
Thrombin circulates as its inactive precursor, pro-
Xa
thrombin, which is activated as the result of a cascade
of enzymatic reactions, each converting an inactive zy-
F-1
2
Prethrombin
(before Xa cleavage)
mogen to an active enzyme and leading finally to the
Thrombin (after Xa cleavage)
conversion of prothrombin to thrombin (Figure 51-1).
At each point in the cascade, feedback mechanisms
Diagrammatic representation (not to
Figure 51-3.
produce a delicate balance of activation and inhibition.
scale) of prothrombin. The amino terminal is to the left;
The concentration of factor XII in plasma is approxi-
region 1 contains all ten Gla residues. The sites of cleav-
mately 30 µg/mL, while that of fibrinogen is 3 mg/mL,
age by factor Xa are shown and the products named.
with intermediate clotting factors increasing in concen-
The site of the catalytically active serine residue is indi-
tration as one proceeds down the cascade, showing that
cated by the solid triangle. The A and B chains of active
the clotting cascade provides amplification. The second
thrombin (shaded) are held together by the disulfide
means of controlling thrombin activity is the inactiva-
bridge.
tion of any thrombin formed by circulating inhibi-
HEMOSTASIS & THROMBOSIS
/
603
FPA
FPB
Aα chain
Figure 51-4. Diagrammatic representation
(not to scale) of fibrinogen showing pairs of Aα,
Bβ chain
γ chain
Bβ, and γ chains linked by disulfide bonds. (FPA,
+
COO-
NH3
COO-
fibrinopeptide A; FPB, fibrinopeptide B.)
tors, the most important of which is antithrombin III
thrombin as well as to its other substrates. This is the
(see below).
basis for the use of heparin in clinical medicine to in-
hibit coagulation. The anticoagulant effects of heparin
can be antagonized by strongly cationic polypeptides
The Activity of Antithrombin III,
such as protamine, which bind strongly to heparin,
an Inhibitor of Thrombin,
thus inhibiting its binding to antithrombin III. Individ-
Is Increased by Heparin
uals with inherited deficiencies of antithrombin III are
Four naturally occurring thrombin inhibitors exist in
prone to develop venous thrombosis, providing evi-
normal plasma. The most important is antithrombin
dence that antithrombin III has a physiologic function
III
(often called simply antithrombin), which con-
and that the coagulation system in humans is normally
tributes approximately 75% of the antithrombin activ-
in a dynamic state.
ity. Antithrombin III can also inhibit the activities of
Thrombin is involved in an additional regulatory
factors IXa, Xa, XIa, XIIa, and VIIa complexed with
mechanism that operates in coagulation. It combines
tissue factor.
with thrombomodulin, a glycoprotein present on the
2-Macroglobulin contributes most of
the remainder of the antithrombin activity, with hep-
surfaces of endothelial cells. The complex activates pro-
arin cofactor II and
tein C. In combination with protein S, activated pro-
1-antitrypsin acting as minor in-
hibitors under physiologic conditions.
tein C (APC) degrades factors Va and VIIIa, limiting
The endogenous activity of antithrombin III is
their actions in coagulation. A genetic deficiency of ei-
greatly potentiated by the presence of acidic proteogly-
ther protein C or protein S can cause venous thrombo-
cans such as heparin (Chapter 48). These bind to a
sis. Furthermore, patients with factor V Leiden (which
specific cationic site of antithrombin III, inducing a
has a glutamine residue in place of an arginine at posi-
conformational change and promoting its binding to
tion 506) have an increased risk of venous thrombotic
A
Thrombin
-
-
+
Arg
Gly
COO-
NH3
-
–
Fibrinopeptide
Fibrin chain
(A or B)
(α or
β)
B
O
Figure 51-5. Formation of a fibrin
clot. A: Thrombin-induced cleavage
Fibrin
CH2
CH2
CH2
CH2
NH3+
H2N
C
CH
2
CH2
Fibrin
of Arg-Gly bonds of the Aα and Bβ
(Lysyl)
(Glutaminyl)
chains of fibrinogen to produce fi-
brinopeptides (left-hand side) and
the α and β chains of fibrin mono-
NH4+
Factor XIIIa (Transglutaminase)
mer (right-hand side). B: Cross-
O
linking of fibrin molecules by acti-
Fibrin
CH2
CH2
CH2
CH2
NH
C
CH2
CH2
Fibrin
vated factor XIII (factor XIIIa).
604
/
CHAPTER 51
disease because factor V Leiden is resistant to inactiva-
In past years, treatment for patients with hemophilia
tion by APC. This condition is termed APC resistance.
A has consisted of administration of cryoprecipitates
(enriched in factor VIII) prepared from individual
donors or lyophilized factor VIII concentrates prepared
from plasma pools of up to 5000 donors. It is now pos-
Coumarin Anticoagulants Inhibit the
sible to prepare factor VIII by recombinant DNA
Vitamin K-Dependent Carboxylation of
technology. Such preparations are free of contaminat-
Factors II, VII, IX, & X
ing viruses (eg, hepatitis A, B, C, or HIV-1) found in
The coumarin drugs (eg, warfarin), which are used as
human plasma but are at present expensive; their use
anticoagulants, inhibit the vitamin K-dependent car-
may increase if cost of production decreases.
boxylation of Glu to Gla residues (see Chapter 45) in
the amino terminal regions of factors II, VII, IX, and X
Fibrin Clots Are Dissolved by Plasmin
and also proteins C and S. These proteins, all of which
are synthesized in the liver, are dependent on the Ca2+-
As stated above, the coagulation system is normally in a
binding properties of the Gla residues for their normal
state of dynamic equilibrium in which fibrin clots are
function in the coagulation pathways. The coumarins
constantly being laid down and dissolved. This latter
act by inhibiting the reduction of the quinone deriva-
process is termed fibrinolysis. Plasmin, the serine pro-
tives of vitamin K to the active hydroquinone forms
tease mainly responsible for degrading fibrin and fi-
(Chapter 45). Thus, the administration of vitamin K
brinogen, circulates in the form of its inactive zymogen,
will bypass the coumarin-induced inhibition and allow
plasminogen (90 kDa), and any small amounts of plas-
maturation of the Gla-containing factors. Reversal of
min that are formed in the fluid phase under physio-
coumarin inhibition by vitamin K requires
12-24
logic conditions are rapidly inactivated by the fast-
hours, whereas reversal of the anticoagulant effects of
acting plasmin inhibitor, α2-antiplasmin. Plasminogen
heparin by protamine is almost instantaneous.
binds to fibrin and thus becomes incorporated in clots
Heparin and warfarin are widely used in the treat-
as they are produced; since plasmin that is formed
ment of thrombotic and thromboembolic conditions,
when bound to fibrin is protected from α2-antiplasmin,
such as deep vein thrombosis and pulmonary embolus.
it remains active. Activators of plasminogen of various
Heparin is administered first, because of its prompt
types are found in most body tissues, and all cleave the
onset of action, whereas warfarin takes several days to
same Arg-Val bond in plasminogen to produce the two-
reach full effect. Their effects are closely monitored by
chain serine protease, plasmin (Figure 51-6).
use of appropriate tests of coagulation (see below) be-
Tissue plasminogen activator (alteplase; t-PA) is a
cause of the risk of producing hemorrhage.
serine protease that is released into the circulation from
vascular endothelium under conditions of injury or
Hemophilia A Is Due to a Genetically
Plasminogen
PLASMINOGEN
Determined Deficiency of Factor VIII
activators
Inherited deficiencies of the clotting system that result
-
in bleeding are found in humans. The most common is
NH3+
Arg -Val
COO
deficiency of factor VIII, causing hemophilia A, an X
S S
chromosome-linked disease that has played a major role
in the history of the royal families of Europe. Hemo-
philia B is due to a deficiency of factor IX; its clinical
features are almost identical to those of hemophilia A,
but the conditions can be separated on the basis of spe-
-
NH3+
Arg Val
COO
cific assays that distinguish between the two factors.
The gene for human factor VIII has been cloned
S S
and is one of the largest so far studied, measuring 186
PLASMIN
kb in length and containing 26 exons. A variety of mu-
tations have been detected leading to diminished activ-
Figure 51-6. Activation of plasminogen. The same
Arg-Val bond is cleaved by all plasminogen activators
ity of factor VIII; these include partial gene deletions
and point mutations resulting in premature chain ter-
to give the two-chain plasmin molecule. The solid trian-
mination. Prenatal diagnosis by DNA analysis after
gle indicates the serine residue of the active site. The
chorionic villus sampling is now possible.
two chains of plasmin are held together by a disulfide
bridge.
HEMOSTASIS & THROMBOSIS
/
605
stress and is catalytically inactive unless bound to fibrin.
slightly better survival rate. Table 51-3 compares some
Upon binding to fibrin, t-PA cleaves plasminogen
thrombolytic features of streptokinase and t-PA.
within the clot to generate plasmin, which in turn di-
There are a number of disorders, including cancer
gests the fibrin to form soluble degradation products
and shock, in which the concentrations of plasmino-
and thus dissolves the clot. Neither plasmin nor the
gen activators increase. In addition, the antiplasmin
plasminogen activator can remain bound to these
activities contributed by α1-antitrypsin and α2-antiplas-
degradation products, and so they are released into the
min may be impaired in diseases such as cirrhosis. Since
fluid phase, where they are inactivated by their natural
certain bacterial products, such as streptokinase, are ca-
inhibitors. Prourokinase is the precursor of a second ac-
pable of activating plasminogen, they may be responsi-
tivator of plasminogen, urokinase. Originally isolated
ble for the diffuse hemorrhage sometimes observed in
from urine, it is now known to be synthesized by cell
patients with disseminated bacterial infections.
types such as monocytes and macrophages, fibroblasts,
and epithelial cells. Its main action is probably in the
degradation of extracellular matrix. Figure 51-7 indi-
Activation of Platelets Involves
cates the sites of action of five proteins that influence
Stimulation of the
the formation and action of plasmin.
Polyphosphoinositide Pathway
Platelets normally circulate in an unstimulated disk-
Recombinant t-PA & Streptokinase Are
shaped form. During hemostasis or thrombosis, they
Used as Clot Busters
become activated and help form hemostatic plugs or
Alteplase (t-PA), produced by recombinant DNA tech-
thrombi. Three major steps are involved: (1) adhesion
nology, is used therapeutically as a fibrinolytic agent, as
to exposed collagen in blood vessels, (2) release of the
is streptokinase. However, the latter is less selective
contents of their granules, and (3) aggregation.
than t-PA, activating plasminogen in the fluid phase
Platelets adhere to collagen via specific receptors on
(where it can degrade circulating fibrinogen) as well as
the platelet surface, including the glycoprotein complex
plasminogen that is bound to a fibrin clot. The amount
GPIa-IIa (α2β1 integrin; Chapter 52), in a reaction
of plasmin produced by therapeutic doses of streptoki-
that involves von Willebrand factor. This is a glyco-
nase may exceed the capacity of the circulating α2-
protein, secreted by endothelial cells into the plasma,
antiplasmin, causing fibrinogen as well as fibrin to be
which stabilizes factor VIII and binds to collagen and
degraded and resulting in the bleeding often encoun-
the subendothelium. Platelets bind to von Willebrand
tered during fibrinolytic therapy. Because of its selec-
factor via a glycoprotein complex (GPIb-V-IX) on the
tivity for degrading fibrin, there is considerable thera-
platelet surface; this interaction is especially important
peutic interest in the use of recombinant t-PA to restore
in platelet adherence to the subendothelium under con-
the patency of coronary arteries following thrombosis.
ditions of high shear stress that occur in small vessels
If administered early enough, before irreversible dam-
and stenosed arteries.
age of heart muscle occurs (about 6 hours after onset of
Platelets adherent to collagen change shape and
thrombosis), t-PA can significantly reduce the mortality
spread out on the subendothelium. They release the
rate from myocardial damage following coronary
contents of their storage granules (the dense granules
thrombosis. t-PA is more effective than streptokinase at
and the alpha granules); secretion is also stimulated by
restoring full patency and also appears to result in a
thrombin.
Streptokinase
-
+
Plasminogen
Streptokinase-plasminogen
complex
Figure 51-7. Scheme of sites of action
Plasminogen
t-PA
Urokinase
of streptokinase, tissue plasminogen acti-
activator
vator (t-PA), urokinase, plasminogen acti-
inhibitor
-
vator inhibitor, and α2-antiplasmin (the
last two proteins exert inhibitory actions).
Streptokinase forms a complex with plas-
Plasmin
α2-Antiplasmin
minogen, which exhibits proteolytic activ-
ity; this cleaves some plasminogen to plas-
Fibrin
Fibrin degradation products
min, initiating fibrinolysis.
606
/
CHAPTER 51
Table 51-3. Comparison of some properties of
Thrombin, formed from the coagulation cascade, is
streptokinase (SK) and tissue plasminogen
the most potent activator of platelets and initiates
platelet activation by interacting with its receptor on
activator (t-PA) with regard to their use as
the plasma membrane
(Figure
51-8). The further
thrombolytic agents.1
events leading to platelet activation are examples of
transmembrane signaling, in which a chemical mes-
SK
t-PA
senger outside the cell generates effector molecules in-
Selective for fibrin clot
−
+
side the cell. In this instance, thrombin acts as the ex-
Produces plasminemia
+
−
ternal chemical messenger (stimulus or agonist). The
Reduces mortality
+
+
interaction of thrombin with its receptor stimulates the
Causes allergic reaction
+
−
activity of an intracellular phospholipase C
. This en-
Causes hypotension
+
−
zyme hydrolyzes the membrane phospholipid phos-
Cost per treatment
Relatively low
Relatively high
phatidylinositol 4,5-bisphosphate (PIP2, a polyphospho-
(approximate)
inositide) to form the two internal effector molecules,
1Data from Webb J, Thompson C: Thrombolysis for acute myocar-
1,2-diacylglycerol and 1,4,5-inositol trisphosphate.
dial infarction. Can Fam Physician 1992;38:1415.
Hydrolysis of PIP2 is also involved in the action of
many hormones and drugs. Diacylglycerol stimulates
Collagen
Prostacyclin
TxA2
Thrombin
ADP
Aggregation
Fibrinogen
GPllb-llla
R1
R2
R3
R4
R5
+
+
+
+
Plasma
membrane
+
PLCβ
PIP2
AC
PLA2
PL
+
PKC
Signaling
cAMP
Arachidonic acid
events
IP
3
DAG
Phosphorylation
TxA2
of pleckstrin
-
Ca2+
Release of contents
of platelet granules
Phosphorylation
(dense and alpha),
of light chain of
including ADP;
Actin
myosin
signaling events
Actomyosin
Change of shape
Figure 51-8.
Diagrammatic representation of platelet activation. The external environ-
ment, the plasma membrane, and the inside of a platelet are depicted from top to bottom.
Thrombin and collagen are the two most important platelet activators. ADP is considered
a weak agonist; it causes aggregation but not granule release. (GP, glycoprotein; R1-R5,
various receptors; AC, adenylyl cyclase; PLA2, phospholipase A2; PL, phospholipids; PLCβ,
phospholipase Cβ; PIP2, phosphatidylinositol 4,5-bisphosphate; cAMP, cyclic AMP; PKC,
protein kinase C; TxA2, thromboxane A2; IP3, inositol 1,4,5-trisphosphate; DAG, 1,2-diacyl-
glycerol. The G proteins that are involved are not shown.)
HEMOSTASIS & THROMBOSIS
/
607
protein kinase C, which phosphorylates the protein
tors, which may help dissolve thrombi. Table 51-4 lists
pleckstrin (47 kDa). This results in aggregation and re-
some molecules produced by endothelial cells that af-
lease of the contents of the storage granules. ADP re-
fect thrombosis and fibrinolysis. Endothelium-derived
leased from dense granules can also activate platelets,
relaxing factor (nitric oxide) is discussed in Chapter 49.
resulting in aggregation of additional platelets. IP3
Analysis of the mechanisms of uptake of atherogenic
causes release of Ca2+
into the cytosol mainly from the
lipoproteins, such as LDL, by endothelial, smooth mus-
dense tubular system (or residual smooth endoplasmic
cle, and monocytic cells of arteries, along with detailed
reticulum from the megakaryocyte), which then inter-
studies of how these lipoproteins damage such cells is a
acts with calmodulin and myosin light chain kinase,
key area of study in elucidating the mechanisms of ath-
leading to phosphorylation of the light chains of
erosclerosis (Chapter 26).
myosin. These chains then interact with actin, causing
changes of platelet shape.
Aspirin Is an Effective Antiplatelet Drug
Collagen-induced activation of a platelet phospholi-
pase A2 by increased levels of cytosolic Ca2+ results in
Certain drugs (antiplatelet drugs) modify the behavior
of platelets. The most important is aspirin (acetylsali-
liberation of arachidonic acid from platelet phospho-
lipids, leading to the formation of thromboxane A2
cylic acid), which irreversibly acetylates and thus in-
hibits the platelet cyclooxygenase system involved in
(Chapter 23), which in turn, in a receptor-mediated
fashion, can further activate phospholipase C, promot-
formation of thromboxane A2 (Chapter 14), a potent
aggregator of platelets and also a vasoconstrictor.
ing platelet aggregation.
Activated platelets, besides forming a platelet aggre-
Platelets are very sensitive to aspirin; as little as 30 mg/d
(one aspirin tablet usually contains 325 mg) effectively
gate, are required, via newly expressed anionic phos-
pholipids on the membrane surface, for acceleration of
eliminates the synthesis of thromboxane A2. Aspirin
also inhibits production of prostacyclin (PGI2, which
the activation of factors X and II in the coagulation cas-
cade (Figure 51-1).
opposes platelet aggregation and is a vasodilator) by en-
All of the aggregating agents, including thrombin,
collagen, ADP, and others such as platelet-activating
factor, modify the platelet surface so that fibrinogen
Table 51-4. Molecules synthesized by
can bind to a glycoprotein complex, GPIIb-IIIa
(αIIbβ3 integrin; Chapter 52), on the activated platelet
endothelial cells that play a role in the regulation
surface. Molecules of divalent fibrinogen then link adja-
of thrombosis and fibrinolysis.1
cent activated platelets to each other, forming a platelet
aggregate. Some agents, including epinephrine, sero-
Molecule
Action
tonin, and vasopressin, exert synergistic effects with
ADPase (an ectoenzyme)
Degrades ADP (an aggregating
other aggregating agents.
agent of platelets) to AMP + Pi
Endothelium-derived relax-
Inhibits platelet adhesion and
Endothelial Cells Synthesize Prostacyclin
ing factor (nitric oxide)
aggregation by elevating lev-
& Other Compounds That Affect
els of cGMP
Clotting & Thrombosis
Heparan sulfate (a glycos-
Anticoagulant; combines with
aminoglycan)
antithrombin III to inhibit
The endothelial cells in the walls of blood vessels make
thrombin
important contributions to the overall regulation of he-
Prostacyclin (PGI2, a prosta-
Inhibits platelet aggregation by
mostasis and thrombosis. As described in Chapter 23,
glandin)
increasing levels of cAMP
these cells synthesize prostacyclin (PGI2), a potent in-
Thrombomodulin (a glyco-
Binds protein C, which is then
hibitor of platelet aggregation, opposing the action of
protein)
cleaved by thrombin to yield
thromboxane A2. Prostacyclin acts by stimulating the
activated protein C; this in
activity of adenylyl cyclase in the surface membranes of
combination with protein S
platelets. The resulting increase of intraplatelet cAMP
degrades factors Va and VIIIa,
opposes the increase in the level of intracellular Ca2+
limiting their actions
produced by IP3 and thus inhibits platelet activation
Tissue plasminogen activa-
Activates plasminogen to plas-
(Figure 51-8). Endothelial cells play other roles in the
tor (t-PA, a protease)
min, which digests fibrin; the
action of t-PA is opposed by
regulation of thrombosis. For instance, these cells pos-
plasminogen activator in-
sess an ADPase, which hydrolyzes ADP, and thus op-
hibitor-1 (PAI-1)
poses its aggregating effect on platelets. In addition,
these cells appear to synthesize heparan sulfate, an anti-
1Adapted from Wu KK: Endothelial cells in hemostasis, thrombosis
coagulant, and they also synthesize plasminogen activa-
and inflammation. Hosp Pract (Off Ed) 1992 Apr; 27:145.
608
/
CHAPTER 51
dothelial cells, but unlike platelets, these cells regenerate
• For activity, factors II, VII, IX, and X and proteins C
cyclooxygenase within a few hours. Thus, the overall
and S require vitamin K-dependent γ-carboxylation
balance between thromboxane A2 and prostacyclin can
of certain glutamate residues, a process that is inhib-
be shifted in favor of the latter, opposing platelet aggre-
ited by the anticoagulant warfarin.
gation. Indications for treatment with aspirin thus in-
• Fibrin is dissolved by plasmin. Plasmin exists as an
clude management of angina and evolving myocardial
inactive precursor, plasminogen, which can be acti-
infarction and also prevention of stroke and death in
vated by tissue plasminogen activator (t-PA). Both
patients with transient cerebral ischemic attacks.
t-PA and streptokinase are widely used to treat early
thrombosis in the coronary arteries.
Laboratory Tests Measure Coagulation
• Thrombin and other agents cause platelet aggrega-
& Thrombolysis
tion, which involves a variety of biochemical and
morphologic events. Stimulation of phospholipase C
A number of laboratory tests are available to measure
and the polyphosphoinositide pathway is a key event
the phases of hemostasis described above. The tests in-
in platelet activation, but other processes are also in-
clude platelet count, bleeding time, activated partial
volved.
thromboplastin time (aPTT or PTT), prothrombin time
• Aspirin is an important antiplatelet drug that acts by
(PT), thrombin time (TT), concentration of fibrin-
ogen, fibrin clot stability, and measurement of fibrin
inhibiting production of thromboxane A2.
degradation products. The platelet count quantitates
the number of platelets, and the bleeding time is an
REFERENCES
overall test of platelet function. aPTT is a measure of
the intrinsic pathway and PT of the extrinsic pathway.
Bennett JS: Mechanisms of platelet adhesion and aggregation: an
PT is used to measure the effectiveness of oral anticoag-
update. Hosp Pract (Off Ed) 1992;27:124.
ulants such as warfarin, and aPTT is used to monitor
Broze GJ: Tissue factor pathway inhibitor and the revised theory of
heparin therapy. The reader is referred to a textbook of
coagulation. Annu Rev Med 1995;46:103.
hematology for a discussion of these tests.
Clemetson KJ: Platelet activation: signal transduction via mem-
brane receptors. Thromb Haemost 1995;74:111.
Collen D, Lijnen HR: Basic and clinical aspects of fibrinolysis and
SUMMARY
thrombolysis. Blood 1991;78:3114.
• Hemostasis and thrombosis are complex processes
Handin RI: Anticoagulant, fibrinolytic and antiplatelet therapy.
involving coagulation factors, platelets, and blood
In: Harrison’s Principles of Internal Medicine, 15th ed. Braun-
wald E et al (editors). McGraw-Hill, 2001.
vessels.
Handin RI: Disorders of coagulation and thrombosis. In: Harri-
• Many coagulation factors are zymogens of serine pro-
son’s Principles of Internal Medicine, 15th ed. Braunwald E et
teases, becoming activated during the overall process.
al (editors). McGraw-Hill, 2001.
• Both intrinsic and extrinsic pathways of coagulation
Handin RI: Disorders of the platelet and vessel wall. In: Harrison’s
exist, the latter initiated by tissue factor. The path-
Principles of Internal Medicine, 15th ed. Braunwald E et al
ways converge at factor Xa, embarking on the com-
(editors). McGraw-Hill, 2001.
mon final pathway resulting in thrombin-catalyzed
Kroll MH, Schafer AI: Biochemical mechanisms of platelet activa-
tion. Blood 1989;74:1181.
conversion of fibrinogen to fibrin, which is strength-
ened by cross-linking, catalyzed by factor XIII.
Roberts HR, Lozier JN: New perspectives on the coagulation cas-
cade. Hosp Pract (Off Ed) 1992;27:97.
• Genetic disorders of coagulation factors occur, and
Roth GJ, Calverley DC: Aspirin, platelets, and thrombosis: theory
the two most common involve factors VIII (hemo-
and practice. Blood 1994;83:885.
philia A) and IX (hemophilia B).
Schmaier AH: Contact activation: a revision. Thromb Haemost
• An important natural inhibitor of coagulation is an-
1997;78:101.
tithrombin III; genetic deficiency of this protein can
Wu KK: Endothelial cells in hemostasis, thrombosis and inflamma-
result in thrombosis.
tion. Hosp Pract (Off Ed) 1992;27:145.
Red & White Blood Cells
52
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
intracellular organelles, such as mitochondria, lyso-
somes, or Golgi apparatus. Human red blood cells, like
Blood cells have been studied intensively because they
most red cells of animals, are nonnucleated. However,
are obtained easily, because of their functional impor-
the red cell is not metabolically inert. ATP is synthe-
tance, and because of their involvement in many disease
sized from glycolysis and is important in processes that
processes. The structure and function of hemoglobin,
help the red blood cell maintain its biconcave shape
the porphyrias, jaundice, and aspects of iron metabo-
and also in the regulation of the transport of ions (eg,
lism are discussed in previous chapters. Reduction of
by the Na+-K+ ATPase and the anion exchange protein
the number of red blood cells and of their content of
[see below]) and of water in and out of the cell. The bi-
hemoglobin is the cause of the anemias, a diverse and
concave shape increases the surface-to-volume ratio of
important group of conditions, some of which are seen
the red blood cell, thus facilitating gas exchange. The
very commonly in clinical practice. Certain of the
red cell contains cytoskeletal components (see below)
blood group systems, present on the membranes of
that play an important role in determining its shape.
erythrocytes and other blood cells, are of extreme im-
portance in relation to blood transfusion and tissue
About Two Million Red Blood Cells Enter
transplantation. Table 52-1 summarizes the causes of a
the Circulation per Second
number of important diseases affecting red blood cells;
some are discussed in this chapter, and the remainder
The life span of the normal red blood cell is 120 days;
are discussed elsewhere in this text. Every organ in the
this means that slightly less than 1% of the population
body can be affected by inflammation; neutrophils play
of red cells (200 billion cells, or 2 million per second) is
a central role in acute inflammation, and other white
replaced daily. The new red cells that appear in the cir-
blood cells, such as lymphocytes, play important roles
culation still contain ribosomes and elements of the en-
in chronic inflammation. Leukemias, defined as malig-
doplasmic reticulum. The RNA of the ribosomes can be
nant neoplasms of blood-forming tissues, can affect
detected by suitable stains (such as cresyl blue), and cells
precursor cells of any of the major classes of white
containing it are termed reticulocytes; they normally
blood cells; common types are acute and chronic my-
number about 1% of the total red blood cell count. The
elocytic leukemia, affecting precursors of the neu-
life span of the red blood cell can be dramatically short-
trophils; and acute and chronic lymphocytic leukemias.
ened in a variety of hemolytic anemias. The number of
Combination chemotherapy, using combinations of
reticulocytes is markedly increased in these conditions,
various chemotherapeutic agents, all of which act at one
as the bone marrow attempts to compensate for rapid
or more biochemical loci, has been remarkably effective
breakdown of red blood cells by increasing the amount
in the treatment of certain of these types of leukemias.
of new, young red cells in the circulation.
Understanding the role of red and white cells in health
and disease requires a knowledge of certain fundamen-
Erythropoietin Regulates Production
tal aspects of their biochemistry.
of Red Blood Cells
Human erythropoietin is a glycoprotein of 166 amino
THE RED BLOOD CELL IS SIMPLE IN
acids (molecular mass about 34 kDa). Its amount in
TERMS OF ITS STRUCTURE & FUNCTION
plasma can be measured by radioimmunoassay. It is the
The major functions of the red blood cell are relatively
major regulator of human erythropoiesis. Erythropoietin
simple, consisting of delivering oxygen to the tissues
is synthesized mainly by the kidney and is released in re-
and of helping in the disposal of carbon dioxide and
sponse to hypoxia into the bloodstream, in which it
protons formed by tissue metabolism. Thus, it has a
travels to the bone marrow. There it interacts with pro-
much simpler structure than most human cells, being
genitors of red blood cells via a specific receptor. The re-
essentially composed of a membrane surrounding a so-
ceptor is a transmembrane protein consisting of two dif-
lution of hemoglobin (this protein forms about 95% of
ferent subunits and a number of domains. It is not a
the intracellular protein of the red cell). There are no
tyrosine kinase, but it stimulates the activities of specific
609
610
/
CHAPTER 52
Table 52-1. Summary of the causes of some
members of this class of enzymes involved in down-
important disorders affecting red blood cells.
stream signal transduction. Erythropoietin interacts
with a red cell progenitor, known as the burst-forming
unit-erythroid (BFU-E), causing it to proliferate and
Disorder
Sole or Major Cause
differentiate. In addition, it interacts with a later pro-
Iron deficiency anemia
Inadequate intake or excessive loss
genitor of the red blood cell, called the colony-forming
of iron
unit-erythroid (CFU-E), also causing it to proliferate
and further differentiate. For these effects, erythropoi-
Methemoglobinemia
Intake of excess oxidants (various
chemicals and drugs)
etin requires the cooperation of other factors (eg, inter-
Genetic deficiency in the NADH-
leukin-3 and insulin-like growth factor; Figure 52-1).
dependent methemoglobin re-
The availability of a cDNA for erythropoietin has
ductase system (MIM 250800)
made it possible to produce substantial amounts of this
Inheritance of HbM (MIM 141800)
hormone for analysis and for therapeutic purposes; pre-
viously the isolation of erythropoietin from human
Sickle cell anemia
Sequence of codon 6 of the β chain
urine yielded very small amounts of the protein. The
(MIM 141900)
changed from GAG in the normal
major use of recombinant erythropoietin has been in
gene to GTG in the sickle cell
the treatment of a small number of anemic states, such
gene, resulting in substitution of
as that due to renal failure.
valine for glutamic acid
α-Thalassemias
Mutations in the α-globin genes,
MANY GROWTH FACTORS REGULATE
(MIM 141800)
mainly unequal crossing-over and
large deletions and less com-
PRODUCTION OF WHITE BLOOD CELLS
monly nonsense and frameshift
A large number of hematopoietic growth factors have
mutations
been identified in recent years in addition to erythro-
β-Thalassemia
A very wide variety of mutations in
poietin. This area of study adds to knowledge about the
(MIM 141900)
the β-globin gene, including dele-
differentiation of blood cells, provides factors that may
tions, nonsense and frameshift
be useful in treatment, and also has implications for un-
mutations, and others affecting
derstanding of the abnormal growth of blood cells (eg,
every aspect of its structure (eg,
the leukemias). Like erythropoietin, most of the growth
splice sites, promoter mutants)
factors isolated have been glycoproteins, are very active
Megaloblastic anemias
Decreased absorption of B12, often
in vivo and in vitro, interact with their target cells via
Deficiency of
due to a deficiency of intrinsic fac-
specific cell surface receptors, and ultimately (via intra-
vitamin B12
tor, normally secreted by gastric
cellular signals) affect gene expression, thereby promot-
parietal cells
ing differentiation. Many have been cloned, permitting
their production in relatively large amounts. Two of
Deficiency of folic
Decreased intake, defective absorp-
particular interest are granulocyte- and granulocyte-
acid
tion, or increased demand (eg, in
macrophage colony-stimulating factors (G-CSF and
pregnancy) for folate
GM-CSF, respectively). G-CSF is relatively specific, in-
Hereditary
Deficiencies in the amount or in the
ducing mainly granulocytes. GM-CSF affects a variety
spherocytosis1
structure of α or β spectrin,
of progenitor cells and induces granulocytes, macro-
ankyrin, band 3 or band 4.1
phages, and eosinophils. When the production of neu-
Glucose-6-phosphate
A variety of mutations in the gene
trophils is severely depressed, this condition is referred
dehydrogenase
(X-linked) for G6PD, mostly single
to as neutropenia. It is particularly likely to occur in
(G6PD) deficiency1
point mutations
patients treated with certain chemotherapeutic regi-
(MIM 305900)
mens and after bone marrow transplantation. These pa-
tients are liable to develop overwhelming infections.
Pyruvate kinase (PK)
Presumably a variety of mutations
G-CSF has been administered to such patients to boost
deficiency1
in the gene for the R (red cell) iso-
production of neutrophils.
(MIM 255200)
zyme of PK
Paroxysmal nocturnal
Mutations in the PIG-A gene, affect-
THE RED BLOOD CELL HAS A UNIQUE &
hemoglobinemia1
ing synthesis of GPI-anchored
(MIM 311770)
proteins
RELATIVELY SIMPLE METABOLISM
1The last four disorders cause hemolytic anemias, as do a number
Various aspects of the metabolism of the red cell, many
of the other disorders listed. Most of the above conditions are dis-
of which are discussed in other chapters of this text, are
cussed in other chapters of this text. MIM numbers apply only to
summarized in Table 52-2.
disorders with a genetic basis.
RED & WHITE BLOOD CELLS
/
611
Interleukins
Interleukins
GM-CSF
GM-CSF
Epo
Pluripotent
Erythroid
Mature
CFU-GEMM
BFU-E
stem cell
precursor
RBCs
(early
and late)
Figure 52-1. Greatly simplified scheme of differentiation of stem cells to red
blood cells. Various interleukins (ILs), such as IL-3, IL-4, IL-9, and IL-11, are involved
at different steps of the overall process. Erythroid precursors include the pronor-
moblast, basophilic, polychromatophilic, and orthochromatophilic normoblasts,
and the reticulocyte. Epo acts on basophilic normoblasts but not on later ery-
throid cells. (CFU-GEMM, colony-forming unit whose cells give rise to granulo-
cytes, erythrocytes, macrophages, and megakaryocytes; BFU-E, burst-forming
unit-erythroid; GM-CSF, granulocyte-macrophage colony-stimulating factor; Epo,
erythropoietin; RBC, red blood cell.)
The Red Blood Cell Has a Glucose
grammed by adding purified mRNAs or whole-cell ex-
Transporter in Its Membrane
tracts of mRNAs, and radioactive proteins are synthe-
sized in the presence of 35S-labeled L-methionine or
The entry rate of glucose into red blood cells is far
other radiolabeled amino acids. The radioactive pro-
greater than would be calculated for simple diffusion.
teins synthesized are separated by SDS-PAGE and de-
Rather, it is an example of facilitated diffusion (Chap-
tected by radioautography.
ter 41). The specific protein involved in this process is
called the glucose transporter or glucose permease.
Some of its properties are summarized in Table 52-3.
Superoxide Dismutase, Catalase,
The process of entry of glucose into red blood cells is of
& Glutathione Protect Blood Cells
major importance because it is the major fuel supply for
From Oxidative Stress & Damage
these cells. About seven different but related glucose
transporters have been isolated from various tissues; un-
Several powerful oxidants are produced during the
like the red cell transporter, some of these are insulin-
course of metabolism, in both blood cells and most
dependent (eg, in muscle and adipose tissue). There is
other cells of the body. These include superoxide (O2⋅ ),
considerable interest in the latter types of transporter
hydrogen peroxide (H2O2), peroxyl radicals (ROO•),
because defects in their recruitment from intracellular
and hydroxyl radicals (OH•). The last is a particularly
sites to the surface of skeletal muscle cells may help ex-
reactive molecule and can react with proteins, nucleic
plain the insulin resistance displayed by patients with
acids, lipids, and other molecules to alter their structure
type 2 diabetes mellitus.
and produce tissue damage. The reactions listed in
Table 52-4 play an important role in forming these ox-
idants and in disposing of them; each of these reactions
Reticulocytes Are Active
will now be considered in turn.
in Protein Synthesis
Superoxide is formed (reaction 1) in the red blood
The mature red blood cell cannot synthesize protein.
cell by the auto-oxidation of hemoglobin to methemo-
Reticulocytes are active in protein synthesis. Once retic-
globin (approximately 3% of hemoglobin in human red
ulocytes enter the circulation, they lose their intracellu-
blood cells has been calculated to auto-oxidize per day);
lar organelles
(ribosomes, mitochondria, etc) within
in other tissues, it is formed by the action of enzymes
about 24 hours, becoming young red blood cells and
such as cytochrome P450 reductase and xanthine oxi-
concomitantly losing their ability to synthesize protein.
dase. When stimulated by contact with bacteria, neu-
Extracts of rabbit reticulocytes (obtained by injecting
trophils exhibit a respiratory burst (see below) and pro-
rabbits with a chemical—phenylhydrazine—that causes
duce superoxide in a reaction catalyzed by NADPH
a severe hemolytic anemia, so that the red cells are al-
oxidase (reaction 2). Superoxide spontaneously dismu-
most completely replaced by reticulocytes) are widely
tates to form H2O2 and O2; however, the rate of this
used as an in vitro system for synthesizing proteins. En-
same reaction is speeded up tremendously by the action
dogenous mRNAs present in these reticulocytes are
of the enzyme superoxide dismutase (reaction 3). Hy-
destroyed by use of a nuclease, whose activity can be in-
drogen peroxide is subject to a number of fates. The en-
hibited by addition of Ca2+. The system is then pro-
zyme catalase, present in many types of cells, converts
612
/
CHAPTER 52
Table 52-2. Summary of important aspects of
Table 52-3. Some properties of the glucose
the metabolism of the red blood cell.
transporter of the membrane of the red
blood cell.
•
The RBC is highly dependent upon glucose as its energy
source; its membrane contains high affinity glucose trans-
• It accounts for about 2% of the protein of the membrane of
porters.
the RBC.
•
Glycolysis, producing lactate, is the site of production of
• It exhibits specificity for glucose and related D-hexoses
ATP.
(L-hexoses are not transported).
•
Because there are no mitochondria in RBCs, there is no pro-
• The transporter functions at approximately 75% of its Vmax
duction of ATP by oxidative phosphorylation.
at the physiologic concentration of blood glucose, is sat-
•
The RBC has a variety of transporters that maintain ionic
urable and can be inhibited by certain analogs of glucose.
and water balance.
• At least seven similar but distinct glucose transporters have
•
Production of 2,3-bisphosphoglycerate, by reactions closely
been detected to date in mammalian tissues, of which the
associated with glycolysis, is important in regulating the
red cell transporter is one.
ability of Hb to transport oxygen.
• It is not dependent upon insulin, unlike the corresponding
•
The pentose phosphate pathway is operative in the RBC (it
carrier in muscle and adipose tissue.
metabolizes about 5-10% of the total flux of glucose) and
• Its complete amino acid sequence (492 amino acids) has
produces NADPH; hemolytic anemia due to a deficiency of
been determined.
the activity of glucose-6-phosphate dehydrogenase is com-
• It transports glucose when inserted into artificial liposomes.
mon.
• It is estimated to contain 12 transmembrane helical seg-
•
Reduced glutathione (GSH) is important in the metabolism
ments.
of the RBC, in part to counteract the action of potentially
• It functions by generating a gated pore in the membrane to
toxic peroxides; the RBC can synthesize GSH and requires
permit passage of glucose; the pore is conformationally de-
NADPH to return oxidized glutathione (G-S-S-G) to the re-
pendent on the presence of glucose and can oscillate
duced state.
rapidly (about 900 times/s).
•
The iron of Hb must be maintained in the ferrous state; fer-
ric iron is reduced to the ferrous state by the action of an
NADH-dependent methemoglobin reductase system in-
8), which also produces OH• and OH−. Superoxide can
volving cytochrome b5 reductase and cytochrome b5.
•
Synthesis of glycogen, fatty acids, protein, and nucleic acids
release iron ions from ferritin. Thus, production of
does not occur in the RBC; however, some lipids (eg, choles-
OH• may be one of the mechanisms involved in tissue
terol) in the red cell membrane can exchange with corre-
injury due to iron overload
(eg, hemochromatosis;
sponding plasma lipids.
Chapter 50).
•
The RBC contains certain enzymes of nucleotide metabo-
Chemical compounds and reactions capable of gen-
lism (eg, adenosine deaminase, pyrimidine nucleotidase,
erating potential toxic oxygen species can be referred to
and adenylyl kinase); deficiencies of these enzymes are in-
as pro-oxidants. On the other hand, compounds and
volved in some cases of hemolytic anemia.
reactions disposing of these species, scavenging them,
•
When RBCs reach the end of their life span, the globin is de-
suppressing their formation, or opposing their actions
graded to amino acids (which are reutilized in the body),
are antioxidants and include compounds such as
the iron is released from heme and also reutilized, and the
NADPH, GSH, ascorbic acid, and vitamin E. In a nor-
tetrapyrrole component of heme is converted to bilirubin,
mal cell, there is an appropriate pro-oxidant:antioxi-
which is mainly excreted into the bowel via the bile.
dant balance. However, this balance can be shifted to-
ward the pro-oxidants when production of oxygen
species is increased greatly (eg, following ingestion of
it to H2O and O2 (reaction 4). Neutrophils possess a
certain chemicals or drugs) or when levels of antioxi-
unique enzyme, myeloperoxidase, that uses H2O2 and
dants are diminished (eg, by inactivation of enzymes in-
halides to produce hypohalous acids (reaction 5); this
volved in disposal of oxygen species and by conditions
subject is discussed further below. The selenium-
that cause low levels of the antioxidants mentioned
containing enzyme glutathione peroxidase (Chapter 20)
above). This state is called “oxidative stress” and can
will also act on reduced glutathione (GSH) and H2O2
result in serious cell damage if the stress is massive or
to produce oxidized glutathione (GSSG) and H2O (re-
prolonged.
action 6); this enzyme can also use other peroxides as
Oxygen species are now thought to play an impor-
substrates. OH• and OH− can be formed from H2O2 in
tant role in many types of cellular injury (eg, resulting
a nonenzymatic reaction catalyzed by Fe2+ (the Fenton
from administration of various toxic chemicals or from
reaction, reaction 7). O2⋅ and H2O2 are the substrates
ischemia), some of which can result in cell death. Indi-
in the iron-catalyzed Haber-Weiss reaction (reaction
rect evidence supporting a role for these species in gen-
RED & WHITE BLOOD CELLS
/
613
Table 52-4. Reactions of importance in relation to oxidative stress in blood cells and
various tissues.
(1) Production of superoxide (by-product of various reactions)
⋅
(2) NADPH-oxidase
2 O2 + NADPH → 2 O2−⋅ + NADP + H+
(3) Superoxide dismutase
⋅ + 2 H+ → H2O2 + O2
(4) Catalase
H2O2 → 2 H2O + O2
(5) Myeloperoxidase
H2O2 + X− + H+ → HOX + H2O (X− = Cl−, Br−, SCN−)
(6) Glutathione peroxidase (Se-dependent)
2 GSH + R-O-OH → GSSG + H2O + ROH
(7) Fenton reaction
Fe2+ + H2O2 → Fe3+ + OH⋅ + OH−
(8) Iron-catalyzed Haber-Weiss reaction
⋅ + H2O2 → O2 + OH⋅ + OH−
(9) Glucose-6-phosphate dehydrogenase (G6PD)
G6P + NADP → 6 Phosphogluconate + NADPH + H+
(10) Glutathione reductase
G-S-S-G + NADPH + H+ → 2 GSH + NADP
erating cell injury is provided if administration of an
sensitive hemolytic anemia] and sulfonamides) and
enzyme such as superoxide dismutase or catalase is
chemicals (eg, naphthalene) precipitate an attack, be-
found to protect against cell injury in the situation
cause their intake leads to generation of H2O2 or O2
⋅.
under study.
Normally, H2O2 is disposed of by catalase and glu-
tathione peroxidase (Table 52-4, reactions 4 and 6),
Deficiency of Glucose-6-Phosphate
the latter causing increased production of GSSG. GSH
is regenerated from GSSG by the action of the enzyme
Dehydrogenase Is Frequent in Certain
glutathione reductase, which depends on the availabil-
Areas & Is an Important Cause
ity of NADPH (reaction 10). The red blood cells of in-
of Hemolytic Anemia
dividuals who are deficient in the activity of glucose-6-
NADPH, produced in the reaction catalyzed by the
phosphate dehydrogenase cannot generate sufficient
X-linked glucose-6-phosphate dehydrogenase
(Table
NADPH to regenerate GSH from GSSG, which in
52-4, reaction 9) in the pentose phosphate pathway
turn impairs their ability to dispose of H2O2 and of
(Chapter 20), plays a key role in supplying reducing
oxygen radicals. These compounds can cause oxidation
equivalents in the red cell and in other cells such as the
of critical SH groups in proteins and possibly peroxida-
hepatocyte. Because the pentose phosphate pathway is
tion of lipids in the membrane of the red cell, causing
virtually its sole means of producing NADPH, the red
lysis of the red cell membrane. Some of the SH groups
blood cell is very sensitive to oxidative damage if the
of hemoglobin become oxidized, and the protein pre-
function of this pathway is impaired (eg, by enzyme de-
cipitates inside the red blood cell, forming Heinz bod-
ficiency). One function of NADPH is to reduce GSSG
ies, which stain purple with cresyl violet. The presence
to GSH, a reaction catalyzed by glutathione reductase
of Heinz bodies indicates that red blood cells have been
(reaction 10).
subjected to oxidative stress. Figure 52-2 summarizes
Deficiency of the activity of glucose-6-phosphate
the possible chain of events in hemolytic anemia due to
dehydrogenase, owing to mutation, is extremely fre-
deficiency of glucose-6-phosphate dehydrogenase.
quent in some regions of the world (eg, tropical Africa,
the Mediterranean, certain parts of Asia, and in North
Methemoglobin Is Useless
America among blacks). It is the most common of all
in Transporting Oxygen
enzymopathies (diseases caused by abnormalities of en-
zymes), and over 300 genetic variants of the enzyme
The ferrous iron of hemoglobin is susceptible to oxida-
have been distinguished; at least 100 million people are
tion by superoxide and other oxidizing agents, forming
deficient in this enzyme owing to these variants. The
methemoglobin, which cannot transport oxygen. Only
disorder resulting from deficiency of glucose-6-phos-
a very small amount of methemoglobin is present in
phate dehydrogenase is hemolytic anemia. Consump-
normal blood, as the red blood cell possesses an effec-
tion of broad beans (Vicia faba) by individuals deficient
tive system (the NADH-cytochrome b5 methemoglobin
in activity of the enzyme can precipitate an attack of
reductase system) for reducing heme Fe3+ back to the
hemolytic anemia (most likely because the beans con-
Fe2+ state. This system consists of NADH (generated
tain potential oxidants). In addition, a number of drugs
by glycolysis), a flavoprotein named cytochrome b5 re-
(eg, the antimalarial drug primaquine [the condition
ductase (also known as methemoglobin reductase), and
caused by intake of primaquine is called primaquine-
cytochrome b5. The Fe3+ of methemoglobin is reduced
614
/
CHAPTER 52
branes due to increased amounts of deoxygenated he-
Mutations in the gene for G6PD
moglobin in arterial blood, or in this case due to in-
creased amounts of methemoglobin) is usually the pre-
Decreased activity of G6PD
senting sign in both types and is evident when over 10%
of hemoglobin is in the “met” form. Diagnosis is made
Decreased levels of NADPH
by spectroscopic analysis of blood, which reveals the
characteristic absorption spectrum of methemoglobin.
Additionally, a sample of blood containing methemo-
Decreased regeneration of GSH from GSSG by
globin cannot be fully reoxygenated by flushing oxygen
glutathione reductase (which uses NADPH)
through it, whereas normal deoxygenated blood can.
Electrophoresis can be used to confirm the presence of
Oxidation, due to decreased levels of GSH and
an abnormal hemoglobin. Ingestion of methylene blue
),
increased levels of intracellular oxidants (eg, O2
•
or ascorbic acid (reducing agents) is used to treat mild
of SH groups of Hb (forming Heinz bodies), and of
methemoglobinemia due to enzyme deficiency. Acute
membrane proteins, altering membrane structure
massive methemoglobinemia (due to ingestion of chem-
and increasing susceptibility to ingestion
icals) should be treated by intravenous injection of
by macrophages (peroxidative damage to lipids
methylene blue.
in the membrane also possible)
Hemolysis
MORE IS KNOWN ABOUT THE MEMBRANE
OF THE HUMAN RED BLOOD CELL THAN
Figure 52-2. Summary of probable events causing
ABOUT THE SURFACE MEMBRANE OF
hemolytic anemia due to deficiency of the activity of
ANY OTHER HUMAN CELL
glucose-6-phosphate dehydrogenase (G6PD) (MIM
305900).
A variety of biochemical approaches have been used to
study the membrane of the red blood cell. These in-
clude analysis of membrane proteins by SDS-PAGE,
the use of specific enzymes (proteinases, glycosidases,
back to the Fe2+ state by the action of reduced cyto-
and others) to determine the location of proteins and
chrome b5:
glycoproteins in the membrane, and various techniques
to study both the lipid composition and disposition of
3+
2+
Hb - Fe
+
Cyt
b
5
red
→Hb - Fe
+
Cyt
b
5 ox
individual lipids. Morphologic
(eg, electron micros-
copy, freeze-fracture electron microscopy) and other
Reduced cytochrome b5 is then regenerated by the ac-
techniques (eg, use of antibodies to specific compon-
ents) have also been widely used. When red blood
tion of cytochrome b5 reductase:
cells are lysed under specific conditions, their mem-
branes will reseal in their original orientation to form
Cyt
b
5
ox
+NADH→Cyt
b
5 red
+NAD
ghosts (right-side-out ghosts). By altering the condi-
tions, ghosts can also be made to reseal with their cy-
tosolic aspect exposed on the exterior
(inside-out
Methemoglobinemia Is Inherited
ghosts). Both types of ghosts have been useful in ana-
or Acquired
lyzing the disposition of specific proteins and lipids in
the membrane. In recent years, cDNAs for many pro-
Methemoglobinemia can be classified as either inherited
teins of this membrane have become available, permit-
or acquired by ingestion of certain drugs and chemicals.
ting the deduction of their amino sequences and do-
Neither type occurs frequently, but physicians must be
mains. All in all, more is known about the membrane
aware of them. The inherited form is usually due to de-
of the red blood cell than about any other membrane of
ficient activity of methemoglobin reductase, transmitted
human cells (Table 52-5).
in an autosomal recessive manner. Certain abnormal he-
moglobins (Hb M) are also rare causes of methemoglo-
binemia. In Hb M, mutation changes the amino acid
Analysis by SDS-PAGE Resolves
residue to which heme is attached, thus altering its affin-
the Proteins of the Membrane
ity for oxygen and favoring its oxidation. Ingestion of
of the Red Blood Cell
certain drugs (eg, sulfonamides) or chemicals (eg, ani-
line) can cause acquired methemoglobinemia. Cyanosis
When the membranes of red blood cells are analyzed
(bluish discoloration of the skin and mucous mem-
by SDS-PAGE, about ten major proteins are resolved
RED & WHITE BLOOD CELLS
/
615
Table 52-5. Summary of biochemical
1
information about the membrane of the human
Spectrin
2
red blood cell.
2.1
Ankyrin
2.2
and
2.3
isoforms
•
The membrane is a bilayer composed of about 50% lipid
2.6
and 50% protein.
Anion exchange protein
3
•
The major lipid classes are phospholipids and cholesterol;
4.1
the major phospholipids are phosphatidylcholine (PC),
4.2
phosphatidylethanolamine (PE), and phosphatidylserine
(PS) along with sphingomyelin (Sph).
Glycophorins
Actin
5
•
The choline-containing phospholipids, PC and Sph, pre-
dominate in the outer leaflet and the amino-containing
G3PD
6
phospholipids (PE and PS) in the inner leaflet.
7
•
Glycosphingolipids (GSLs) (neutral GSLs, gangliosides, and
complex species, including the ABO blood group sub-
Globin
stances) constitute about 5-10% of the total lipid.
•
Analysis by SDS-PAGE shows that the membrane contains
about 10 major proteins and more than 100 minor species.
•
The major proteins (which include spectrin, ankyrin, the
anion exchange protein, actin, and band 4.1) have been
studied intensively, and the principal features of their dis-
position (eg, integral or peripheral), structure, and function
Coomassie
PAS stain
have been established.
blue stain
•
Many of the proteins are glycoproteins (eg, the glyco-
phorins) containing O- or N-linked (or both) oligosaccharide
Figure 52-3. Diagrammatic representation of the
chains located on the external surface of the membrane.
major proteins of the membrane of the human red
blood cell separated by SDS-PAGE. The bands detected
by staining with Coomassie blue are shown in the two
left-hand channels, and the glycoproteins detected by
(Figure 52-3), several of which have been shown to be
glycoproteins. Their migration on SDS-PAGE was
staining with periodic acid-Schiff (PAS) reagent are
used to name these proteins, with the slowest migrating
shown in the right-hand channel. (Reproduced, with
(and hence highest molecular mass) being designated
permission, from Beck WS, Tepper RI: Hemolytic anemias
band 1 or spectrin. All these major proteins have been
III: membrane disorders. In: Hematology, 5th ed. Beck WS
isolated, most of them have been identified, and con-
[editor]. The MIT Press, 1991.)
siderable insight has been obtained about their func-
tions
(Table
52-6). Many of their amino acid se-
quences also have been established. In addition, it has
been determined which are integral or peripheral mem-
bilayer at least ten times. It probably exists as a dimer in
brane proteins, which are situated on the external sur-
the membrane, in which it forms a tunnel, permitting
face, which are on the cytosolic surface, and which span
the exchange of chloride for bicarbonate. Carbon diox-
the membrane (Figure 52-4). Many minor compo-
ide, formed in the tissues, enters the red cell as bicar-
nents can also be detected in the red cell membrane by
bonate, which is exchanged for chloride in the lungs,
use of sensitive staining methods or two-dimensional
where carbon dioxide is exhaled. The amino terminal
gel electrophoresis. One of these is the glucose trans-
end binds many proteins, including hemoglobin, pro-
porter described above.
teins 4.1 and 4.2, ankyrin, and several glycolytic en-
zymes. Purified band 3 has been added to lipid vesicles
in vitro and has been shown to perform its transport
The Major Integral Proteins of the Red
functions in this reconstituted system.
Blood Cell Membrane Are the Anion
Glycophorins A, B, and C are also transmembrane
Exchange Protein & the Glycophorins
glycoproteins but of the single-pass type, extending
The anion exchange protein (band 3) is a transmem-
across the membrane only once. A is the major gly-
brane glycoprotein, with its carboxyl terminal end on
cophorin, is made up of 131 amino acids, and is heavily
the external surface of the membrane and its amino ter-
glycosylated (about 60% of its mass). Its amino terminal
minal end on the cytoplasmic surface. It is an example
end, which contains 16 oligosaccharide chains (15 of
of a multipass membrane protein, extending across the
which are O-glycans), extrudes out from the surface of
616
/
CHAPTER 52
Table 52-6. Principal proteins of the red cell membrane.1
Integral (I) or
Approximate
Band Number2
Protein
Peripheral (P)
Molecular Mass (kDa)
1
Spectrin (α)
P
240
2
Spectrin (β)
P
220
2.1
Ankyrin
P
210
2.2
“
P
195
2.3
“
P
175
2.6
“
P
145
3
Anion exchange protein
I
100
4.1
Unnamed
P
80
5
Actin
P
43
6
Glyceraldehyde-3-phosphate dehydrogenase
P
35
7
Tropomyosin
P
29
8
Unnamed
P
23
Glycophorins A, B, and C
I
31, 23, and 28
1Adapted from Lux DE, Becker PS: Disorders of the red cell membrane skeleton: hereditary spherocytosis and
hereditary elliptocytosis. Chapter 95 in: The Metabolic Basis of Inherited Disease, 6th ed. Scriver CR et al (editors).
McGraw-Hill, 1989.
2The band number refers to the position of migration on SDS-PAGE (see Figure 52-3). The glycophorins are de-
tected by staining with the periodic acid-Schiff reagent. A number of other components (eg, 4.2 and 4.9) are not
listed. Native spectrin is α2β2.
the red blood cell. Approximately 90% of the sialic acid
of the red cell membrane is located in this protein. Its
transmembrane segment (23 amino acids) is α-helical.
The carboxyl terminal end extends into the cytosol and
binds to protein 4.1, which in turn binds to spectrin.
Polymorphism of this protein is the basis of the MN
SPECTRIN-
SPECTRIN-
blood group system (see below). Glycophorin A con-
ANKYRIN-3
ACTIN-4.1
tains binding sites for influenza virus and for Plasmo-
INTERACTION
INTERACTION
dium falciparum, the cause of one form of malaria. In-
triguingly, the function of red blood cells of individuals
who lack glycophorin A does not appear to be affected.
Glycophorin
Outside
Lipid bilayer
Spectrin, Ankyrin, & Other Peripheral
Inside
3
Alpha
4.1
Membrane Proteins Help Determine the
Spectrin
Shape & Flexibility of the Red Blood Cell
Ankyrin
The red blood cell must be able to squeeze through
Actin
some tight spots in the microcirculation during its nu-
Beta
merous passages around the body; the sinusoids of the
SPECTRIN
spleen are of special importance in this regard. For the
SELF-
ASSOCIATION
red cell to be easily and reversibly deformable, its mem-
brane must be both fluid and flexible; it should also
Diagrammatic representation of the
Figure 52-4.
preserve its biconcave shape, since this facilitates gas ex-
interaction of cytoskeletal proteins with each other and
change. Membrane lipids help determine membrane
with certain integral proteins of the membrane of the
fluidity. Attached to the inner aspect of the membrane
red blood cell. (Reproduced, with permission, from Beck
of the red blood cell are a number of peripheral cy-
WS, Tepper RI: Hemolytic anemias III: membrane disor-
toskeletal proteins
(Table 52-6) that play important
ders. In: Hematology, 5th ed. Beck WS [editor]. The MIT
roles in respect to preserving shape and flexibility; these
Press, 1991.)
will now be described.
RED & WHITE BLOOD CELLS
/
617
Spectrin is the major protein of the cytoskeleton. It
0.85 g/dL. When exposed to a concentration of NaCl
is composed of two polypeptides: spectrin 1 (α chain)
of 0.5 g/dL, very few normal red blood cells are he-
and spectrin 2 (β chain). These chains, measuring ap-
molyzed, whereas approximately 50% of spherocytes
proximately 100 nm in length, are aligned in an an-
would lyse under these conditions. The explanation is
tiparallel manner and are loosely intertwined, forming a
that the spherocyte, being almost circular, has little po-
dimer. Both chains are made up of segments of 106
tential extra volume to accommodate additional water
amino acids that appear to fold into triple-stranded
and thus lyses readily when exposed to a slightly lower
α-helical coils joined by nonhelical segments. One
osmotic pressure than is normal.
dimer interacts with another, forming a head-to-head
One cause of hereditary spherocytosis (Figure 52-5)
tetramer. The overall shape confers flexibility on the
is a deficiency in the amount of spectrin or abnormali-
protein and in turn on the membrane of the red blood
ties of its structure, so that it no longer tightly binds the
cell. At least four binding sites can be defined in spec-
other proteins with which it normally interacts. This
trin: (1) for self-association, (2) for ankyrin (bands 2.1,
weakens the membrane and leads to the spherocytic
etc), (3) for actin (band 5), and (4) for protein 4.1.
shape. Abnormalities of ankyrin and of bands 3 and 4.1
Ankyrin is a pyramid-shaped protein that binds
are involved in other cases.
spectrin. In turn, ankyrin binds tightly to band 3, se-
Hereditary elliptocytosis is a genetic disorder that
curing attachment of spectrin to the membrane. Anky-
is similar to hereditary spherocytosis except that af-
rin is sensitive to proteolysis, accounting for the appear-
fected red blood cells assume an elliptic, disk-like shape,
ance of bands 2.2, 2.3, and 2.6, all of which are derived
recognizable by microscopy. It is also due to abnormali-
from band 2.1.
ties in spectrin; some cases reflect abnormalities of band
Actin (band 5) exists in red blood cells as short, dou-
4.1 or of glycophorin C.
ble-helical filaments of F-actin. The tail end of spectrin
dimers binds to actin. Actin also binds to protein 4.1.
THE BIOCHEMICAL BASES OF THE
Protein 4.1, a globular protein, binds tightly to the
ABO BLOOD GROUP SYSTEM
tail end of spectrin, near the actin-binding site of the
HAVE BEEN ESTABLISHED
latter, and thus is part of a protein 4.1-spectrin-actin
ternary complex. Protein 4.1 also binds to the integral
At least 21 human blood group systems are recognized,
proteins, glycophorins A and C, thereby attaching the
the best known of which are the ABO, Rh (Rhesus),
ternary complex to the membrane. In addition, protein
and MN systems. The term “blood group” applies to a
4.1 may interact with certain membrane phospholipids,
defined system of red blood cell antigens (blood group
thus connecting the lipid bilayer to the cytoskeleton.
substances) controlled by a genetic locus having a vari-
Certain other proteins (4.9, adducin, and tropo-
able number of alleles (eg, A, B, and O in the ABO sys-
myosin) also participate in cytoskeletal assembly.
tem). The term “blood type” refers to the antigenic
phenotype, usually recognized by the use of appropriate
Abnormalities in the Amount or Structure
of Spectrin Cause Hereditary
Spherocytosis & Elliptocytosis
Mutations in DNA affecting the amount or structure
of α or β spectrin or of certain other cytoskeletal
Hereditary spherocytosis is a genetic disease, transmitted
proteins (eg, ankyrin, band 3, band 4.1)
as an autosomal dominant, that affects about 1:5000
North Americans. It is characterized by the presence of
spherocytes (spherical red blood cells, with a low sur-
Weakens interactions among the peripheral and
face-to-volume ratio) in the peripheral blood, by a he-
integral proteins of the red cell membrane
molytic anemia, and by splenomegaly. The spherocytes
are not as deformable as are normal red blood cells, and
Weakens the structure of the red cell membrane
they are subject to destruction in the spleen, thus greatly
shortening their life in the circulation. Hereditary sphe-
Adopts spherocytic shape and is subject to
rocytosis is curable by splenectomy because the sphero-
destruction in the spleen
cytes can persist in the circulation if the spleen is absent.
The spherocytes are much more susceptible to os-
motic lysis than are normal red blood cells. This is as-
Hemolytic anemia
sessed in the osmotic fragility test, in which red blood
cells are exposed in vitro to decreasing concentrations
Figure 52-5. Summary of the causation of heredi-
of NaCl. The physiologic concentration of NaCl is
tary spherocytosis (MIM 182900).
618
/
CHAPTER 52
antibodies. For purposes of blood transfusion, it is par-
golipids, whereas in secretions the same oligosaccha-
ticularly important to know the basics of the ABO and
rides are present in glycoproteins. Their presence in se-
Rh systems. However, knowledge of blood group sys-
cretions is determined by a gene designated Se (for se-
tems is also of biochemical, genetic, immunologic, an-
cretor), which codes for a specific fucosyl
(Fuc)
thropologic, obstetric, pathologic, and forensic interest.
transferase in secretory organs, such as the exocrine
Here, we shall discuss only some key features of the
glands, but which is not active in red blood cells. Indi-
ABO system. From a biochemical viewpoint, the major
viduals of SeSe or Sese genotypes secrete A or B antigens
interests in the ABO substances have been in isolating
(or both), whereas individuals of the sese genotype do
and determining their structures, elucidating their
not secrete A or B substances, but their red blood cells
pathways of biosynthesis, and determining the natures
can express the A and B antigens.
of the products of the A, B, and O genes.
H Substance Is the Biosynthetic Precursor
The ABO System Is of Crucial Importance
of Both the A & B Substances
in Blood Transfusion
The ABO substances have been isolated and their struc-
This system was first discovered by Landsteiner in 1900
tures determined; simplified versions, showing only
when investigating the basis of compatible and incom-
their nonreducing ends, are presented in Figure 52-6.
patible transfusions in humans. The membranes of the
It is important to first appreciate the structure of the H
red blood cells of most individuals contain one blood
substance, since it is the precursor of both the A and B
group substance of type A, type B, type AB, or type O.
substances and is the blood group substance found in
Individuals of type A have anti-B antibodies in their
persons of type O. H substance itself is formed by the
plasma and will thus agglutinate type B or type AB
action of a fucosyltransferase, which catalyzes the ad-
blood. Individuals of type B have anti-A antibodies and
dition of the terminal fucose in α1 → 2 linkage onto
will agglutinate type A or type AB blood. Type AB
the terminal Gal residue of its precursor:
blood has neither anti-A nor anti-B antibodies and has
been designated the universal recipient. Type O blood
GDP Fuc+Gal β R→Fuc
-
α1,2
-
Gal
-
β
-
R+GDP
has neither A nor B substances and has been designated
Pr
ecursor
H substance
the universal donor. The explanation of these findings
is related to the fact that the body does not usually pro-
The H locus codes for this fucosyltransferase. The h al-
duce antibodies to its own constituents. Thus, individ-
lele of the H locus codes for an inactive fucosyltrans-
uals of type A do not produce antibodies to their own
ferase; therefore, individuals of the hh genotype cannot
blood group substance, A, but do possess antibodies to
generate H substance, the precursor of the A and B
the foreign blood group substance, B, possibly because
antigens. Thus, individuals of the hh genotype will have
similar structures are present in microorganisms to
red blood cells of type O, even though they may possess
which the body is exposed early in life. Since individu-
the enzymes necessary to make the A or B substances
als of type O have neither A nor B substances, they pos-
(see below).
sess antibodies to both these foreign substances. The
above description has been simplified considerably; eg,
The A Gene Encodes a GalNAc Transferase,
there are two subgroups of type A: A1 and A2.
the B Gene a Gal Transferase, & the O Gene
The genes responsible for production of the ABO
an Inactive Product
substances are present on the long arm of chromo-
some 9. There are three alleles, two of which are
In comparison with H substance (Figure 52-6), A sub-
codominant (A and B) and the third (O) recessive;
stance contains an additional GalNAc and B substance
these ultimately determine the four phenotypic prod-
an additional Gal, linked as indicated. Anti-A antibod-
ucts: the A, B, AB, and O substances.
ies are directed to the additional GalNAc residue found
in the A substance, and anti-B antibodies are directed
toward the additional Gal residue found in the B sub-
The ABO Substances Are
stance. Thus, GalNAc is the immunodominant sugar
Glycosphingolipids & Glycoproteins
(ie, the one determining the specificity of the antibody
Sharing Common Oligosaccharide Chains
formed) of blood group A substance, whereas Gal is the
The ABO substances are complex oligosaccharides pre-
immunodominant sugar of the B substance. In view of
sent in most cells of the body and in certain secretions.
the structural findings, it is not surprising that A sub-
On membranes of red blood cells, the oligosaccharides
stance can be synthesized in vitro from O substance in a
that determine the specific natures of the ABO sub-
reaction catalyzed by a GalNAc transferase, employing
stances appear to be mostly present in glycosphin-
UDP-GalNAc as the sugar donor. Similarly, blood
RED & WHITE BLOOD CELLS
/
619
Fucα1
2Galβ1
4GlcNAc - R
α1
3
Fucα1
2Galβ1
4GlcNAc - R
GalNAc
A substance
H (or O) substance
Fucα1
2Galβ1
4GlcNAc - R
α1
3
Gal
B substance
Figure 52-6. Diagrammatic representation of the structures of the H, A, and B
blood group substances. R represents a long complex oligosaccharide chain,
joined either to ceramide where the substances are glycosphingolipids, or to the
polypeptide backbone of a protein via a serine or threonine residue where the
substances are glycoproteins. Note that the blood group substances are bianten-
nary; ie, they have two arms, formed at a branch point (not indicated) between the
GlcNAc—R, and only one arm of the branch is shown. Thus, the H, A, and B sub-
stances each contain two of their respective short oligosaccharide chains shown
above. The AB substance contains one type A chain and one type B chain.
group B can be synthesized from O substance by the
Immunologic abnormalities (eg, transfusion reactions,
action of a Gal transferase, employing UDP-Gal. It is
the presence in plasma of warm and cold antibodies
crucial to appreciate that the product of the A gene is
that lyse red blood cells, and unusual sensitivity to com-
the GalNAc transferase that adds the terminal GalNAc
plement) also fall in this class, as do toxins released by
to the O substance. Similarly, the product of the B gene
various infectious agents, such as certain bacteria (eg,
is the Gal transferase adding the Gal residue to the O
clostridium). Some snakes release venoms that act to
substance. Individuals of type AB possess both enzymes
lyse the red cell membrane (eg, via the action of phos-
and thus have two oligosaccharide chains
(Figure
pholipases or proteinases).
52-6), one terminated by a GalNAc and the other by a
Causes within the membrane include abnormalities
Gal. Individuals of type O apparently synthesize an in-
of proteins. The most important conditions are heredi-
active protein, detectable by immunologic means; thus,
tary spherocytosis and hereditary elliptocytosis, princi-
H substance is their ABO blood group substance.
pally caused by abnormalities in the amount or struc-
In 1990, a study using cloning and sequencing tech-
ture of spectrin (see above).
nology described the nature of the differences between
Causes inside the red blood cell include hemoglo-
the glycosyltransferase products of the A, B, and O
binopathies and enzymopathies. Sickle cell anemia is
genes. A difference of four nucleotides is apparently re-
the most important hemoglobinopathy. Abnormalities
sponsible for the distinct specificities of the A and B
of enzymes in the pentose phosphate pathway and in
glycosyltransferases. On the other hand, the O allele has
glycolysis are the most frequent enzymopathies in-
a single base-pair mutation, causing a frameshift muta-
volved, particularly the former. Deficiency of glucose-
tion resulting in a protein lacking transferase activity.
6-phosphate dehydrogenase is prevalent in certain parts
of the world and is a frequent cause of hemolytic ane-
HEMOLYTIC ANEMIAS ARE CAUSED
mia (see above). Deficiency of pyruvate kinase is not
BY ABNORMALITIES OUTSIDE,
frequent, but it is the second commonest enzyme defi-
ciency resulting in hemolytic anemia; the mechanism
WITHIN, OR INSIDE THE RED
appears to be due to impairment of glycolysis, resulting
BLOOD CELL MEMBRANE
in decreased formation of ATP, affecting various as-
Causes outside the membrane include hypersplenism, a
pects of membrane integrity.
condition in which the spleen is enlarged from a variety
Laboratory investigations that aid in the diagnosis of
of causes and red blood cells become sequestered in it.
hemolytic anemia are listed in Table 52-7.
620
/
CHAPTER 52
Table 52-7. Laboratory investigations that assist
known as the “acute inflammatory response.” They in-
in the diagnosis of hemolytic anemia.
clude (1) increase of vascular permeability, (2) entry of
activated neutrophils into the tissues, (3) activation of
platelets, and (4) spontaneous subsidence (resolution) if
General tests and findings
the invading microorganisms have been dealt with suc-
Increased nonconjugated (indirect) bilirubin
cessfully.
Shortened red cell survival time as measured by injection
A variety of molecules are released from cells and
of autologous 51Cr-labeled red cells
plasma proteins during acute inflammation whose net
Reticulocytosis
Hemoglobinemia
overall effect is to increase vascular permeability, result-
Low level of plasma haptoglobin
ing in tissue edema (Table 52-10).
Specific tests and findings
In acute inflammation, neutrophils are recruited from
Hb electrophoresis (eg, HbS)
the bloodstream into the tissues to help eliminate the for-
Red cell enzymes (eg, G6PD or PK deficiency)
eign invaders. The neutrophils are attracted into the tis-
Osmotic fragility (eg, hereditary spherocytosis)
sues by chemotactic factors, including complement
1
Coombs test
fragment C5a, small peptides derived from bacteria (eg,
Cold agglutinins
N-formyl-methionyl-leucyl-phenylalanine), and a num-
1The direct Coombs test detects the presence of antibodies on
ber of leukotrienes. To reach the tissues, circulating neu-
red cells, whereas the indirect test detects the presence of circu-
trophils must pass through the capillaries. To achieve
lating antibodies to antigens present on red cells.
this, they marginate along the vessel walls and then ad-
here to endothelial (lining) cells of the capillaries.
Integrins Mediate Adhesion of Neutrophils
NEUTROPHILS HAVE AN ACTIVE
to Endothelial Cells
METABOLISM & CONTAIN SEVERAL
UNIQUE ENZYMES & PROTEINS
Adhesion of neutrophils to endothelial cells employs
specific adhesive proteins (integrins) located on their
The major biochemical features of neutrophils are sum-
surface and also specific receptor proteins in the en-
marized in Table 52-8. Prominent features are active
dothelial cells. (See also the discussion of selectins in
aerobic glycolysis, active pentose phosphate pathway,
Chapter 47.)
moderately active oxidative phosphorylation (because
The integrins are a superfamily of surface proteins
mitochondria are relatively sparse), and a high content
present on a wide variety of cells. They are involved in
of lysosomal enzymes. Many of the enzymes listed in
the adhesion of cells to other cells or to specific compo-
Table 52-4 are also of importance in the oxidative me-
nents of the extracellular matrix. They are heterodimers,
tabolism of neutrophils (see below). Table 52-9 sum-
containing an α and a β subunit linked noncovalently.
marizes the functions of some proteins that are rela-
The subunits contain extracellular, transmembrane,
tively unique to neutrophils.
and intracellular segments. The extracellular segments
bind to a variety of ligands such as specific proteins of
Neutrophils Are Key Players in the Body’s
the extracellular matrix and of the surfaces of other
Defense Against Bacterial Infection
cells. These ligands often contain Arg-Gly-Asp (R-G-
Neutrophils are motile phagocytic cells that play a key
D) sequences. The intracellular domains bind to vari-
role in acute inflammation. When bacteria enter tissues,
ous proteins of the cytoskeleton, such as actin and vin-
a number of phenomena result that are collectively
culin. The integrins are proteins that link the outsides
of cells to their insides, thereby helping to integrate re-
sponses of cells
(eg, movement, phagocytosis) to
changes in the environment.
Table 52-8. Summary of major biochemical
Three subfamilies of integrins were recognized ini-
features of neutrophils.
tially. Members of each subfamily were distinguished
by containing a common β subunit, but they differed
• Active glycolysis
in their α subunits. However, more than three β sub-
• Active pentose phosphate pathway
units have now been identified, and the classification of
• Moderate oxidative phosphorylation
integrins has become rather complex. Some integrins of
• Rich in lysosomes and their degradative enzymes
specific interest with regard to neutrophils are listed in
• Contain certain unique enzymes (eg, myeloperoxidase and
Table 52-11.
NADPH-oxidase) and proteins
A deficiency of the β2
subunit
(also designated
• Contain CD 11/CD18 integrins in plasma membrane
CD18) of LFA-1 and of two related integrins found in
RED & WHITE BLOOD CELLS
/
621
Table 52-9. Some important enzymes and proteins of neutrophils.1
Enzyme or Protein
Reaction Catalyzed or Function
Comment
Myeloperoxidase
H2O2 +X− (halide) + H+ →HOX + H2O
Responsible for the green color of pus
(MPO)
(where X− = Cl−, HOX = hypochlorous
Genetic deficiency can cause recurrent infections
acid)
NADPH-oxidase
2O2 + NADPH → 2O2−⋅ + NADP + H+
Key component of the respiratory burst
Deficient in chronic granulomatous disease
Lysozyme
Hydrolyzes link between N-acetylmura-
Abundant in macrophages
mic acid and N-acetyl-D-glucosamine
found in certain bacterial cell walls
Defensins
Basic antibiotic peptides of 20-33 amino
Apparently kill bacteria by causing membrane
acids
damage
Lactoferrin
Iron-binding protein
May inhibit growth of certain bacteria by binding
iron and may be involved in regulation of prolif-
eration of myeloid cells
CD11a/CD18, CD11b/CD18,
Adhesion molecules (members of the
Deficient in leukocyte adhesion deficiency type I
CD11c/CD182
integrin family)
(MIM 116920)
Receptors for Fc fragments of IgGs
Bind Fc fragments of IgG molecules
Target antigen-antibody complexes to myeloid
and lymphoid cells, eliciting phagocytosis and
other responses
1The expression of many of these molecules has been studied during various stages of differentiation of normal neutrophils and also of
corresponding leukemic cells employing molecular biology techniques (eg, measurements of their specific mRNAs). For the majority,
cDNAs have been isolated and sequenced, amino acid sequences deduced, genes have been localized to specific chromosomal locations,
and exons and intron sequences have been defined. Some important proteinases of neutrophils are listed in Table 52-12.
2CD = cluster of differentiation. This refers to a uniform system of nomenclature that has been adopted to name surface markers of leuko-
cytes. A specific surface protein (marker) that identifies a particular lineage or differentiation stage of leukocytes and that is recognized by
a group of monoclonal antibodies is called a member of a cluster of differentiation. The system is particularly helpful in categorizing sub-
classes of lymphocytes. Many CD antigens are involved in cell-cell interactions, adhesion, and transmembrane signaling.
neutrophils and macrophages, Mac-1 (CD11b/CD18)
to turn on many of the metabolic processes involved in
and p150,95 (CD11c/CD18), causes type 1 leukocyte
phagocytosis and killing of bacteria.
adhesion deficiency, a disease characterized by recur-
rent bacterial and fungal infections. Among various re-
Activation of Neutrophils Is Similar
sults of this deficiency, the adhesion of affected white
to Activation of Platelets
blood cells to endothelial cells is diminished, and lower
& Involves Hydrolysis of
numbers of neutrophils thus enter the tissues to combat
Phosphatidylinositol Bisphosphate
infection.
Once having passed through the walls of small
The mechanisms involved in platelet activation are dis-
blood vessels, the neutrophils migrate toward the high-
cussed in Chapter 51 (see Figure 51-8). The process in-
est concentrations of the chemotactic factors, encounter
volves interaction of the stimulus (eg, thrombin) with a
the invading bacteria, and attempt to attack and de-
receptor, activation of G proteins, stimulation of phos-
stroy them. The neutrophils must be activated in order
pholipase C, and liberation from phosphatidylinositol
Table 52-10. Sources of biomolecules with vasoactive properties involved in acute inflammation.
Mast Cells and Basophils
Platelets
Neutrophils
Plasma Proteins
Histamine
Serotonin
Platelet-activating factor (PAF)
C3a, C4a, and C5a from the complement system
Eicosanoids (various prostaglan-
Bradykinin and fibrin split products from the coagu-
dins and leukotrienes)
lation system
622
/
CHAPTER 52
Table 52-11. Examples of integrins that are important in the function of neutrophils, of other white
blood cells, and of platelets.1
Integrin
Cell
Subunit
Ligand
Function
VLA-1 (CD49a)
WBCs, others
α1β1
Collagen, laminin
Cell-ECM adhesion
VLA-5 (CD49e)
WBCs, others
α5β1
Fibronectin
Cell-ECM adhesion
VLA-6 (CD49f)
WBCs, others
α6β1
Laminin
Cell-ECM adhesion
LFA-1 (CD11a)
WBCs
αLβ2
ICAM-1
Adhesion of WBCs
Glycoprotein llb/llla
Platelets
αllbβ3
ICAM-2
Platelet adhesion and aggregation
Fibrinogen, fibronectin, von Willebrand
factor
1LFA-1, lymphocyte function-associated antigen 1; VLA, very late antigen; CD, cluster of differentiation; ICAM, intercellular adhesion mole-
cule; ECM, extracellular matrix. A deficiency of LFA-1 and related integrins is found in type I leukocyte adhesion deficiency (MIM 116290).
A deficiency of platelet glycoprotein llb/llla complex is found in Glanzmann thrombasthenia (MIM 273800), a condition characterized by a
history of bleeding, a normal platelet count, and abnormal clot retraction. These findings illustrate how fundamental knowledge of cell
surface adhesion proteins is shedding light on the causation of a number of diseases.
bisphosphate of inositol triphosphate and diacylglyc-
22 kDa. When the system is activated (see below), two
erol. These two second messengers result in an eleva-
cytoplasmic polypeptides of 47 kDa and 67 kDa are re-
tion of intracellular Ca2+ and activation of protein ki-
cruited to the plasma membrane and, together with cy-
nase C. In addition, activation of phospholipase A2
tochrome b558, form the NADPH oxidase responsible
produces arachidonic acid that can be converted to a
for the respiratory burst. The reaction catalyzed by
variety of biologically active eicosanoids.
NADPH oxidase, involving formation of superoxide
The process of activation of neutrophils is essentially
anion, is shown in Table 52-4 (reaction 2). This system
similar. They are activated, via specific receptors, by in-
catalyzes the one-electron reduction of oxygen to super-
teraction with bacteria, binding of chemotactic factors,
oxide anion. The NADPH is generated mainly by the
or antibody-antigen complexes. The resultant rise in in-
pentose phosphate cycle, whose activity increases mark-
tracellular Ca2+ affects many processes in neutrophils,
edly during phagocytosis.
such as assembly of microtubules and the actin-myosin
The above reaction is followed by the spontaneous
system. These processes are respectively involved in se-
production (by spontaneous dismutation) of hydrogen
cretion of contents of granules and in motility, which
peroxide from two molecules of superoxide:
enables neutrophils to seek out the invaders. The acti-
vated neutrophils are now ready to destroy the invaders
−⋅
2
O ⋅+O
2
+
2
H+→H O +O
2
2
2
by mechanisms that include production of active deriv-
atives of oxygen.
The superoxide ion is discharged to the outside of
the cell or into phagolysosomes, where it encounters in-
The Respiratory Burst of Phagocytic
gested bacteria. Killing of bacteria within phagolyso-
Cells Involves NADPH Oxidase
somes appears to depend on the combined action of
& Helps Kill Bacteria
elevated pH, superoxide ion, or further oxygen deriva-
When neutrophils and other phagocytic cells engulf
tives (H2O2, OH•, and HOCl [hypochlorous acid; see
bacteria, they exhibit a rapid increase in oxygen con-
below]) and on the action of certain bactericidal pep-
sumption known as the respiratory burst. This phe-
tides (defensins) and other proteins (eg, cathepsin G and
nomenon reflects the rapid utilization of oxygen (fol-
certain cationic proteins) present in phagocytic cells.
lowing a lag of 15-60 seconds) and production from it
Any superoxide that enters the cytosol of the phagocytic
of large amounts of reactive derivatives, such as O2
⋅,
cell is converted to H2O2 by the action of superoxide
H2O2, OH•, and OCl− (hypochlorite ion). Some of
dismutase, which catalyzes the same reaction as the
these products are potent microbicidal agents.
spontaneous dismutation shown above. In turn, H2O2 is
The electron transport chain system responsible
used by myeloperoxidase (see below) or disposed of by
for the respiratory burst (named NADPH oxidase) is
the action of glutathione peroxidase or catalase.
composed of several components. One is cytochrome
NADPH oxidase is inactive in resting phagocytic
b558, located in the plasma membrane; it is a het-
cells and is activated upon contact with various ligands
erodimer, containing two polypeptides of 91 kDa and
(complement fragment C5a, chemotactic peptides, etc)
RED & WHITE BLOOD CELLS
/
623
with receptors in the plasma membrane. The events re-
Mutations in genes for the polypeptide components
sulting in activation of the oxidase system have been
of the NADPH oxidase system
much studied and are similar to those described above
for the process of activation of neutrophils. They in-
Diminished production of superoxide ion
volve G proteins, activation of phospholipase C, and
and other active derivatives of oxygen
generation of inositol 1,4,5-triphosphate (IP3). The
last mediates a transient increase in the level of cytosolic
Ca2+, which is essential for induction of the respiratory
Diminished killing of certain bacteria
burst. Diacylglycerol is also generated and induces the
translocation of protein kinase C into the plasma mem-
Recurrent infections and formation of tissue
brane from the cytosol, where it catalyzes the phosphor-
granulomas in order to wall off surviving bacteria
ylation of various proteins, some of which are compo-
nents of the oxidase system. A second pathway of
Figure 52-7. Simplified scheme of the sequence of
also operates.
activation not involving Ca2+
events involved in the causation of chronic granuloma-
tous disease (MIM 306400). Mutations in any of the
Mutations in the Genes for Components
genes for the four polypeptides involved (two are com-
of the NADPH Oxidase System Cause
ponents of cytochrome b558 and two are derived from
Chronic Granulomatous Disease
the cytoplasm) can cause the disease. The polypeptide
of 91 kDa is encoded by a gene in the X chromosome;
The importance of the NADPH oxidase system was
approximately 60% of cases of chronic granulomatous
clearly shown when it was observed that the respiratory
disease are X-linked, with the remainder being inher-
burst was defective in chronic granulomatous disease, a
ited in an autosomal recessive fashion.
relatively uncommon condition characterized by recur-
rent infections and widespread granulomas (chronic in-
flammatory lesions) in the skin, lungs, and lymph
nodes. The granulomas form as attempts to wall off
cause it reacts with primary or secondary amines pre-
bacteria that have not been killed, owing to genetic de-
sent in neutrophils and tissues to produce various nitro-
ficiencies in the NADPH oxidase system. The disorder
gen-chlorine derivatives; these chloramines are also oxi-
is due to mutations in the genes encoding the four
dants, though less powerful than HOCl, and act as
polypeptides that constitute the NADPH oxidase sys-
microbicidal agents (eg, in sterilizing wounds) without
tem. Some patients have responded to treatment with
causing tissue damage.
gamma interferon, which may increase transcription of
the 91-kDa component if it is affected. The probable
The Proteinases of Neutrophils
sequence of events involved in the causation of chronic
Can Cause Serious Tissue Damage
granulomatous disease is shown in Figure 52-7.
If Their Actions Are Not Checked
Neutrophils Contain Myeloperoxidase,
Neutrophils contain a number of proteinases (Table
52-12) that can hydrolyze elastin, various types of col-
Which Catalyzes the Production
lagens, and other proteins present in the extracellular
of Chlorinated Oxidants
matrix. Such enzymatic action, if allowed to proceed
The enzyme myeloperoxidase, present in large amounts
unopposed, can result in serious damage to tissues.
in neutrophil granules and responsible for the green color
Most of these proteinases are lysosomal enzymes and
of pus, can act on H2O2 to produce hypohalous acids:
exist mainly as inactive precursors in normal neu-
trophils. Small amounts of these enzymes are released
MYELOPEROXIDASE
into normal tissues, with the amounts increasing
H2O2 + X— + H+
HOX + H2O
markedly during inflammation. The activities of elas-
(X— = Cl—, Br—, I— or SCN—; HOCl = hypochlorous acid)
tase and other proteinases are normally kept in check
by a number of antiproteinases (also listed in Table
The H2O2 used as substrate is generated by the
52-12) present in plasma and the extracellular fluid.
NADPH oxidase system. Cl- is the halide usually em-
Each of them can combine—usually forming a nonco-
ployed, since it is present in relatively high concentra-
valent complex—with one or more specific proteinases
tion in plasma and body fluids. HOCl, the active ingre-
and thus cause inhibition. In Chapter 50 it was shown
dient of household liquid bleach, is a powerful oxidant
that a genetic deficiency of
1-antiproteinase inhi-
and is highly microbicidal. When applied to normal tis-
bitor (α1-antitrypsin) permits elastase to act unopposed
sues, its potential for causing damage is diminished be-
and digest pulmonary tissue, thereby participating in
624
/
CHAPTER 52
Table 52-12. Proteinases of neutrophils and
ter
51) have been greatly clarified by investigations
antiproteinases of plasma and tissues.1
using cloning and sequencing. The study of oncogenes
and chromosomal translocations has advanced under-
standing of the leukemias. As discussed above, cloning
Proteinases
Antiproteinases
techniques have made available therapeutic amounts of
Elastase
α1-Antiproteinase (α1-antitrypsin)
erythropoietin and other growth factors. Deficiency of
Collagenase
α2-Macroglobulin
adenosine deaminase, which affects lymphocytes partic-
Gelatinase
Secretory leukoproteinase inhibitor
ularly, is the first disease to be treated by gene therapy.
Cathepsin G
α1-Antichymotrypsin
Like many other areas of biology and medicine, hema-
Plasminogen activator
Plasminogen activator inhibitor-1
tology has been and will continue to be revolutionized
Tissue inhibitor of metalloproteinase
by this technology.
1The table lists some of the important proteinases of neutrophils
and some of the proteins that can inhibit their actions. Most of
the proteinases listed exist inside neutrophils as precursors. Plas-
SUMMARY
minogen activator is not a proteinase, but it is included because it
•
The red blood cell is simple in terms of its structure
influences the activity of plasmin, which is a proteinase. The pro-
teinases listed can digest many proteins of the extracellular ma-
and function, consisting principally of a concentrated
trix, causing tissue damage. The overall balance of proteinase:an-
solution of hemoglobin surrounded by a membrane.
tiproteinase action can be altered by activating the precursors of
•
The production of red cells is regulated by erythro-
the proteinases, or by inactivating the antiproteinases. The latter
poietin, whereas other growth factors (eg, granulo-
can be caused by proteolytic degradation or chemical modifica-
cyte- and granulocyte-macrophage colony-stimulat-
tion, eg, Met-358 of α1-antiproteinase inhibitor is oxidized by cig-
ing factors) regulate the production of white blood
arette smoke.
cells.
•
The red cell contains a battery of cytosolic enzymes,
such as superoxide dismutase, catalase, and glu-
the causation of emphysema.
2-Macroglobulin is a
tathione peroxidase, to dispose of powerful oxidants
plasma protein that plays an important role in the
generated during its metabolism.
body’s defense against excessive action of proteases; it
•
Genetically determined deficiency of the activity of
combines with and thus neutralizes the activities of a
glucose-6-phosphate dehydrogenase, which produces
number of important proteases (Chapter 50).
NADPH, is an important cause of hemolytic anemia.
When increased amounts of chlorinated oxidants are
•
Methemoglobin is unable to transport oxygen; both
formed during inflammation, they affect the pro-
genetic and acquired causes of methemoglobinemia
teinase:antiproteinase equilibrium, tilting it in favor of
are recognized. Considerable information has accu-
the former. For instance, certain of the proteinases
mulated concerning the proteins and lipids of the red
listed in Table 52-12 are activated by HOCl, whereas
cell membrane. A number of cytoskeletal proteins,
certain of the antiproteinases are inactivated by this
such as spectrin, ankyrin, and actin, interact with spe-
compound. In addition, the tissue inhibitor of metallo-
cific integral membrane proteins to help regulate the
proteinases and α1-antichymotrypsin can be hydrolyzed
shape and flexibility of the membrane.
by activated elastase, and α1-antiproteinase inhibitor
•
Deficiency of spectrin results in hereditary spherocy-
can be hydrolyzed by activated collagenase and gelati-
tosis, another important cause of hemolytic anemia.
nase. In most circumstances, an appropriate balance
of proteinases and antiproteinases is achieved. How-
•
The ABO blood group substances in the red cell
membrane are complex glycosphingolipids; the im-
ever, in certain instances, such as in the lung when
α1-antiproteinase inhibitor is deficient or when large
munodominant sugar of A substance is N-acetyl-
galactosamine, whereas that of the B substance is
amounts of neutrophils accumulate in tissues because of
inadequate drainage, considerable tissue damage can re-
galactose.
sult from the unopposed action of proteinases.
•
Neutrophils play a major role in the body’s defense
mechanisms. Integrins on their surface membranes
determine specific interactions with various cell and
RECOMBINANT DNA TECHNOLOGY
tissue components.
HAS HAD A PROFOUND IMPACT
•
Leukocytes are activated on exposure to bacteria and
ON HEMATOLOGY
other stimuli; NADPH oxidase plays a key role in the
Recombinant DNA technology has had a major impact
process of activation (the respiratory burst). Muta-
on many aspects of hematology. The bases of the tha-
tions in this enzyme and associated proteins cause
lassemias and of many disorders of coagulation (Chap-
chronic granulomatous disease.
RED & WHITE BLOOD CELLS
/
625
• The proteinases of neutrophils can digest many tissue
Israels LG, Israels ED: Mechanisms in Hematology, 2nd ed. Univ
Manitoba Press, 1997. (Includes an excellent interactive CD.)
proteins; normally, this is kept in check by a battery
Jaffe ER, Hultquist DE: Cytochrome b5 reductase deficiency and
of antiproteinases. However, this defense mechanism
enzymopenic hereditary methemoglobinemia. In: The Meta-
can be overcome in certain circumstances, resulting
bolic and Molecular Bases of Inherited Disease, 8th ed. Scriver
in extensive tissue damage.
CR et al (editors). McGraw-Hill, 2001.
• The application of recombinant DNA technology is
Lekstrom-Hunes JA, Gallin JI: Immunodeficiency diseases caused
revolutionizing the field of hematology.
by defects in granulocytes. N Engl J Med 2000;343:1703.
Luzzato L et al: Glucose-6-phosphate dehydrogenase. In: The
Metabolic and Molecular Bases of Inherited Disease, 8th ed.
REFERENCES
Scriver CR et al (editors). McGraw-Hill, 2001.
Borregaard N, Cowland JB: Granules of the human neutrophilic
Rosse WF et al: New Views of Sickle Cell Disease Pathophysiology
polymorphonuclear leukocyte. Blood 1997;89:3503.
and Treatment. The American Society of Hematology.
Daniels G: A century of human blood groups. Wien Klin Wochen-
schr 2001;113:781.
Tse WT, Lux SE: Hereditary spherocytosis and hereditary ellipto-
Goodnough LT et al: Erythropoietin, iron, and erythropoiesis.
cytosis. In: The Molecular Bases of Inherited Disease, 8th ed.
Blood 2000;96:823.
Scriver CR et al (editors). McGraw-Hill, 2001.
Hirono A et al: Pyruvate kinase deficiency and other enzy-
Weatherall DJ et al: The hemoglobinopathies. In: The Metabolic
mopathies of the erythrocyte. In: The Metabolic and Molecu-
and Molecular Bases of Inherited Disease, 8th ed. Scriver CR et
lar Bases of Inherited Disease, 8th ed. Scriver CR et al (edi-
al (editors). McGraw-Hill, 2001.
tors). McGraw-Hill, 2001.
Metabolism of Xenobiotics
53
Robert K. Murray, MD, PhD
BIOMEDICAL IMPORTANCE
In phase 2, the hydroxylated or other compounds
produced in phase 1 are converted by specific enzymes
Increasingly, humans are subjected to exposure to vari-
to various polar metabolites by conjugation with glu-
ous foreign chemicals (xenobiotics)—drugs, food addi-
curonic acid, sulfate, acetate, glutathione, or certain
tives, pollutants, etc. The situation is well summarized
amino acids, or by methylation.
in the following quotation from Rachel Carson: “As
The overall purpose of the two phases of metabo-
crude a weapon as the cave man’s club, the chemical
lism of xenobiotics is to increase their water solubility
barrage has been hurled against the fabric of life.” Un-
(polarity) and thus excretion from the body. Very hy-
derstanding how xenobiotics are handled at the cellular
drophobic xenobiotics would persist in adipose tissue
level is important in learning how to cope with the
almost indefinitely if they were not converted to more
chemical onslaught.
polar forms. In certain cases, phase 1 metabolic reac-
Knowledge of the metabolism of xenobiotics is basic
tions convert xenobiotics from inactive to biologically
to a rational understanding of pharmacology and thera-
active compounds. In these instances, the original
peutics, pharmacy, toxicology, management of cancer,
xenobiotics are referred to as “prodrugs” or “procar-
and drug addiction. All these areas involve administra-
cinogens.” In other cases, additional phase 1 reactions
tion of, or exposure to, xenobiotics.
(eg, further hydroxylation reactions) convert the active
compounds to less active or inactive forms prior to con-
HUMANS ENCOUNTER THOUSANDS
jugation. In yet other cases, it is the conjugation reac-
OF XENOBIOTICS THAT MUST BE
tions themselves that convert the active products of
METABOLIZED BEFORE BEING EXCRETED
phase 1 reactions to less active or inactive species, which
are subsequently excreted in the urine or bile. In a very
A xenobiotic (Gk xenos “stranger”) is a compound that
few cases, conjugation may actually increase the bio-
is foreign to the body. The principal classes of xenobi-
logic activity of a xenobiotic.
otics of medical relevance are drugs, chemical carcino-
The term “detoxification” is sometimes used for
gens, and various compounds that have found their way
many of the reactions involved in the metabolism of
into our environment by one route or another, such as
xenobiotics. However, the term is not always appropri-
polychlorinated biphenyls (PCBs) and certain insecti-
ate because, as mentioned above, in some cases the reac-
cides. More than 200,000 manufactured environmental
tions to which xenobiotics are subject actually increase
chemicals exist. Most of these compounds are subject to
their biologic activity and toxicity.
metabolism (chemical alteration) in the human body,
with the liver being the main organ involved; occasion-
ally, a xenobiotic may be excreted unchanged. At least
ISOFORMS OF CYTOCHROME
30 different enzymes catalyze reactions involved in
P450 HYDROXYLATE A MYRIAD
xenobiotic metabolism; however, this chapter will only
OF XENOBIOTICS IN PHASE 1
cover a selected group of them.
It is convenient to consider the metabolism of xeno-
OF THEIR METABOLISM
biotics in two phases. In phase 1, the major reaction in-
Hydroxylation is the chief reaction involved in phase
volved is hydroxylation, catalyzed by members of a
1. The responsible enzymes are called monooxygenases
class of enzymes referred to as monooxygenases or cy-
or cytochrome P450s; the human genome encodes at
tochrome P450s. Hydroxylation may terminate the ac-
least
14 families of these enzymes. Estimates of the
tion of a drug, though this is not always the case. In ad-
number of distinct cytochrome P450s in human tissues
dition to hydroxylation, these enzymes catalyze a wide
range from approximately 35 to 60. The reaction cat-
range of reactions, including those involving deamina-
alyzed by a monooxygenase (cytochrome P450) is as
tion, dehalogenation, desulfuration, epoxidation, per-
follows:
oxygenation, and reduction. Reactions involving hy-
drolysis (eg, catalyzed by esterases) and certain other
+
non-P450-catalyzed reactions also occur in phase 1.
2
RH + O + NADPH + H
→R—OH+H O+NADP
2
626
METABOLISM OF XENOBIOTICS
/
627
RH above can represent a very wide variety of xenobi-
described above except that italics are used; thus, the
otics, including drugs, carcinogens, pesticides, petro-
gene encoding CYP1A1 is CYP1A1.
leum products, and pollutants (such as a mixture of
(2) Like hemoglobin, they are hemoproteins.
PCBs). In addition, endogenous compounds, such as
(3) They are widely distributed across species. Bac-
certain steroids, eicosanoids, fatty acids, and retinoids,
teria possess cytochrome P450s, and P450cam (involved
are also substrates. The substrates are generally lip-
in the metabolism of camphor) of Pseudomonas putida
ophilic and are rendered more hydrophilic by hydroxy-
is the only P450 isoform whose crystal structure has
lation.
been established.
Cytochrome P450 is considered the most versatile
(4) They are present in highest amount in liver and
biocatalyst known. The actual reaction mechanism is
small intestine but are probably present in all tissues.
complex and has been briefly described previously (Fig-
In liver and most other tissues, they are present mainly
ure 11-6). It has been shown by the use of 18O2 that
in the membranes of the smooth endoplasmic reticu-
one atom of oxygen enters ROH and one atom en-
lum, which constitute part of the microsomal fraction
ters water. This dual fate of the oxygen accounts for the
when tissue is subjected to subcellular fractionation. In
former naming of monooxygenases as
“mixed-
hepatic microsomes, cytochrome P450s can comprise
function oxidases.” The reaction catalyzed by cy-
as much as 20% of the total protein. P450s are found
tochrome P450 can also be represented as follows:
in most tissues, though often in low amounts compared
with liver. In the adrenal, they are found in mitochon-
Reduced cytochrome P450 Oxidized cytochrome P450
dria as well as in the endoplasmic reticulum; the vari-
RH + O2 → R—OH + H2O
ous hydroxylases present in that organ play an impor-
tant role in cholesterol and steroid biosynthesis. The
The major monooxygenases in the endoplasmic reticu-
mitochondrial cytochrome P450 system differs from
lum are cytochrome P450s—so named because the en-
the microsomal system in that it uses an NADPH-
zyme was discovered when it was noted that prepara-
linked flavoprotein, adrenodoxin reductase, and a
tions of microsomes that had been chemically reduced
nonheme iron-sulfur protein, adrenodoxin. In addi-
and then exposed to carbon monoxide exhibited a dis-
tion, the specific P450 isoforms involved in steroid
tinct peak at 450 nm. Among reasons that this enzyme
biosynthesis are generally much more restricted in their
is important is the fact that approximately 50% of the
substrate specificity.
drugs humans ingest are metabolized by isoforms of cy-
(5) At least six isoforms of cytochrome P450 are
tochrome P450; these enzymes also act on various car-
present in the endoplasmic reticulum of human liver,
cinogens and pollutants.
each with wide and somewhat overlapping substrate
specificities and acting on both xenobiotics and en-
Isoforms of Cytochrome P450 Make Up a
dogenous compounds. The genes for many isoforms of
Superfamily of Heme-Containing Enzymes
P450 (from both humans and animals such as the rat)
have been isolated and studied in detail in recent years.
The following are important points concerning cy-
(6) NADPH, not NADH, is involved in the reac-
tochrome P450s.
tion mechanism of cytochrome P450. The enzyme that
(1) Because of the large number of isoforms (about
uses NADPH to yield the reduced cytochrome P450,
150) that have been discovered, it became important to
shown at the left-hand side of the above equation, is
have a systematic nomenclature for isoforms of P450
called NADPH-cytochrome P450 reductase. Elec-
and for their genes. This is now available and in wide
trons are transferred from NADPH to NADPH-
use and is based on structural homology. The abbrevi-
cytochrome P450 reductase and then to cytochrome
ated root symbol CYP denotes a cytochrome P450.
P450. This leads to the reductive activation of molec-
This is followed by an Arabic number designating the
ular oxygen, and one atom of oxygen is subsequently
family; cytochrome P450s are included in the same
inserted into the substrate. Cytochrome b5, another
family if they exhibit 40% or more sequence identity.
hemoprotein found in the membranes of the smooth
The Arabic number is followed by a capital letter indi-
endoplasmic reticulum (Chapter 11), may be involved
cating the subfamily, if two or more members exist;
as an electron donor in some cases.
P450s are in the same subfamily if they exhibit greater
(7) Lipids are also components of the cytochrome
than 55% sequence identity. The individual P450s
P450 system. The preferred lipid is phosphatidyl-
are then arbitrarily assigned Arabic numerals. Thus,
choline, which is the major lipid found in membranes
CYP1A1 denotes a cytochrome P450 that is a member
of the endoplasmic reticulum.
of family 1 and subfamily A and is the first individual
(8) Most isoforms of cytochrome P450 are in-
member of that subfamily. The nomenclature for the
ducible. For instance, the administration of phenobar-
genes encoding cytochrome P450s is identical to that
bital or of many other drugs causes hypertrophy of the
628
/
CHAPTER 53
smooth endoplasmic reticulum and a three- to fourfold
potentially altering the quantities of metabolites of
increase in the amount of cytochrome P450 within 4-5
PAHs (some of which could be harmful) to which the
days. The mechanism of induction has been studied ex-
fetus is exposed.
tensively and in most cases involves increased transcrip-
(10) Certain cytochrome P450s exist in polymor-
tion of mRNA for cytochrome P450. However, certain
phic forms (genetic isoforms), some of which exhibit
cases of induction involve stabilization of mRNA, en-
low catalytic activity. These observations are one im-
zyme stabilization, or other mechanisms (eg, an effect
portant explanation for the variations in drug responses
on translation).
noted among many patients. One P450 exhibiting
polymorphism is CYP2D6, which is involved in the
Induction of cytochrome P450 has important clini-
metabolism of debrisoquin (an antihypertensive drug;
cal implications, since it is a biochemical mechanism of
see Table 53-2) and sparteine (an antiarrhythmic and
drug interaction. A drug interaction has occurred
oxytocic drug). Certain polymorphisms of CYP2D6
when the effects of one drug are altered by prior, con-
cause poor metabolism of these and a variety of other
current, or later administration of another. As an illus-
drugs so that they can accumulate in the body, resulting
tration, consider the situation when a patient is taking
in untoward consequences. Another interesting poly-
the anticoagulant warfarin to prevent blood clotting.
morphism is that of CYP2A6, which is involved in the
This drug is metabolized by CYP2C9. Concomitantly,
metabolism of nicotine to conitine. Three CYP2A6 al-
the patient is started on phenobarbital (an inducer of
leles have been identified: a wild type and two null or
this P450) to treat a certain type of epilepsy, but the
inactive alleles. It has been reported that individuals
dose of warfarin is not changed. After 5 days or so, the
with the null alleles, who have impaired metabolism of
level of CYP2C9 in the patient’s liver will be elevated
nicotine, are apparently protected against becoming to-
three- to fourfold. This in turn means that warfarin will
bacco-dependent smokers (Table 53-2). These individ-
be metabolized much more quickly than before, and its
uals smoke less, presumably because their blood and
dosage will have become inadequate. Therefore, the
brain concentrations of nicotine remain elevated longer
dose must be increased if warfarin is to be therapeuti-
than those of individuals with the wild-type allele. It
cally effective. To pursue this example further, a prob-
has been speculated that inhibiting CYP2A6 may be a
lem could arise later on if the phenobarbital is discon-
novel way to help prevent and to treat smoking.
tinued but the increased dosage of warfarin stays the
same. The patient will be at risk of bleeding, since the
Table 53-1 summarizes some principal features of
high dose of warfarin will be even more active than be-
cytochrome P450s.
fore, because the level of CYP2C9 will decline once
phenobarbital has been stopped.
Another example of enzyme induction involves
CONJUGATION REACTIONS PREPARE
CYP2E1, which is induced by consumption of
XENOBIOTICS FOR EXCRETION IN
ethanol. This is a matter for concern, because this
PHASE 2 OF THEIR METABOLISM
P450 metabolizes certain widely used solvents and also
components found in tobacco smoke, many of which
In phase 1 reactions, xenobiotics are generally con-
are established carcinogens. Thus, if the activity of
verted to more polar, hydroxylated derivatives. In phase
CYP2E1 is elevated by induction, this may increase the
2 reactions, these derivatives are conjugated with mole-
risk of carcinogenicity developing from exposure to
cules such as glucuronic acid, sulfate, or glutathione.
such compounds.
This renders them even more water-soluble, and they
are eventually excreted in the urine or bile.
(9) Certain isoforms of cytochrome P450
(eg,
CYP1A1) are particularly involved in the metabolism of
polycyclic aromatic hydrocarbons (PAHs) and related
Five Types of Phase 2 Reactions
molecules; for this reason they were formerly called aro-
Are Described Here
matic hydrocarbon hydroxylases (AHHs). This enzyme
A. GLUCURONIDATION
is important in the metabolism of PAHs and in car-
cinogenesis produced by these agents. For example, in
The glucuronidation of bilirubin is discussed in Chap-
the lung it may be involved in the conversion of inac-
ter
32; the reactions whereby xenobiotics are glu-
tive PAHs (procarcinogens), inhaled by smoking, to ac-
curonidated are essentially similar. UDP-glucuronic
tive carcinogens by hydroxylation reactions. Smokers
acid is the glucuronyl donor, and a variety of glu-
have higher levels of this enzyme in some of their cells
curonosyltransferases, present in both the endoplasmic
and tissues than do nonsmokers. Some reports have in-
reticulum and cytosol, are the catalysts. Molecules such
dicated that the activity of this enzyme may be elevated
as 2-acetylaminofluorene (a carcinogen), aniline, ben-
(induced) in the placenta of a woman who smokes, thus
zoic acid, meprobamate (a tranquilizer), phenol, and
METABOLISM OF XENOBIOTICS
/
629
Table 53-1. Some properties of human
GSH (because of the sulfhydryl group of its cysteine,
cytochrome P450s.
which is the business part of the molecule). A number
of potentially toxic electrophilic xenobiotics (such as
certain carcinogens) are conjugated to the nucleophilic
•
Involved in phase I of the metabolism of innumerable
GSH in reactions that can be represented as follows:
xenobiotics, including perhaps 50% of the drugs adminis-
tered to humans
R+ GSH→R—S—G
•
Involved in the metabolism of many endogenous com-
pounds (eg, steroids)
where R = an electrophilic xenobiotic. The enzymes
•
All are hemoproteins
catalyzing these reactions are called glutathione S-
•
Often exhibit broad substrate specificity, thus acting on
transferases and are present in high amounts in liver
many compounds; consequently, different P450s may cat-
cytosol and in lower amounts in other tissues. A variety
alyze formation of the same product
of glutathione S-transferases are present in human tis-
•
Extremely versatile catalysts, perhaps catalyzing about 60
sue. They exhibit different substrate specificities and
types of reactions
can be separated by electrophoretic and other tech-
•
However, basically they catalyze reactions involving intro-
niques. If the potentially toxic xenobiotics were not
duction of one atom of oxygen into the substrate and one
conjugated to GSH, they would be free to combine co-
into water
valently with DNA, RNA, or cell protein and could
•
Their hydroxylated products are more water-soluble than
thus lead to serious cell damage. GSH is therefore an
their generally lipophilic substrates, facilitating excretion
important defense mechanism against certain toxic
•
Liver contains highest amounts, but found in most if not
compounds, such as some drugs and carcinogens. If the
all tissues, including small intestine, brain, and lung
•
Located in the smooth endoplasmic reticulum or in mito-
levels of GSH in a tissue such as liver are lowered (as
chondria (steroidogenic hormones)
can be achieved by the administration to rats of certain
•
In some cases, their products are mutagenic or carcino-
compounds that react with GSH), then that tissue can
genic
be shown to be more susceptible to injury by various
•
Many have a molecular mass of about 55 kDa
chemicals that would normally be conjugated to GSH.
•
Many are inducible, resulting in one cause of drug interac-
Glutathione conjugates are subjected to further metab-
tions
olism before excretion. The glutamyl and glycinyl
•
Many are inhibited by various drugs or their metabolic
groups belonging to glutathione are removed by spe-
products, providing another cause of drug interactions
cific enzymes, and an acetyl group (donated by acetyl-
•
Some exhibit genetic polymorphisms, which can result in
CoA) is added to the amino group of the remaining
atypical drug metabolism
cysteinyl moiety. The resulting compound is a mercap-
•
Their activities may be altered in diseased tissues (eg, cir-
turic acid, a conjugate of L-acetylcysteine, which is
rhosis), affecting drug metabolism
then excreted in the urine.
•
Genotyping the P450 profile of patients (eg, to detect
Glutathione has other important functions in
polymorphisms) may in the future permit individualization
human cells apart from its role in xenobiotic metabo-
of drug therapy
lism.
1. It participates in the decomposition of potentially
toxic hydrogen peroxide in the reaction cat-
many steroids are excreted as glucuronides. The glu-
alyzed by glutathione peroxidase (Chapter 20).
curonide may be attached to oxygen, nitrogen, or sulfur
2. It is an important intracellular reductant, help-
groups of the substrates. Glucuronidation is probably
the most frequent conjugation reaction.
ing to maintain essential SH groups of enzymes in
their reduced state. This role is discussed in Chap-
B. SULFATION
ter 20, and its involvement in the hemolytic ane-
Some alcohols, arylamines, and phenols are sulfated.
mia caused by deficiency of glucose-6-phosphate
The sulfate donor in these and other biologic sulfation
dehydrogenase is discussed in Chapters 20 and
reactions (eg, sulfation of steroids, glycosaminoglycans,
52.
glycolipids, and glycoproteins) is adenosine 3 -phos-
3. A metabolic cycle involving GSH as a carrier has
phate-5 -phosphosulfate (PAPS) (Chapter 24); this
been implicated in the transport of certain
compound is called “active sulfate.”
amino acids across membranes in the kidney.
The first reaction of the cycle is shown below.
C. CONJUGATION WITH GLUTATHIONE
Glutathione (γ-glutamyl-cysteinylglycine) is a tripep-
Amino acid + GSH
→γ
- Glutamyl amino acid+
tide consisting of glutamic acid, cysteine, and glycine
Cysteinylglycine
(Figure
3-3). Glutathione is commonly abbreviated
630
/
CHAPTER 53
This reaction helps transfer certain amino acids across
Again, this can affect the doses of certain drugs that are
the plasma membrane, the amino acid being subse-
administered to patients. Various diseases (eg, cirrhosis
quently hydrolyzed from its complex with GSH and
of the liver) can affect the activities of drug-metaboliz-
the GSH being resynthesized from cysteinylglycine.
ing enzymes, sometimes necessitating adjustment of
The enzyme catalyzing the above reaction is
-glu-
dosages of various drugs for patients with these disor-
tamyltransferase (GGT). It is present in the plasma
ders.
membrane of renal tubular cells and bile ductule cells,
and in the endoplasmic reticulum of hepatocytes. The
enzyme has diagnostic value because it is released into
RESPONSES TO XENOBIOTICS
the blood from hepatic cells in various hepatobiliary
INCLUDE PHARMACOLOGIC,
diseases.
TOXIC, IMMUNOLOGIC,
D. OTHER REACTIONS
& CARCINOGENIC EFFECTS
The two most important other reactions are acetylation
Xenobiotics are metabolized in the body by the reac-
and methylation.
tions described above. When the xenobiotic is a drug,
1. Acetylation—Acetylation is represented by
phase 1 reactions may produce its active form or may
diminish or terminate its action if it is pharmacologi-
X+Acetyl-CoA →Acetyl-X+ CoA
cally active in the body without prior metabolism. The
where X represents a xenobiotic. As for other acetyla-
diverse effects produced by drugs comprise the area of
tion reactions, acetyl-CoA (active acetate) is the acetyl
study of pharmacology; here it is important to appreci-
donor. These reactions are catalyzed by acetyltrans-
ate that drugs act primarily through biochemical mech-
ferases present in the cytosol of various tissues, particu-
anisms. Table 53-2 summarizes four important reac-
larly liver. The drug isoniazid, used in the treatment of
tions to drugs that reflect genetically determined
tuberculosis, is subject to acetylation. Polymorphic
differences in enzyme and protein structure among in-
types of acetyltransferases exist, resulting in individuals
dividuals—part of the field of study known as pharma-
who are classified as slow or fast acetylators, and influ-
cogenetics (see below).
ence the rate of clearance of drugs such as isoniazid
from blood. Slow acetylators are more subject to certain
toxic effects of isoniazid because the drug persists
Table 53-2. Some important drug reactions due
longer in these individuals.
to mutant or polymorphic forms of enzymes or
2. Methylation—A few xenobiotics are subject to
proteins.1
methylation by methyltransferases, employing S-adeno-
sylmethionine (Figure 30-17) as the methyl donor.
Enzyme or Protein
THE ACTIVITIES OF XENOBIOTIC-
Affected
Reaction or Consequence
METABOLIZING ENZYMES ARE
Glucose-6-phosphate
Hemolytic anemia following in-
AFFECTED BY AGE, SEX,
dehydrogenase (G6PD)
gestion of drugs such as prim-
[mutations] (MIM 305900)
aquine
& OTHER FACTORS
Ca2+ release channel (ryan-
Malignant hyperthermia (MIM
Various factors affect the activities of the enzymes me-
odine receptor) in the
145600) following administra-
tabolizing xenobiotics. The activities of these enzymes
sarcoplasmic reticulum
tion of certain anesthetics (eg,
may differ substantially among species. Thus, for exam-
[mutations] (MIM 180901)
halothane)
ple, the possible toxicity or carcinogenicity of xenobi-
otics cannot be extrapolated freely from one species to
CYP2D6 [polymorphisms]
Slow metabolism of certain
(MIM 124030)
drugs (eg, debrisoquin), result-
another. There are significant differences in enzyme ac-
ing in their accumulation
tivities among individuals, many of which appear to be
due to genetic factors. The activities of some of these
CYP2A6 [polymorphisms]
Impaired metabolism of nico-
enzymes vary according to age and sex.
(MIM 122720)
tine, resulting in protection
Intake of various xenobiotics such as phenobarbital,
against becoming a tobacco-
PCBs, or certain hydrocarbons can cause enzyme in-
dependent smoker
duction. It is thus important to know whether or not
1G6PD deficiency is discussed in Chapters 20 and 52 and malig-
an individual has been exposed to these inducing agents
nant hyperthermia in Chapter 49. At least one gene other than
in evaluating biochemical responses to xenobiotics.
that encoding the ryanodine receptor is involved in certain cases
Metabolites of certain xenobiotics can inhibit or stimu-
of malignant hypertension. Many other examples of drug reac-
late the activities of xenobiotic-metabolizing enzymes.
tions based on polymorphism or mutation are available.
METABOLISM OF XENOBIOTICS
/
631
Certain xenobiotics are very toxic even at low levels
in the endoplasmic reticulum to become carcinogenic
(eg, cyanide). On the other hand, there are few xenobi-
(they are thus called indirect carcinogens). The activi-
otics, including drugs, that do not exert some toxic ef-
ties of the monooxygenases and of other xenobiotic-
fects if sufficient amounts are administered. The toxic
metabolizing enzymes present in the endoplasmic retic-
effects of xenobiotics cover a wide spectrum, but the
ulum thus help to determine whether such compounds
major effects can be considered under three general
become carcinogenic or are “detoxified.” Other chemi-
headings (Figure 53-1).
cals (eg, various alkylating agents) can react directly (di-
The first is cell injury (cytotoxicity), which can be
rect carcinogens) with DNA without undergoing intra-
severe enough to result in cell death. There are many
cellular chemical activation.
mechanisms by which xenobiotics injure cells. The one
The enzyme epoxide hydrolase is of interest be-
considered here is covalent binding to cell macromol-
cause it can exert a protective effect against certain car-
ecules of reactive species of xenobiotics produced by
cinogens. The products of the action of certain
metabolism. These macromolecular targets include
monooxygenases on some procarcinogen substrates are
DNA, RNA, and protein. If the macromolecule to
epoxides. Epoxides are highly reactive and mutagenic
which the reactive xenobiotic binds is essential for
or carcinogenic or both. Epoxide hydrolase—like cy-
short-term cell survival, eg, a protein or enzyme in-
tochrome P450, also present in the membranes of the
volved in some critical cellular function such as oxida-
endoplasmic reticulum—acts on these compounds,
tive phosphorylation or regulation of the permeability
converting them into much less reactive dihydrodiols.
of the plasma membrane, then severe effects on cellular
The reaction catalyzed by epoxide hydrolase can be rep-
function could become evident quite rapidly.
resented as follows:
Second, the reactive species of a xenobiotic may
bind to a protein, altering its antigenicity. The xenobi-
C C
+
H2O
C C
otic is said to act as a hapten, ie, a small molecule that
O
by itself does not stimulate antibody synthesis but will
HO
OH
combine with antibody once formed. The resulting an-
Epoxide
Dihydrodiol
tibodies can then damage the cell by several immuno-
logic mechanisms that grossly perturb normal cellular
PHARMACOGENOMICS WILL DRIVE THE
biochemical processes.
DEVELOPMENT OF NEW & SAFER DRUGS
Third, reactions of activated species of chemical car-
cinogens with DNA are thought to be of great impor-
As indicated above, pharmacogenetics is the study of
tance in chemical carcinogenesis. Some chemicals (eg,
the roles of genetic variations in the responses to drugs.
benzo[α]pyrene) require activation by monooxygenases
As a result of the progress made in sequencing the
GSH S-transferase or
Cytochrome P450
epoxide hydrolase
Xenobiotic
Reactive metabolite
Nontoxic metabolite
Covalent binding to
macromolecules
Cell injury
Hapten
Mutation
Antibody production
Cancer
Cell injury
Figure 53-1. Simplified scheme showing how metabolism of a xenobiotic can result in cell injury, immuno-
logic damage, or cancer. In this instance, the conversion of the xenobiotic to a reactive metabolite is catalyzed
by a cytochrome P450, and the conversion of the reactive metabolite (eg, an epoxide) to a nontoxic metabolite
is catalyzed either by a GSH S-transferase or by epoxide hydrolase.
632
/
CHAPTER 53
human genome, a new field of study—pharmacoge-
•
Cytochrome P450s are generally located in the endo-
nomics—has developed recently. It includes pharmaco-
plasmic reticulum of cells and are particularly en-
genetics but covers a much wider sphere of activity. In-
riched in liver.
formation from genomics, proteomics, bioinformatics,
•
Many cytochrome P450s are inducible. This has im-
and other disciplines such as biochemistry and toxicol-
portant implications in phenomena such as drug in-
ogy will be integrated to make possible the synthesis of
teraction.
newer and safer drugs. As the sequences of all our genes
•
Mitochondrial cytochrome P450s also exist and are
and their encoded proteins are determined, this will re-
involved in cholesterol and steroid biosynthesis.
veal many new targets for drug actions. It will also re-
They use a nonheme iron-containing sulfur protein,
veal polymorphisms (this term is briefly discussed in
adrenodoxin, not required by microsomal isoforms.
Chapter 50) of enzymes and proteins related to drug
•
Cytochrome P450s, because of their catalytic activi-
metabolism, action, and toxicity. DNA probes capable
ties, play major roles in the reactions of cells to
of detecting them will be synthesized, permitting
chemical compounds and in chemical carcinogenesis.
screening of individuals for potentially harmful poly-
•
Phase 2 reactions are catalyzed by enzymes such as
morphisms prior to the start of drug therapy. As the
structures of relevant proteins and their polymorphisms
glucuronosyltransferases, sulfotransferases, and glu-
tathione S-transferases, using UDP-glucuronic acid,
are revealed, model building and other techniques will
permit the design of drugs that take into account both
PAPS (active sulfate), and glutathione, respectively,
as donors.
normal protein targets and their polymorphisms. At
least to some extent, drugs will be tailor-made for indi-
•
Glutathione not only plays an important role in
viduals based on their genetic profiles. A new era of ra-
phase 2 reactions but is also an intracellular reducing
tional drug design built on information derived from
agent and is involved in the transport of certain
genomics and proteomics has already commenced.
amino acids into cells.
•
Xenobiotics can produce a variety of biologic effects,
SUMMARY
including pharmacologic responses, toxicity, immuno-
logic reactions, and cancer.
•
Xenobiotics are chemical compounds foreign to the
body, such as drugs, food additives, and environmen-
•
Catalyzed by the progress made in sequencing the
tal pollutants; more than 200,000 have been identi-
human genome, the new field of pharmacogenomics
fied.
offers the promise of being able to make available a
host of new rationally designed, safer drugs.
•
Xenobiotics are metabolized in two phases. The
major reaction of phase 1 is hydroxylation catalyzed
by a variety of monooxygenases, also known as the
REFERENCES
cytochrome P450s. In phase 2, the hydroxylated
species are conjugated with a variety of hydrophilic
Evans WE, Johnson JA: Pharmacogenomics: the inherited basis for
compounds such as glucuronic acid, sulfate, or glu-
interindividual differences in drug response. Annu Rev Ge-
nomics Hum Genet 2001;2:9.
tathione. The combined operation of these two
Guengerich FP: Common and uncommon cytochrome P450 reac-
phases renders lipophilic compounds into water-
tions related to metabolism and chemical toxicity. Chem Res
soluble compounds that can be eliminated from the
Toxicol 2001;14:611.
body.
Honkakakoski P, Negishi M: Regulation of cytochrome P450
•
Cytochrome P450s catalyze reactions that introduce
(CYP) genes by nuclear receptors. Biochem J 2000;347:321.
one atom of oxygen derived from molecular oxygen
Kalow W, Grant DM: Pharmacogenetics. In: The Metabolic and
into the substrate, yielding a hydroxylated product.
Molecular Bases of Inherited Disease, 8th ed. Scriver CR et al
NADPH and NADPH-cytochrome P450 reductase
(editors). McGraw-Hill, 2001.
are involved in the complex reaction mechanism.
Katzung BG (editor): Basic & Clinical Pharmacology, 8th ed. Mc-
Graw-Hill, 2001.
•
All cytochrome P450s are hemoproteins and gener-
McLeod HL, Evans WE: Pharmacogenomics: unlocking the
ally have a wide substrate specificity, acting on many
human genome for better drug therapy. Annu Rev Pharmacol
exogenous and endogenous substrates. They repre-
Toxicol 2001;41:101.
sent the most versatile biocatalyst known.
Nelson DR et al: P450 superfamily: update on new sequences, gene
•
Members of at least 11 families of cytochrome P450
mapping, accession numbers and nomenclature. Pharmacoge-
are found in human tissue.
netics 1996;6:1.
The Human Genome Project
54
Robert K. Murray, MD, PhD
BIOMEDICAL SIGNIFICANCE
rizes the differences between a genetic map, a cytoge-
netic map, and a physical map of a chromosome. These
The information deriving from determination of the se-
and other initial goals were achieved and surpassed by
quences of the human genome and those of other or-
the mid-nineties. In 1998, new goals for the United
ganisms will change biology and medicine for all time.
States wing of the HGP were announced. These in-
For example, with reference to the human genome, new
cluded the aim of completing the entire sequence by
information on our origins, on disease genes, on diag-
the end of 2003 or sooner. Other specific objectives
nosis, and possible approaches to therapy are already
concerned sequencing technology, comparative ge-
flooding in. Progress in fields such as genomics, pro-
nomics, bioinformatics, ethical considerations, and
teomics, bioinformatics, biotechnology, and pharma-
other issues. By the fall of
1998, about 6% of the
cogenomics is accelerating rapidly.
human genome sequence had been completed and the
The aims of this chapter are to briefly summarize
foundations for future work laid. Further progress was
the major findings of the Human Genome Project
catalyzed by the announcement that a second group,
(HGP) and indicate their implications for biology and
the private company Celera Genomics, led by Craig
medicine.
Venter, had undertaken the objective of sequencing the
human genome. Venter and colleagues had published
THE HUMAN GENOME PROJECT
in 1995 the entire genome sequences of Haemophilus
influenzae and Mycoplasma genitalium, the first of many
HAS A VARIETY OF GOALS
species to have their genomic sequences determined. An
The HGP, which started in 1990, is an international
important factor in the success of these workers was the
effort whose principal goals were to sequence the entire
use of a shotgun approach, ie, sonicating the DNA, se-
human genome and the genomes of several other model
quencing the fragments, and reassembling the se-
organisms that have been basic to the study of genetics
quence, based on overlaps. For comparison, a variety of
(eg, Escherichia coli, Saccharomyces cerevisiae [a yeast],
approaches that have been used at different times to
Drosophila melanogaster [the fruit fly], Caenorhabditis
study normal and disease genes are listed in Table 54-1.
elegans [the roundworm], and Mus musculus [the com-
mon house mouse]). Most of these goals have been ac-
A Draft Sequence of the Human Genome
complished. In the United States, the National Center
Was Announced in June 2000
for Human Genome Research (NCHGR) was estab-
lished in 1989, initially directed by James D. Watson
In June 2000, leaders of the IHGSC and the personnel
and subsequently by Francis Collins. The NCHGR
at Celera Genomics announced completion of working
played a leading role in directing the United States ef-
drafts of the sequence of the human genome, covering
fort in the HGP. In 1997, it became the National
more than 90% of it. The principal findings of the two
Human Genome Research Institute (NHGRI). The in-
groups were published separately in February 2001 in
ternational collaboration—involving groups from the
special issues of Nature (the IHGSC) and Science (Cel-
USA, UK, Japan, France, Germany, and China—came
era). The draft published by the Consortium was the
to be known as the International Human Genome Se-
product of at least 10 years of work involving 20 se-
quencing Consortium (IHGSC). Initially, a number of
quencing centers located in six countries. That pub-
short-term goals were established for the United States
lished by Celera and associates was the product of some
effort—eg, producing a human genetic map with mark-
3 years or less of work; it relied in part on data obtained
ers
2-5 centimorgans (cM) apart and constructing a
by the IHGSC. The combined achievement has been
physical map of all 24 human chromosomes (22 auto-
hailed, among other descriptions, as providing a Library
somal plus X and Y) with markers spaced at approxi-
of Life, supplying a Periodic Table of Life, and finding
mately 100,000 base pairs (bp). Figure 54-1 summa-
the Holy Grail of Human Genetics.
633
634
/
CHAPTER 54
Genetic map
20
30
30
20
25
cM
Cytogenetic
map
Physical map
25
50
75
100
125
150 Mb
EcoRI Hind III NotI
Restriction map
STS map
Contig map
Figure 54-1. Principal methods used to identify and isolate normal
and disease genes. For the genetic map, the positions of several hypo-
thetical genetic markers are shown, along with the genetic distances in
centimorgans between them. The circle shows the position of the cen-
tromere. For the cytogenetic map, the classic banding pattern of a hypo-
thetical chromosome is shown. For the physical map, the approximate
physical positions of the above genetic markers are shown, along with
the relative physical distances in megabase pairs. Examples of a restric-
tion map, a contig mark, and an STS map are also shown. (Reproduced,
with permission, from Green ED, Waterston RH: The Human Genome
Project: Prospects and implications for clinical medicine. JAMA 1991;266:
1966. Copyright © 1991 by the American Medical Association.)
Different Approaches Were Used
(STSs), whose locations had been already determined.
by the Two Groups
STSs are short (usually < 500 bp), unique genomic loci
for which a PCR assay is available. Clones of the BACs
We shall summarize the major findings reported in the
were then broken into small fragments (shotgunning).
two drafts and comment on their implications. While
Each fragment was then sequenced, and computer algo-
there are differences between the drafts, they will not be
rithms were used that recognized matching sequence
dwelt on here, as the areas of general agreement are
information from overlapping fragments to piece to-
much more extensive. It is worthwhile, however, to
gether the complete sequence.
summarize the different approaches used by the two
Celera used the whole genome shotgun approach,
groups. Basically, the IHGSC employed a map first,
in effect bypassing the mapping step. Shotgun frag-
sequence later approach. In part, this was because se-
ments were assembled by algorithms onto large scaf-
quencing was a slow process when the public project
folds, and the correct position of these scaffolds in the
started, and the strategy of the Consortium evolved
genome was determined using STSs. A scaffold is a se-
over time as advances were made in sequencing and
ries of “contigs” that are in the right order but not nec-
other techniques. The overall approach, referred to as
essarily connected in one continuous sequence. Contigs
hierarchical shotgun sequencing, consisted of frag-
are contiguous sequences of DNA made by assembling
menting the entire genome into pieces of approxi-
overlapping sequenced fragments of a natural chromo-
mately 100-200 kb and inserting them into bacterial
some or a BAC. The availability of high-throughput
artificial chromosomes (BACs). The BACs were then
sequenators, powerful computer programs, the ele-
positioned on individual chromosomes by looking for
ment of competition, and other factors accounted for the
marker sequences known as sequence-tagged sites
rapid progress made by both groups from 1998 onward.
THE HUMAN GENOME PROJECT
/
635
Table 54-1. Principal methods used to identify and isolate normal and disease genes.
Procedure
Comments
Detection of specific cytogenetic
For instance, a small deletion of band Xp21.2 was important in cloning the gene involved in
abnormalities
Duchenne muscular dystrophy.
Extensive linkage studies
Large families with defined pedigrees are desirable. Dominant genes are easier to recognize
than recessives.
Use of probes to define marker
Probes identify STSs, RFLPs, SNPs,1 etc; thousands, covering all the chromosomes, are now
loci
available. It is desirable to flank the gene on both sides, clearly delineating it.
Radiation hybrid mapping2
Now the most rapid method of localizing a gene or DNA fragment to a subregion of a human
chromosome and constructing a physical map.
Use of rodent or human somatic
Permits assignment of a gene to one specific chromosome but not to a subregion.
cell hybrids
Fluorescence in situ hybridization
Permits localization of a gene to one chromosomal band.
Use of pulsed-field gel
Permits isolation of large DNA fragments obtained by use of restriction endonucleases (rare
electrophoresis (PFGE) to
cutters) that result in very limited cutting of DNA.
separate large DNA fragments
Chromosome walking
Involves repeated cloning of overlapping DNA segments; the procedure is laborious and can
usually cover only 100-200 kb.
Chromosome jumping
By cutting DNA into relatively large fragments and circularizing it, one can move more quickly
and cover greater lengths of DNA than with chromosomal walking.
Cloning via YACs, BACs, cosmids,
Permits isolation of fragments of varying lengths.
phages, and plasmids
Detection of expression of
The mRNA should be expressed in affected tissues.
mRNAs in tissues by Northern
blotting using one or more
fragments of the gene as a
probe
PCR
Can be used to amplify fragments of the gene; also many other applications.
DNA sequencing
Establishes the highest resolution physical map. Identifies open reading frame. Facilities with
many high throughput instruments could sequence millions of base pairs per day.
Databases
Comparison of DNA and protein sequences obtained from unknown gene with known se-
quences in databases can facilitate gene identification.
Abbreviations: STS, sequence tagged site; RFLP, restriction fragment linked polymorphism; SNP, single nucleotide polymorphism; YAC,
yeast artificial chromosome; BAC, bacterial artificial chromosome; PCR, polymerase chain reaction.
1Many single nucleotide polymorphisms (SNPs) are being detected and catalogued. These are stable and frequent, and their detection
can be automated. It is anticipated that they will be particularly useful for mapping complex traits such as diabetes mellitus.
panel of somatic cell hybrids, with each cell line containing a random set of irradiated human genomic DNA in a hamster background.
Briefly, the radiation fragments the DNA into small pieces of variable length; if a gene is located close to another known gene, it is likely
that the two will remain linked (compare genetic linkage) on the same fragment. An STS marker is typed against a radiation hybrid panel
by using its two oligonucleotide primers to perform a PCR assay against the DNA from each hybrid cell line of the panel. If enough mark-
ers are typed on one panel, continuous linkage can be established along each arm of a chromosome, and the markers can be assembled
into the map as a single linkage group.
636
/
CHAPTER 54
DETERMINATION OF THE SEQUENCE OF
fly (13,061). The figures suggest that the complexity of
humans compared with that of the two simpler organ-
THE HUMAN GENOME HAS PRODUCED A
isms must have explanations other than strictly gene
WEALTH OF NEW FINDINGS
number.
Only a small fraction of the findings can be covered
here. The interested reader is referred to the original ar-
Only 1.1-1.5% of the Human
ticles. Table 54-2 summarizes a number of the high-
Genome Encodes Proteins
lights, which can now be described.
Analyses of the available data reveal that 1.1-1.5% of
Most of the Human Genome
the genome consists of exons. About 24% consists of
Has Been Sequenced
introns, and 75% of sequences lying between genes (in-
tergenic). Comparisons with the data on the round-
Over 90% of the human genome had been sequenced
worm and fruit fly have shown that exon size across the
by July 2000. This is by far the largest genome se-
three species is relatively constant (mean size of 145 bp
quenced, with an estimated size of approximately 3.2
in humans). However, intron size in humans is much
gigabases (Gb). Prior to the human genome, that of the
more variable (mean size of over 3300 bp), resulting in
fruit fly had been the largest (~180 Mb) sequenced.
great variation in gene size.
Gaps still exist, small and large, and the quality of some
of the sequencing data will be refined since some of the
findings are probably not exactly right.
The Landscape of Human Chromosomes
Varies Widely
The Human Genome Is Estimated to
There are marked differences among individual chro-
Encode About 30,000-40,000 Proteins
mosomes in many features, such as gene number per
The greatest surprise provided by the results to date has
megabase, density of single nucleotide polymorphisms
been the apparently low number of genes encoding pro-
(SNPs), GC content, number of transposable elements
teins, estimated to lie between 30,000 and 40,000. The
and CpG islands, and recombination rate. To take one
higher number could increase as new data are obtained.
example, chromosome 19 has the richest gene content
This number is approximately twice that found in the
(23 genes per megabase), whereas chromosome 13 and
roundworm (19,099) and three times that of the fruit
the Y chromosome have the sparsest content (5 genes
per megabase). Explanations for these variations are not
apparent at this time.
Table 54-2. Major findings reported in the rough
drafts of the human genome.
Human Genes Do More Work Than
Those of Simpler Organisms
• More than 90% of the genome has been sequenced; gaps,
Alternative splicing appears to be more prevalent in hu-
large and small, remain to be filled in.
mans, involving at least 35% of their genes. Data indi-
• Estimated number of protein-coding genes ranges from
cate that the average number of distinct transcripts per
30,000 to 40,000.
gene for chromosomes 22 and 19 were 2.6 and 3.2, re-
• Only 1.1-1.5% of the genome codes for proteins.
spectively. These figures are higher than for the round-
• There are wide variations in features of individual chromo-
worm, where only 12.2% of genes appear to be alterna-
somes (eg, in gene number per Mb, SNP density, GC con-
tively spliced and only 1.34 splice variants per gene
tent, numbers of transposable elements and CpG islands,
were noted.
recombination rate).
• Human genes do more work than those of the roundworm
or fruit fly (eg, alternative splicing is used more frequently).
The Human Proteome Is More Complex
• The human proteome is more complex than that found in
Than That of Invertebrates
invertebrates.
• Repeat sequences probably constitute more than 50% of
Relatively few new protein domains appear to have
the genome.
emerged among vertebrates. However, the number of
• Approximately 100 coding regions have been copied and
distinct domain architectures (~1800) in human pro-
moved by RNA-based transposons.
teins is 1.8 times that of the roundworm or fruit fly.
• Approximately 200 genes may be derived from bacteria by
About 90 vertebrate-specific families of proteins have
lateral transfer.
been identified, and these have been found to be en-
• More than 3 million SNPs have been identified.
riched in proteins of the immune and nervous systems.
THE HUMAN GENOME PROJECT
/
637
The results of the two drafts are rich in information
Segmental duplications have been found to be much
about protein families and classes. One example is
more common than in the roundworm or fruit fly. It is
shown in Table 54-3, in which the major classes of
possible that these structures may be involved in exon
proteins encoded by human genes are listed. As can be
shuffling and the increased diversity of proteins found
seen, the largest class is “unknown.” Identification of
in humans.
these unknown proteins will be a major focus of activity
for many laboratories.
Other Findings of Interest
The last three major points of interest listed in Table
Repeat Sequences Probably Constitute
54-2 will be briefly described together.
More Than 50% of the Human Genome
Approximately 100 coding regions are estimated to
Repeat sequences probably account for at least half of
have been copied and moved by RNA-based trans-
the genome. They fall into five classes: (1) transposon-
posons (retrotransposons). It is possible that some of
derived repeats
(interspersed repeats);
(2) processed
these genes may adopt new roles in the course of time.
pseudogenes; (3) simple sequence repeats; (4) segmental
A surprising finding is that over 200 genes may be de-
duplications, made up of 10-300 kb that have been
rived from bacteria by lateral transfer. None of these
copied from one region of the genome into another;
genes are present in nonvertebrate eukaryotes. More
and (5) blocks of tandemly repeated sequences, found
than 3 million SNPs have been identified. It is likely
at centromeres, telomeres, and other areas. Consider-
that they will prove invaluable for certain aspects of
able information on most of the above classes of repeat
gene mapping.
sequences—of great value in understanding the archi-
It is stressed that the findings listed here are only a
tecture and development of the human genome—is re-
few of those reported in the drafts, and the reader is
ported in the drafts. Only two points of interest will be
urged to consult the original reports (see References,
mentioned here. It is speculated that Alu elements, the
below).
most prominent members (about 10% of the total
genome) of the short interspersed elements (SINEs),
FURTHER WORK IS PLANNED ON THE
may be present in GC-rich areas because of positive se-
HUMAN & OTHER GENOMES
lection, implying that they are of benefit to the host.
The IHGSC has indicated that it will determine the
complete sequence, it is hoped, by 2003. The task in-
Table 54-3. Major classes of proteins encoded by
volves filling in the gaps and identifying new genes,
human genes.1
their locations, and functions. Regulatory regions will
be identified, and the sequences of other large genomes
(eg, of the house mouse; of Rattus norvegicus, the Nor-
Class of Protein
Number (%)2
way rat; of Danio rerio, the zebra fish; of Fugu rubripes,
Unknown
12,809 (41%)
the tiger puffer fish; and of one or more primates) will
be obtained; indeed, a draft version of the genome of
Nucleic acid enzymes
2,308 (7.5%)
the tiger puffer fish was published in 2002. Additional
Transcription factors
1,850 (6%)
SNPs will be identified; a complete catalog of these
Receptors
1,543 (5%)
variants is expected to be of great value in mapping
genes associated with complex traits and for other uses
Hydrolases
1,227 (4.0%)
as well. Along with the above, existing databases will be
Select regulatory molecules (eg,
988 (3.2%)
added to as new information flows in, and new data-
G proteins, cell cycle regulators)
bases will probably be established to serve specific pur-
poses. A variety of studies in functional genomics (ie,
Protooncogenes
902 (2.9%)
the study of genomes to determine the functions of all
Cytoskeletal structural proteins
876 (2.8%)
their genes and their products) will also be undertaken.
Kinases
868 (2.8%)
IMPLICATIONS FOR PROTEOMICS,
1Data from Venter JC et al: The sequence of the human genome.
Science 2001;291:1304.
BIOTECHNOLOGY, & BIOINFORMATICS
2The percentages are derived from a total of 26,383 genes re-
ported in the rough draft by Celera Genomics. Classes containing
Many fields will be influenced by knowledge of the
more than 2.5% of the total proteins encoded by the genes iden-
human genome. Only a few are briefly discussed here.
tified when this rough draft was written are arbitrarily listed as
Proteomics (see Chapter 4) in its broadest sense is
major.
the study of all the proteins encoded in an organism (ie,
638
/
CHAPTER 54
the proteome), including their structures, modifica-
effects on health services and the diagnosis and treat-
tions, functions, and interactions. In a narrower sense,
ment of disease.
it involves the identification and study of multiple pro-
teins linked through cellular actions—but not necessar-
SUMMARY
ily the entire proteome. With regard to humans, many
• Determination of the complete sequence of the
individual proteins will be identified and characterized;
human genome, now almost completed, is one of the
their interactions and levels will be determined in phys-
most significant scientific achievements of all time.
iologic and pathologic states, and the resulting informa-
tion will be entered into appropriate databases. Tech-
• Many important findings have already emerged. The
niques such as two-dimensional electrophoresis, a
one to date that has generated the most discussion is
variety of modes of mass spectrometry, and antibody
that the number of human genes may be only two to
arrays will be central to expansion of this rapidly grow-
three times that estimated for the roundworm and
ing field. Overall, proteomics will greatly advance our
the fruit fly.
knowledge of proteins at the basic level and will also
• Information flowing from the Human Genome Proj-
nourish biotechnology as new proteins that are likely
ect is having major influences in fields such as pro-
to have diagnostic, therapeutic, and other uses are dis-
teomics, bioinformatics, biotechnology, and phar-
covered and methods for their economic production are
macogenomics as well as all areas of biology and
developed. Specialists in bioinformatics will be in de-
medicine.
mand, as this field rapidly gears up to manage, analyze,
• It is hoped that the knowledge derived will be used
and utilize the flood of data from genomic and pro-
wisely and fairly and that the benefits that will ensue
teomic studies.
regarding health, disease, and other matters will be
made available to all people everywhere.
IMPLICATIONS FOR MEDICINE
REFERENCES
Practically every area of medicine will be affected by the
Collins FS, McKusick VA: Implications of the Human Genome
new information accruing from knowledge of the
Project for medical science. JAMA 2001;285:540. (The Feb-
human genome. The tracking of disease genes will be
ruary 7, 2001, issue describes opportunities for medical re-
enormously facilitated. As mentioned above, SNP maps
search in the 21st century. Many articles of interest.)
should greatly assist determination of genes involved in
Hedges SB, Kumar S: Vertebrate genomes compared. Science
complex diseases. Probes for any gene will be available
2002;297:1283. (The same issue—No. 5585, August 23—
contains a draft version of the genome of the tiger puffer
if needed, leading to improved diagnostic testing for
fish.)
disease susceptibility genes and for genes directly in-
McKusick VA: The anatomy of the human genome: a neo-Vesalian
volved in the causation of specific diseases. The field of
basis for medicine in the
21st century. JAMA 2001;286:
pharmacogenomics (see Chapter 53) is already ex-
2289. (The November 14, 2001, issue contains a number of
panding greatly, and it is possible that in the future
other excellent articles—eg, on clinical proteomics, pharma-
drugs will be tailored to accommodate the variations in
cogenomics—relating to the Human Genome Project and its
enzymes and other proteins involved in drug action and
impact on medicine.)
metabolism found among individuals. Studies of genes
Nature
2001;409(6822)
(February
15), and Science
2001;291
involved in behavior may lead to new insights into the
(5507) (February 16). (These two issues present the rough
drafts prepared by the IHGSC and Celera, respectively, along
causation and possible treatment of psychiatric disor-
with many other articles analyzing the meaning and signifi-
ders. Many ethical issues—eg, privacy concerns and
cance of the findings.)
the use of genomic information for commercial pur-
Science 2001;294(5540) (October 5). (This issue contains a num-
poses—will have to be addressed. It will also be impor-
ber of articles under the title Genome: Unlocking Biology’s
tant that medical and economic benefits accrue to indi-
Storehouse. They describe new ideas, approaches, and re-
viduals in Third World countries from the anticipated
search related to genome information.)
APPENDIX
SELECTED WORLD WIDE WEB SITES
(Maintains the EMBL Nucleotide and SWISS-PROT databases as
well as other databases.)
The following is a list of Web sites that readers may
find useful. The sites have been visited at various times
(A database of human genes, their products, and their involvements
by one of the authors (RKM). Most are located in the
in diseases. From the Weizmann Institute of Science.)
USA, but many provide extensive links to international
sites and to databases (eg, for protein and nucleic acid
(A medical genetics information resource with comprehensive arti-
sequences) and online journals. RKM would be grateful
cles on many genetic diseases.)
if readers who find other useful sites would notify him
of their URLs by e-mail (rmurray6745@rogers. com) so
(Coverage of the genetic basis of many different types of diseases.)
that they may be considered for inclusion in future edi-
tions of this text.
tgn_side.htm
Readers should note that URLs may change or cease
(TGN is an informal worldwide grouping of scientists who share an
to exist.
interest in carbohydrates. The site contains considerable in-
formation on carbohydrates and an extensive list of links to
other sites dealing with sugar-containing molecules).
ACCESS TO THE BIOMEDICAL
(An excellent site for following current biomedical research. Con-
tains a comprehensive Research News Archive.)
LITERATURE
uwcm/mg/hgmd0.html
(An extensive tabulation of mutations in human genes from the In-
stitute of Medical Genetics in Cardiff, Wales.)
(Extensive lists of various classes of journals—biology, medicine,
etc—and offers also the most extensive list of journals with
free online access.)
(From the Human Genome Program of the United States Depart-
ment of Energy.)
(Free access to Medline via PubMed.)
(Sequences of various bacterial genomes and other information.)
GENERAL RESOURCE SITES
Karolinska Institute Nutritional and Metabolic Diseases: http://
The Biology Project
(from the University of Arizona): http://
(Access to information on many nutritional and metabolic disor-
ders.)
Harvard Department of Molecular & Cellular Biology Links:
(A human mitochondrial genome database.)
SITES ON SPECIFIC TOPICS
nih.gov/
(Information on molecular biology and how molecular processes af-
fect human health and disease.)
(Useful information on nutrition, on the role of various biomole-
cules—eg, cholesterol, lipoproteins—in heart disease, and on
the major cardiovascular diseases.)
gov/
(Extensive information about the Human Genome Project.)
nih.gov/ncicgap
(An interdisciplinary program to generate the information and
(Includes links to the separate Institutes and Centers that constitute
technical tools needed to decipher the molecular anatomy of
NIH, covering a wide range of biomedical research.)
the cancer cell.)
(A comprehensive list of neuroscience resources; part of the World-
home.html
Wide Web Virtual Library.)
639
640
/
APPENDIX
main.html
(Access to information on more than 7000 rare diseases, including
Chapter 6
current research.)
OMIM Home Page—Online Mendelian Inheritance in Man:
(An extensive catalog of human genetic disorders, updated daily.
Lists access to various allied resources.)
Chapter 7
(A worldwide repository for the processing and distribution of
three-dimensional biologic macromolecular structure data.)
shtml
(Information on the protein kinase family of enzymes.)
Chapter 8
(An extensive list of Web resources for protein scientists.)
(A one-stop overview for the specialist or nonspecialist of what is
happening in cell signaling.)
Chapter 9
(Aims to promote advancement of public education in endocrinol-
ogy. Contains a number of articles on endocrinology and a
list of links to other relevant sites.)
LEC06/Lec6.htm
tbase—the Transgenic/Targeted Mutation Database at the Jackson
(An attempt to organize information on transgenic animals and tar-
geted mutations generated and analyzed worldwide.)
Chapter 22
(A genome research center whose purpose it is to increase knowl-
edge of genomes, particularly through large-scale sequencing
and analysis,)
Whitehead Institute/MIT Center for Genome Research: http://
Chapter 28
(Access to various databases and articles entitled “What’s New in
Genome Research.”)
Chapter 2
Chapter 29
Chapter 3
Chapter 30
Chapter 4
Chapter 34
Chapter 5
APPENDIX
/
641
BIOCHEMICAL JOURNALS AND REVIEWS
Biochimica et Biophysica Acta (Biochim Biophys
Acta)
The following is a partial list of biochemistry journals
Biochimie (Biochimie)
and review series and of some biomedical journals that
European Journal of Biochemistry (Eur J Biochem)
contain biochemical articles. Biochemistry and biology
journals now usually have Web sites, often with useful
Indian Journal of Biochemistry and Biophysics (In-
links, and some journals are fully accessible without
dian J Biochem Biophys)
charge. The reader can obtain the URLs for the follow-
Journal of Biochemistry
(Tokyo)
(J Biochem
ing by using a search engine.
[Tokyo])
Journal of Biological Chemistry (J Biol Chem)
Annual Reviews of Biochemistry, Cell and Develop-
Journal of Clinical Investigation (J Clin Invest)
mental Biology, Genetics, Genomics and Human
Journal of Lipid Research (J Lipid Res)
Genetics
Nature (Nature)
Archives of Biochemistry and Biophysics
(Arch
Biochem Biophys)
Nature Genetics (Nat Genet)
Biochemical and Biophysical Research Communica-
Proceedings of the National Academy of Sciences
tions (Biochem Biophys Res Commun)
USA (Proc Natl Acad Sci USA)
Biochemical Journal (Biochem J)
Science (Science)
Biochemistry (Biochemistry)
Trends in Biochemical Sciences (Trends Biochem
Sci)
Biochemistry (Moscow) (Biochemistry [Mosc])
Index
Note: Page numbers in bold face type indicate a major discussion. A t following a page number indicates tabular ma-
terial and an f following a page number indicates a figure.
A bands, 556, 557f, 558f
formation of, 254f, 255-259, 255f, 256f,
molecular structure affecting strength of,
A blood group substance, 618, 619f
257f, 258f, 259f
12, 12t
A cyclins, 333, 334f, 335t
lipogenesis and, 173-177, 174f, 175f
polyfunctional, nucleotides as, 290
A gene, GalNAc transferase encoded by,
as fatty acid building block, 176-177
as proton donors, 9
618-619
pyruvate dehydrogenase regulated by,
strong, 9
A (aminoacyl/acceptor) site, aminoacyl-
141-142, 142f, 178
weak. See Weak acids
tRNA binding to, in protein
pyruvate oxidation to, 134, 135f,
Aciduria
synthesis, 368, 368f
140-142, 141f, 142f, 143t
dicarboxylic, 188
ABC-1. See ATP-binding cassette
xenobiotic metabolism and, 630
methylmalonic, 155
transporter-1
Acetyl-CoA carboxylase, 156t, 179
orotic, 300, 301
Abetalipoproteinemia, 207, 228t
in lipogenesis regulation, 156t, 173,
urocanic, 250
ABO blood group system, biochemical basis
174f, 178, 178f, 179
Aconitase (aconitate hydratase), 130
of, 617-619, 619f
N-Acetylgalactosamine (GalNAc), in
ACP. See Acyl carrier protein
Absorption, 474-480
glycoproteins, 515, 516t
Acrosomal reaction, glycoproteins in, 528
Absorption chromatography, for
N-Acetylglucosamine (GlcNAc), in
ACTH. See Adrenocorticotropic hormone
protein/peptide purification, 22
glycoproteins, 516t
Actin, 557, 559, 560
Absorption spectra, of porphyrins,
N-Acetylglucosamine phosphotransferase
decoration of, 561, 561f
273-274, 277f
(GlcNAc phosphotransferase)
fibronectin receptor interacting with,
Absorptive pinocytosis, 430
in I-cell disease, 532
540, 541f
ACAT (acyl-CoA:cholesterol acyltrans-
in pseudo-Hurler polydystrophy, 532
in muscle contraction, 557-559, 558f,
ferase), 223
N-Acetylglutamate, in urea biosynthesis,
561-562, 561f, 562f
Accelerator (Ac-) globulin (factor V), 600t,
245, 246f
regulation of striated muscle and,
601, 602f
Acetyl hexosamines, in glycoproteins, 109t
562-563
Acceptor (A/aminoacyl) site, aminoacyl-
N-Acetyl lactosamines, on N-linked glycan
in nonmuscle cells, 576-577
tRNA binding to, in protein
chains, 521
in red cell membranes, 615f, 616f, 616t,
synthesis, 368, 368f
Acetyl (acyl)-malonyl enzyme, 173, 175f
617
Acceptor arm, of tRNA, 310, 312f, 360,
N-Acetyl neuraminic acid, 169, 171f
in striated vs. smooth muscle, 572t
361f
in gangliosides, 201, 203f
β-Actin, 577
Aceruloplasminemia, 589
in glycoproteins, 169, 171f, 515, 516t
F-Actin, 559, 559f, 561
ACEs. See Angiotensin-converting enzyme
in mucins, 519f, 520
in nonmuscle cells, 576, 577
inhibitors
Acetyl transacylase, 173, 174f, 175f
G-Actin, 559, 559f
Acetal links, 105-106
Acetyltransferases, xenobiotic metabolism
in nonmuscle cells, 576
Acetic acid, 112t
and, 630
γ-Actin, 577
pK/pKa value of, 12t
Acholuric jaundice, 282
Actin-filament capping protein, 540, 541f
Acetoacetate, 183-184, 184f
Achondroplasia, 432t, 551t, 553-554, 554f
Actin (thin) filaments, 557, 558f, 559f
in tyrosine catabolism, 254f, 255
Acid anhydride bonds, 287
α-Actinin, 540, 566t
Acetoacetyl-CoA synthetase, in mevalonate
Acid anhydrides, group transfer potential
Activated protein C, in blood coagulation,
synthesis, 219, 220f
for, 289-290, 289f, 290f, 290t
603
Acetone, 183
Acid-base balance, ammonia metabolism in,
Activation energy, 61, 63
Acetone bodies. See Ketone bodies
245
Activation energy barrier, enzymes affecting,
Acetylation
Acid-base catalysis, 51-52
63
in covalent modification, mass increases
HIV protease in, 52, 53f
Activation function 1, 460
and, 27t
α1-Acid glycoprotein (orosomucoid), 583t
Activation function 2, 460
of xenobiotics, 630
Acid hydrolysis, for polypeptide cleavage,
Activation reaction, 456, 458f
Acetyl-CoA, 122, 122f
26t
Activator-recruited cofactor (ARC), 472t,
carbohydrate metabolism and, 122, 122f,
Acid phosphatase, diagnostic significance
473
123f
of, 57t
Activators
catabolism of, 130-135, 131f, 132f. See
Acidemia, isovaleric, 259, 259-262
in regulation of gene expression, 374,
also Citric acid cycle
Acidosis
376. See also Enhancers
cholesterol synthesis and, 219-220, 220f,
lactic. See Lactic acidosis
transcription, 351, 351t
221f, 222f
metabolic, ammonia in, 245
Active chromatin, 316-318, 318f, 383
fatty acid oxidation to, 123-124, 123f,
Acids
Active site, 51, 51f. See also Catalytic site
181-183, 181f, 182f
conjugate, 10
∆GF and, 63
643
644
/
INDEX
Active sulfate (adenosine 3′-phosphate-
metabolism in, 214-215, 214f, 235t
Alanine (alanine-pyruvate) aminotransferase
5′-phosphosulfate), 289, 289f, 629
control of, 216-217
(ALT/SGPT)
Active transport, 423, 423t, 424f, 426-427,
ADP, 287f
diagnostic significance of, 57t
427-428, 428f
free energy of hydrolysis of, 82t
in urea synthesis, 243-244, 244f
in bilirubin secretion, 280, 281f
mitochondrial respiratory rate and,
β-Alanyl dipeptides, 264, 265f
Activity (physical), energy expenditure and,
94-95, 97t, 98f
Albumin, 580, 581f, 583-584, 583t
478
myosin, muscle contraction and, 561,
conjugated bilirubin binding to, 283
Actomyosin, 560
561f
free fatty acids in combination with, 180,
ACTR coactivator, 472, 472t
in platelet activation, 606f, 607
206, 206t, 584
Acute fatty liver of pregnancy, 188
in respiratory control, 94-95, 97, 97t,
glomerular membrane permeability to,
Acute inflammatory response, neutrophils
98f, 134-135
540-541
in, 620
structure of, 83f
Albuminuria, 542
Acute phase proteins, 583, 583t
ADP/ATP transporter, 95, 98f
Alcohol, ethyl. See Ethanol
negative, vitamin A as, 483-484
ADPase, 607, 607t
Alcohol dehydrogenase, in fatty liver, 212
Acylcarnitine, 180-181, 181f
ADP-chaperone complex, 508. See also
Alcoholism
Acyl carrier protein (ACP), 173, 174f
Chaperones
cirrhosis and, 212
synthesis of, from pantothenic acid, 173,
ADP-ribose, NAD as source of, 490
fatty liver and, 212-214
495
ADP-ribosylation, 490
Aldehyde dehydrogenase, 87
Acyl-CoA:cholesterol acyltransferase
Adrenal cortical hormones. See also specific
in fatty liver, 212
(ACAT), 223
hormone and Glucocorticoids;
Aldolase A, deficiency of, 143
Acyl-CoA dehydrogenase, 87, 181, 182f
Mineralocorticoids
Aldolase B, 167, 168f
medium-chain, deficiency of, 188
synthesis of, 438-442, 440f, 441f
deficiency of, 171
Acyl-CoA synthetase (thiokinase)
Adrenal gland, cytochrome P450 isoforms
Aldolases, in glycolysis, 137, 138f
in fatty acid activation, 180, 181f
in, 627
Aldose-ketose isomerism, 103f, 104, 104f
in triacylglycerol synthesis, 199, 214f, 215
Adrenal medulla, catecholamines produced
Aldose reductase, 167, 169f, 172
Acylglycerols, 197
in, 445
Aldoses, 102, 102t, 104t
metabolism of, 197-201
Adrenergic receptors, in glycogenolysis, 148
ring structure of, 104
catabolism, 197
Adrenocorticotropic hormone (ACTH),
Aldosterone
clinical aspects of, 202
437, 438, 439f, 453, 453f
binding of, 455
synthesis, 197-201, 197f, 198f
Adrenodoxin, 627
synthesis of, 438-440, 441f
in endoplasmic reticulum, 126, 127f
Adrenodoxin reductase, 627
angiotensin affecting, 451, 452
Adapter proteins, in absorptive pinocytosis,
Adrenogenital syndrome, 442
Aldosterone synthase (18-hydroxylase), in
430
Adrenoleukodystrophy, neonatal, 503, 503t
steroid synthesis, 440, 441f
Adenine, 288f, 288t
Aerobic glycolysis, 139
Alkaline phosphatase
Adenosine, 287f, 288t
as muscle ATP source, 575, 575f, 575t
in bone mineralization, 550
base pairing of in DNA, 303, 304, 305f
Aerobic respiration
isozymes of, diagnostic significance of,
in uric acid formation, 299, 299f
citric acid cycle and, 130
57t
Adenosine deaminase
AF-1. See Activation function 1
in recombinant DNA technology, 400t
deficiency of, 300
AF-2. See Activation function 2
Alkalosis, metabolic, ammonia in, 245
localization of gene for, 407t
AF-2 domain, 470
Alkaptonuria, 255
Adenosine diphosphate. See ADP
Affinity chromatography
Allergic reactions, peptide absorption
Adenosine monophosphate. See AMP
for protein/peptide purification, 23
causing, 474
Adenosine 3′-phosphate-5′-phosphosulfate,
in recombinant fusion protein
Allopurinol, 290, 297
289, 289f, 629
purification, 58, 59f
Allosteric activators, 157
Adenosine triphosphate. See ATP
AFP. See Alpha-fetoprotein
Allosteric effectors/modifiers, 129
S-Adenosylmethionine, 258f, 259, 264,
Agammaglobulinemia, 595
in gluconeogenesis regulation, 157
266f, 289, 290f, 290t
Age, xenobiotic-metabolizing enzymes
negative, 74. See also Feedback inhibition
Adenylic acid, as second messenger, 457
affected by, 630
second messengers as, 76
Adenylyl cyclase
Aggrecan, 542, 551t, 553, 553f
Allosteric enzymes, 75, 129
in cAMP-dependent signal transduction,
Aggregates, formation of after denaturation,
aspartate transcarbamoylase as model of,
458-459, 460t
36
75
cAMP derived from, 147
Aging, glycosaminoglycans and, 549
Allosteric properties of hemoglobin, 42-46
in lipolysis, 215, 216f
Aglycone, 105, 106
Allosteric regulation, of enzymatic catalysis,
Adenylyl kinase (myokinase), 84
AHG. See Antihemophilic factor A/globulin
74, 74-76, 75f, 128f, 129
deficiencies of, 151-152
AIB1 coactivator, 472, 472t
gluconeogenesis regulation and, 157
in gluconeogenesis regulation, 157
ALA. See Aminolevulinate
Allosteric site, 74, 75
as source of ATP in muscle, 573, 575f
Alanine, 15t
Alpha-adrenergic receptors, in
Adhesion molecules, 529, 529t. See also Cell
pI of, 17
glycogenolysis, 148
adhesion
in pyruvate formation, 250, 252f
Alpha-amino acids. See also Amino acids
Adipose tissue, 111, 214-215, 214f
synthesis of, 237, 238f
genetic code specifying, 14, 15-16t
brown, 217, 217f
β-Alanine, 264, 300, 301f
in proteins, 14
INDEX
/
645
Alpha-amino nitrogen. See Amino acid
absorption of, 477
Aminolevulinate dehydratase, 270, 273f
nitrogen
analysis/identification of, 20
in porphyria, 274, 277t
Alpha anomers, 104
blood glucose and, 159
Aminolevulinate synthase, 270, 272-273,
Alpha1 antiproteinase/antitrypsin. See
branched chain, catabolism of, 259,
273f, 276f
α1-Antiproteinase
260f, 261f, 262f
in porphyria, 274, 277f, 278
Alpha- (α) carotene, 482
disorders of, 259-262
Aminopeptidases, 477
Alpha-fetoprotein, 583t
in catalysis, conservation of, 54, 55t
Aminophospholipids, membrane
Alpha-globin gene, localization of, 407t
chemical reactions of, functional groups
asymmetry and, 420
Alpha (α) helix, 31-32, 32f, 33f
dictating, 18-20
Aminoproteinase, procollagen, 537
amphipathic, 31-32
deamination of. See Deamination
Amino sugars (hexosamines), 106, 106f
in myoglobin, 40, 41f
deficiency of, 237, 480
glucose as precursor of, 169, 171f
Alpha (α) ketoglutarate. See
excitatory. See also Aspartate; Glutamate
in glycosaminoglycans, 109, 169, 171f
α-Ketoglutarate
glucogenic, 231-232
in glycosphingolipids, 169, 171f
Alpha-lipoproteins, 205. See also High-
in gluconeogenesis, 133-134, 134f
interrelationships in metabolism of,
density lipoproteins
interconvertability of, 231-232
171f
familial deficiency of, 228t
keto acid replacement of in diet, 240
Aminotransferases (transaminases),
Alpha-R groups, amino acid properties
ketogenic, 232
133-134, 134f
affected by, 18
melting point of, 18
diagnostic significance of, 57t
Alpha (α) thalassemia, 47
metabolism of, 122f, 124, 124f, 125f. See
in urea biosynthesis, 243-244, 244f
Alpha-tocopherol. See Tocopherol
also Amino acid carbon skeletons,
Ammonia
Alport syndrome, 538, 538t
catabolism of; Amino acid
in acid-base balance, 245
ALT. See Alanine aminotransferase
nitrogen, catabolism of
detoxification of, 242
Alteplase (tissue plasminogen activator/t-PA),
pyridoxal phosphate in, 491
excess of, 247
604-605, 605f, 606t, 607t
net charge of, 16-17, 17f
glutamine synthase fixing, 245, 245f
Alternative pathway, of complement
nutritionally essential, 124
nitrogen removed as, 244, 244f
activation, 596
nutritionally nonessential, 124
Ammonia intoxication, 244
Altitude, high, adaptation to, 46
synthesis of, 237-241
Ammonium ion, pK/pKa value of, 12t
Alu family, 321-322
in peptides, 14, 19, 19f
Amobarbital, oxidative phosphorylation/
Alzheimer disease, amyloid in, 37, 590
pK/pKa values of, 15-16t, 17, 17f
respiratory chain affected by, 92,
α-Amanitin, RNA polymerases affected by,
environment affecting, 18, 18t
95, 96f
343
products derived from, 264-269. See also
AMP, 287f, 288f, 288t, 297f
Ambiguity, genetic code and, 359
specific product
coenzyme derivatives of, 290t
Amino acid carbon skeletons, catabolism of,
properties of, 14-18
cyclic. See Cyclic AMP
249-263
α-R group determining, 18
free energy of hydrolysis of, 82t
acetyl-CoA formation and, 254f,
protein degradation and, 242, 243f
IMP conversion to, 293, 296f
255-259, 255f, 256f, 257f, 258f,
in proteins, 14
feedback-regulation of, 294, 296f
259f
removal of ammonia from, 244, 244f
PRPP glutamyl amidotransferase
branched-chain, 259, 260f, 261f, 262f
requirements for, 480
regulated by, 294
disorders of, 259-262
sequence of, primary structure
structure of, 83f, 288f
pyruvate formation and, 250-255, 252f,
determined by, 18-19
Amp resistance genes, 402, 403f
253f
solubility point of, 18
Amphibolic pathways/processes, 122
transamination in initiation of, 249-250,
substitutions of, missense mutations
citric acid cycle and, 133
249f, 250f, 251f
caused by, 362-363, 362f
Amphipathic α-helix, 31-32
Amino acid nitrogen
synthesis of, 237-241
Amphipathic lipids, 119-121, 120f
catabolism of, 242-248
in carbohydrate metabolism, 123
in lipoproteins, 205, 207f
in amino acid carbon skeleton
citric acid cycle in, 133, 134f
in membranes, 119, 120f, 417-418, 417f
catabolism, 249, 249f
transamination of. See Transamination
Amphipathic molecules, folding and, 6
end products of, 242-243
transporter/carrier systems for, 99
Ampicillin resistance genes, 402, 403f
urea as, 245-247, 246f
glutathione and, 629-630
Amplification, gene, in gene expression
L-glutamate dehydrogenase in,
hormones affecting, 427
regulation, 392-393, 393f
244-245, 244f, 245f
Aminoacyl residues, 18-19
Amylases
transamination of, 243-244, 243f
peptide structure and, 19
diagnostic significance of, 57t
L-Amino acid oxidase, 86-87
Aminoacyl (A/acceptor) site, aminoacyl-
in hydrolysis of starch, 474
in nitrogen metabolism, 244, 244f
tRNA binding to, in protein
β-Amyloid, in Alzheimer disease, 37, 590
Amino acid sequences. See also Protein
synthesis, 368, 368f
Amyloid-associated protein, 590
sequencing
Aminoacyl-tRNA, in protein synthesis, 368
Amyloid precursor proteins, 590
determination of, for glycoproteins, 515t
Aminoacyl-tRNA synthetases, 360, 360f
in Alzheimer disease, 37, 590
primary structure determined by, 18-19
γ-Aminobutyrate, 267, 268f
Amyloidosis, 590-591
repeating, in mucins, 519, 520f
β-Aminoisobutyrate, 300, 301f
Amylopectin, 107, 108f
Amino acids, 2, 14-20, 15-16t. See also
Aminolevulinate (ALA), 270, 273f
Amylopectinosis, 152t
Peptides
in porphyria, 278
Amylose, 107, 108f
646
/
INDEX
Anabolic pathways/anabolism, 81, 122. See
Anti conformers, 287, 287f
Apo A-IV, 206, 206t
also Endergonic reaction;
Antibiotics
Apo B-48, 206, 206t
Metabolism
amino sugars in, 106
Apo B-100, 206, 206t
Anaerobic glycolysis, 136, 137f, 139
bacterial protein synthesis affected by,
in LDL metabolism, 209, 210f
as muscle ATP source, 574-576, 575f,
371-372
regulation of, 223
575t
folate inhibitors as, 494
Apo C-I, 206, 206t
Analbuminemia, 584
Antibodies, 580, 581. See also
Apo C-II, 206, 206t
Anaphylaxis, slow-reacting substance of,
Immunoglobulins
in lipoprotein lipase activity, 207-208
196
monoclonal, hybridomas in production
Apo C-III, 206, 206t
Anaplerotic reactions, in citric acid cycle,
of, 595-596, 596f
lipoprotein lipase affected by, 208
133
in xenobiotic cell injury, 631, 631f
Apo D, 206, 206t
Anchorin, in cartilage, 551t
Antibody diversity, 591
Apo E, 206, 206t
Andersen’s disease, 152t
DNA/gene rearrangement and, 593-594
receptors for
Androgen response element, 459t
Antichymotrypsin, 583t
in chylomicron remnant uptake,
Androgens
Anticoagulants, coumarin, 604
208-209, 209f
estrogens produced from, 442-445, 444f
Anticodon region, of tRNA, 359, 360, 360f
in LDL metabolism, 209, 210f
receptors for, 471
Antifolate drugs, purine nucleotide
Apoferritin, 586
synthesis of, 440-442, 441f, 443f
synthesis affected by, 293
Apolipoproteins/apoproteins, 205,
Androstenedione
Antigenic determinant (epitope), 33, 591
205-206
estrone produced from, 444f, 445
Antigenicity, xenobiotics altering, cell injury
distribution of, 205-206, 206t
synthesis of, 442, 443f
and, 631, 631f
hemoglobin; oxygenation affecting, 42
Anemias, 47
Antigens, 591
Apomyoglobin, hindered environment for
Fanconi’s, 338
Antihemophilic factor A/globulin (factor
heme iron and, 41, 41f
hemolytic, 136, 143, 609, 619, 620t
VIII), 599f, 600, 600t
Apoproteins. See Apolipoproteins/
glucose-6-phosphate dehydrogenase
deficiency of, 604
apoproteins
deficiency causing, 163,
Antihemophilic factor B (factor IX), 599f,
Apoptosis, 201
169-170, 613, 614f, 619
600, 600t
p53 and, 339
haptoglobin levels in, 584
coumarin drugs affecting, 604
Apo-transketolase, activation of, in thiamin
hyperbilirubinemia/jaundice in, 282,
deficiency of, 604
nutritional status assessment, 489
284, 284t
Antimalarial drugs, folate inhibitors as,
APP. See Amyloid precursor protein
pentose phosphate pathway/
494
Apurinic endonuclease, in base excision-
glutathione peroxidase and, 166,
Antimycin A, respiratory chain affected by,
repair, 337
167f, 169-170
95, 96f
Apyrimidinic endonuclease, in base
primaquine-sensitive, 613
Antioxidants, 91, 119, 611-613, 613t
excision-repair, 337
red cell membrane abnormalities
retinoids and carotenoids as, 119, 482t
Aquaporins, 424-426
causing, 619
vitamin C as, 119
D-Arabinose, 104f, 105t
iron deficiency, 478, 497, 586, 610t
vitamin E as, 91, 119, 486, 487f
Arabinosyl cytosine (cytarabine), 290
megaloblastic
Antiparallel loops, mRNA and tRNA, 360
Arachidonic acid/arachidonate, 113t, 190,
folate deficiency causing, 482t, 492,
Antiparallel β sheet, 32, 33f
190f
494
Antiparallel strands, DNA, 303
eicosanoid formation and, 192, 193f,
vitamin B12 deficiency causing, 482t,
Antiport systems, 426, 426f
194, 194f, 195f
492, 610t
for nucleotide sugars, 517
for essential fatty acid deficiency,
pernicious, 482t, 492
α1-Antiproteinase (α1-antitrypsin), 583t,
191-192
recombinant erythropoietin for, 526, 610
589
ARC, 472t, 473
sickle cell. See Sickle cell disease
deficiency of, 589-590, 589f, 590f,
ARE. See Androgen response element
Angiotensin II, 437, 451, 452f
623
Argentaffinoma (carcinoid), serotonin in,
synthesis of, 451-452, 452f
as thrombin inhibitor, 603
266-267
Angiotensin III, 452, 452f
Antiproteinases, 623, 624t
Arginase
Angiotensin-converting enzyme, 451-452,
Antithrombin/antithrombin III, 583t,
in periodic hyperlysinemia, 258
452f
603-604
in urea synthesis, 246f, 247
Angiotensin-converting enzyme inhibitors,
heparin binding to, 547, 603-604
Arginine, 16t, 265, 266f
451-452
α1-Antitrypsin (α1-antiproteinase), 583t,
catabolism of, 250, 251f
Angiotensinogen, 451, 452f
589
in urea synthesis, 246f, 247
Anion exchange protein, 615, 615f, 616t
deficiency of, 589-590, 589f, 590f,
Arginosuccinase
Ankyrin, 615f, 616f, 616t, 617
623-624
deficiency of, 248
Anomeric carbon atom, 104
as thrombin inhibitor, 603
in urea synthesis, 246f, 247
Anomers, α and β, 104
APC. See Activated protein C
Arginosuccinate, in urea synthesis, 245,
Anserine, 264, 265, 265f
Apo A-I, 206, 206t, 224
246f, 247
Antennae (oligosaccharide branches), 521
deficiencies of, 228t
Arginosuccinate synthase, 246f, 247
Anterior pituitary gland hormones, blood
Apo A-II, 206t
deficiency of, 247
glucose affected by, 161
lipoprotein lipase affected by, 207-208
Arginosuccinicaciduria, 248
INDEX
/
647
Aromatase enzyme complex, 442, 444f, 445
Asymmetry
ATP synthase, membrane-located, 96, 97f,
ARS (autonomously replicating sequences),
importin binding and, 501
98f
326, 413
lipid and protein, membrane assembly
Atractyloside, respiratory chain affected by,
Arsenate, oxidation and phosphorylation
and, 511, 512f
95
affected by, 137, 142
in membranes, 416, 419-420
Attachment proteins, 540, 541f
Arterial wall, intima of, proteoglycans in,
Ataxia-telangiectasia, 338
Autoantibodies, in myasthenia gravis, 431
548
ATCase. See Aspartate transcarbamoylase
Autonomously replicating sequences (ARS),
Arthritis
Atherosclerosis, 205, 607
326, 413
gouty, 299
cholesterol and, 117, 219, 227
Auto-oxidation. See Peroxidation
proteoglycans in, 548
HDL and, 210-211
Autoradiography, definition of, 413
rheumatoid, glycosylation alterations in,
hyperhomocysteinemia and, folic acid
Autotrophic organisms, 82
533
supplements in prevention of,
Avidin, biotin deficiency caused by, 494
Artificial membranes, 421-422
494
Axial ratios, 30
Ascorbate, 167, 168f
LDL receptor deficiency in, 209
Axonemal dyneins, 577
Ascorbic acid (vitamin C), 163, 482t,
lysophosphatidylcholine (lysolecithin)
5- or 6-Azacytidine, 290
495-496, 496f
and, 116
8-Azaguanine, 290, 291f
as antioxidant, 119
Atorvastatin, 229
Azathioprine, 290
in collagen synthesis, 38, 496, 535
ATP, 82, 82-85, 287f, 289
5- or 6-Azauridine, 290, 291f
deficiency of, 482t, 496
in active transport, 427-428, 428f
collagen affected in, 38-39, 496,
in coupling, 82, 84
538-539
fatty acid oxidation producing, 182
B blood group substance, 618, 619f
iron absorption and, 478, 496
free energy of hydrolysis of, 82-83, 82t
B (β) cells, pancreatic, insulin produced by,
supplemental, 496
in free energy transfer from exergonic to
160
Asialoglycoprotein receptors
endergonic processes, 82-83, 82f
B cyclins, 333, 334f, 335t
in cotranslational insertion, 506, 506f
hydrolysis of
B gene, Gal transferase encoded by,
in glycoprotein clearance, 517
in muscle contraction, 561-562, 561f
618-619
Asn-GlcNAc linkage
by NSF, 509, 510f
B lymphocytes, 591
in glycoproteins, 521
inorganic pyrophosphate production
in hybridoma production, 595-596, 596f
in glycosaminoglycans, 543
and, 85
B vitamins. See Vitamin B complex
Asparaginase, in amino acid nitrogen
in mitochondrial protein synthesis and
BAC vector. See Bacterial artificial
catabolism, 245, 245f
import, 499
chromosome (BAC) vector
Asparagine, 15t
in muscle/muscle contraction, 556,
Bacteria
in amino acid nitrogen catabolism, 245
561-562, 561f
intestinal, in bilirubin deconjugation, 281
catabolism of, 249, 250f
decrease in availability of, 564
transcription cycle in, 342-343, 342f
synthesis of, 237-238, 238f
sources of, 573-574, 574-576, 575f,
Bacterial artificial chromosome (BAC)
Asparagine synthetase, 238, 238f
575t
vector, 401-402, 402t
Aspartate
in purine synthesis, 293-294, 295f
for cloning in gene isolation, 635t
catabolism of, 249, 250f
respiratory control in maintenance of
in Human Genome project, 634
synthesis of, 237-238, 238f
supply of, 94-95, 97, 97t, 98f,
Bacterial gyrase, 306
in urea synthesis, 246f, 247
134-135
Bacterial promoters, in transcription,
Aspartate 102, in covalent catalysis, 53-54,
structure of, 83f
345-346, 345f
54f
synthesis of
Bacteriophage, definition of, 413
Aspartate aminotransferase (AST/SGOT),
ATP synthase in, 96, 97f, 98f
Bacteriophage lambda (λ), 378-383, 379f,
diagnostic significance of, 57t
in citric acid cycle, 131f, 133, 142,
380f, 381f, 382f
Aspartate transcarbamoylase, 75
143t
BAL. See Dimercaprol
in pyrimidine synthesis, 298f, 299
glucose oxidation yielding, 142, 143t
BAL 31 nuclease, in recombinant DNA
Aspartic acid, 15t
respiratory chain in, 93-95, 98f
technology, 400t
pI of, 17
ATP/ADP cycle, 83, 84f
Balanced chemical equations, 60
Aspartic protease family, in acid-base
ATPase
BamHI, 398, 399t
catalysis, 52, 53f
in active transport, 427-428, 428f
Barbiturates, respiratory chain affected by,
Aspartylglycosaminuria, 532-533, 533t
chaperones exhibiting activity of, 508
95, 96f
Aspirin
copper-binding P-type, mutations in
Basal lamina, laminin as component of,
antiplatelet actions of, 607-608
gene for
540-542
cyclooxygenase affected by, 193
Menkes diseases caused by, 588
Basal metabolic rate, 478
prostaglandins affected by, 190
Wilson disease caused by, 588-589
Base excision-repair of DNA, 336t, 337,
Assembly particles, in absorptive
ATP-binding cassette transporter-1, 210,
337f
pinocytosis, 430
211f
Base pairing in DNA, 7, 303, 304, 305f
AST. See Aspartate aminotransferase
ATP-chaperone complex, 508. See also
matching of for renaturation, 305-306
Asthma, leukotrienes in, 112
Chaperones
recombinant DNA technology and,
Asymmetric substitution, in porphyrins,
ATP-citrate lyase, 134, 135f, 156t, 157
396-397
270, 271f
acetyl-CoA for lipogenesis and, 177
replication/synthesis and, 328-330, 330f
648
/
INDEX
Base substitution, mutations occurring by,
Bilirubin
Blood cells, 609-625. See also Erythrocytes;
361, 361f, 362
accumulation of (hyperbilirubinemia),
Neutrophils; Platelets
Basement membranes, collagen in, 537
281-284, 284t
Blood clotting. See Coagulation
Bases
conjugated
Blood glucose
conjugate, 10
binding to albumin and, 283
normal, 145
as proton acceptors, 9
reduction of to urobilinogen, 281,
regulation of
strong, 9
282f
clinical aspects of, 161-162, 161f
weak, 9
conjugation of, 280, 280f, 281f
diet/gluconeogenesis/glycogenolysis in,
Bence Jones protein, 595
fecal, in jaundice, 284t
158-161, 159f, 160f
Bends (protein conformation), 32-33,
heme catabolism producing, 278-280,
glucagon in, 160-161
34f
279f
glucokinase in, 159-160, 160f
Beriberi, 482t, 489
liver uptake of, 280-281, 280f, 281f,
glycogen in, 145
Beta-alanine. See β-Alanine
282f
insulin in, 160
Beta anomers, 104
normal values for, 284t
limits of, 158
Beta- (β) carotene, 482, 482t, 483, 483f.
secretion of into bile, 280, 281f
metabolic and hormonal mechanisms
See also Vitamin A
unconjugated, disorders occurring in,
in, 159, 160t, 161
as antioxidant, 119, 482t
282-283
Blood group substances, 618, 619f
Beta-endorphins, 453, 453f
urine, in jaundice, 284, 284t
glycoproteins as, 514, 618
Beta-globin gene
Biliverdin, 278, 279f
Blood group systems, 617-619, 619f
localization of, 407t
Biliverdin reductase, 278
Blood plasma. See Plasma
recombinant DNA technology in
Bimolecular membrane layer, 418-419. See
Blood type, 617-618
detection of variations in,
also Lipid bilayer
Blood vessels, nitric oxide affecting,
407-408, 408f, 409t
Binding constant, Michaelis constant (Km)
571-573, 573f, 574t
Beta-lipoproteins, 205. See also low density
approximating, 66
Blot transfer techniques, 403, 404f
lipoproteins
Binding proteins, 454-455, 454t, 455t, 583t
Blunt end ligation/blunt-ended DNA, 398,
Beta-oxidation of fatty acids, 181-183,
Biochemistry
399-400, 400f, 413
181f, 182f
as basis of health/disease, 2-4, 3t
BMR. See Basal metabolic rate
ketogenesis regulation and, 186-187,
definition of, 1
Body mass index, 478
187f, 188f
Human Genome Project and, 3-4
Body water. See Water
modified, 183, 183f
methods and preparations used in, 1, 2t
Bohr effect, 44, 45f
Beta (β) sheet, 32, 33f
relationship of to medicine, 1-4, 3f
in hemoglobin M, 46
Beta subunits of hemoglobin, myoglobin
Biocytin, 494, 495f
Bone, 549-550, 549f, 550f
and, 42
Bioenergetics, 80-85. See also ATP
metabolic and genetic disorders
Beta (β) thalassemia, 47
Bioinformatics, 412, 638
involving, 551-552, 551t
Beta (β) turn, 32, 34f
protein function and, 28-29
proteins in, 548t, 549
BFU-E. See Burst-forming unit-erythroid
Biologic oxidation. See also Oxidation
Bone Gla protein, 548t
BgIII, 399t
Biomolecules. See also specific type
Bone marrow, heme synthesis in, 272
BHA. See Butylated hydroxyanisole
stabilization of, 7
Bone matrix Gla protein, 488
BHT. See Butylated hydroxytoluene
water affecting structure of, 6-7, 6t
Bone morphogenetic proteins, 548t
Bi-Bi reactions, 69-70, 69f, 70f
Biotechnology, Human Genome Project
Bone sialoprotein, 548t
Michaelis-Menten kinetics and, 70, 70f
affecting, 638
Bone SPARC protein, 548t
Bicarbonate, in extracellular and
Biotin, 482t, 494-495, 495f
Botulinum B toxin, 511
intracellular fluid, 416t
deficiency of, 482t, 494
Bovine spongiform encephalopathy, 37
Biglycan
in malonyl-CoA synthesis, 173, 174f
BPG. See 1,3-Bisphosphoglycerate;
in bone, 548t
as prosthetic group, 50
2,3-Bisphosphoglycerate
in cartilage, 551t
BiP. See Immunoglobulin heavy chain
Bradykinin, in inflammation, 621
Bilayers, lipid, 418-419, 418f, 419f
binding protein
Brain, metabolism in, 235t
membrane proteins and, 419
1,3-Bisphosphoglycerate (BPG), free energy
glucose as necessity for, 232
Bile, bilirubin secretion into, 280, 281f
of hydrolysis of, 82t
Branched chain amino acids, catabolism of,
Bile acids (salts), 225-227
2,3-Bisphosphoglycerate (BPG), T structure
259, 260f, 261f, 262f
enterohepatic circulation of, 227
of hemoglobin stabilized by, 45,
disorders of, 259-262
in lipid digestion and absorption, 475,
45f
Branched-chain α-keto acid dehydrogenase,
476f
Bisphosphoglycerate mutase, in glycolysis in
259
secondary, 226f, 227
erythrocytes, 140, 140f
Branched chain ketonuria (maple syrup
synthesis of, 225-227, 226f
2,3-Bisphosphoglycerate phosphatase, in
urine disease), 259
regulation of, 226, 226f, 227
erythrocytes, 140, 140f
Branching enzymes
Bile pigments, 278-284, 282f. See also
Blindness, vitamin A deficiency causing, 483
absence of, 152t
Bilirubin
Blood
in glycogen biosynthesis, 145, 147f
Biliary obstruction,
coagulation of, 598-608. See also
Brefeldin A, 510-511
hyperbilirubinemia/jaundice
Coagulation; Coagulation factors
Brittle bones (osteogenesis imperfecta), 551t
caused by, 283, 284, 284t
functions of, 580, 581t
Broad beta disease, 228t
INDEX
/
649
Bronze diabetes, 587
in platelet activation, 606f, 607
in urea synthesis, 245, 246-247, 246f,
Brown adipose tissue, 217, 217f
as second messenger, 436-437, 437t,
247
Brush border enzymes, 475
457, 463-465, 463t
Carbamoyl phosphate synthase I
BSE. See Bovine spongiform encephalopathy
phosphatidylinositide metabolism
deficiency of, 247
Buffers
affecting, 464-465, 464f, 465f
in urea synthesis, 245-246, 246f
Henderson-Hasselbalch equation
vitamin D metabolism affected by,
Carbamoyl phosphate synthase II, in pyrim-
describing behavior of, 11, 12f
485-486
idine synthesis, 296, 298f, 299
weak acids and their salts as, 11-12, 12f
Calcium ATPase, 463, 568
Carbohydrates, 102-110. See also specific
“Bulk flow,” of membrane proteins, 507
Calcium-binding proteins, vitamin K and
type and Glucose; Sugars
Burst-forming unit-erythroid, 610, 611f
glutamate carboxylation and
in cell membranes, 110
Butylated hydroxyanisole (BHA), as
postsynthetic modification
cell surface, glycolipids and, 116
antioxidant/food preservative, 119
and, 487-488, 488f
classification of, 102, 102t
Butylated hydroxytoluene (BHT), as
synthesis and, 488, 604
complex (glycoconjugate), glycoproteins
antioxidant/food preservative, 119
Calcium/calmodulin, 463
as, 514
Butyric acid, 112t
Calcium/calmodulin-sensitive phosphorylase
digestion and absorption of, 474-475,
kinase, in glycogenolysis, 148
475f
Calcium channels, 463. See also Calcium
interconvertibility of, 231
C1-9 (complement proteins), 596
release channel
isomerism of, 102-104, 103f
C-peptide, 449, 450f
in cardiac muscle, 566-567
in lipoproteins, 110
C20 polyunsaturated acids, eicosanoids
Calcium-induced calcium release, in cardiac
metabolism of, 122-123, 122f, 123f,
formed from, 190, 192, 193f,
muscle, 567
124-125, 125f
194f
Calcium pump, 463, 568
diseases associated with, 102
C-reactive protein, 583, 583t
Calcium release channel
vitamin B1 (thiamin) in, 488-489,
C regions/segments. See Constant
dihydropyridine receptor and, 563-564,
489f
regions/segments
563f
in proteoglycans, 542, 543, 543f
Ca2+ ATPase, 463
mutations in gene for, malignant
Carbon dioxide
Ca2+-Na+ exchanger, 463, 567-568
hyperthermia caused by,
citric acid cycle in production of,
Cachexia, cancer, 136, 479
564-565, 565f, 630t
130-133, 132f
Caffeine, 289, 289f
Calcium release channel, 563, 564f
transport of, by hemoglobin, 44, 45f
hormonal regulation of lipolysis and, 215
Calcium-sodium exchanger, 463
Carbon monoxide
Calbindin, 477
Caldesmon, 571
heme catabolism producing, 278
Calcidiol (25-hydroxycholecalciferol), in
Calmodulin, 463, 463t, 562
oxidative phosphorylation/respiratory
vitamin D metabolism, 484, 485f
muscle phosphorylase and, 148, 149f
chain affected by, 92, 95, 96f
Calciferol. See Vitamin D
Calmodulin-4Ca2+, in smooth muscle
Carbon skeleton, amino acid. See Amino
Calcineurin, 566t
contraction, 570-571, 571f
acid carbon skeletons
Calcinosis, 486
Calnexin, 508, 526
Carbonic acid, pK/pKa value of, 12t
Calcitonin, 437
Calreticulin, 508, 526
Carbonic anhydrase, in osteopetrosis, 552
Calcitriol (1,25(OH)2-D3), 437, 439f, 485
Calsequestrin, 563, 563f
Carboxybiotin, 494, 495f
calcium concentration regulated by, 485
cAMP. See Cyclic AMP
γ-Carboxyglutamate, vitamin K in synthesis
storage/secretion of, 453, 454t
Cancer/cancer cells. See also
of, 487, 488f
synthesis of, 445, 446f, 484, 485f
Carcinogenesis/carcinogens
Carboxyl terminal repeat domain, 350
Calcium, 496t
cyclins and, 334
Carboxylase enzymes, biotin as coenzyme
absorption of, 477
glycoproteins and, 514, 526, 530t, 531
of, 494-495
vitamin D metabolism and, 477, 484,
hormone-dependent, vitamin B6
Carboxypeptidases, 477
484-485
deficiency and, 491
Carboxyproteinase, procollagen, 537
in blood coagulation, 599f, 600, 600t,
membrane abnormalities and, 432t
Carcinogenesis/carcinogens, 631
601
mucins produced by, 520
chemical, 631
in bone, 549
Cancer cachexia, 136, 479
cytochrome P450 induction and, 628
in extracellular fluid, 416, 416t
Cancer chemotherapy
indirect, 631
in intracellular fluid, 416, 416t
folate inhibitors in, 494
Carcinoid (argentaffinoma), serotonin in,
iron absorption affected by, 478
neutropenia caused by, 610
266-267
in malignant hyperthermia, 564-565,
synthetic nucleotide analogs in,
Carcinoid syndrome, 490
565f
290-291, 291f
Cardiac developmental defects, 570
metabolism of, 463
Cancer phototherapy, porphyrins in,
Cardiac glycosides, 106
vitamin D metabolism and, 484-485
273
Cardiac muscle, 556, 566-570, 568t, 569t
in muscle contraction, 562
CAP. See Catabolite gene activator protein
Cardiolipin, 115, 115f
in cardiac muscle, 566-568
Caproic acid, 112t
synthesis of, 197, 197f, 199, 199f
phosphorylase activation and, 148
Carbamates, hemoglobin, 44
Cardiomyopathies, 556, 569-570, 569t
sarcoplasmic reticulum and, 563-564,
Carbamoyl phosphate
Cargo proteins/molecules, 510
563f, 564f
excess, 301
in export, 503
in smooth muscle, 570, 571
free energy of hydrolysis of, 82t
in import, 501, 502f
650
/
INDEX
Carnitine
factors affecting rates of, 61-63, 62f,
CDK-cyclin inhibitor/CDKI, DNA/-
deficiency of, 180, 187
63-64, 64f
chromosome integrity and, 339
in fatty acid transport, 180-181, 181f
free energy changes and, 60-61
CDKs. See Cyclin-dependent protein
Carnitine-acylcarnitine translocase, 180,
initial velocity and, 64
kinases
181f
multiple substrates and, 69-70, 69f,
cDNA, 413
Carnitine palmitoyltransferase, deficiency
70f
sequencing, in glycoprotein analysis, 515t
of, 180
substrate concentration and, 64, 64f,
cDNA library, 402, 413
Carnitine palmitoyltransferase-I, 180,
65f
CDRs. See Complementarity-determining
181f
models of, 65-67, 66f, 67f
regions
deficiency of, 187
transition states and, 61
Celiac disease, 474
in ketogenesis regulation, 186-187, 187f,
mechanisms of, 51-52, 52f
Cell, 1
188f
prosthetic groups/cofactors/coenzymes
injury to
Carnitine palmitoyltransferase-II, 181, 181f
in, 50-51, 51f
oxygen species causing, 611-613,
deficiency of, 187-188
site-directed mutagenesis in study of,
613t
Carnosinase deficiency, 264
58
xenobiotics causing, 631, 631f
Carnosine, 264, 265, 265f
oxaloacetate and, 130
lysis of, complement in, 596
Carnosinuria, 264
ping-pong, 69-70, 69f
in macromolecule transport, 428-431,
β-Carotene, 482, 482t, 483, 483f. See also
prosthetic groups in, 50-51, 51f
429f, 430f
Vitamin A
by proximity, 51
Cell adhesion
as antioxidant, 119, 482t
regulation of, 72-79, 128f, 129
fibronectin in, 540, 541f
Carotene dioxygenase, 482-483, 483f
active and passive processes in, 72, 73f
glycosphingolipids in, 202
Carotenoids, 482-484, 483f, 484f. See also
allosteric, 74, 74-76, 75f, 128f, 129
integrins in, 620-621, 622t
Vitamin A
compartmentation in, 72-73
selectins in, 528-529, 529t, 530f
Carrier proteins/systems, 426, 426f
covalent, 74, 76, 77-78, 78f
Cell biology, 1
for nucleotide sugars, 517
enzyme quantity and, 73-74
Cell-cell interactions, 415
Cartilage, 543, 551t, 552-553, 553f
feedback inhibition and, 74-76, 75f,
mucins in, 520
chondrodysplasia affecting, 553-554
76, 129
Cell cycle, S phase of, DNA synthesis
Catabolic pathways/catabolism, 81, 122.
feedback regulation and, 76, 129
during, 333-335, 334f, 335t
See also Exergonic reaction;
metabolite flow and, 72, 73f
Cell death, 201
Metabolism
Michaelis constant (Km) in, 72, 73f
Cell-free systems, vesicles studied in, 509
Catabolite gene activator protein (cyclic
phosphorylation-dephosphorylation
Cell fusion, 595
AMP regulatory protein), 376,
in, 78-79, 78t
Cell-mediated immunity, 591
378
proteolysis in, 76-77, 77f
Cell membrane. See Plasma membrane
Catabolite regulatory protein, 460
second messengers in, 76
Cell migration, fibronectin in, 540
Catalase, 88-89
RNA and, 356
Cell recognition, glycosphingolipids in, 202
as antioxidant, 119, 611-613, 613t
sequential (single) displacement, 69, 69f
Cell sap. See Cytosol
in nitrogen metabolism, 244, 244f
specificity of, 49, 50f
Cell surface carbohydrates, glycolipids and,
Catalysis/catalytic reactions (enzymatic). See
by strain, 52
116
also Metabolism
substrate concentration affecting rate of,
Cell surfaces, heparan sulfate on, 545
acid-base, 51-52
64, 64f, 65f
Cellulose, 109
HIV protease in, 52, 53f
Hill model of, 66-67, 67f
Cellulose acetate zone electrophoresis, 580,
at active site, 51, 51f
Michaelis-Menten model of, 65-66,
582f
Bi-Bi, 69-70, 69f, 70f
66f
Central core disease, 565, 569t
Michaelis-Menten kinetics and, 70,
Catalytic residues, conserved, 54, 55t
Central nervous system, glucose as
70f
Catalytic site, 75. See also Active site
metabolic necessity for, 232
coenzymes/cofactors in, 50-51, 51f
Cataracts, diabetic, 172
Centromere, 318, 319f
conservation of residues and, 54, 55t
Catecholamines. See also specific type
Cephalin (phosphatidylethanolamine), 115,
covalent, 52, 52f, 63
receptors for, 436
115f
chymotrypsin in, 52-54, 54f, 63
storage/secretion of, 453, 454t
membrane asymmetry and, 420
fructose-2,6-bisphosphatase in, 54, 55f
synthesis of, 445-447, 447f
synthesis of, 197, 197f
double displacement, 69-70, 69f
Cathepsins, in acid-base catalysis, 52
Ceramide, 116, 116f, 201-202, 202f, 203f
enzyme detection facilitated by, 55-56,
Caveolae, 422
synthesis of, 201-202, 202f
56f
Caveolin-1, 422
Cerebrohepatorenal (Zellweger) syndrome,
equilibrium constant and, 63
CBG. See Corticosteroid-binding globulin
188, 503, 503t
isozymes and, 54-55
CBP/CBP/p300 (CREB-binding protein),
Cerebrosides, 201
kinetics of, 63-70
461, 468, 469, 469f, 471-472,
Ceruloplasmin, 583t, 587, 588
activation energy and, 61, 63
472t
deficiency of, 589
balanced equations and, 60
CD11a-c/CD18, in neutrophils, 621, 621t
diagnostic significance of, 57t, 587
competitive versus noncompetitive
CD18, 620-621
Cervonic acid, 113t
inhibition and, 67-69, 67f,
CD49a/e/f, 622t
CFTR. See Cystic fibrosis transmembrane
68f, 69f
CD59, 531
regulator
INDEX
/
651
CFU-E. See Colony-forming unit-erythroid
in lipoprotein, 205, 207f
reconstitution of, in DNA replication,
Chain elongation. See also Elongation
in membranes, 417
333
by DNA polymerase, 328
fluid mosaic model and, 422
remodeling of in gene expression,
in glycosaminoglycan synthesis, 543
metabolism of, 123-124, 123f
383-384
in transcription cycle, 342, 342f
clinical aspects of, 227-229, 228t
Chromatography. See also specific type
Chain initiation. See also Initiation
diurnal variations in, 220
affinity
in transcription cycle, 342, 342f
high-density lipoproteins in,
for protein/peptide purification, 23
Chain termination. See also Termination
209-211, 211f
for recombinant fusion protein
in glycosaminoglycan synthesis, 543
plasma levels of
purification, 58, 59f
in transcription cycle, 342, 342f
atherosclerosis and coronary heart
for protein/peptide purification, 21-24
Channeling, in citric acid cycle, 130
disease and, 227
Sepharose-lectin column, for
Channelopathies, 568, 569t
dietary changes affecting, 227
glycoprotein analysis, 515t
Chaperones, 36-37, 507-508, 508t
drug therapy affecting, 229
Chromium, 496t
ATPase activity of, 508
lifestyle changes affecting, 227-229
Chromosomal integration, 324, 324f
ATP-dependent protein binding to, 499,
normal, 223
Chromosomal recombination, 323-324,
508
synthesis of, 219-220, 220f, 221f, 222f
323f, 324f
histone, 315
acetyl-CoA in, 123f, 124, 219-220,
Chromosomal transposition, 324-325
in protein sorting, 499, 508t
220f, 221f, 222f
Chromosome jumping, 635t
Chaperonins, 36-37
carbohydrate metabolism and, 123
Chromosome walking, 411, 411f, 635t
Charging, in protein synthesis, 360, 360f
HMG-CoA reductase in regulation of,
Chromosomes, 318-319, 319f, 319t, 320f,
Checkpoint controls, 339
220, 223f
321f
Chédiak-Higashi syndrome, 512t
in tissues, 118, 119f
integrity of, monitoring, 339
Chemical carcinogenesis/carcinogens, 631
factors affecting balance of, 220-223,
interphase, chromatin fibers in, 316
Chemiosmotic theory, 92, 95-97, 97f
224f
metaphase, 317f, 318, 319t
experimental findings in support of, 96
transport of, 223-224, 225f
polytene, 318, 318f
respiratory control and uncouplers and,
reverse, 210, 211f, 219, 224
variations in, 636
97
Cholesteryl ester hydrolase, 223
Chronic granulomatous disease, 623, 623f
Chemotactic factors, 620
Cholesteryl ester transfer protein, 224, 225f
Chyle, 207
Chemotherapy, cancer
Cholesteryl esters, 118, 205, 224
Chylomicron remnants, 206t, 208, 209f
folate inhibitors in, 494
in lipoprotein core, 205, 207f
liver uptake of, 208-209
neutropenia caused by, 610
Cholestyramine resins, for
Chylomicrons, 125, 205, 206t
synthetic nucleotide analogs in,
hypercholesterolemia, 229
apolipoproteins of, 206, 206t
290-291, 291f
Cholic acid, 225
metabolism of, 125, 126f, 207-209,
Chenodeoxycholic acid, 225, 226f
Choline, 114-115, 115f
209f
Chenodeoxycholyl CoA, 226, 226f
deficiency of, fatty liver and, 212
in triacylglycerol transport, 207, 208f,
Chimeric gene approach, 385-386, 387f,
in glycine synthesis, 238, 239f
209f
388f
membrane asymmetry and, 420
Chymotrypsin, 477
Chimeric molecules, 397-406, 413
Cholinesterase. See Acetylcholinesterase
conserved residues and, 55t
restriction enzymes and DNA ligase in
Choluric jaundice, 282
in covalent catalysis, 52-54, 54f
preparation of, 399-400, 401f
Cholyl CoA, in bile acid synthesis, 226,
in digestion, 477
Chips, gene array, protein expression and,
226f
for polypeptide cleavage, 26t
28
Chondrodysplasias, 551t, 553-554, 554f
Chymotrypsinogen, 477
Chitin, 109, 109f
Chondroitin sulfates, 109, 109f, 538,
cI repressor protein/cI repressor gene,
Chloride
543-545, 544f, 544t
379-383, 380f, 381f, 382f
in extracellular and intracellular fluid,
functions of, 547
CICR. See Calcium-induced calcium release
416, 416t
Chondronectin, 551t, 553
Cirrhosis of liver, 130, 212
permeability coefficient of, 419f
Chorionic gonadotropin, human (hCG),
in α1-antitrypsin deficiency, 590
Chlorophyll, 270
438
Cistron, 375-376
Cholecalciferol (vitamin D3)
Christmas factor (factor IX), 599f, 600, 600t
Citrate
synthesis of in skin, 445, 446f, 484, 485f
coumarin drugs affecting, 604
in citric acid cycle, 130, 131f
in vitamin D metabolism, 484, 485f
deficiency of, 604
in lipogenesis regulation, 178
Cholestatic jaundice, 283
Chromatids
Citrate synthase, 130, 132f
Cholesterol, 117, 118, 119f, 205, 219-230
nucleoprotein packing in, 318, 319t,
Citric acid, pK/pKa value of, 12t
in bile acid synthesis, 225-227, 226f
320f
Citric acid cycle (Krebs/tricarboxylic acid
in calcitriol (1,25(OH)2-D3) synthesis,
sister, 318, 319f
cycle), 83, 130-135, 131f, 132f
445, 446f
exchanges between, 325, 325f
ATP generated by, 131f, 133, 142, 143t
dietary, 219
Chromatin, 314-316, 315f, 315t
carbon dioxide liberated by, 130-133,
excess of. See Hypercholesterolemia
active vs. inactive regions of, 316-318,
132f
excretion of, 225-227, 226f
318f
deamination and, 133-134
in hormone synthesis, 438, 438-445,
higher order structure/compaction of,
gluconeogenesis and, 133, 134f,
439t, 440f
316, 317f
153-155, 154f
652
/
INDEX
Citric acid cycle (cont.)
vitamin K in, 486-488, 488f
secretion of, 537
in metabolism, 122, 122f, 123f, 124f,
coumarin anticoagulants affecting, 604
triple helix structure of, 38, 38f,
126, 127f, 130, 133-135, 134f
Coagulation factors, 600t. See also specific
535-539, 536f
amino acid, 122f, 124f
type under Factor
types of, 535, 536t
carbohydrate, 122-123, 122f, 123f,
vitamin K in synthesis of, 486-488, 488f
Collision-induced dissociation, in mass
133-134, 134f
Coat proteins, recruitment of, 509, 510f
spectrometry, 27
lipid/fatty acid, 122f, 123, 123f, 134,
Coated pits, in absorptive pinocytosis, 429f,
Collision (kinetic) theory, 61
135f
430
Colon cancer. See Colorectal cancer
at subcellular level, 126, 127f
Coating, vesicle, 509, 510f
Colony-forming unit-erythroid, 610, 611f
in mitochondria, 126, 127f
brefeldin A affecting, 510-511
Colony hybridization, 403-404. See also
reducing equivalents liberated by,
Cobalamin (vitamin B12), 482t, 491-492,
Hybridization
130-133, 132f
492f
Colorectal cancer, mismatch repair genes in,
regulation of, 134-135
absorption of, 491-492
336
respiratory chain substrates provided by,
intrinsic factor in, 477, 491-492
Coltranslational glycosylation, 504
130, 131f
deficiency of, 482t, 492
Column chromatography, for protein/
transamination and, 133-134, 134f
functional folate deficiency and, 492,
peptide purification, 21, 22f
vitamins in, 133
494
Combinatorial diversity, 592
Citrulline, in urea synthesis, 245, 246-247,
in methylmalonic aciduria, 155
Compartmentation, 72-73
246f, 247
Cobalophilin, 492
Competitive inhibition, noncompetitive
Citrullinemia, 247
Cobalt, 496t
inhibition differentiated from,
CJD. See Creutzfeldt-Jakob disease
in vitamin B12
67-69, 67f, 68f, 69f
Cl. See Chloride
Cobamide, coenzymes derived from, 51
Complement, 583t, 596-597
Class B scavenger receptor B1, 210, 211f
Coding regions, 319, 321f, 637
in inflammation, 596, 621t
Class (isotype) switching, 594
in recombinant DNA technology, 397,
Complementarity
Classic pathway, of complement activation,
398f
of DNA, 306, 307f
596
Coding strand, 304
recombinant DNA technology and,
Clathrin, 429f, 430
in RNA synthesis, 341
396-397
Clathrin-coated vesicles, 510
Codon usage tables, 359-360
of RNA, 306, 309f
Cleavage, in protein sequencing, 25, 26t
Codons, 358, 359t
Complementarity-determining regions,
CLIP, 453, 453f
amino acid sequence of encoded protein
591-592, 594f
Clofibrate, 229
specified by, 358
Complementary DNA (cDNA), 413
Clones
nonsense, 359
Complementary DNA (cDNA) library,
definition of, 413
Coenzyme A, synthesis of from pantothenic
402, 413
library of, 402, 413
acid, 495, 495f
Complex (glycoconjugate) carbohydrates,
in monoclonal antibody production, 596
Coenzyme Q (Q/ubiquinone), 92, 95f
glycoproteins as, 514
Cloning, 400-402, 401f, 402t, 403f
Coenzymes, 50
Complex oligosaccharide chains, 521, 522f
in gene isolation, 635t
in catalysis, 50-51, 51f
formation of, 521, 524
Cloning vectors, 400-402, 401f, 402t,
nucleotide derivatives, 290, 290t
Concanavalin A (ConA), 110, 518t
403f, 414
Cofactors, 50
Conformational diseases, 590
Closed complex, 345
in blood coagulation, 600, 600t, 603
Congenital disorders of glycosylation
Clotting factors, 600t. See also specific type
in catalysis, 50-51, 51f
(CDG), 530t, 531
under Factor
in citric acid cycle regulation, 134-135
Congenital long QT syndrome, 432t
vitamin K in synthesis of, 486-488, 488f
Colipase, 475
Congenital nonhemolytic jaundice (type I
CMP, 288t
Collagen, 37-39, 371, 535-539, 536t
Crigler-Najjar syndrome), 283
CMP-NeuAc, 516t, 517
in bone, 548t, 549
Conjugate acid, 10
CNBr. See Cyanogen bromide
in cartilage, 551t, 552, 553f
Conjugate base, 10
CO. See Carbon monoxide
classification of, 535, 536t
Conjugation
CO2. See Carbon dioxide
elastin differentiated from, 539t
of bilirubin, 280, 280f, 281f
CoA. See Coenzyme A
fibril formation by, 535-539, 536f, 537t
of xenobiotics, 626, 628-630
Coactivators, transcription, 351, 351t
genes for, 535, 536t
Connective tissue, 535
Coagulation (blood), 598-608
diseases caused by mutations in, 39,
bone as, 549-550
endothelial cell products in, 607, 607t
538-539, 538t
keratan sulfate I in, 545
extrinsic pathway of, 598, 599f, 601
chondrodysplasias, 551t, 553
Connexin, 431
fibrin formation in, 598-601, 599f
osteogenesis imperfecta, 551
Consensus sequences, 353, 353f
final common pathway in, 598, 599,
maturation/synthesis of, 38-39
Kozak, 365
601, 602f
ascorbic acid (vitamin C) in, 38, 496
Conservation of energy, 83
intrinsic pathway of, 598, 599f, 600-601
disorders of, 38-39
Conserved residues, 54, 55t
laboratory tests in evaluation of, 608
O-glycosidic linkage in, 518
Constant regions/segments, 593
prostaglandins in, 190
in platelet activation, 605, 606f, 607
gene for, 593
proteins involved in, 599-600, 600t. See
posttranslational modification of,
DNA rearrangement and, 325-326,
also Coagulation factors
537-538, 537t
393, 593-594
INDEX
/
653
immunoglobulin heavy chain, 591, 592f
Corticotropin. See Adrenocorticotropic
binding of to DNA, by helix-turn-helix
immunoglobulin light chain, 325-326,
hormone
motif, 389-390, 389f
393, 591, 592f
Cortisol, 439f, 440f
Cross-bridges, 557, 557-559, 558f, 562f
Constitutive enzymes, 75
binding of, 454, 455, 455t
Cross-links, covalent in collagen, 537
Constitutive gene expression, 376, 378
synthesis of, 440, 441f
Crossing-over, in chromosomal recombina-
Constitutive heterochromatin, 316
Cos sites, 401
tion, 323-324, 323f, 324f
Constitutive mutation, 376
Cosmids, 401, 402t, 413
Crouzon syndrome, 551t
Constitutive secretion, 498
for cloning in gene isolation, 635t
CRP. See C-reactive protein; Catabolite
Contig map, 634f
Cothromboplastin (factor VII), 599f, 600t,
regulatory protein; Cyclic AMP
Contractility/contraction. See Muscle
601
regulatory protein
contraction
coumarin drugs affecting, 604
Cryoprecipitates, recombinant DNA tech-
Cooperative binding
Cotranslational insertion, 504, 505-506
nology in production of, 604
hemoglobin, 42
Cotransport systems, 426, 426f
Cryptoxanthin, 482
Bohr effect and, 44, 45f
Coulomb’s law, 5
Crystallography, x-ray, protein structure
Hill equation describing, 66-67, 67f
Coumarin, 604
demonstrated by, 35
COPI vesicles, 510
Coupling, 81-82, 81f, 82f
CS-PG I/II/III, in bone, 548t
COPII vesicles, 510
ATP in, 82, 84
CT. See Calcitonin
Coplanar atoms, partial double-bond
hormone receptor-effector, 435-436
CTD. See Carboxyl terminal repeat domain
character and, 19, 20f
Coupling domains, on hormone receptors,
CTP, 290
Copper, 496t
435-436
in phosphorylation, 85
ceruloplasmin in binding of, 587, 588
Covalent bonds
CY282Y mutation, in hemochromatosis, 587
as cofactor, 588, 588t
biologic molecules stabilized by, 6, 6t
Cyanide, oxidative phosphorylation/
enzymes containing, 588t
membrane lipid-protein interaction and,
respiratory chain affected by, 92,
in Menkes disease, 588
419
95, 96f
metallothioneins in regulation of, 588
xenobiotic cell injury and, 631, 631f
Cyanogen bromide, for polypeptide cleav-
in oxidases, 86
Covalent catalysis, 52, 52f, 63
age, 25, 26t
tests for disorders of metabolism of, 588,
chymotrypsin in, 52-54, 54f, 63
Cyclic AMP, 147, 148f, 289, 289f, 290t,
589t
fructose-2,6-bisphosphatase in, 54, 55f
458-462, 460t, 462f
in Wilson disease, 587, 588-589
Covalent cross-links, collagen, 537
adenylyl cyclase affecting, 147, 458-459,
Copper-binding P-type ATPase, mutations
Covalent modification
460t
in gene for
mass spectrometry in detection of, 27,
in cardiac muscle regulation, 566
Menkes diseases caused by, 588
27f, 27t
in gluconeogenesis, 158, 158f
Wilson disease caused by, 588-589
in protein maturation, 37-39
in glycogen metabolism regulation,
Copper toxicosis, 588. See also Wilson
in regulation of enzymatic catalysis, 74,
147-150, 148f, 149f, 150f
disease
76, 77-78, 78f. See also
as second messenger, 147, 436, 437t,
Coproporphyrinogen I, 271, 275f
Phosphorylation; Proteolysis
457, 458-462, 460t, 462f
Coproporphyrinogen III, 271, 275f
gluconeogenesis regulation and, 157
smooth muscle contraction affected by,
Coproporphyrinogen oxidase, 271, 275f,
irreversible, 76-77, 77f
571
276f
metabolite flow and, 79
Cyclic AMP-dependent protein kinase. See
in porphyria, 277t
reversible, 77-79, 78f, 78t
Protein kinases
Coproporphyrins, 272f
CPT-I. See Carnitine palmitoyl
Cyclic AMP regulatory protein (catabolite
spectrophotometry in detection of,
transferase-I
gene activator protein), 376, 378
273-274
CRE. See Cyclic AMP response element
Cyclic AMP response element, 459t, 461
Coprostanol (coprosterol), 225
Creatine, 267, 268f
Cyclic AMP response element binding
Core proteins, 542, 543f
Creatine kinase, diagnostic significance of,
protein, 461
in glycosaminoglycan synthesis, 542-543
57t
Cyclic GMP, 289f, 290
Coregulator proteins, 469, 471-473, 472f
Creatine phosphate, 267, 268f
as second messenger, 290, 436, 437t,
Corepressors, 472t, 473
free energy of hydrolysis of, 82t
457, 462-463
Cori cycle, 159, 159f
in muscle, 573-574, 574-576, 575f,
role in smooth muscle, 573f
Cori’s disease, 152t
575t
Cyclic 3′,5′-nucleotide phosphodiesterase,
Cornea, keratan sulfate I in, 545, 546, 547
Creatine phosphate shuttle, 100, 101f
in lipolysis, 215
Coronary (ischemic) heart disease. See also
Creatinine, 267, 268f
Cyclin-dependent protein kinases, 333,
Atherosclerosis
CREB, 461
334f, 335t
cholesterol and, 227
CREB-binding protein, 461, 469, 469f, 471
inhibition of, DNA/chromosome
Corrinoids, 491. See also Cobalamin
Creutzfeldt-Jakob disease, 37
integrity and, 339
Corticosteroid-binding globulin (CBG/
Crigler-Najjar syndrome
Cyclins, 333-335, 334f, 335t
transcortin), 454-455, 455t, 583t
type I (congenital nonhemolytic
Cycloheximide, 372
cyclooxygenases affected by, 193
jaundice), 283
Cyclooxygenase, 192
Corticosterone
type II, 283
as “suicide enzyme,” 194
binding of, 454, 455t
Cro protein/cro gene, 379-383, 380f, 381f,
Cyclooxygenase pathway, 192, 192-194,
synthesis of, 438, 440, 441f
382f
193f, 194f
654
/
INDEX
CYP nomenclature, for cytochrome P450
Cytoskeleton/cytoskeletal proteins, 556,
Delta4 (∆4) (progesterone) pathway, 442,
isoforms, 627
576-578
443f
CYP2A6, polymorphism of, 628, 630t
red cell, 615f, 616-617, 616f, 616t
Delta5 (∆5) (dehydroepiandrosterone)
CYP2C9, in warfarin-phenobarbital
Cytosol
pathway, 442, 443f
interaction, 628
ALA synthesis in, 270, 273f
Delta9 (∆9) desaturase
CYP2D6, polymorphism of, 628, 630t
glycolysis in, 126, 127f, 136
in monounsaturated fatty acid synthesis,
CYP2E1, enzyme induction and, 628
lipogenesis in, 173-177, 174f, 175f
191, 191f
Cysteine, 15t, 265
pentose phosphate pathway reactions in,
in polyunsaturated fatty acid synthesis,
metabolism of, 250, 252f, 253f
163
191, 191f
abnormalities of, 250-255, 253f
pyrimidine synthesis in, 296, 298f
Denaturation
in pyruvate formation, 250, 252f
Cytosolic branch, for protein sorting, 498,
DNA structure analysis and, 304-305
requirements for, 480
499f
protein refolding and, 36
synthesis of, 238-239, 239f
Cytosolic dynein, 577
temperature and, 63
Cystic fibrosis, 431-432, 432t, 474, 569t
Cytosolic proteins, O-glycosidic linkages in,
Deoxoynojirimycin, 527, 527t
Cystic fibrosis transmembrane regulator
518
Deoxyadenylate, 303
(CFTR), 431, 431f, 432t
Cytotoxicity, xenobiotic, 631, 631f
Deoxycholic acid, synthesis of, 226
Cystine reductase, 250, 252f
Deoxycorticosterone
Cystinosis (cystine storage disease),
binding of, 454-455
250-255
D-amino acids, free, 14
synthesis of, 438, 441f
Cystinuria (cystine-lysinuria), 250
D arm, of tRNA, 310, 312f, 360, 361f
11-Deoxycortisol, synthesis of, 440, 441f
Cytarabine (arabinosyl cytosine), 290
D cyclins, 333, 334f, 335t
Deoxycytidine residues, methylation of,
Cytidine, 287f, 288t
cancer and, 334
gene expression affected by, 383
Cytidine monophosphate (CMP), 288t
D isomerism, 102-104, 103f
Deoxycytidylate, 303
Cytidine monophosphate
DAF. See Decay accelerating factor
Deoxyguanylate, 303
N-acetylneuraminic acid
dAMP, 288f
Deoxyhemoglobin, proton binding by, 44,
(CMP-NeuAc), 516t, 517
Dantrolene, for malignant hyperthermia,
45f
Cytidine triphosphate (CTP), 290
564
Deoxyhemoglobin A, “sticky patch” recep-
in phosphorylation, 85
DBD. See DNA-binding domain
tor on, 46
Cytochrome b5, 89, 627
DBH. See Dopamine-β-hydroxylase
Deoxyhemoglobin S, “sticky patch”
Cytochrome b558, 622
Deamination, 124, 124f
receptor on, 46
Cytochrome c, 93
citric acid cycle in, 133-134
Deoxynucleotides, 303-304, 304f, 305f
Cytochrome oxidase/cytochrome aa3, 86,
liver in, 125
Deoxyribonucleases (DNase)/DNase I, 312
93
Debranching enzymes
active chromatin and, 316
Cytochrome P450-dependent microsomal
absence of, 152t
Deoxyribonucleic acid. See DNA
ethanol oxidizing system,
in glycogenolysis, 146-147, 148f
Deoxyribonucleoside diphosphates
212-214
Debrisoquin, CYP2D6 in metabolism of,
(dNDPs), reduction of NDPs to,
Cytochrome P450 side chain cleavage en-
628
294, 297f
zyme (P450scc), 438, 440f, 442
Decay accelerating factor, 531
Deoxyribonucleosides, 286
Cytochrome P450 system, 86, 89-90, 90f,
Decorin
in pyrimidine synthesis, 296
626
in bone, 548t
Deoxyribose, 102, 106, 106f
ALA synthase affected by, 272, 278
in cartilage, 551t
Deoxy sugars, 106, 106f
enzyme induction and, 272-273,
Defensins, 621t
3-Deoxyuridine, 290
627-628
Degeneracy, of genetic code, 359
Dephosphorylation. See also
genes encoding, nomenclature for, 627
Degradation, rate of (kdeg), 74
Phosphorylation
isoforms of, 627-628
Dehydrocholesterol, in vitamin D
in covalent modification, 78-79, 78t
in metabolism of xenobiotics, 626-628,
metabolism, 484, 485f
Depolarization, in nerve impulse
629t
Dehydroepiandrosterone (DHEA),
transmission, 428
membrane insertion, 504
synthesis of, 440, 441f
Depurination, DNA, base excision-repair
mitochondrial, 89-90
Dehydroepiandrosterone (∆5) pathway, 442,
and, 337
nomenclature system for, 627
443f
Dermatan sulfate, 544f, 544t, 545
in xenobiotic cell injury, 631, 631f
Dehydrogenases, 86, 87-88, 88f
functions of, 547
Cytochromes, as dehydrogenases, 88
in enzyme detection, 56, 56f
∆9 Desaturase
Cytogenetic abnormalities, detection of,
nicotinamide coenzyme-dependent, 87,
in monounsaturated fatty acid synthesis,
635t
89f
191, 191f
Cytogenetic map, 633, 634f
in respiratory chain, 87
in polyunsaturated fatty acid synthesis,
Cytokines, α2-macroglobulin binding of,
riboflavin-dependent, 87
191, 191f
590
Deletions, DNA, recombinant DNA tech-
Desmin, 566t, 577t
Cytosine, 288t
nology in detection of, 409,
Desmosines, 539
base pairing of in DNA, 303, 304, 305f
409t
Desmosterol, in cholesterol synthesis, 220,
deoxyribonucleosides of, in pyrimidine
Delta5,4 (∆5,4) isomerase, 438, 441f, 442,
222f
synthesis, 296-297, 298f
443f
Detergents, 417-418
INDEX
/
655
Detoxification, 626
Dihydrobiopterin, defect in synthesis of, 255
Diversity
cytochrome P450 system in, 89-90, 90f,
Dihydrobiopterin reductase, defect in, 255
antibody, 592, 593-594
626-628, 629t
Dihydrofolate/dihydrofolate reductase,
combinatorial, 592
Dextrinosis, limit, 152t
methotrexate affecting, 296-297,
in gene expression, 387, 388f
Dextrins, 109
494
junctional, 593-594
Dextrose, 104
Dihydrolipoyl dehydrogenase, 140, 141f
Diversity segment, DNA rearrangement
DHA. See Docosahexaenoic acid
Dihydrolipoyl transacetylase, 140, 141f
and, 593-594
DHEA. See Dehydroepiandrosterone
Dihydropyridine receptor, 563-564, 563f,
DNA, 303, 303-306, 314 -340
DHPR. See Dihydropyridine receptor
564f
base excision-repair of, 336t, 337, 337f
DHT. See Dihydrotestosterone
Dihydrotestosterone, 442, 444f
base pairing in, 7, 303, 304, 305f
Diabetes mellitus, 102, 161-162
binding of, 455t
matching of for renaturation, 305-306
fatty liver and, 212
Dihydroxyacetone, 106f
recombinant DNA technology and,
free fatty acid levels in, 206
Dihydroxyacetone phosphate, in glycolysis,
396-397
hemochromatosis and, 587
197, 197f, 198f
replication/synthesis and, 328-330,
insulin resistance and, 611
1,25-Dihydroxyvitamin D3. See Calcitriol
330f
ketosis/ketoacidosis in, 188
24,25-Dihydroxyvitamin D3 (24-hydroxy-
binding of to regulatory proteins, motifs
lipid transport and storage disorders and,
calcidiol), in vitamin D
for, 387-390, 388t, 389f, 390f,
205
metabolism, 484, 485f
391f
lipogenesis in, 173
Diiodotyrosine (DIT), 447, 448f, 449
blunt-ended, 398, 399-400, 400f, 413
as metabolic disease, 122, 231
Dilated cardiomyopathy, 570
in chromatin, 314 -318, 315f, 315t,
starvation and, 236
Dimercaprol (BAL), respiratory chain
317f, 318f
Diabetic cataract, 172
affected by, 95, 96f
chromosomal, 318-319, 319f, 319t,
Diacylglycerol, 115, 475, 476f
Dimeric proteins, 34
320f, 321f
in calcium-dependent signal
Dimers
relationship of to mRNA, 321f
transduction, 464, 465f
Cro protein, 380, 381f
coding regions of, 319, 321f, 637
formation of, 197f, 198f
histone, 315
complementarity of, 306, 307f
in platelet activation, 606, 606f
lambda repressor (cI) protein, 380, 381f
recombinant DNA technology and,
in respiratory burst, 623
Dimethylallyl diphosphate, in cholesterol
396-397
Diacylglycerol acyltransferase, 198f, 199
synthesis, 219, 221f
damage to, 335, 335t
Diagnostic enzymology, 57, 57t
Dimethylaminoadenine, 289f
repair of, 335-339, 335t, 336t
Dicarboxylate anions, transporter systems
Dinitrophenol, respiratory chain affected
ADP-ribosylation for, 490
for, 98-99
by, 95, 96f
deletions in, recombinant DNA technol-
Dicarboxylic aciduria, 188
Dinucleotide, 291
ogy in detection of, 409, 409t
Dicumarol (4-hydroxydicoumarin), 486
Dioxygenases, 89
depurination of, base excision-repair and,
Dielectric constant, of water, 5
Dipalmitoyl lecithin, 115
337
Diet. See also Nutrition
Dipeptidases, 477
double-strand break repair of, 336t,
blood glucose regulation and, 159-161
Diphosphates, nucleoside, 287, 287f
337-338, 338f
cholesterol levels affected by, 227
Diphosphatidylglycerol. See Cardiolipin
double-stranded, 304
hepatic VLDL secretion and, 211-212,
Diphtheria toxin, 372
flanking sequence, 397
213f
Dipoles, water forming, 5, 6f
genetic information contained in,
high-fat, fatty liver and, 212
Disaccharidases, 102, 475
303 -306
Diet-induced thermogenesis, 217, 478
Disaccharides, 106-107, 107f, 107t. See
grooves in, 305f, 306
Diethylenetriaminepentaacetate (DTPA),
also specific type
insertions in, recombinant DNA
as preventive antioxidant, 119
Disease
technology in detection of, 409
Diffusion
biochemical basis of, 2, 3t
integrity of, monitoring, 339
facilitated, 423, 423t, 424f, 426 - 427,
Human Genome Project and, 3- 4
“jumping,” 325
427, 427f
major causes of, 3t
mismatch repair of, 36f, 336, 336f,
of bilirubin, 280
Displacement reactions
336t
of glucose. See also Glucose
double, 69-70, 69f
mitochondrial, 322-323, 322f, 323t
transporters
sequential (single), 69, 69f
mutations in, 314, 323 -326, 323f, 324f,
insulin affecting, 427
Dissociation, of water, 8-9
325f. See also Mutations
in red cell membrane, 611
Dissociation constant, 8-9
in nucleosomes, 315-316, 316f
hormones in regulation of, 427
Michaelis constant (Km) and, 66
nucleotide excision-repair of, 336, 337,
“Ping-Pong” model of, 427, 427f
in pH calculation, 10
338f
net, 423, 424f
of weak acids, 10 -11, 12
rearrangements of
passive, 423, 423t, 424f
Distal histidine (histidine E7), in oxygen
in antibody diversity, 325-326, 393,
simple, 423, 423t, 424f
binding, 40, 41f
593-594
Digestion, 474-480
Disulfide bonds, protein folding and, 37
recombinant DNA technology in
Digitalis
DIT. See Diiodotyrosine
detection of, 409, 409t
Ca2+-Na+ exchanger in action of, 568
Diurnal rhythm, in cholesterol synthesis,
recombinant. See Recombinant DNA/
Na+-K+ ATPase affected by, 428, 568
220
recombinant DNA technology
656
/
INDEX
DNA (cont.)
DNA-PK. See DNA-dependent protein
Dopamine, 446, 447f. See also Cate-
relaxed form of, 306
kinase
cholamines
renaturation of, base pair matching and,
DNA polymerases, 326, 327-328, 327f,
synthesis of, 267, 267f, 445-447, 447f
305-306
328, 328t
Dopamine-β-hydroxylase, 447
repair of, 335-339, 335t, 336t
in recombinant DNA technology, 400t
vitamin C as coenzyme for, 495-496
repetitive-sequence, 320-322
DNA primase, 327, 327f, 328t
Dopamine β-oxidase, 267, 267f
replication/synthesis of, 306, 307f,
DNA probes, 402, 414
Double displacement reactions, 69-70, 69f
326-339, 326t, 327f, 328t
for gene isolation, 635t
Double helix, of DNA structure, 7, 303,
DNA polymerase complex in, 328,
library searched with, 402
304, 305f
328t
in porphyria diagnosis, 274
recombinant DNA technology and, 396,
DNA primer in, 328, 329f, 330f
DNA-protein interactions, bacteriophage
397
initiation of, 328-330, 329f, 330f,
lambda as paradigm for,
Double reciprocal plot
331f
378-383, 379f, 380f, 381f, 382f
inhibitor evaluation and, 68, 68f, 69f
origin of, 326
DNA sequences
Km and Vmax estimated from, 66, 66f
polarity of, 330-331
amplification of by PCR, 405-406,
Double-strand break repair of DNA, 336t,
proteins involved in, 328t
406f
337-338, 338f
reconstitution of chromatin structure
determination of, 404, 405f
Double-stranded DNA, 304, 314
and, 333
in gene isolation, 635t
unwinding
repair during, 335-339, 335t, 336t
protein sequencing and, 25-26
for replication, 326, 326-327
replication bubble formation and,
DNA topoisomerases, 306, 328t, 332,
RNA synthesis and, 344
331-333, 331f, 332f, 333f
332f
Downstream promoter element, 346-348,
replication fork formation and,
DNA transfection, identification of
347f
327-328, 327f
enhancers/regulatory elements
DPE. See Downstream promoter element
ribonucleoside diphosphate reduction
and, 386
DRIPs, 472t, 473
and, 294, 297f
DNA unwinding element, 326
Drug detoxification/interactions,
in S phase of cell cycle, 333-335,
DNase (deoxyribonuclease)/DNase I, 312
cytochromes P450 and,
334f, 335t
active chromatin and, 316
89-90, 90f, 628
semiconservative nature of, 306, 307f
in recombinant DNA technology, 400t
Drug development, pharmacogenetics and,
semidiscontinuous, 327f, 331, 331f
dNDPs. See Deoxyribonucleoside
631-632
unwinding and, 326, 326-327
diphosphates
Drug resistance, gene amplification in, 393
in RNA synthesis, 341-343, 342f, 343t
DOC. See 11-Deoxycorticosterone
DS-PG I/DS-PG II, in cartilage, 551t
stabilization of, 7
Docking, in nuclear import, 501, 502f
dsDNA. See Double-stranded DNA
structure of, 303-306, 304f, 305f
Docking protein, 504
DTPA, as preventive antioxidant, 119
denaturation in analysis of, 304-305
Docosahexaenoic acid, 191-192
Dubin-Johnson syndrome, 283
double-helical, 7, 303, 304, 305f
Dolichol, 118, 119f, 522, 523f
Duchenne muscular dystrophy, 556,
recombinant DNA technology and,
in cholesterol synthesis, 220, 221f
565-566, 566f
396, 397
in N-glycosylation, 522
DUE. See DNA unwinding element
supercoiled, 306, 332, 333f
Dolichol kinase, 522
Dwarfism, 551t, 553-554
transcription of, 306
Dolichol-P-P-GlcNAc, 522-523
Dynamin, in absorptive pinocytosis, 430,
transposition of, 325
Dolichol-P-P-oligosaccharide (dolichol-
577
unique-sequence (nonrepetitive), 320,
pyrophosphate-oligosaccharide),
Dyneins, 577
320-321
521, 524f
Dysbetalipoproteinemia, familial, 228t
unwinding of, 326, 326-327
in N-glycosylation, 521-524, 523f
Dyslipoproteinemias, 228t, 229
RNA synthesis and, 344
Dolichol phosphate, 522
Dystrophin, 556, 565-566, 566t, 567f
xenobiotic cell injury and, 631
Domains. See also specific type
mutation in gene for, in muscular
DNA binding domains, 390-391, 392f, 470
albumin, 584
dystrophy, 565-566, 566f
DNA binding motifs, 387-390, 388t, 389f,
carboxyl terminal repeat, 350
390f, 391f
chromatin, 316, 318, 319f
DNA-dependent protein kinase, in
coupling, on hormone receptors, 435-436
E0. See Redox (oxidation-reduction)
double-strand break repair, 338
DNA binding, 390-391, 392f, 470
potential
DNA-dependent RNA polymerases,
fibronectin, 540, 541f
Eact. See Activation energy
342-343, 342f, 343t
protein, 33-34
E coli, lactose metabolism in, operon
DNA elements, gene expression affected by,
Src homology 2 (SH2)
hypothesis and, 376-378,
384-385, 384f, 385t, 386f
in insulin signal transmission, 465,
376f, 377f
diversity and, 387, 388f
466f, 467
E coli bacteriophage P1-based (PAC) vector,
DNA fingerprinting, 413
in Jak/STAT pathway, 467, 467f
401-402, 402t, 413
DNA footprinting, 413
trans-activation, of regulatory proteins,
E cyclins, 333, 334f, 335t
DNA helicase, 326-327, 327f, 328, 328t
390-391, 392f
E-selectin, 529t
DNA ligase, 328t, 330
transcription, 387
E (exit) site, in protein synthesis, 38f, 368
in recombinant DNA technology,
L-Dopa, 446, 447f
ECF. See Extracellular fluid
399-400, 400t, 401f
Dopa decarboxylase, 267, 267f, 446, 447f
ECM. See Extracellular matrix
INDEX
/
657
EcoRI, 398, 399t, 401f
in glycosaminoglycan synthesis, 543
neutrophil interaction and
EcoRII, 399t
in protein synthesis, 367-370, 368f
integrins in, 529t, 620-621, 622t
Edema
in RNA synthesis, 342, 342f, 344
selectins in, 528-529, 529t, 530f
in kwashiorkor, 479
Elongation arrest, 504
Endothelium-derived relaxing factor, 572,
plasma protein concentration and, 580
Elongation factor 2, in protein synthesis,
607t. See also Nitric oxide
in thiamin deficiency, 489
368, 368f
Energy
Edman reaction, for peptide/protein
Elongation factor EF1A, in protein
activation, 61, 63
sequencing, 25, 26f
synthesis, 368, 368f
conservation of, 83
Edman reagent (phenylisothiocyanate), in
Elongation factors, in protein synthesis,
free. See Free energy
protein sequencing, 25, 26f
367, 368, 368f
in muscle, creatine phosphate as reserve
EDRF. See Endothelium-derived relaxing
Emelin, 539
for, 573-574, 575f
factor
Emphysema, α1-antiproteinase deficiency
nutritional requirement for, 478
EDTA, as preventive antioxidant, 119
and, 589, 589f, 623-624
transduction of
EFA. See Essential fatty acids
Emulsions, amphipathic lipids forming,
membranes in, 415
EFs. See Elongation factors
120f, 121
in muscle, 556-559
EGF. See Epidermal growth factor
Encephalopathies
Energy balance, 478-479
Egg white, uncooked, biotin deficiency
hyperbilirubinemia causing (kernicterus),
Energy capture, 82-83, 82f, 83
caused by, 494
282, 283
Energy expenditure, 478
Ehlers-Danlos syndrome, 538, 538t
mitochondrial, with lactic acidosis and
Energy transfer, 82-83, 82f
Eicosanoids, 112, 190, 192, 193f, 194f,
stroke (MELAS), 100-101
Enhanceosome, 385, 386f
621t
spongiform (prion diseases), 37
Enhancers/enhancer elements, 348
Eicosapentaenoic acid, 190f
Wernicke’s, 489
in gene expression, 384-385, 384f, 385t
eIF-4E complex, in protein synthesis, 367,
Endergonic reaction, 80, 81
tissue-specific expression and, 385
367f
coupling and, 81-82, 81f, 82f
recombinant DNA technology and, 397
eIFs, in protein synthesis, 365
ATP in, 82, 84
reporter genes in definition of, 385-386,
80S initiation complex, in protein synthesis,
Endocrine system, 434 - 455. See also
387f, 388f
366f, 367
Hormones
Enolase, in glycolysis, 137, 138f
Elaidic acid, 112, 113, 113t, 114f
diversity of, 437- 438
∆2Enoyl-CoA hydratase, 181, 182f
Elastase, in digestion, 477
Endocytosis, 428, 429-430, 429f
∆3cis-Enoyl-CoA isomerase, 183
Elastin, 539, 539t
receptor-mediated, 429f
∆2trans-Enoyl-CoA isomerase, 183
Electrochemical potential difference, in
Endoglycosidase F, 517
Entactin, in basal lamina, 540
oxidative phosphorylation, 96
Endoglycosidase H, 517
Enterohepatic circulation, 227
Electron carriers, flavin coenzymes as, 490
Endoglycosidases, in glycoprotein analysis,
lipid absorption and, 475
Electron movement, in active transport, 427
515t, 517, 517t
Enterohepatic urobilinogen cycle, 281
Electron-transferring flavoprotein, 87, 181
Endonucleases, 312, 413
Enteropeptidase, 477
Electron transport chain system, 622. See
apurinic and apyrimidinic, in base
Enthalpy, 80
also Respiratory chain
excision-repair, 337
Entropy, 80
Electrophiles, 7
restriction, 312, 397-399, 399t, 400f,
Enzyme induction, 630
Electrophoresis
414
cytochrome P450 and, 272-273,
for plasma protein analysis, 580
in recombinant DNA technology,
627-628
polyacrylamide, for protein/peptide
399- 400, 399t, 400f, 400t, 401f
in gluconeogenesis regulation, 155-157,
purification, 24, 24f, 25f
Endopeptidases, 477
156t
pulsed-field gel, for gene isolation, 635t
Endoplasmic reticulum, 370
Enzyme-linked immunoassays (ELISAs), 55
two-dimensional, protein expression and,
acylglycerol synthesis and, 126, 127f
Enzymes, 7-8
28
core protein synthesis in, 543
active sites of, 51, 51f
Electrospray dispersion, in mass
fatty acid chain elongation in, 177, 177f
assay of, 55-56, 56f
spectrometry, 27
rough
branching, in glycogen biosynthesis, 145,
Electrostatic bonds/interactions, 7. See also
glycosylation in, 524-525, 525f
147f
Salt (electrostatic) bonds
in protein sorting, 498, 499f, 500f
catalytic activity of, 49, 50f. See also
oxygen binding rupturing, Bohr effect
protein synthesis and, 370
Catalysis
protons and, 44-45, 45f
routes of protein insertion into,
detection facilitated by, 55-56, 56f
ELISAs. See Enzyme-linked immunoassays
505-507, 506f
kinetics of, 63-70. See also Kinetics
Elliptocytosis, hereditary, 617
signal hypothesis of polyribosome
(enzyme)
Elongase, 177, 177f
binding to, 503-505, 504t, 505f
regulation of, 72-79, 128f, 129
in polyunsaturated fatty acid synthesis,
smooth, cytochrome P450 isoforms in,
RNA and, 356
191, 191f, 192f
627
specificity of, 49, 50f
Elongation
Endoproteinase, for polypeptide cleavage,
classification of, 49-50
chain
26t
constitutive, 75
in fatty acid synthesis, 177, 177f
Endorphins, 453, 453f
debranching
in transcription cycle, 342, 342f
Endothelial cells
absence of, 152t
in DNA synthesis, 328
in clotting and thrombosis, 607, 607t
in glycogenolysis, 146-147, 148f
658
/
INDEX
Enzymes (cont.)
Erythrocytes, 609-610, 610-619
iron absorption and, 478
degradation of, control of, 74
2,3-bisphosphoglycerate pathway in,
thiamin deficiency and, 489
in disease diagnosis/prognosis, 56 -57,
140, 140f
Ethylenediaminetetraacetate (EDTA), as
57, 57t, 58f, 580
disorders affecting, 609, 610t
preventive antioxidant, 119
in DNA repair, 335, 336t, 338-339
erythropoietin in regulation of, 609-610,
Euchromatin, 316
hydrolysis rate affected by, 7-8
611f
Eukaryotic gene expression, 383-387,
irreversible inhibition (“poisoning”) of,
glucose-6-phosphate dehydrogenase defi-
391-395, 392t. See also Gene
69
ciency affecting, 613, 614f, 619
expression
isozymes and, 54 -55
glucose as metabolic necessity for, 232
chromatic remodeling in, 383-384
kinetics of, 60-71. See also Kinetics
glycolysis in, 140, 140f
diversity of, 387, 388f
(enzyme)
hemoglobin S “sticky patch” affecting, 46
DNA elements affecting, 384-385, 384f,
mechanisms of action of, 49-59
hemolysis of, pentose phosphate
385t, 386f
membranes in localization of, 415
pathway/glutathione peroxidase
DNA-protein interactions in, bacterio-
metal-activated, 50
and, 166, 167f, 169-170
phage lambda as paradigm for,
as mitochondrial compartment markers,
life span of, 609
378-383, 379f, 380f, 381f, 382f
92
membranes of, 614-617, 615f, 615t,
locus control regions and insulators in,
plasma, diagnostic significance of, 57, 57t
616f, 616t
387
quantity of, catalytic capacity affected by,
glucose transporter of, 611, 612t
prokaryotic gene expression compared
73-74
hemolytic anemias and, 619, 620t
with, 391-395, 392t
rate of degradation of (kdeg), control of,
metabolism of, 235t, 610-614, 612t
reporter genes and, 385-386, 387f, 388f
74
oxidants produced during, 611-613,
tissue-specific, 385
rate of synthesis of (ks), control of, 74
613t
Eukaryotic promoters, in transcription,
recombinant DNA technology in study
recombinant DNA technology in study
346-349, 347f, 348f, 349f
of, 58, 59f
of, 624
Eukaryotic transcription complex,
regulatory, 126-129, 128f
structure and function of, 609-610
350-352, 351t
restriction. See Restriction endonucleases
Erythroid ALA synthase (ALAS2), 272, 273
Exchange transporters, 98-100, 98f, 99f
specificity of, 49, 50f
in porphyria, 274, 277t
Excitation-response coupling, membranes
substrates affecting conformation of, 52,
Erythropoiesis, 609-610, 611f
in, 415
53f
Erythropoietin/recombinant erythropoietin
Exergonic reaction, 80, 81
Enzymopathies, 619
(epoitin alfa/EPO), 526, 583t,
coupling and, 81-82, 81f, 82f
Epidermal growth factor (EGF), receptor
609-610, 611f
ATP in, 82, 84
for, 436
D-Erythrose, 104f
Exinuclease, in DNA repair, 337, 337f
Epidermolysis bullosa, 538, 538t
Escherichia coli, lactose metabolism in,
Exit (E) site, in protein synthesis, 368,
Epimerases
operon hypothesis and, 376-378,
368f
in galactose metabolism, 167, 170f
376f, 377f
Exocytosis, 429, 430-431, 430f
in glycosaminoglycan synthesis, 543
Escherichia coli bacteriophage P1-based
in insulin synthesis, 430-431
in pentose phosphate pathway, 163, 165f
(PAC) vector, 401-402, 402t,
Exocytotic (secretory) pathway, 498
Epimers, 104, 104f
413
Exoglycosidases, in glycoprotein analysis,
Epinephrine, 439f, 447, 447f. See also
Essential amino acids. See Nutritionally
515t, 517, 517t
Catecholamines
essential amino acids
Exons, 319, 358, 413
blood glucose affected by, 161
Essential fatty acids, 190, 190f, 193
interruptions in. See Introns
in gluconeogenesis regulation, 157
abnormal metabolism of, 195-196
in recombinant DNA technology, 397,
in lipogenesis regulation, 178
deficiency of, 191-192, 194-195
398f
synthesis of, 267, 267f, 445-447, 447f
prostaglandin production and, 190
splicing, 352-354, 414
Epitope (antigenic determinant), 33, 591
Essential fructosuria, 163, 171-172
alternative, in regulation of gene
Epoxide hydrolase, 631
Essential pentosuria, 163, 170
expression, 354, 354f,
Epoxides, 631
Estradiol/17β-Estradiol, 439f, 440f
393-394, 636
Equilibrium constant (Keq), 62-63
binding of, 455, 455t
recombinant DNA technology and,
in enzymatic catalysis, 63
synthesis of, 442-445, 444f
397, 398f
free energy changes and, 60-61
Estriol, synthesis of, 442, 444f
Exonucleases, 312, 413
ER. See Estrogens, receptors for
Estrogen response element, 459t
in recombinant DNA technology, 400t
Ercalcitriol, 484
Estrogens
Exopeptidases, 477
ERE. See Estrogen response element
binding of, 455, 455t
Exportins, 503
eRF. See Releasing factors
receptors for, 471
Expression vector, 402
Ergocalciferol (vitamin D2), 484
synthesis of, 442-445, 444f, 445f
Extra arm, of tRNA, 310, 312f
Ergosterol, 118, 119f
Estrone
Extracellular environment, membranes in
Erythrocyte aminotransferases, in vitamin
binding of, 455t
maintenance of, 415-416, 416t
B6 status assessment, 491
synthesis of, 442, 444f
Extracellular fluid (ECF), 415-416, 416,
Erythrocyte transketolase activation, in
Ethanol
416t
thiamin nutritional status
CYP2E1 induction and, 628
Extracellular matrix, 535-555. See also spe-
assessment, 489
fatty liver and, 212-214
cific component and under Matrix
INDEX
/
659
Extrinsic pathway of blood coagulation,
Farber’s disease, 203t
trans, 113-114, 192
598, 599f, 601
Farnesoid X receptor, in bile acid synthesis
transport of, carnitine in, 180-181, 181f
Eye, fructose and sorbitol in, diabetic
regulation, 227
triacylglycerols (triglycerides) as storage
cataract and, 172
Farnesyl diphosphate, in cholesterol/
form of, 114, 115f
polyisoprenoid synthesis,
unesterified (free). See Free fatty acids
219, 220, 221f
unsaturated. See Unsaturated fatty acids
F0, in ATP synthesis, 96, 97f, 98f
Fast acetylators, 630
Fatty liver
F1, in ATP synthesis, 96, 97f, 98f
Fast (white) twitch fibers, 574-576, 575t
alcoholism and, 212-214
Fab region, 591, 592f
Fat tissue. See Adipose tissue
of pregnancy, 188
Fabry’s disease, 203t
Fatal infantile mitochondrial myopathy and
triacylglycerol metabolism imbalance
Facilitated diffusion/transport system, 423,
renal dysfunction, oxidoreductase
and, 212
423t, 424f, 426-427, 427, 427f
deficiency causing, 100
Favism, 170
for bilirubin, 280
Fatigue (muscle), 136
Fc fragment, 591, 592f
for glucose. See also Glucose transporters
Fats, 111. See also Lipids
receptors for, in neutrophils, 621t
insulin affecting, 427
diets high in, fatty liver and, 212
Fe. See Iron
in red cell membrane, 611
metabolism of, 122f, 123-124, 123f,
Fed state, metabolic fuel reserves and, 232,
hormones in regulation of, 427
125-126, 126f
234t
“Ping-Pong” model of, 427, 427f
Fatty acid-binding protein, 180, 207
Feedback inhibition, in allosteric regulation,
Factor I (fibrinogen), 580, 600, 600t
Fatty acid chains, elongation of, 177, 177f
74-76, 75f, 76, 129
conversion of to fibrin, 601-602
Fatty acid elongase system, 177, 177f
Feedback regulation
Factor II (prothrombin), 600t, 601
in polyunsaturated fatty acid synthesis,
in allosteric regulation, 76, 129
coumarin drugs affecting, 487, 604
191, 191f, 192f
thrombin levels controlled by, 602
vitamin K in synthesis of, 487
Fatty acid oxidase, 181, 182f
Fenton reaction, 612
Factor III (tissue factor), 599f, 600t, 601
Fatty acid synthase, 156t, 173
Ferric iron, 278
Factor IV. See Calcium
Fatty acid synthase complex, 173-176,
in methemoglobinemia, 46
Factor V (proaccelerin/labile factor/accelera-
174f, 175f, 179
Ferrireductase, 585
tor globulin), 600t, 601, 602f
Fatty acid-transport protein, membrane,
Ferritin, 478, 585, 586
Factor V Leiden, 603-604
207
protein synthesis affected by, 370
Factor VII (proconvertin/serum
Fatty acids, 2, 111-114
Ferritin receptor, 586
prothrombin conversion accelera-
activation of, 180-181, 181f
Ferrochelatase (heme synthase), 271, 272f
tor/cothromboplastin), 599f,
calcium absorption affected by, 477
in porphyria, 277t
600t, 601
eicosanoids formed from, 190, 192,
Ferrous iron
coumarin drugs affecting, 604
193f, 194f
incorporation of into protoporphyrin,
Factor VIII (antihemophilic factor
essential, 190, 190f, 193
271-272, 272f
A/globulin), 599f, 600, 600t
abnormal metabolism of, 195-196
in oxygen transport, 40-41
deficiency of, 604
deficiency of, 191-192, 194-195
Fertilization, glycoproteins in, 528
Factor VIII concentrates, recombinant
prostaglandin production and, 190
FeS. See Iron sulfur protein complex
DNA technology in production
free. See Free fatty acids
Fetal hemoglobin, P50 of, 42
of, 604
interconvertibility of, 231
Fetal warfarin syndrome, 488
Factor IX (antihemophilic factor B/Christ-
in membranes, 417, 418f
α-Fetoprotein, 583t
mas factor/plasma thromboplas-
metabolism of, 123-124, 123f
FFA. See Free fatty acids
tin component), 599f, 600, 600t
nomenclature of, 111-112, 112f
FGFs. See Fibroblast growth factors
coumarin drugs affecting, 604
oxidation of, 180-189. See also
Fibrillin, 535, 539
deficiency of, 604
Ketogenesis
Marfan syndrome caused by mutations in
Factor X (Stuart-Prower factor), 599f, 600,
acetyl-CoA release and, 123-124,
gene for, 539-540, 540f
600t
123f, 181-183, 181f, 182f
Fibrils, collagen, 535-539, 536f, 537t
activation of, 599f, 600-601
β, 181-183, 181f, 182f
Fibrin
coumarin drugs affecting, 604
ketogenesis regulation and,
dissolution of by plasmin, 604-605,
Factor XI (plasma thromboplastin
186-187, 187f, 188f
604f
antecedent), 599f, 600, 600t
modified, 183,183f
formation of, 598-601, 599f
deficiency of, 601
clinical aspects of, 187-189
thrombin in, 601- 602, 603f
Factor XII (Hageman factor), 599f, 600,
hypoglycemia caused by impairment
in thrombi, 598
600t
of, 187-188
Fibrin deposit, 598
Factor XIII (fibrin stabilizing factor/
in mitochondria, 180-181, 181f
Fibrin mesh, formation of, 598
fibrinoligase), 600t
physical/physiologic properties of, 114
Fibrin split products, in inflammation, 621t
Facultative heterochromatin, 316
saturated, 111, 112, 112t
Fibrin stabilizing factor (factor XIII), 600t
FAD. See Flavin adenine dinucleotide
synthesis of, 173-179, 174f, 175f. See
Fibrinogen (factor I), 580, 600, 600t
FADH2, fatty acid oxidation yielding, 181
also Lipogenesis
conversion of to fibrin, 601-602
Familial hypertrophic cardiomyopathy,
carbohydrate metabolism and, 123
Fibrinoligase (factor XIII), 600t
569-570, 570f
citric acid cycle in, 133, 134, 135f
Fibrinolysis, 604-605
Fanconi’s anemia, 338
extramitochondrial, 173
Fibrinopeptides A and B, 602, 603f
660
/
INDEX
Fibroblast growth factor receptor 3, achon-
Follicle-stimulating hormone (FSH), 437,
Fructose
droplasia caused by mutation
438, 439f
absorption of, 475, 475f
in gene for, 551t, 554, 554f
Footprinting, DNA, 413
in diabetic cataract, 172
Fibroblast growth factor receptors, chon-
Forbes’ disease, 152t
glycemic index of, 474
drodysplasias caused by mutation
Forensic medicine
hepatic
in gene for, 551t, 554, 554f
polymerase chain reaction (PCR) in,
hyperlipidemia/hyperuricemia and,
Fibroblast growth factors (FGFs), 554
405
170-171
Fibronectin, 535, 537-538, 540, 541f
restriction fragment length
metabolism affected by, 167, 169f
Fibrous proteins, 30
polymorphisms (RFLPs) in, 411
iron absorption affected by, 478
collagen as, 38
variable numbers of tandemly repeated
metabolism of, 167, 169f
Figlu. See Formiminoglutamate
units (VNTRs) in, 411
defects in, 171-172
Final common pathway of blood
Formic acid, pK/pKa value of, 12t
pyranose and furanose forms of, 103f
coagulation, 599, 601, 602f
Formiminoglutamate, in histidine
D-Fructose, 105t, 106f
Fingerprinting, DNA, 413
catabolism, 250, 251f
Fructose-1,6-bisphosphatase, 156t, 166
FISH. See Fluorescence in situ hybridization
Formyl-tetrahydrofolate, 493, 493f, 494
deficiency of, 171-172
Fish-eye disease, 228t
43S initiation complex, in protein synthesis,
Fructose-2,6-bisphosphatase, 157, 158f
5′ cap, mRNA modification and, 355
365, 366f
in covalent catalysis, 54, 55f
Flanking-sequence DNA, 397
43S preinitiation complex, in protein
Fructose-1,6-bisphosphate
Flavin adenine dinucleotide (FAD), 86-87,
synthesis, 365, 366f
in gluconeogenesis, 153, 154f
290t, 489
FPA/FPB. See Fibrinopeptides A and B
in glycolysis, 137, 138f
in citric acid cycle, 133
Frameshift mutations, 363, 364f
Fructose-2,6-bisphosphate, 157-158,
Flavin mononucleotide (FMN), 50, 86-87,
ABO blood group and, 619
158f
489
Framework regions, 592
Fructose intolerance, hereditary, 171
Flavoproteins
Free amino acids, absorption of, 477
Fructose 6-phosphate
electron-transferring, 87
Free energy
free energy of hydrolysis of, 82t
as oxidases, 86-87, 88f
changes in, 80-81
in gluconeogenesis, 153, 154f
Flip-flop rate, phospholipid, membrane
chemical reaction direction and,
in glycolysis, 137, 138f
asymmetry and, 420
60-61
Fructosuria, essential, 163, 171-172
Flippases, membrane asymmetry and, 420
coupling and, 81-82, 81f, 82f
FSF. See Fibrin stabilizing factor
Fluid mosaic model, 421f, 422
enzymes affecting, 63
FSH. See Follicle-stimulating hormone
Fluid-phase pinocytosis, 429-430, 429f
equilibrium state and, 60-61
Fucose, in glycoproteins, 516t
Fluidity, membrane, 422
redox potential and, 86, 87t
Fucosidosis, 532-533, 533t
Fluorescence, of porphyrins, 273-274,
transition states and, 61
Fucosylated oligosaccharides, selectins
277f
of hydrolysis of ATP, 82-83, 82t
binding, 530
Fluorescence in situ hybridization, in gene
liberation of as heat, 95
Fucosyltransferase/fucosyl (Fuc) transferase,
mapping, 406- 407, 407t,
Free fatty acids, 111, 180, 205, 206t
618
635t
in fatty liver, 212
Fumarase (fumarate hydratase), 132f, 133
Fluoride, 496t
glucose metabolism affecting, 215
Fumarate, 132f, 133
Fluoroacetate, 130, 132f
insulin affecting, 215
in tyrosine catabolism, 254, 255f
1-Fluoro-2,4-dinitrobenzene (Sanger’s
ketogenesis regulation and, 186-187,
in urea synthesis, 246f, 247
reagent), for polypeptide
187f, 188f
Fumarylacetoacetate, in tyrosine catabolism,
sequencing, 25
lipogenesis affected by, 177-178, 177f
254f, 255
5-Fluorouracil, 290, 291f, 297
metabolism of, 206-207
Fumarylacetoacetate hydrolase, defect at, in
Flux-generating reaction, 129
starvation and, 232-234, 234f, 234t
tyrosinemia, 255
FMN. See Flavin mononucleotide
Free polyribosomes, protein synthesis on,
Functional groups
Folate. See Folic acid
498, 506. See also
amino acid chemical reactions affected
Folate trap, 492f, 494
Polyribosomes
by, 18-20
Folding
Free radicals (reactive oxygen species). See
amino acid properties affected by, 18
polar and charged group positioning and,
also Antioxidants
physiologic significance of, 10-11
6
hydroperoxidases in protection against,
pK of, medium affecting, 13
protein, 36-37, 37f
88
Functional plasma enzymes, 57. See also
after denaturation, 36
in kwashiorkor, 479
Enzymes
Folic acid (folate/pteroylglutamic acid),
lipid peroxidation producing, 118-119,
Furanose ring structures, 103f, 104
482t, 492-494, 493f
120f
Fusion proteins, recombinant, in enzyme
coenzymes derived from, 51
in oxygen toxicity, 90-91, 611-613,
study, 58, 59f
deficiency of, 250, 482t, 494
613t
FXR. See Farnesoid X receptor
functional, 492, 494
xenobiotic cell injury and, 631, 631f
forms of in diet, 492, 493f
D-Fructofuranose, 103f
inhibitors of metabolism of, 494
Fructokinase, 167, 169f
∆G. See Free energy
supplemental, 494
deficiency of, 171-172
∆G0, 60, 61, 81
Folinic acid, 493
D-Fructopyranose, 103f
∆GD, 61
INDEX
/
661
∆GF, 61
Gastroenteropathy, protein-losing, 582
disease causing, recombinant DNA
enzymes affecting, 63
Gated ion channels, 424
technology in detection of,
G-CSF. See Granulocyte colony-stimulating
Gaucher’s disease, 203t
407-408, 408f, 409t
factor
GDH. See Glutamate dehydrogenase/
human genome information and, 638
G protein-coupled receptors (GPCRs), 458,
L-Glutamate dehydrogenase
heterogeneous nuclear RNA processing
459, 460f
GDP-Fuc, 516t
in regulation of, 354
G proteins, 459, 461t
GDP-Man, 516t, 517
housekeeping, 376
in calcium-dependent signal
GEFs. See Guanine nucleotide exchange
immunoglobulin, DNA rearrangement
transduction, 464f, 465
factors
and, 325-326, 393, 593-594
in cAMP-dependent signal transduction,
Gel electrophoresis
double-strand break repair and,
436, 459, 461t
polyacrylamide, for protein/peptide
337-338
in Jak/STAT pathway, 467
purification, 24, 24f, 25f
inducible, 376
in respiratory burst, 623
pulsed-field, for gene isolation, 635t
knockout, 412
GABA. See γ-Aminobutyrate
Gel filtration, for protein/peptide
processed, 325
GAGs. See Glycosaminoglycans
purification, 21-22, 23f
reporter, 385-386, 387f, 388f
Gal-Gal-Xyl-Ser trisaccharide, 518
Gemfibrozil, 229
targeted disruption of, 412
Gal-hydroxylysine (Hyl) linkage, 518
Gender, xenobiotic-metabolizing enzymes
variations in
Gal transferase, 618-619, 619f
affected by, 630
methods for isolation of, 635t
Galactokinase, 167, 170f
Gene. See Genes; Genome
normal, recombinant DNA techniques
inherited defects in, 172
Gene array chips, protein expression and,
for identification of, 407
Galactosamine, 169, 171f
28
size/complexity and, 397, 399t
D-Galactosamine (chondrosamine), 106
Gene conversion, 325
Genetic code, 303, 358-363, 359t
Galactose, 102, 167-169, 170f
Gene disruption/knockout, targeted, 412
features of, 358-359, 360t
absorption of, 475, 475f
Gene expression
L-α-amino acids encoded by, 14, 15-16t
glycemic index of, 474
constitutive, 376, 378
Genetic diseases
in glycoproteins, 516t
in pyrimidine nucleotide synthesis,
diagnosis of
metabolism of, 167-169, 170f
regulation of, 297-299
enzymes in, 57
enzyme deficiencies and, 172
regulation of, 374-395
recombinant DNA technology in,
D-Galactose, 104f, 105t
alternative splicing and, 354, 354f,
407-412, 408f, 409t, 410f, 411f
α-D-Galactose, 104f
393-394, 636
gene therapy for, 411
Galactose 1-phosphate uridyl transferase,
eukaryotic transcription and, 383-387
Genetic linkage. See Linkage analysis
167, 170f
hormones in, 458f
Genetic mapping, 633, 634f. See also
Galactosemia, 102, 163, 172
negative versus positive, 374, 375t,
Human Genome Project
Galactosidases, in glycoprotein analysis,
378, 380
Genetics. See also Human Genome Project
517
in prokaryotes versus eukaryotes,
molecular, 1
Galactoside, 106
391-395, 392t
xenobiotic-metabolizing enzymes affected
Galactosylceramide, 116, 117f, 201, 203f
regulatory protein-DNA binding
by, 630
GalCer. See Galactosylceramide
motifs and, 387-390, 388t,
Genevan system, for fatty acid
Gallstones, 474
389f, 390f, 391f
nomenclature, 111
cholesterol, 219
regulatory protein DNA binding and
Genome
GalNAc, in glycoproteins, 515, 516t
trans-activation domains and,
redundancy in, 320-322
GalNAc-Ser(Thr) linkage
390-391, 392f
removal of gene from (targeted gene
in glycoproteins, 518, 519f
retinoic acid in, 483
disruption/knockout), 412
in glycosaminoglycans, 543
temporal responses and, 374-375,
sequencing. See also Human Genome
GalNAc transferase, in ABO system,
375f
Project
618-619, 619f
Gene mapping, 319, 406-407, 407t, 633,
approaches used in, 634, 635t
Gamma- (γ) aminobutyrate. See
634f, 635t
results of, 636-637, 636t, 637t
γ-Aminobutyrate
Gene products, diseases associated with
working draft of, 633
Gamma- (γ) carotene, 482
deficiency of, 407t
Genomic library, 402, 413
Gamma-globulin, 581f
gene therapy for, 411
Genomic technology, 396. See also
Gamma- (γ) glutamyltransferase, 630
Gene therapy, 411
Recombinant DNA/recombinant
Gamma- (γ) glutamyl transpeptidase
for α1-antitrypsin deficiency emphysema,
DNA technology
(γ-glutamyltransferase),
589
Genomics, protein sequencing and, 28
diagnostic significance of, 57t
for urea biosynthesis defects, 248
Geometric isomerism, of unsaturated fatty
Gangliosides, 116
Gene transcription. See Transcription
acids, 112-114, 114f
amino sugars in, 106, 169, 171f
General acid/base catalysis, 52
Geranyl diphosphate, in cholesterol
sialic acids in, 110
Genes
synthesis, 219, 221f
synthesis of, 201-202, 203f
alteration of, 323-326, 324f, 325f
GGT. See γ-Glutamyltransferase
Gap junctions, 431
amplification of, in gene expression
Ghosts, red cell membrane analysis and, 614
GAPs. See Guanine activating proteins
regulation, 323-326, 324f,
Gibbs free energy/Gibbs energy. See Free
Gastric lipase, 475
325f
energy
662
/
INDEX
Gilbert syndrome, 283
regulation of, 155-158, 156t, 158f
Glucose 1-phosphate
GK (glucokinase) gene, regulation of, 355,
allosteric modification in, 157
free energy of hydrolysis of, 82t
355f
covalent modification in, 157
in gluconeogenesis, 154f, 155
Gla. See γ-Carboxyglutamate
enzyme induction/repression in,
Glucose 6-phosphate
GlcCer. See Glucosylceramide
155-157, 156t
free energy of hydrolysis of, 82t
GlcNAc. See N-Acetylglucosamine
fructose 2,6-bisphosphate in,
in gluconeogenesis, 153, 154f
Glial fibrillary acid protein, 577t
157-158, 158f
in glycogen biosynthesis, 145, 146f
Glibenclamide. See Glyburide
substrate (futile) cycles in, 158
in glycolysis, 137, 138f
Globin, 278
thermodynamic barriers to glycolysis
Glucose-6-phosphate dehydrogenase
α-Globin gene, localization of, 407t
and, 153-155, 154f
deficiency of, 163, 169-170, 613, 614f,
β-Globin gene
Gluconolactone hydrolase, 163, 165f
619, 630t
localization of, 407t
D-Glucopyranose, 103f
in pentose phosphate pathway, 156t,
recombinant DNA technology in
αD-Glucopyranose (α-anomer), 103f, 104
163, 164f, 165f
detection of variations in,
βD-Glucopyranose (β-anomer), 103f, 104
Glucose tolerance, 161-162, 161f
407-408, 408f, 409t
Glucosamine, 106f, 169, 171f
Glucose transporters, 159, 160t
Globular proteins, 30
in heparin, 545
in blood glucose regulation, 159, 160
β-turns in, 32, 34f
Glucosan (glucan), 107
insulin affecting, 427
β1-Globulin, 581f
Glucose, 102, 102-106
red cell membrane, 611, 612t
transferrin as, 586
absorption of, 474, 475, 475f
Glucoside, 106
γ-Globulin, 581f
as amino sugar precursor, 169, 171f
Glucosuria, 161
Globulins, 580
blood levels of. See Blood glucose
Glucosylceramide, 116, 201, 203f
Glomerular filtration, basal lamina in, 540
epimers of, 104, 104f
Glucuronate/glucuronic acid, 166-167,
Glomerular membrane, laminin in, 540-542
in extracellular and intracellular fluid,
168f
Glomerulonephritis, 541
416, 416t
bilirubin conjugation with, 280, 280f,
Glomerulus, renal, laminin in, 540-542
furanose forms of, 103f, 104
281f
Glucagon, 148, 160-161
galactose conversion to, 167-169,
D-Glucuronate, 105, 106f
in gluconeogenesis regulation, 157
170f
β-Glucuronidases, 281
in lipogenesis regulation, 178, 178f
glycemic index of, 474
Glucuronidation
Glucagon/insulin ratio, in ketogenesis
in glycogen biosynthesis, 145, 146f
of bilirubin, 280, 280f, 281f
regulation, 187
in glycoproteins, 516t
of xenobiotics, 628-629
Glucagon-like peptide, 437
insulin secretion and, 160, 161-162
Glucuronides, 163
Glucan (glucosan), 107
interconvertibility of, 231
GLUT 1-4. See Glucose transporters
Glucan transferase, in glycogenolysis, 146,
isomers of, 102-104, 103f
Glutamate
146f, 148f
as metabolic necessity, 232
carboxylation of, vitamin K as cofactor
Glucocorticoid receptor-interacting protein
metabolism of, 122-123, 122f, 123f,
for, 487-488, 488f
(GRIP1 coactivator), 472, 472t
124-125, 125f, 136-140, 138f,
catabolism of, 249, 250f
Glucocorticoid response element (GRE),
139f, 140f, 159, 159f. See also
in proline synthesis, 238, 239f
456, 458f, 459t
Gluconeogenesis; Glycolysis
synthesis of, 237, 238f
Glucocorticoids, 437. See also specific type
ATP generated by, 142, 143t
transamination and, 243-244, 243f,
in amino acid transport, 427
in fed state, 232
244f
blood glucose affected by, 161
free fatty acids and, 215
in urea biosynthesis, 243-244, 243f,
in lipolysis, 215, 216f
insulin affecting, 160, 161-162
244f
NF-κB pathway regulated by, 468, 468f
by pentose phosphate pathway, 123,
L-Glutamate decarboxylase, 267, 268f
receptors for, 471
163-166, 164f, 165f, 167f
Glutamate dehydrogenase/L-glutamate
synthesis of, 440, 441f
starvation and, 232-234, 234f, 234t,
dehydrogenase, 237, 238f
transport of, 454-455, 455t
236
in nitrogen metabolism, 244-245, 244f,
D-Glucofuranose, 103f
permeability coefficient of, 419f
245f
Glucogenic amino acids, 231-232
pyranose forms of, 103f, 104
Glutamate-α-ketoglutarate transaminase
Glucokinase, 156t
renal threshold for, 161
(glutamate transaminase), in urea
in blood glucose regulation, 159-160,
structure of, 102, 103f
biosynthesis, 243-244, 244f
160f
transport of, 159, 160t, 428, 429f, 475,
Glutamate-γ-semialdehyde dehydrogenase,
in glycogen biosynthesis, 145, 146f, 156t
475f
block at in hyperprolinemia,
in glycolysis, 137, 138f, 156t
insulin affecting, 427
250
Glucokinase gene, regulation of, 355, 355f
D-Glucose, 103f, 104f, 105t
Glutamic acid, 15t
Gluconeogenesis, 123, 125, 153-162, 154f
α-D-Glucose, 104f
Glutaminase, in amino acid nitrogen
blood glucose regulation and, 158-161,
L-Glucose, 103f
catabolism, 245, 245f
159f, 160f
Glucose-alanine cycle, 159
Glutamine, 15t
citric acid cycle in, 133-134, 134f,
Glucose-6-phosphatase
in amino acid nitrogen catabolism, 245,
153-155, 154f
deficiency of, 152t, 300
245f
glycolysis and, 136-140, 138f, 139f,
in gluconeogenesis, 156t
catabolism of, 249, 250f
153-155, 154f
in glycogenolysis, 147
synthesis of, 237, 238f
INDEX
/
663
Glutamine analogs, purine nucleotide
Glycine, 15t, 264
glycogen synthase and phosphorylase in
synthesis affected by, 293
catabolism of, pyruvate formation and,
regulation of, 148-150,
Glutamine synthetase/synthase, 237, 238f,
250, 252f
150-151, 150f, 151f
245, 245f
in collagen, 535
Glycolipid storage diseases, 197
Glutamyl amidotransferase, PRPP,
in heme synthesis, 264, 270-273, 273f,
Glycolipids (glycosphingolipids), 111, 116,
regulation of, 294, 295f
274f, 275f, 276f
117f
γ-Glutamyltransferase, 630
synthesis of, 238, 239f
ABO blood group and, 618
γ-Glutamyl transpeptidase
Glycine synthase complex, 250
amino sugars in, 169, 171f
(γ-glutamyltransferase),
Glycinuria, 250
galactose in synthesis of, 167-169, 170f
diagnostic significance of, 57t
Glycocalyx, 110
Glycolysis, 83, 122, 123f, 136-144, 137f
Glutaric acid, pK/pKa value of, 12t
Glycochenodeoxycholic acid, synthesis of,
aerobic, 139
Glutathione
226f
anaerobic, 136, 137f, 139
as antioxidant, 611-613, 613t
Glycocholic acid, synthesis of, 226f
as muscle ATP source, 574-576, 575f,
in conjugation of xenobiotics,
Glycoconjugate (complex) carbohydrates,
575t
629-630
glycoproteins as, 514
ATP generated by, 142, 143t
as defense mechanism, 629
Glycoforms, 514
clinical aspects of, 142-143
functions of, 629-630
Glycogen, 102, 107, 108f
in erythrocytes, 140, 140f
Glutathione peroxidase, 88, 166, 167f, 170,
in carbohydrate metabolism, 123, 123f,
glucose utilization/gluconeogenesis and,
612, 613t
155
136-140, 138f, 139f, 153-155,
Glutathione reductase, erythrocyte
carbohydrate storage and, 145, 146t
154f. See also Gluconeogenesis
pentose phosphate pathway and, 166,
metabolism of, 145-152. See also
pathway of, 136-140, 138f, 139f, 140f
167f
Glycogenesis; Glycogenolysis
pyruvate oxidation and, 134, 135f,
riboflavin status and, 490
branching in, 145, 147f
140-142, 141f, 142f, 143t
Glutathione S-transferases, 629
clinical aspects of, 151-152, 152f
regulation of, 140
in enzyme study, 58, 59f
regulation of
enzymes in, 156t
Glyburide (glibenclamide), 188
cyclic AMP in, 147-150, 148f,
fructose 2,6-bisphosphate in,
Glycan intermediates, formation of during
149f, 150f
157-158, 158f
N-glycosylation, 526
glycogen synthase and
gluconeogenesis and, 140, 155-158,
Glycemic index, 474
phosphorylase in, 150-151, 151f
156t, 158f
Glyceraldehyde (glycerose), D and L isomers
in starvation, 234
at subcellular level, 126, 127f
of, 103f, 104f
muscle, 145, 146t, 573, 575f
thermodynamic barriers to reversal of,
Glyceraldehyde 3-phosphate
Glycogen phosphorylase, 145-146, 146f
153-155
in glycolysis, 137, 138f
pyridoxal phosphate as cofactor for,
Glycolytic enzymes, in muscle, 556
oxidation of, 137, 139f
491
Glycomics, 533
Glyceraldehyde 3-phosphate dehydrogenase
regulation of, 148-150, 150-151, 150f,
Glycophorins, 110, 518, 615-616, 615f,
in glycolysis, 137, 138f
151f
616f, 616t
in red cell membranes, 615f, 616t
Glycogen storage diseases, 102, 145,
Glycoprotein IIb-IIIa, in platelet activation,
Glycerol, 114
151-152, 152t
607, 622t
in lactic acid cycle, 159
Glycogen synthase, in glycogen metabolism,
Glycoprotein glycosyltransferases, 520
permeability coefficient of, 419f
145, 146f, 155, 156t
Glycoproteins, 30, 109-110, 109t, 439f,
synthesis of, 155
regulation of, 148-150, 150-151, 150f,
514-534, 580, 581-582. See also
Glycerol ether phospholipids, synthesis of,
151f
specific type and Plasma proteins
199, 200f
Glycogen synthase a, 148-150, 150f
amino sugars in, 106, 169, 171f
Glycerol kinase, 155, 197, 198f, 214
Glycogen synthase b, 150, 150f
asialoglycoprotein receptor in clearance
Glycerol moiety, of triacylglycerols, 123
Glycogenesis, 124-125, 145, 146f
of, 517
Glycerol phosphate pathway, 198f
regulation of
as blood group substances, 514, 618
Glycerol-3-phosphate
cyclic AMP in, 147-150, 148f, 149f,
carbohydrates in, 109t
acylglycerol biosynthesis and, 197, 197f,
150f
classes of, 518, 519f
198f
enzymes in, 156t
complex, 521, 522f
free energy of hydrolysis of, 82t
glycogen synthase and phosphorylase
formation of, 521, 524
triacylglycerol esterification and,
in, 148-150, 150-151, 150f,
diseases associated with abnormalities of,
214-215, 214f
151f
530, 530t, 531f, 531t, 532f, 533t
Glycerol-3-phosphate acyltransferase, 198f,
Glycogenin, 145, 146f
extracellular, absorptive pinocytosis of,
199
Glycogenolysis, 125, 145-147, 146f
430
Glycerol-3-phosphate dehydrogenase, 155,
blood glucose regulation and, 158-161,
in fertilization, 528
198f, 199
159f, 160f
functions of, 514, 515t, 528-533, 529t
mitochondrial, 87
cyclic AMP-independent, 148
galactose in synthesis of, 167-169, 170f
Glycerophosphate shuttle, 99, 100f
cyclic AMP in regulation of, 147-150,
glycosylphosphatidylinositol-anchored,
Glycerophospholipids, 111
148f, 149f, 150f
518, 519f, 527-528, 528t
Glycerose (glyceraldehyde), D and L isomers
debranching enzymes in, 146-147,
high-mannose, 521, 522f
of, 103f, 104f
148f
formation of, 521, 524
664
/
INDEX
Glycoproteins (cont.)
Glycosylphosphatidylinositol-anchored
GTP-γ-S, vesicle coating and, 510
hybrid, 521, 522f
(GPI-anchored/GPI-linked)
Guanine, 288t
formation of, 521
glycoproteins, 518, 519f,
Guanine activating proteins, 501, 502f
immunoglobulins as, 593
527-528, 528t
Guanine nucleotide exchange factors, 501,
membrane asymmetry and, 420
in paroxysmal nocturnal hemoglobinuria,
502f
N-linked, 518, 519f, 521-527
531, 531f
Guanosine, 287f, 288t
nucleotide sugars, 516-517, 516t
Glycosyltransferases, glycoprotein, 520,
base pairing of in DNA, 303, 304, 305f
O-linked, 518, 518-520, 519f, 520f,
526-527
in uric acid formation, 299, 299f
520t, 521t
Glypiation, 528
Guanosine diphosphate fucose (GDP-Fuc),
oligosaccharide chains of, 514
GM-CSF. See Granulocyte-macrophage
516t
red cell membrane, 615, 615f
colony-stimulating factor
Guanosine diphosphate mannose
sugars in, 515-517, 516t
GM1 ganglioside, 116, 117f
(GDP-Man), 516t, 517
techniques for study of, 514, 515t
GM3 ganglioside, 116
Guanosine monophosphate. See GMP
asialoglycoprotein receptor in, 517
GMP, 288t, 297f
Guanylyl cyclase, 462, 463
glycosidases in, 517, 517t
cyclic, 289f, 290
Gyrase, bacterial, 306
lectins in, 517-518, 518t
as second messenger, 290, 436, 437t,
Gyrate atrophy of retina, 250
in zona pellucida, 528
457, 462-463
Glycosaminoglycans, 109, 109f, 542,
IMP conversion to, 293, 296f
542-547. See also specific type
feedback-regulation of, 294, 296f
H bands, 556, 557f, 558f
amino sugars in, 106
PRPP glutamyl amidotransferase
H blood group substance, 618, 619f
deficiencies of enzymes in degradation of,
regulated by, 294
H chains. See Heavy chains
545-547, 546t, 547f
Golgi apparatus
H1 histones, 314, 315f
disease associations of, 548-549
core protein synthesis in, 543
H2A histones, 314, 315
distributions of, 543-545, 544f, 544t
glycosylation and, 509, 524-525, 525f
H2B histones, 314, 315
functions of, 547-549, 548t
in membrane synthesis, 509
H3 histones, 314, 314-315
structural differences among, 543-545,
in protein sorting, 498, 500f, 509
H4 histones, 314, 314-315
544f, 544t, 545f
in VLDL formation. 213f
H2S. See Hydrogen sulfide
synthesis of, 542-543
retrograde transport from, 507
H substance, ABO blood group and, 618
Glycosidases, in glycoprotein analysis, 517
Gout/gouty arthritis, 299
Haber-Weiss reaction, 612
Glycosides, 105-106
GPIIb-IIIa, in platelet activation, 607, 622t
Hageman factor (factor XII), 599f, 600,
β-N-Glycosidic bond, 286, 287f
GPCRs. See G protein-coupled receptors
600t
Glycosphingolipids (glycolipids), 111, 116,
GPI-anchored/linked glycoproteins, 518,
Hairpin, 306, 309f, 413
117f, 201-202, 203f
519f, 527-528, 528t
Half life
ABO blood group and, 618
in paroxysmal nocturnal hemoglobinuria,
enzyme, 242
amino sugars in, 169, 171f
531, 531f
protein, 242
galactose in synthesis of, 167-169, 170f
Granulocyte-colony stimulating factor, 610
plasma protein, 582
in membranes, 417
Granulocyte-macrophage colony-
Halt-transfer signal, 506
membrane asymmetry and, 420
stimulating factor, 610
Hapten, in xenobiotic cell injury, 631,
Glycosuria, 161
Granulomatous disease, chronic, 623, 623f
631f
N-Glycosylases, in base excision-repair, 337,
Granulosa cells, hormones produced by,
Haptoglobin, 583t, 584, 584f
337f
442
Hartnup disease, 258, 490
Glycosylated hemoglobin (HbA1c), 47
Gratuitous inducers, 378
HAT activity. See Histone acetyltransferase
Glycosylation
GRE. See Glucocorticoid response element
activity
of collagen, 537
GRIP1 coactivator, 472, 472t
HbA (hemoglobin A), P50 of, 42
congenital disorders of, 531, 531t
Group transfer potential, 83, 83f
HbA1c (glycosylated hemoglobin), 47
in covalent modification, mass increases
of nucleoside triphosphates, 289-290,
HbF (fetal hemoglobin), P50 of, 42
and, 27t
289f, 290f, 290t
HbM (hemoglobin M), 46, 363, 614
inhibitors of, 527, 527t
Group transfer reactions, 8
HbS (hemoglobin S), 46, 46f, 363
nucleotide sugars in, 516-517, 516t
Growth factors, hematopoietic, 610
hCG. See Human chorionic gonadotropin
N-Glycosylation, 521-527, 523f, 524f,
Growth hormone, 437, 438
HDL. See High-density lipoproteins
525f, 526t
amino acid transport affected by, 427
Health, normal biochemical processes as
dolichol-P-P-oligosaccharide in,
localization of gene for, 407t
basis of, 2-4, 3t
521-524, 523f
receptor for, 436
Heart
in endoplasmic reticulum, 524-525,
GSH. See Glutathione
developmental defects of, 570
525f
GSLs. See Glycosphingolipids
metabolism in, 235t
glycan intermediates formed during, 526
GST (glutathione S-transferase) tag, in
thiamin deficiency affecting, 489
in Golgi apparatus, 524-525, 525f
enzyme study, 58, 59f
Heart disease, coronary (ischemic). See also
inhibition of, 527, 527t
GTP, 290
Atherosclerosis
regulation of, 526-527, 527t
cyclic GMP formed from, 462
cholesterol and, 227
tunicamycin affecting, 527, 527t
in phosphorylation, 85
Heart failure, 556
O-Glycosylation, 520, 521f
GTPases, 459
in thiamin deficiency, 489
INDEX
/
665
Heat, free energy liberated as, 95
oxygen dissociation curve for, 41-42,
Heparin, 109, 109f, 544f, 544t, 545, 545f,
Heat-shock proteins, as chaperones, 36-37
42f
603-604
Heavy chains
in oxygen transport, 40-41
antithrombin III activity affected by,
immunoglobulin, 591, 592f
oxygenation of
547, 603-604
genes producing, 593
conformational changes and, 43, 43f,
in basal lamina, 540
myosin, 560
44f
binding of, fibronectin in, 540, 541f
familial hypertrophic cardiomyopathy
apoprotein, 42
functions of, 547
caused by mutations in gene for,
2,3-bisphosphoglycerate stabilizing,
lipoprotein and hepatic lipases affected
569-570, 570f
45, 45f
by, 207
Heavy meromyosin, 560f, 561, 561f
high altitude adaptation and, 46
Heparin cofactor II, as thrombin inhibitor,
Heinz bodies, 613
mutant hemoglobins and, 46
603
Helicases, DNA, 326-327, 327f, 328, 328t
in proton transport, 44
Hepatic ALA synthase (ALAS1), 272
Helicobacter pylori, ulcers associated with,
tetrameric structure of, 42
in porphyria, 277t, 278
474
changes in during development,
Hepatic lipase, 207
Helix
42-43, 43f
in chylomicron remnant uptake, 209,
Double, of DNA structure, 7, 303, 304,
Hemoglobin A (HbA), P50 of, 42
209f
305f
Hemoglobin A1c (glycosylated hemoglobin),
deficiency of, 228t
recombinant DNA technology and,
47
Hepatic portal system, 158
396, 397
Hemoglobin A2
in metabolite circulation, 124, 125f
triple, of collagen structure, 38, 38f,
Hemoglobin Bristol, 362
Hepatitis, 130
535-539, 536f
Hemoglobin Chesapeake, 46
in α1-antitrypsin deficiency, 590
α-Helix, 31-32, 32f, 33f
Hemoglobin F(fetal hemoglobin), P50 of, 42
jaundice in, 284t
amphipathic, 31-32
Hemoglobin Hikari, 362-363
Hepatocytes
in myoglobin, 40, 41f
Hemoglobin M, 46, 363, 614
glycoprotein clearance from, asialoglyco-
Helix-loop-helix motifs, 33
Hemoglobin Milwaukee, 362
protein receptor in, 517
Helix-turn-helix motif, 387, 388t,
Hemoglobin S, 46, 46f, 363
heme synthesis in, 272
389-390, 389f
Hemoglobin Sydney, 362
ALA synthase in regulation of,
Hemagglutinin, influenza virus, calnexin
Hemoglobinopathies, 46, 619
272-273, 276f
binding to, 526
Hemoglobinuria, paroxysmal nocturnal,
Hepatolenticular degeneration (Wilson
Hematology, recombinant DNA
432t, 528, 530t, 531, 531f
disease), 432t, 587, 588-589
technology affecting, 624
Hemolytic anemias, 136, 143, 609, 619,
ceruloplasmin levels in, 587
Hematopoietic growth factors, 610
620t
gene mutations in, 432t, 588-589
Heme, 40, 41f, 270
glucose-6-phosphate dehydrogenase
Heptoses, 102, 102t
catabolism of, bilirubin produced by,
deficiency causing, 163,
Hereditary elliptocytosis, 617
278-280, 279f
169-170, 613, 614f, 619
Hereditary erythroblastic multinuclearity
in proteins, 270. See also Heme proteins
haptoglobin levels in, 584
with a positive acidified lysis test
synthesis of, 270-273, 273f, 274f, 275f,
hyperbilirubinemia/jaundice in, 282,
(HEMPAS), 530t, 531
276f
284, 284t
Hereditary hemochromatosis, 586-587,
disorders of (porphyrias), 274-278,
pentose phosphate pathway/glutathione
587f
277f, 277t
peroxidase and, 166, 167f,
Hereditary nonpolyposis colon cancer,
Heme iron, 278, 585
169-170
mismatch repair genes in, 336
absorption of, 478, 585, 585f
primaquine-sensitive, 613
Hereditary spherocytosis, 432t, 617, 617f
hindered environment for, 41, 41f
red cell membrane abnormalities causing,
Hermansky-Pudlak syndrome, 512t
Heme oxygenase system, 278, 279f
619
Hers’ disease, 152t
Heme proteins (hemoproteins), 270, 271t.
Hemopexin, 583t
Heterochromatin, 316
See also Hemoglobin; Myoglobin
Hemophilia A, 604
Heterogeneous nuclear RNA (hnRNA), 310
cytochrome P450 isoforms as, 627
Hemophilia B, 604
processing of, gene regulation and, 354
Heme synthase (ferrochelatase), 271, 272f
Hemoproteins. See Heme proteins
Heterotrophic organisms, 82
in porphyria, 277t
Hemosiderin, 586
Hexapeptide, in albumin synthesis, 583
Hemin, 278, 279f
Hemostasis, 598-608. See also Coagulation
Hexokinase, 156t
Hemochromatosis, 478, 586-587, 587f
laboratory tests in evaluation of, 608
in blood glucose regulation, 159, 160f
Hemoglobin, 40-48, 581f
phases of, 598
in fructose metabolism, 167, 169f
allosteric properties of, 42-46
HEMPAS. See Hereditary erythroblastic
in glycogen biosynthesis, 145, 156t
β subunits of, myoglobin and, 42
multinuclearity with a positive
in glycolysis, 136-137, 138f, 156t
bilirubin synthesis and, 278-280, 279f
acidified lysis test
as flux-generating reaction, 129
in carbon dioxide transport, 44, 45f
Henderson-Hasselbalch equation, 11
regulation and, 140
extracorpuscular, haptoglobin binding of,
Heparan sulfate, 538, 544f, 544t, 545,
Hexosamines (amino sugars), 106, 106f
583f, 584
547-548
glucose as precursor of, 169, 171f
glycosylated (HbA1c), 47
in basal lamina, 540
in glycosaminoglycans, 109, 169, 171f
mutant, 46, 362-363
clotting/thrombosis affected by, 607,
in glycosphingolipids, 169, 171f
oxygen affinities (P50) and, 42-43, 43f
607t
interrelationships in metabolism of, 171f
666
/
INDEX
Hexose monophosphate shunt. See Pentose
Histidine F8
receptors for, 435- 436, 436f, 471
phosphate pathway
in oxygen binding, 40, 41f
proteins as, 436
Hexoses, 102, 102t
replacement of in hemoglobin M, 46
recognition and coupling domains on,
in glycoproteins, 109t
Histidinemia, 250
435-436
metabolism of, 163-166, 164f, 165f,
Histone acetyltransferase activity, of
specificity/selectivity of, 435, 436f
167f. See also Pentose phosphate
coactivators, 383, 472, 473
signal transduction and, 456 -473
pathway
Histone chaperones, 315
intracellular messengers and,
clinical aspects of, 169-172
Histone dimer, 315
457-468, 461t, 463t
physiologic importance of, 105, 105t
Histone octamer, 315, 315f
response to stimulus and, 456, 457f
HFE mutations, in hemochromatosis,
Histone tetramer, 314-315, 315
signal generation and, 456-457, 458f,
586-587
Histones, 314, 314-315, 315f, 315t
459f, 459t
HGP. See Human Genome Project
acetylation and deacetylation of, gene
transcription modulation and,
HhaI, 399t
expression affected by, 383
468-473, 470f, 471f, 472t
Hierarchical shotgun sequencing, 634
HIV-I, glycoproteins in attachment of, 533
stimulus recognition by, 456, 457f
High altitude, adaptation to, 46
HIV protease, in acid-base catalysis, 52, 53f
storage/secretion of, 453, 454t
High-density lipoproteins, 205, 206t
HMG-CoA. See 3-Hydroxy-3-methyl-
synthesis of
apolipoproteins of, 205-206, 206t
glutaryl-CoA
chemical diversity of, 438, 439f
atherosclerosis and, 210-211, 227
HMM. See Heavy meromyosin
cholesterol in, 438, 438-445, 439t,
metabolism of, 209-211, 211f
hMSH1/hMSH2, in colon cancer, 336
440f
ratio of to low-density lipoproteins, 227
HNCC. See Hereditary nonpolyposis colon
peptide precursors and, 449-453
receptor for, 210, 211f
cancer
specialization of, 437
High-density microarray technology, 412
hnRNA. See Heterogeneous nuclear RNA
tyrosine in, 438, 439-449, 439t
High-energy phosphates, 83. See also ATP
Holocarboxylase synthetase, biotin as
target cells for, 434-435, 435t
in energy capture and transfer, 82-83,
coenzyme of, 494
transport of, 454-455, 454t, 455t
82f, 82t, 83f
Homeostasis
vitamin D as, 484-486
as “energy currency” of cell, 83-85, 84f,
blood in maintenance of, 580
Housekeeping genes, 376
85f
hormone signal transduction in
Hp. See Haptoglobin
symbol designating, 83
regulation of, 456, 457f
HpaI, 399t
transport of, creatine phosphate shuttle
Homocarnosine, 264, 265f
HPETE. See Hydroperoxides
in, 100, 101f
Homocarnosinosis, 264
HPLC. See High-performance liquid
High-mannose oligosaccharides, 521,
Homocysteine
chromatography
522f
in cysteine and homoserine synthesis,
HREs. See Hormone response elements
formation of, 521, 524
238-239, 239f
HRPT. See Hypoxanthine-guanine
High-molecular-weight kininogen, 599f, 600
functional folate deficiency and, 494
phosphoribosyl transferase
High-performance liquid chromatography,
Homocystinurias, 250
hsp60/hsp70, as chaperones, 36-37
reversed phase, for protein/
vitamin B12 deficiency/functional folate
5HT (5-hydroxytryptamine). See Serotonin
peptide purification, 23-24
deficiency and, 492f, 494
Human chorionic gonadotropin (hCG), 438
Hill coefficient, 67
Homodimers, 34
Human Genome Project, 3-4, 633-641
Hill equation, 66-67, 67f
Homogentisate, in tyrosine catabolism,
approaches used in elucidation of, 634,
Hindered environment, for heme iron, 41,
254f, 255
635t
41f
Homogentisate dioxygenase/oxidase, 89
future work and, 637
HindIII, 399t
deficiency of, in alkaptonuria, 255
goals of, 633-635
Hinge region
Homology
implications of, 637-638
immunoglobulin, 591, 592f
conserved residues and, 54, 55t
major findings of, 636-637, 636t, 637t
nuclear receptor protein, 460
in protein classification, 30
protein sequencing and, 28
Hippuric acid/hippurate, synthesis of, 264,
Homopolymer tailing, 399
Human immunodeficiency virus (HIV-I),
265f
Homoserine, synthesis of, 239, 239f
glycoproteins in attachment of,
Histamine, 621t
Hormone-dependent cancer, vitamin B6
533
formation of, 265
deficiency and, 491
Hunter syndrome, 546t
Histidase, impaired, 250
Hormone response elements, 349, 386,
Hurler syndrome, 546t
Histidine, 16t, 265, 265f
388f, 456-457, 459t, 469, 470f
Hurler-Scheie syndrome, 546t
β-alanyl dipeptides and, 264, 265f
Hormone-sensitive lipase, 214-215, 214f
Hyaluronic acid, 109, 109f, 543, 544f, 544t
catabolism of, 250, 251f
insulin affecting, 215
disease associations and, 548
conserved residues and, 55t
Hormones. See also specific type
functions of, 547
decarboxylation of, 265
in blood glucose regulation, 159
Hyaluronidase, 547
in oxygen binding, 40, 41f
classification of, 436-437, 437t
Hybrid glycoproteins, 521, 522f
requirements for, 480
facilitated diffusion regulated by, 427
formation of, 521
Histidine 57, in covalent catalysis, 53-54,
glycoproteins as, 514
Hybrid mapping, radiation, 635t
54f
lipid metabolism regulated by, 215-217,
Hybridization, 397, 403-404, 413
Histidine E7, in oxygen binding,
216f
in situ, in gene mapping, 406-407, 407t,
40, 41f
in metabolic control, 128f, 129
635t
INDEX
/
667
Hybridomas, 595-596, 596f
17α-Hydroxylase, in steroid synthesis, 440,
Hyperglycemia. See also Diabetes mellitus
Hydrocortisone. See Cortisol
441f, 442, 443f
glucagon causing, 161
Hydrogen bonds, 5, 6f
18-Hydroxylase, in steroid synthesis, 440,
insulin release in response to, 466f
in DNA, 303, 304, 305f
441f
Hyperhomocysteinemia, folic acid supple-
Hydrogen ion concentration. See also pH
21-Hydroxylase, in steroid synthesis, 440,
ments in prevention of, 494
enzyme-catalyzed reaction rate affected
441f
Hyperhydroxyprolinemia, 255
by, 64, 64f
27-Hydroxylase, sterol, 226
Hyperkalemic periodic paralysis, 569t
Hydrogen peroxide
Hydroxylase cycle, 89, 90f
Hyperlacticacidemia, 212
glutathione in decomposition of, 629
Hydroxylases, 89-90
Hyperlipidemia, 170-171, 490
as hydroperoxidase substrate, 88-89
in steroid synthesis, 438, 440, 441f
Hyperlipoproteinemias, 205, 228t, 229
production of in respiratory burst, 622
Hydroxylation
familial, 228t
Hydrogen sulfide, respiratory chain affected
in collagen processing, 537
Hyperlysinemia, periodic, 258
by, 95, 96f
in covalent modification, mass increases
Hypermetabolism, 136, 479
Hydrolases, 50
and, 27t
Hyperornithinemia-hyperammonemia
cholesteryl ester, 223
of xenobiotics, 626, 626-628, 629t
syndrome, 250
fumarylacetoacetate, defect at, in
Hydroxylysine, synthesis of, 240
Hyperoxaluria, primary, 250
tyrosinemia, 255
5-Hydroxymethylcytosine, 287, 289f
Hyperparathyroidism, bone and cartilage
gluconolactone, 163, 165f
3-Hydroxy-3-methylglutaryl-CoA
affected in, 551t
lysosomal, deficiencies of, 532-533, 533t
(HMG-CoA)
Hyperphenylalaninemias, 255
Hydrolysis (hydrolytic reactions), 7-8. See
in ketogenesis, 184-185, 185f
Hyperprolinemias, types I and II, 249-250
also specific reaction
in mevalonate synthesis, 219, 220f
Hypersensitive sites, chromatin, 316
free energy of, 82-83, 82t
3-Hydroxy-3-methylglutaryl-CoA
Hypersplenism, in hemolytic anemia, 619
in glycogenolysis, 146, 146f, 148f
(HMG-CoA) lyase
Hypertension, hyperhomocysteinemia and,
of triacylglycerols, 197
deficiency of, 188
folic acid supplements in
Hydropathy plot, 419
in ketogenesis, 185, 185f
prevention of, 494
Hydroperoxidases, 86, 88-89
3-Hydroxy-3-methylglutaryl-CoA
Hyperthermia, malignant, 556, 564-565,
Hydroperoxides, formation of, 194, 195f
(HMG-CoA) reductase
565f, 569t
Hydrophilic compounds, hydroxylation
cholesterol synthesis controlled by, 220,
Hypertriacylglycerolemia
producing, 627
223f
in diabetes mellitus, 205
Hydrophilic portion of lipid molecule, 119,
in mevalonate synthesis, 219, 220f
familial, 228t
120f
3-Hydroxy-3-methylglutaryl-CoA
Hypertrophic cardiomyopathy, familial,
Hydrophobic effect, in lipid bilayer self-
(HMG-CoA) synthase
569-570, 570f
assembly, 418
in ketogenesis, 185, 185f
Hyperuricemia, 170-171, 300
Hydrophobic interaction chromatography,
in mevalonate synthesis, 219, 220f
Hypervariable regions, 591-592, 594f
for protein/peptide purification,
p-Hydroxyphenylpyruvate, in tyrosine
Hypoglycemia, 153
23
catabolism, 254f, 255
fatty acid oxidation and, 180, 187-188
Hydrophobic interactions, 6-7
17-Hydroxypregnenolone, 440, 441f
fructose-induced, 171-172
Hydrophobic portion of lipid molecule,
17-Hydroxyprogesterone, 440, 441f
insulin excess causing, 162
119, 120f
Hydroxyproline
during pregnancy and in neonate, 161
Hydrostatic pressure, 580
synthesis of, 240, 240f, 535-537
Hypoglycin, 180, 188
L(+)-3-Hydroxyacyl-CoA dehydrogenase,
tropoelastin hydroxylation and, 539
Hypokalemic periodic paralysis, 569t
181, 182f
4-Hydroxyproline, catabolism of, 253f,
Hypolipidemic drugs, 229
3-Hydroxyanthranilate dioxygenase/
255
Hypolipoproteinemia, 205, 228t, 229
oxygenase, 89
4-Hydroxyproline dehydrogenase, defect in,
Hypouricemia, 300
Hydroxyapatite, 549
in hyperhydroxyprolinemia,
Hypoxanthine, 289
D(-)-3-Hydroxybutyrate (β-hydroxy-
255
Hypoxanthine-guanine phosphoribosyl
butyrate), 183-184, 184f
15-Hydroxyprostaglandin dehydrogenase,
transferase (HRPT)
D(-)-3-Hydroxybutyrate dehydrogenase,
194
defect of in Lesch-Nyhan syndrome, 300
184, 184f
3β-Hydroxysteroid dehydrogenase, 438,
localization of gene for, 407t
24-Hydroxycalcidiol (24,25-dihydroxyvita-
441f, 442, 443f
Hypoxia, lactate production and, 136, 137f,
min D3), in vitamin D
17β-Hydroxysteroid dehydrogenase, 442,
139-140
metabolism, 484, 485f
443f
25-Hydroxycholecalciferol (calcidiol), in
5-Hydroxytryptamine. See Serotonin
vitamin D metabolism, 484, 485f
Hyperalphalipoproteinemia, familial, 228t
I. See Iodine/iodide
4-Hydroxydicoumarin (dicumarol), 486
Hyperammonemia, types 1 and 2, 247
I bands, 556, 557f, 558f
Hydroxylamine, for polypeptide cleavage,
Hyperargininemia, 248
I-cell disease, 431, 432t, 512t, 524, 530t,
26t
Hyperbilirubinemia, 281-284, 284t
531-532, 532t, 546-547, 546t
7α-Hydroxylase, bile acid synthesis
Hypercholesterolemia, 205
Ibuprofen, cyclooxygenases affected by, 193
regulated by, 226, 226f, 227
familial, 1, 228t, 432t
ICAM-1, 529, 529t
11β-Hydroxylase, in steroid synthesis, 440,
LDL receptor deficiency in, 209, 432t
ICAM-2, 529, 529t
441f
Hyperchromicity of denaturation, 304-305
ICF. See Intracellular fluid
668
/
INDEX
Icterus (jaundice), 270, 281-284, 284t
in gluconeogenesis regulation,
Insert/insertions, DNA, 413
IDDM. See Insulin-dependent diabetes
155-157
recombinant DNA technology in
mellitus
gratuitous, 378
detection of, 409
Idiotypes, 594
in regulation of gene expression, 376
Inside-outside asymmetry, membrane,
IDL. See Intermediate-density lipoproteins
Inducible gene, 376
419-420
L-Iduronate, 105, 106f
Infantile Refsum disease, 188, 503, 503t
Insulators, 387
IEF. See Isoelectric focusing
Infection
nonpolar lipids as, 111
IgA, 591, 594t, 595f
glycoprotein hydrolase deficiencies and,
Insulin, 107-109, 438, 449, 450f
IgD, 591, 594t
533
adipose tissue metabolism affected by,
IgE, 591, 594t
neutrophils in, 620, 621t
216-217
IGF-I. See Insulin-like growth factor-I
protein loss and, 480
in blood glucose regulation, 160-162
IgG, 591, 592f, 594t
respiratory burst in, 622-623
deficiency of, 161. See also Diabetes
deficiency of, 595
Inflammation, 190
mellitus
hypervariable regions of, 591-592,
acute phase proteins in, 583, 583t
free fatty acids affected by, 215
594f
complement in, 596
gene for, localization of, 407t
IgM, 591, 594t, 595f
neutrophils in, 620, 621t
glucagon opposing actions of, 160-161
Immune response, class/isotype switching
integrins and, 529t, 620-621, 622t
in glucose transport, 427
and, 594
selectins and, 528-529, 529t, 530f
in glycolysis, 137, 155-157
Immunoglobulin genes, 593
NF-κB in, 468
initiation of protein synthesis affected by,
DNA rearrangement and, 325-326, 393,
prostaglandins in, 190
367, 367f
593-594
selectins in, 528-530, 529f, 529t, 530f
in lipogenesis regulation, 178-179
double-strand break repair and,
Influenza virus
in lipolysis regulation, 178-179, 215,
337-338
hemagglutinin in, calnexin binding to,
216f
Immunoglobulin heavy chain binding
526
phosphorylase b affected by, 148
protein, 508
neuraminidase in, 533
receptor for, 436, 465, 466f
Immunoglobulin heavy chains, 591, 592f
Information pathway, 457, 459f
signal transmission by, 465-467, 466f
genes producing, 593
Inhibition
storage/secretion of, 453, 454t
Immunoglobulin light chains, 591, 592f
competitive versus noncompetitive,
synthesis of, 449, 450f
in amyloidosis, 590
67-69, 67f, 68f, 69f
Insulin-dependent diabetes mellitus
genes producing, 593
feedback, in allosteric regulation, 74-76,
(IDDM/type 1), 161-162. See
DNA rearrangement and, 325-326,
75f
also Diabetes mellitus
393, 593-594
irreversible, 69
Insulin/glucagon ratio, in ketogenesis
Immunoglobulins, 583t, 591-597, 593t.
Inhibitor-1, 148, 149f, 151, 151f
regulation, 187
See also specific type under Ig
Initial velocity, 64
Insulin-like growth factor I, receptor for, 436
class switching and, 594
inhibitors affecting, 68, 68f, 69f
Insulin resistance, 611
classes of, 591, 593t, 594t
Initiation
Integral proteins, 30, 420, 421f
diseases caused by over- and
in DNA synthesis, 328-330, 329f, 330f,
as receptors, 431
underproduction of, 594-595
331f
red cell membrane, 615-616, 615f, 616f,
functions of, 593, 594t
in protein synthesis, 365-367, 366f
616t
genes for. See Immunoglobulin genes
in RNA synthesis, 342, 342f, 343-344
Integration, chromosomal, 324, 324f
hybridomas as sources of, 595-596, 596f
Initiation complexes, in protein synthesis,
Integrins, neutrophil interactions and, 529t,
structure of, 591, 592, 592f, 594f, 595f
365, 366f, 367
620-621, 622t
IMP (inosine monophosphate)
Initiator sequence, 346-348, 347f
Intercellular junctions, 431
conversion of to AMP and GMP, 293,
Inner mitochondrial membrane, 92, 93f
Intermediate-density lipoproteins, 206t
296f
impermeability of, exchange transporters
Intermediate filaments, 577-578, 577t
feedback regulation of, 294, 296f
and, 98-100, 98f, 99f
Intermembrane space, proteins in, 501
synthesis of, 293-294, 295f, 296f, 297f
protein insertion in, 501
Intermittent branched-chain ketonuria, 259
Importins, 501, 502f
Inorganic pyrophosphatase, in fatty acid
Internal presequences, 501
In situ hybridization/fluorescence in situ
activation, 85
Internal ribosomal entry site, 371, 371f
hybridization, in gene mapping,
Inosine monophosphate (IMP)
Interphase chromosomes, chromatin fibers
406-407, 407t, 635t
conversion of to AMP and GMP, 293,
in, 316
Inactive chromatin, 316-318, 383
296f
Intervening sequences. See Introns
Inborn errors of metabolism, 1, 249, 545
feedback-regulation of, 294, 296f
Intestinal bacteria, in bilirubin conjugation,
Inclusion cell (I-cell) disease, 431, 432t,
synthesis of, 293-294, 295f, 296f, 297f
281
512t, 524, 530t, 531-532, 532t
Inositol hexaphosphate (phytic acid), calcium
Intracellular environment, membranes in
Indole, permeability coefficient of, 419f
absorption affected by, 477
maintenance of, 415-416, 416t
Indomethacin, cyclooxygenases affected by,
Inositol trisphosphate, 464-465, 464f, 465f
Intracellular fluid (ICF), 415, 416, 416t
193
in platelet activation, 606, 606f, 607
Intracellular membranes, 415
Induced fit model, 52, 53f
in respiratory burst, 623
Intracellular messengers, 457-468, 461t,
Inducers
Inotropic effects, 566
463t. See also specific type and
enzyme synthesis affected by, 74
Inr. See Initiator sequence
Second messengers
INDEX
/
669
Intracellular signals, 457-458
metabolism of, 585, 585f
J chain, 595f
Intracellular traffic, 498-513. See also
disorders of, 586, 587t
Jackson-Weiss syndrome, 551t
Protein sorting
nonheme, 92, 95f, 585
JAK kinases, 436, 467, 467f
disorders due to mutations in genes
transferrin in transport of, 584-586,
Jak-STAT pathway, 436, 467, 467f
encoding, 512t, 513
585f, 585t
Jamaican vomiting sickness, 188
Intrinsic factor, 477, 491-492
Iron-binding capacity, total, 586
Jaundice (icterus), 270, 281-284, 284t
in pernicious anemia, 492
Iron deficiency/iron deficiency anemia, 478,
Joining region, gene for, 593
Intrinsic pathway of blood coagulation,
497, 586
DNA rearrangement and, 393, 593-594
598, 599f, 600-601
Iron porphyrins, 270
“Jumping DNA,” 325
Introns (intervening sequences), 319,
Iron regulatory protein, 585
Junctional diversity, 593-594
352-354, 353f, 358, 413
Iron response elements, 586
Juxtaglomerular cells, in renin-angiotensin
in recombinant DNA technology, 397,
Iron-sulfur protein complex, 92, 95f
system, 451
398f
Irreversible covalent modifications, 76-77,
removal of from primary transcript,
77f
352-354, 353f
Irreversible inhibition, enzyme, 69
K. See Dissociation constant
Inulin, glomerular membrane permeability
IRS 1-4, in insulin signal transmission,
K. See Potassium
to, 540
465, 466f
k. See Rate constant
“Invert sugar,” 107
Ischemia, 136, 431
Kd. See Dissociation constant
Iodine/iodide, 496t
Islets of Langerhans, insulin produced by,
kdeg. See Rate of degradation
deficiency of, 447-449
160
Keq. See Equilibrium constant
in thyroid hormone synthesis, 447, 448f,
Isocitrate dehydrogenase, 130-131, 132f
Km. See Michaelis constant
449
in NADPH production, 176, 176f
ks. See Rate of synthesis
5-Iodo-2′-deoxyuridine, 291f
Isoelectric focusing, for protein/peptide
Kw. See Ion product
Iodopsin, 483
purification, 24, 25f
Kappa (κ) chains, 591
o-Iodosobenzene, for polypeptide cleavage,
Isoelectric pH (pI), amino acid net charge
Kartagener syndrome, 577
26t
and, 17
Karyotype, 320f
Iodothyronyl residues, 447. See also
Isoenzymes. See Isozymes
Kayser-Fleischer ring, 588
Thyroxine; Triiodothyronine
Isoleucine, 15t
KDEL-containing proteins, 506-507,
5-Iodouracil, 290
catabolism of, 259, 260f, 261f
508t
Ion channels, 415, 423-424, 425f, 426t,
interconversion of, 240
Keratan sulfates, 544f, 544t, 545
568t
requirements for, 480
functions of, 547
in cardiac muscle, 566-567, 568, 568t
∆5,4-Isomerase, 438, 441f, 442, 443f
Keratins, 577t, 578
diseases associated with disorders of, 568,
Isomerases, 50
Kernicterus, 282, 283
569t
in steroid synthesis, 438, 441f, 442,
α-Keto acid decarboxylase, defect/absence
Ion exchange chromatography, for protein/
443f
of, in maple syrup urine disease
peptide purification, 22-23
Isomerism
(branched-chain ketonuria), 259
Ion product, 8-9
geometric, of unsaturated fatty acids,
α-Keto acid dehydrogenase,
Ion transport, in mitochondria, 99
112-114, 114f
branched-chain, 259
Ionizing radiation, nucleotide excision-
of steroids, 117, 118f
thiamin diphosphate as coenzyme for,
repair of DNA damage caused
of sugars, 102-104, 103f
488-489
by, 337
Isoniazid, acetylation of, 630
α-Keto acids
Ionophores, 99, 424
Isopentenyl diphosphate, in cholesterol
amino acids in diet replaced by, 240
IP3. See Inositol trisphosphate
synthesis, 219, 221f
oxidative carboxylation of, 259, 260f,
IPTG. See Isopropylthiogalactoside
Isoprene units, polyprenoids synthesized
261f, 262f
IREG1. See Iron regulatory protein
from, 118, 119f
Ketoacidosis, 180, 188-189
IRES. See Internal ribosomal entry site
Isoprenoids, synthesis of, in cholesterol
3-Ketoacyl-CoA thiolase deficiency, 188
Iron, 496t
synthesis, 219, 221f, 222f
3-Ketoacyl synthase, 173, 175f
absorption of, 478, 584-586, 585f,
Isopropylthiogalactoside, 378
Ketogenesis, 125-126, 126f, 183-187
585t
Isoprostanes (prostanoids), 112, 119
high rates of fatty acid oxidation and,
in hemochromatosis, 478
cyclooxygenase pathway in synthesis of,
183-186, 184f
vitamin C and ethanol affecting, 478,
192, 192-194, 193f, 194f
HMG-CoA in, 184-185, 185f
496
Isothermic systems, biologic systems as,
regulation of, 186-187, 187f, 188f
deficiency of, 497
80
Ketogenic amino acids, 232
distribution of, 585t
Isotopes. See also specific type
α-Ketoglutarate, 131
ferrous, in oxygen transport, 40-41
in plasma protein analysis, 581
in amino acid carbon skeleton
heme, 278, 585
Isotype (class) switching, 594
catabolism, 249, 250, 250f,
absorption of, 478, 585, 585f
Isotypes, 594
251f
hindered environment for, 41, 41f
Isovaleric acidemia, 259, 259-262
in glutamate synthesis, 237, 238f, 243,
in methemoglobinemia, 46
Isovaleryl-CoA dehydrogenase, in isovaleric
243f, 244f
incorporation of into protoporphyrin,
acidemia, 259-262
transporter systems for, 99
271-272, 272f
Isozymes, 54-55
in urea synthesis, 244, 244f
670
/
INDEX
α-Ketoglutarate dehydrogenase complex,
Kynurenine-anthranilate pathway, for
Lauric acid, 112t
131, 132f
tryptophan catabolism, 257f, 258
Laws of thermodynamics, 80-81
regulation of, 135
Kynurenine formylase, 257f, 258
hydrophobic interactions and, 7
thiamin diphosphate as coenzyme for,
LBD. See Ligand-binding domain
488-489
LCAT. See Lecithin:cholesterol
Ketone bodies, 124, 125-126, 126f, 180,
L-α-amino acids, 14. See also Amino acids
acyltransferase
183-184, 184f
genetic code specifying, 14, 15-16t
LCRs. See Locus control regions
free fatty acids as precursors of, 186
in proteins, 14
LDH. See Lactate dehydrogenase isozymes
as fuel for extrahepatic tissues, 185-186,
L chains. See Light chains
LDL. See Low-density lipoproteins
186f
L-Dopa, 446, 447f
LDL:HDL cholesterol ratio, 227
in starvation, 232-234, 234f, 234t
L isomerism, 102-104, 103f
Lead poisoning, ALA dehydratase inhibition
Ketonemia, 186, 188
L-selectin, 529f, 529t
and, 270, 278
Ketonuria, 188
L-type calcium channel, 567
Leader sequence. See Signal peptide
branched chain (maple syrup urine
Labile factor (factor V), 600t, 601, 602f
Leading (forward) strand, in DNA
disease), 259
lacA gene, 376, 376f, 377f, 378
replication, 327f, 328, 330
Ketoses (sugars), 102, 102t
lacI gene, 377, 377f, 378
Lecithin:cholesterol acyltransferase (LCAT),
Ketosis, 180, 186, 188
lac operon, 375, 376-378, 376f, 377f
200-201, 209-210, 211f, 223,
in cattle
lac repressor, 377, 377f
224
fatty liver and, 212
lacY gene, 376, 376f, 377f, 378
familial deficiency of, 228t
lactation and, 188
lacZ gene, 376, 376f, 377f, 378
Lecithins (phosphatidylcholines), 114-115,
in diabetes mellitus, 188
Lactase, 475
115f
ketoacidosis caused by, 188-189
deficiency of (lactose/milk intolerance),
in cytochrome P450 system, 617
nonpathologic, 188-189
102, 474, 475
membrane asymmetry and, 420
in starvation, 188
Lactate
synthesis of, 197, 197f, 198f
Kidney
anaerobic glycolysis and, 136, 137f,
Lectins, 110, 517-518, 518t
glycogenolysis in, 147
139-140
in glycoprotein analysis, 515t, 517-518,
metabolism in, 235t
hypoxia and, 137f, 139-140
518t
in renin-angiotensin system, 451
Lactate dehydrogenase, in anaerobic
Leiden factor V, 603
vitamin D3 synthesis in, 445, 446f, 484
glycolysis, 139
Lens of eye, fructose and sorbitol in,
Kinases, protein. See Protein kinases
Lactate dehydrogenase isozymes, 57, 139
diabetic cataract and, 172
Kinesin, 577
diagnostic significance of, 57, 57t, 58f
Leptin, 215-216
Kinetic (collision) theory, 61
Lactic acid, pK/pKa value of, 12t
Lesch-Nyhan syndrome, 300
Kinetics (enzyme), 60-71. See also Catalysis
Lactic acid cycle, 159, 159f
Leucine, 15t
activation energy affecting, 61, 63
Lactic acidosis, 92, 136
catabolism of, 259, 260f, 261f
balanced equations and, 60
with mitochondrial encephalopathy and
interconversion of, 240
competitive versus noncompetitive inhi-
stroke-like episodes (MELAS),
requirements for, 480
bition and, 67-69, 67f, 68f, 69f
100-101
Leucine aminomutase, 492
factors affecting reaction rate and,
pyruvate metabolism and, 142-143
Leucine zipper motif, 387-388, 388t, 390,
61-63, 62f, 63-64, 64f
thiamin deficiency and, 489
391f
free energy changes affecting, 60-61
Lactoferrin, 621t
Leucovorin, 493
initial velocity and, 64
Lactogenic hormone. See Prolactin
Leukocyte adhesion deficiency
multisubstrate enzymes and, 69-70, 69f,
Lactose, 106-107, 107f, 107t
type I, 621
70f
galactose in synthesis of, 167-169, 170f
type II, 530t, 531
saturation, 64f, 66
metabolism of, operon hypothesis and,
Leukocytes, 620-624
sigmoid (Hill equation), 66-67, 67f
376-378, 376f, 377f
growth factors regulating production of,
substrate concentration and, 64, 64f, 65f
Lactose (milk) intolerance, 102, 474, 475
610
models of effects of, 65-67, 66f, 67f
Lactose synthase, 167, 170f
recombinant DNA technology in study
transition states and, 61
Lagging (retrograde) strand, in DNA
of, 624
Kinetochore, 318
replication, 327f, 328, 330-331
Leukodystrophy, metachromatic, 203t
Kininogen, high-molecular-weight, 599f,
Lambda (λ) chains, 591
Leukotriene A4, 114f
600
Lambda (λ) phage, 378-383, 379f, 380f,
Leukotrienes, 112, 114f, 190, 192
Kinky hair disease (Menkes disease), 588
381f, 382f
clinical significance of, 196
Knockout genes, 412
Lambda repressor (cI) protein/gene,
lipoxygenase pathway in formation of,
Korsakoff’s psychosis, 489
379-383, 380f, 381f, 382f
192, 193f, 194, 195f
Kozak consensus sequences, 365
Laminin, 535, 540-542, 541f
LFA-1, 529, 529t, 620, 622t
Krabbe’s disease, 203t
Lamins, 577t, 578
LH. See Luteinizing hormone
Krebs cycle. See Citric acid cycle
Langerhans, islets of, insulin produced by,
Library, 402, 413
Ku, in double-strand break repair, 338,
160
Lifestyle changes, cholesterol levels affected
338f
Lanosterol, in cholesterol synthesis, 219,
by, 227-228
Kwashiorkor, 237, 478, 478-479
220, 222f
Ligand-binding domain, 470
Kynureinase, 257f, 258
Latch state, 571
Ligand-gated channels, 424, 568t
INDEX
/
671
Ligand-receptor complex, in signal
disorders associated with abnormalities
remnant, 206t, 208, 209f
generation, 456-457
of, 431
liver uptake of, 208-209
Ligases, 50
fatty acids, 111-114
Liposomes, 421
DNA, 328t, 330, 332, 332f
glycolipids, 111, 116, 117f
amphipathic lipids forming, 119-121,
Ligation, 413
interconvertibility of, 231
120f
in RNA processing, 352
in membranes, 416-418
artificial membranes and, 421
Light, in active transport, 427
ratio of to protein, 416, 416f
Lipotropic factor, 212
Light chains
metabolism of, 122f, 123-124, 123f,
β-Lipotropin, 453, 453f
immunoglobulin, 591, 592f
125-126, 126f. See also Lipolysis
Lipoxins, 112, 114f, 190, 192
in amyloidosis, 590
in fed state, 232
clinical significance of, 196
genes producing, 593
in liver, 211-212, 213f
lipoxygenase pathway in formation of,
DNA rearrangement and, 325-326,
neutral, 111
192, 193f, 194, 195f
393, 593-594
peroxidation of, 118-119, 120f
Lipoxygenase, 119, 194, 195f
myosin, 560
phospholipids, 111, 114-116, 115f
reactive species produced by, 119
in smooth muscle contraction, 570
precursor, 111
5-Lipoxygenase, 194, 195f
Light meromyosin, 560-561, 560f
simple, 111
Lipoxygenase pathway, 192, 193f, 194,
Limit dextrinosis, 152t
steroids, 117-118, 117f, 118f, 119f
195f
Lines, definition of, 413
transport and storage of, 205-218
Liquid chromatography, high-performance
LINEs. See Long interspersed repeat
adipose tissue and, 214-215, 214f
reversed-phase, for peptide
sequences
brown adipose tissue and, 217, 217f
separation, 23-24
Lineweaver-Burk plot
clinical aspects of, 212-214
Lithium, 496t
inhibitor evaluation and, 68, 68f, 69f
fatty acid deficiency and, 194-195
Lithocholic acid, synthesis of, 226f
Km and Vmax estimated from, 66, 66f
as lipoproteins, 205-206, 206t, 207f
Liver
Lingual lipase, 475
liver in, 211-212, 213f
angiotensinogen made in, 451
Link trisaccharide, in glycosaminoglycan
triacylglycerols (triglycerides), 114, 115f
bilirubin uptake by, 280-281, 280f,
synthesis, 543
turnover of, membranes and, 511-512
281f, 282f
Linkage analysis, 635t
Lipogenesis, 125, 173-177, 174f, 175f
cirrhosis of, 130, 212
in glycoprotein study, 515t
acetyl-CoA for, 176-177
in α1-antitrypsin deficiency, 590
Linoleic acid/linoleate, 113t, 190, 190f,
fatty acid synthase complex in, 173-176,
cytochrome P450 isoforms in, 627
192
174f, 175f
disorders of, in α1-antitrypsin deficiency,
in essential fatty acid deficiency, 191
malonyl-CoA production in, 173, 174f
589-590, 590f
synthesis of, 191f
NADPH for, 175f, 176, 176f
fatty
α-Linolenic acid/α-linolenate, 113t, 190,
regulation of, 178-179, 178f
alcoholism and, 212-214
190f, 192
enzymes in, 156t, 173, 174f, 178,
of pregnancy, 188
in essential fatty acid deficiency, 191
178f
triacylglycerol metabolism imbalance
synthesis of, 191, 191f
nutritional state in, 177-178
and, 212
γ-Linolenic acid/γ-linolenate, 113t
Lipolysis, 125, 126f, 216-217, 216f. See
fructose overload and, 170-171
in essential fatty acid deficiency, 191
also Lipids, metabolism of
glycogen in, 145, 146t
in polyunsaturated fatty acid synthesis,
hormone-sensitive lipase in, 214-215,
heme synthesis in, 272
191, 192f
214f
ALA synthase in regulation of,
Lipases
hormones affecting, 215-216, 216f
272-273, 276f
diagnostic significance of, 57t
insulin affecting, 178-179
ketone bodies produced by, 183-184,
in digestion, 475, 476f
triacylglycerol, 197
184f, 186
in triacylglycerol metabolism, 197,
Lipophilic compounds, cytochrome P450
metabolism in, 124-125, 125f, 126f,
214-215, 214f, 475, 476f
isoforms in hydroxylation of, 627
130, 235t
Lipid bilayer, 418-419, 418f, 419f
Lipoprotein lipase, 125, 126f, 207-208,
fatty acid oxidation, ketogenesis and,
membrane proteins and, 419
209f, 210f
183-186, 184f
Lipid core, of lipoprotein, 205
familial deficiency of, 228t
fructose, 167, 169f
Lipid rafts, 422
involvement in remnant uptake, 208,
glucose, 154f, 159, 159-160,
Lipid storage disorders (lipidoses),
209f
159f
202-203, 203t
α1-Lipoprotein, 581f
fructose 2,6-bisphosphate in
Lipids, 111-121. See also specific type
β1-Lipoprotein, 581f
regulation of, 157-158, 158f
amphipathic, 119-121, 120f
Lipoprotein(a) excess, familial, 228t
glycogen, 145-147, 145, 146f, 148
asymmetry of, membrane assembly and,
Lipoproteins, 30, 111, 125, 205-206, 206t,
lipid, 211-212, 213f
511, 512f
207f, 580, 583t. See also specific
plasma protein synthesis in, 125, 581
classification of, 111
type
vitamin D3 synthesis in, 445, 446f, 484,
complex, 111
carbohydrates in, 110
485f
in cytochrome P450 system, 627
in cholesterol transport, 223-224, 225f
Liver phosphorylase, 147
derived, 111
classification of, 205, 206t
deficiency of, 152t
digestion and absorption of, 475-477,
deficiency of, fatty liver and, 212
LMM. See Light meromyosin
476f
disorders of, 228t, 229
Lock and key model, 52
672
/
INDEX
Locus control regions, 387
Lysosomal enzymes, 623
D-Mannose, 104f, 105t
Long interspersed repeat sequences
in I-cell disease, 431, 432t, 531-532,
α-D-Mannose, 104f
(LINEs), 321-322
532f
Mannose-binding protein, deficiency of, 533
Looped domains, chromatin, 316, 318,
Lysosomal hydrolases, deficiencies of,
Mannose 6-phosphate/mannose 6-P signal,
319f
532-533, 533t
526
Loops (protein conformation), 32-33
Lysosomes
in I-cell disease, 531, 532, 532f
Loose connective tissue, keratan sulfate I in,
in oligosaccharide processing, 524
in protein flow, 507, 508t
545
protein entry into, 507, 507f, 508t
Mannosidosis, 532-533, 533t
Low-density lipoprotein receptor-related
disorders associated with defects in,
MAP (mitogen-activated protein) kinase
protein, 206
512t, 513
in insulin signal transmission, 466f, 467
in chylomicron remnant uptake,
Lysozyme, 621t
in Jak/STAT pathway, 467
208-209, 209f
Lysyl hydroxylase
Maple syrup urine disease (branched-chain
Low-density lipoproteins, 205, 206t
diseases caused by deficiency of, 538t
ketonuria), 259
apolipoproteins of, 206, 206t
in hydroxylysine synthesis, 240, 537
Marasmus, 80, 237, 478, 478-479
metabolism of, 209, 210f
Lysyl oxidase, 537, 539
Marble bone disease (osteopetrosis), 552
ratio of to high-density lipoproteins,
Lytic pathway, 379, 379f
Marfan syndrome, fibrillin mutations
atherosclerosis and, 227
D-Lyxose, 104f, 105t
causing, 539-540, 540f
receptors for, 209
Maroteaux-Lamy syndrome, 546t
in chylomicron remnant uptake,
Mass spectrometry, 27, 27f
208-209, 209f
Mac-1, 529, 529t
covalent modifications detected by, 27,
in cotranslational insertion, 505-506,
α2-Macroglobulin, 583t, 590, 624
27f, 27t
506f
antithrombin activity of, 603
for glycoprotein analysis, 514, 515t
regulation of, 223
Macromolecules, cellular transport of,
tandem, 27
Low-energy phosphates, 83
428-431, 429f, 430f
transcript-protein profiling and, 412
β-LPH. See β-Lipotropin
Mad cow disease (bovine spongiform
Mast cells, heparin in, 545
LRP. See Low-density lipoprotein
encephalopathy), 37
Matrix
receptor-related protein
Magnesium, 496t
extracellular, 535-555. See also specific
L-tryptophan dioxygenase (tryptophan
in chlorophyll, 270
component
pyrrolase), 89
in extracellular and intracellular fluid,
mitochondrial, 92, 93f, 130
LTs. See Leukotrienes
416, 416t
Matrix-assisted laser-desorption (MALDI),
Lung surfactant, 115, 197
Major groove, in DNA, 305f, 306
in mass spectrometry, 27
deficiency of, 115, 202
operon model and, 378
Matrix-processing peptidase, 499
Luteinizing hormone (LH), 437, 438, 439f
Malate, 132f, 133
Matrix proteins, 499
LXs. See Lipoxins
Malate dehydrogenase, 132f, 133
diseases caused by defects in import of,
LXXLL motifs, nuclear receptor
Malate shuttle, 99, 100f
503
coregulators, 473
MALDI. See Matrix-assisted laser-desorption
Maxam and Gilbert’s method, for DNA
Lyases, 50
Maleylacetoacetate, in tyrosine catabolism,
sequencing, 404-405
in steroid synthesis, 440-442, 441f,
254f, 255
Maximal velocity (Vmax)
443f
Malic enzyme, 156t, 157
allosteric effects on, 75-76
Lymphocyte homing, selectins in, 528-530,
in NADPH production, 176, 176f
inhibitors affecting, 68, 68f, 69f
529f, 529t, 530f
Malignancy/malignant cells. See
Michaelis-Menten equation in
Lymphocytes. See also B lymphocytes;
Cancer/cancer cells
determination of, 65-66, 66f
T lymphocytes
Malignant hyperthermia, 556, 564-565,
substrate concentration and, 64, 64f
recombinant DNA technology in study
565f, 569t
McArdle’s disease/syndrome, 152t, 573
of, 624
Malonate
Mechanically gated ion channels, 568t
Lysine, 16t
respiratory chain affected by, 95, 96f
Mediator-related proteins, 472t, 473
catabolism of, 256f, 258
succinate dehydrogenase inhibition by,
Medicine
pI of, 17
67-68, 67f
preventive, biochemical research
requirements for, 480
Malonyl-CoA, in fatty acid synthesis, 173,
affecting, 2
Lysine hydroxylase, vitamin C as coenzyme
174f
relationship of to biochemistry, 1-4, 3f
for, 496
Malonyl transacylase, 173, 174f, 175f
Medium-chain acyl-CoA dehydrogenase,
Lysis, cell, complement in, 596
Maltase, 475
deficiency of, 188
Lysogenic pathway, 379, 379f
Maltose, 106-107, 107f, 107t
Megaloblastic anemia
Lysolecithin (lysophosphatidylcholine),
Mammalian target of rapamycin (mTOR),
folate deficiency causing, 482t, 492, 610t
116, 116f
in insulin signal transmission,
vitamin B12 deficiency causing, 482t,
metabolism of, 200-201, 201f
466f, 467
492, 494, 610t
Lysophosphatidylcholine. See Lysolecithin
Mammotropin. See Prolactin
Melanocyte-stimulating hormone (MSH),
Lysophospholipase, 200, 201f
Manganese, 496t
453, 453f
Lysophospholipids, 116, 116f
Mannosamine, 169, 171f
MELAS (mitochondrial encephalomyopa-
Lysosomal degradation pathway, defect in
D-Mannosamine, 106
thy with lactic acidosis and
in lipidoses, 203
Mannose, in glycoproteins, 516t
stroke-like episodes), 100-101
INDEX
/
673
Melting point, of amino acids, 18
6-Mercaptopurine, 290, 291f
control of quantity and, 73-74
Melting temperature/transition
Mercapturic acid, 629
covalent modification and, 74, 76,
temperature, 305, 422
Mercuric ions, pyruvate metabolism
77-78, 78f
Membrane attack complex, 596
affected by, 142
rate-limiting reactions and, 73
Membrane fatty acid-transport protein, 207
Meromyosin
at subcellular level, 126, 127f
Membrane proteins, 419, 420t, 514. See
heavy, 560f, 561, 561f
at tissue and organ levels, 124-126, 125f,
also Glycoproteins
light, 560-561, 560f
126f, 235t
association of with lipid bilayer, 419
Messenger RNA (mRNA), 307, 309-310,
of xenobiotics, 626-632
flow of, 507, 507f, 508t
310f, 311f, 341, 342t, 359. See
Metachromatic leukodystrophy, 203t
integral, 30, 420, 421f
also RNA
Metal-activated enzymes, 50
mutations affecting, diseases caused by,
alternative splicing and, 354, 354f,
Metal ions, in enzymatic reactions, 50
431-432, 432f, 432t
393-394, 636
Metalloenzymes, 50
peripheral, 420-421, 421f
codon assignments in, 358, 359t
Metalloflavoproteins, 86-87
red cell, 614-617, 615f, 616f, 616t
editing of, 356
Metalloproteins, 30
structure of, dynamic, 419
expression of, detection of in gene
Metallothioneins, 588
Membrane transport, 423, 423t, 424f,
isolation, 635t
Metaphase chromosomes, 317f, 318, 319t
426-431, 426f. See also specific
modification of, 355-356
Metastasis
mechanism
nucleotide sequence of, 358
glycoproteins and, 514, 526, 530t, 531
Membranes, 415-433
mutations caused by changes in,
membrane abnormalities and, 432t
artificial, 421-422
361-363, 361f, 362f, 364f
Methacrylyl-CoA, catabolism of, 262f
assembly of, 511-513, 512f, 512t
polycistronic, 376
Methemoglobin, 46, 363, 613-614
asymmetry of, 416, 419-420
recombinant DNA technology and, 397
Methemoglobinemia, 46, 614
bilayers of, 418-419, 418f, 419f
relationship of to chromosomal DNA,
Methionine, 15t, 264, 266f
membrane protein association and, 419
321f
active (S-adenosylmethionine), 258f,
biogenesis of, 511-513, 512f, 512t
stability of, regulation of gene expression
259, 264, 266f, 289, 290f, 290t
cholesterol in, 417
and, 394-395, 394f
catabolism of, 258f, 259, 259f
fluid mosaic model and, 422
transcription starting point and, 342
requirements for, 480
depolarization of, in nerve impulse
variations in size/complexity of, 397,
Methionine synthase, 492, 494
transmission, 428
399t
Methotrexate, 296-297, 494
function of, 415-416, 421-422
Metabolic acidosis, ammonia in, 245
dihydrofolate/dihydrofolate reductase
fluidity affecting, 422
Metabolic alkalosis, ammonia in, 245
affected by, 296-297, 494
glycosphingolipids in, 417
Metabolic fuels, 231-236. See also Digestion
Methylation
Golgi apparatus in synthesis of, 509
clinical aspects of, 236
in covalent modification, mass increases
intracellular, 415
diet providing, 474, 478
and, 27t
lipids in, 416-418
in fed and starving states, 232-234,
of deoxycytidine residues, gene
amphipathic, 119, 120f, 417-418, 417f
233f, 234f, 234t
expression affected by, 383
mutations affecting, diseases caused by,
interconvertability of, 231-232
in glycoprotein analysis, 515t
431-432, 432f, 432t
Metabolic pathway/metabolite flow, 122,
of xenobiotics, 626, 630
phospholipids in, 114-116, 115f,
122-124. See also specific type
β-Methylcrotonyl-CoA, catabolism of, 261f
416-417, 417f
and Metabolism
5-Methylcytosine, 287, 289f
plasma. See Plasma membrane
flux-generating reactions in, 129
α-Methyldopa, 446
protein:lipid ratio in, 416, 416f
nonequilibrium reactions in, 128-129
Methylene tetrahydrofolate, 493, 493f
proteins in, 419, 420t. See also
regulation of, 72, 73f, 126-129, 128f
in folate trap, 493f, 494
Membrane proteins
covalent modification in, 79
7-Methylguanine, 289f
red cell, 614-617, 615f, 615t, 616f, 616t
unidirectional nature of, 72, 73f
Methylhistidine, 576
hemolytic anemias and, 619, 620t
Metabolism, 81, 122-129, 235t. See also
in Wilson’s disease, 265
selectivity of, 415, 423-426, 423t, 424f,
specific type and Catalysis;
Methylmalonic aciduria, 155
425f, 426t
Metabolic pathway
Methylmalonyl-CoA, accumulation of in
sterols in, 417
blood circulation and, 124-126, 125f,
vitamin B12 deficiency, 492
structure of, 416-421, 416f
126f
Methylmalonyl-CoA isomerase (mutase), in
asymmetry and, 416, 419-420
group transfer reactions in, 8
propionate metabolism, 155,
fluid mosaic model of, 421f, 422
inborn errors of, 1, 249
155f, 492
Menadiol, 486, 487f
integration of, metabolic fuels and,
Methylmalonyl-CoA mutase (isomerase),
Menadiol diacetate, 486, 488f
231-236
155, 155f, 492
Menadione, 486. See also Vitamin K
regulation of, 72, 73f, 126-129, 128f
Methylmalonyl-CoA racemase, in propi-
Menaquinone, 482t, 486, 488f. See also
allosteric and hormonal mechanisms
onate metabolism, 155, 155f
Vitamin K
in, 74, 74-76, 75f, 128f, 129
Methyl pentose, in glycoproteins, 109t
Menkes disease, 588
enzymes in, 126-129, 128f
Methyl-tetrahydrofolate, in folate trap,
MEOS. See Cytochrome P450-dependent
allosteric regulation and, 74,
493f, 494
microsomal ethanol oxidizing
74-76, 75f, 128f, 129
Mevalonate, synthesis of, in cholesterol
system
compartmentation and, 72-73
synthesis, 219, 220f, 221f, 222f
674
/
INDEX
Mg. See Magnesium
Mitochondrial encephalomyopathies, with
Monounsaturated fatty acids, 112, 113t. See
Micelles, 418, 418f
lactic acidosis and stroke-like
also Fatty acids; Unsaturated fatty
amphipathic lipids forming, 119, 120f,
episodes (MELAS), 100
acids
418, 418f
Mitochondrial genome, 499
dietary, cholesterol levels affected by, 227
in lipid absorption, 475
Mitochondrial glycerol-3-phosphate
synthesis of, 191, 191f
Michaelis constant (Km), 65
dehydrogenase, 87
Morquio syndrome, 546t
allosteric effects on, 75-76
Mitochondrial membrane proteins, muta-
MPP. See Matrix-processing peptidase
binding constant approximated by, 66
tions of, 431
MPS. See Mucopolysaccharidoses
enzymatic catalysis rate and, 65-66, 66f,
Mitochondrial membranes, 92, 93f
MRE. See Mineralocorticoid response
72, 73f
enzymes as markers and, 92
element
inhibitors affecting, 68, 69f
exchange transporters and, 98-100, 98f,
mRNA. See Messenger RNA
Michaelis-Menten equation in
99f
MRP2. See Multidrug resistance-like
determination of, 65-66, 66f
protein insertion in, 501
protein 2
Michaelis-Menten equation, 65
Mitochondrial myopathies, fatal infantile,
MSH. See Melanocyte-stimulating hormone
Bi-Bi reactions and, 70, 70f
and renal dysfunction, oxidore-
MstII, 399t
regulation of metabolite flow and, 72,
ductase deficiency causing, 100
in sickle cell disease, 409, 410f
73f
Mitogen-activated protein (MAP) kinase
mtDNA. See Mitochondrial DNA
Microfilaments, 576-577
in insulin signal transmission, 466f,
mTOR, in insulin signal transmission,
α2-Microglobulin, 583t
467
466f, 467
Microsatellite instability, 322
in Jak/STAT pathway, 467
Mucins, 519-520, 520t
Microsatellite polymorphism, 322, 411,
Mitotic spindle, microtubules in formation
genes for, 520
413
of, 577
O-glycosidic linkages in, 518, 519-520,
Microsatellite repeat sequences, 322, 413
Mixed-function oxidases, 89-90, 627. See
519f
Microsomal elongase system, 177, 177f
also Cytochrome P450 system
repeating amino acid sequences in, 519,
Microsomal fraction, cytochrome P450
ML. See Mucolipidoses
520f
isoforms in, 627
MOAT. See Multispecific organic anion
Mucolipidoses, 546-547, 546t
Microtubules, 577
transporter
Mucopolysaccharides, 109, 109f
Migration, cell, fibronectin in, 540
Modeling, molecular, in protein structure
Mucopolysaccharidoses, 545-547, 546t,
Milk (lactose) intolerance, 102, 474, 475
analysis, 36
547f
Mineralocorticoid response element, 459t
Molecular biology, 1. See also Recombinant
Mucoproteins. See Glycoproteins
Mineralocorticoids, 437
DNA/recombinant DNA
Mucus, 519-520
receptor for, 471
technology
Multidrug resistance-like protein 2, in
synthesis of, 438-440, 441f
in primary structure determination,
bilirubin secretion, 280
Minerals, 2, 496-497, 496t
25-26
Multipass membrane protein, anion
digestion and absorption of, 477-478
Molecular chaperones. See Chaperones
exchange protein as, 615, 615f,
Minor groove, in DNA, 305f, 306
Molecular genetics, 1, 396. See also
616t
Mismatch repair of DNA, 336, 336f, 336t
Recombinant DNA/recombinant
Multiple myeloma, 595
colon cancer and, 336
DNA technology
Multiple sclerosis, 202
Missense mutations, 361, 362-363, 362f
Molecular modeling, in protein structure
Multiple sulfatase deficiency, 203
familial hypertrophic cardiomyopathy
analysis, 36
Multisite phosphorylation, in glycogen
caused by, 569-570, 570f
Molecular motors, 577
metabolism, 151
MIT. See Monoiodotyrosine
Molybdenum, 496t
Multispecific organic anion transporter, in
Mitchell’s chemiosmotic theory. See
Monoacylglycerol acyltransferase, 198f, 199
bilirubin secretion, 280
Chemiosmotic theory
Monoacylglycerol pathway, 198f, 199,
Muscle, 556-576, 557f. See also Cardiac
Mitochondria
475-477, 476f
muscle; Skeletal muscle
ALA synthesis in, 270, 273f
2-Monoacylglycerols, 198f, 199
ATP in, 556, 561-562, 573-574, 575f
citric acid cycle in, 122, 122f, 123f, 124f,
Monoclonal antibodies, hybridomas in
contraction of. See Muscle contraction
126, 127f, 130, 133-135, 134f
production of, 595-596, 596f
in energy transduction, 556-559, 557f,
fatty acid oxidation in, 180-181, 181f
Monoglycosylated core structure, calnexin
558f, 559f
ion transport in, 99
binding and, 526
fibers in, 556
protein synthesis and import by,
Monoiodotyrosine (MIT), 447, 448f, 449
glycogen in, 145, 146t
499-501, 501t
Monomeric proteins, 34
metabolism in, 125, 125f, 235t, 576t
respiration rate of, ADP in control of,
Mononucleotides, 287
glycogen, 145
94-95, 97t, 98f
“salvage” reactions and, 294, 295f, 297f
lactate production and, 139
respiratory chain in, 92. See also
Monooxygenases, 89-90. See also
as protein reserve, 576
Respiratory chain
Cytochrome P450 system
proteins of, 566t. See also Actin; Myosin;
Mitochondrial cytochrome P450, 89-90,
in metabolism of xenobiotics, 626
Titin
627. See also Cytochrome P450
Monosaccharides, 102. See also specific type
Muscle contraction, 556, 558f, 561-565,
system
and Glucose
564t
Mitochondrial DNA, 322-323, 322f,
absorption of, 475, 475f
ATP hydrolysis in, 561-562, 561f
323t
physiologic importance of, 104-105, 105t
in cardiac muscle, 566-568
INDEX
/
675
regulation of
Myocardial infarction, lactate
regulation of, 526-527, 527t
actin-based, 562-563
dehydrogenase isoenzymes in
tunicamycin affecting, 527, 527t
calcium in, 562
diagnosis of, 57, 57t, 58f
Na. See Sodium
in cardiac muscle, 566-568
Myofibrils, 556, 557f, 558f
Na+-Ca2+ exchanger, 463
sarcoplasmic reticulum and,
Myoglobin, 40-48
Na+-K+ ATPase, 427-428, 428f
563-564, 563f, 564f
α-helical regions of, 40, 41f
in glucose transport, 428, 429f
in smooth muscle, 570-571, 571f
β subunits of hemoglobin and, 42
NAD+ (nicotinamide adenine dinucleotide),
myosin-based, 570
oxygen dissociation curve for, 41-42, 42f
87, 490, 490f
myosin light chain kinase in,
oxygen stored by, 40, 41-42, 42f, 573
absorption spectrum of, 56, 56f
570-571, 571f
Myoglobinuria, 47
in citric acid cycle, 133
relaxation phase of, 561, 564, 564t
Myokinase (adenylyl kinase), 84
as coenzyme, 87, 89f, 290t
in smooth muscle
deficiencies of, 151-152
NADH
calcium in, 571
in gluconeogenesis regulation, 157
absorption spectrum of, 56, 56f
nitric oxide in, 571-573, 573f
as source of ATP in muscle, 573, 575f
extramitochondrial, oxidation of, 99,
sliding filament cross-bridge model of,
Myopathies, 92
100f
557-559, 558f
mitochondrial, 100-101
fatty acid oxidation yielding, 181
in smooth muscle, 570-573
fatal infantile, and renal dysfunction,
in pyruvate dehydrogenase regulation,
tropomyosin and troponin in, 562
oxidoreductase deficiency
141-142, 142f
Muscle fatigue, 136
causing, 100
NADH dehydrogenase, 87, 93
Muscle phosphorylase, 147
Myophosphorylase deficiency, 152t
NADP+ (nicotinamide adenine dinucleotide
absence of, 152t
Myosin, 557, 559, 560f
phosphate), 87, 490
activation of
in muscle contraction, 557-559, 558f,
as coenzyme, 87, 89f, 290t
calcium/muscle contraction and,
561-562, 561f, 562f
in pentose phosphate pathway, 163,
148
regulation of smooth muscle
164f, 165f
cAMP and, 147-148, 149f
contraction and, 570
NAD(P)+-dependent dehydrogenases, in
Muscular dystrophy, Duchenne, 556,
in striated versus smooth muscle, 572t
enzyme detection, 56
565-566, 566f
structure and function of, 560-561, 560f
NADPH
Mutagenesis, site-directed, in enzyme study,
Myosin-binding protein C, 566t
in cytochrome P450 reactions, 90f, 627
58
Myosin (thick) filaments, 557, 558f
intramitochondrial, proton-translocating
Mutations, 314, 323-326, 323f, 324f,
Myosin head, 560, 560f
transhydrogenase and, 99
325f
conformational changes in, in muscle
for lipogenesis, 175f, 176, 176f
base substitution, 361, 361f, 362
contraction, 561
pentose phosphate pathway and, 163,
constitutive, 376
Myosin heavy chains, 560
164f, 165f, 169
frameshift, 363, 364f
familial hypertrophic cardiomyopathy
NADPH-cytochrome P450 reductase, 627
ABO blood group and, 619
caused by mutations in gene for,
NADPH oxidase, 621t, 622-623
gene conversion and, 325
569-570, 570f
chronic granulomatous disease associated
integration and, 324, 324f
Myosin light chain kinase, 570-571, 571f
with mutations in, 623, 623f
of membrane proteins, diseases caused
Myosin light chains, 560
NCoA-1/NCoA-2 coactivators, 472, 472t
by, 431-432, 432f, 432t
in smooth muscle contraction, 570
NCoR, 472t, 473
missense, 361, 362-363, 362f
Myotonia congenita, 569t
NDPs. See Ribonucleoside diphosphates
familial hypertrophic cardiomyopathy
Myristic acid, 112t, 510
Nebulin, 566t
caused by, 569-570, 570f
Myristylation, 510
NEFA (nonesterified fatty acids). See Free
mRNA nucleotide sequence changes caus-
in covalent modification, mass increases
fatty acids
ing, 361-363, 361f, 362f, 364f
and, 27t
Negative nitrogen balance, 479
nonsense, 362
Negative regulators, of gene expression,
point, 361
374, 375t, 378, 380
recombinant DNA technology in
N-acetyl neuraminic acid, 169, 171f
Negative supercoils, DNA, 306
detection of, 408-409, 408f
in gangliosides, 201, 203f
NEM-sensitive factor (NSF), 509, 510f
recombination and, 323-324, 323f, 324f
in glycoproteins, 169, 171f, 515, 516t
Neonatal adrenoleukodystrophy, 503,
silent, 361
in mucins, 519f, 520
503t
sister chromatid exchanges and, 325, 325f
N-linked glycoproteins, 518, 519f,
Neonatal (physiologic) jaundice, 282-283
suppressor, 363
521-527
Neonatal tyrosinemia, 255
transition, 361, 361f
classes of, 521, 522f
Neonate, hypoglycemia in, 151
transposition and, 324-325
synthesis of, 521-527, 523f, 524f, 525f,
Nerve cells. See Neurons
transversion, 361, 361f
526t
Nerve impulses, 428
Myasthenia gravis, 431
dolichol-P-P-oligosaccharide in,
Nervous system
Myelin sheets, 428
521-524, 523f
glucose as metabolic necessity for, 232
Myeloma, 595
in endoplasmic reticulum and Golgi
thiamin deficiency affecting, 489
Myeloma cells, hybridomas grown from,
apparatus, 524-525, 526t
NESs. See Nuclear export signals
596, 596f
glycan intermediates formed during,
Net charge, of amino acid, 16-17, 17f
Myeloperoxidase, 612, 621t, 623
526
Net diffusion, 423
676
/
INDEX
NeuAc. See N-Acetylneuraminic acid
Night blindness, vitamin A deficiency
Nuclear genes, proteins encoded by, 499
Neural tube defects, folic acid supplements
causing, 482t, 483
Nuclear localization signal (NLS), 501,
in prevention of, 494
Nitric oxide, 556, 571-573, 573f, 574t,
502f, 508t
Neuraminic acid, 110, 116
607t
Nuclear magnetic resonance (NMR)
Neuraminidases
clotting/thrombosis affected by, 607,
spectroscopy
deficiency of, 532-533, 533t
607t
for glycoprotein analysis, 514, 515t
in glycoprotein analysis, 517
Nitric oxide synthases, 572-573, 573f, 574t
protein structure demonstrated by,
influenza virus, 533
Nitrite, nitric oxide formation from, 572
35-36
Neurofilaments, 577t
Nitrogen, amino acid (α-amino)
Nuclear pore complexes, 501
Neurologic diseases, protein conformation
catabolism of, 242-248
Nuclear proteins, O-glycosidic linkages in,
alterations and, 37
end products of, 242-243
518
Neurons, membranes of
urea as, 242-243, 245-247, 246f
Nuclear receptor coactivators
impulses transmitted along, 428
L-glutamate dehydrogenase in,
(NCoA-1/NCoA-2), 472, 472t
ion channels in, 424, 425f
244-245, 244f, 245f
Nuclear receptor corepressor (NCoR), 472t,
synaptic vesicle fusion with, 511
Nitrogen balance, 479
473
Neuropathy, sensory, in vitamin B6 excess,
Nitroglycerin, 572
Nuclear receptor superfamily, 436, 469,
491
NLS. See Nuclear localization signal
469-471, 471f, 472t
Neutral lipids, 111
NMR. See Nuclear magnetic resonance
Nucleases, 8, 312
Neutropenia, 610
(NMR) spectroscopy
active chromatin and, 316
Neutrophils, 620-624
NO. See Nitric oxide
Nucleic acids. See also DNA; RNA
activation of, 621-622
NO synthase. See Nitric oxide synthase
bases of, 287-289, 288t
biochemical features of, 620t
Noncoding regions, in recombinant DNA
dietarily nonessential, 293
enzymes and proteins of, 621t
technology, 397, 398f
digestion of, 312
in infection, 620
Noncoding strand, 304
structure and function of, 303-313
in inflammation, 620, 621t
Noncompetitive inhibition, competitive
Nucleolytic processing, of RNA, 352
integrins and, 620-621, 622t
inhibition differentiated from,
Nucleophile, water as, 7-8
selectins and, 528-529, 529t, 530f
67-69, 67f, 68f, 69f
Nucleophilic attack, in DNA synthesis,
proteinases of, 623-624, 624t
Noncovalent assemblies, in membranes, 416
328, 329f
respiratory burst and, 622-623
Noncovalent forces
Nucleoplasmin, 315
NF-κB pathway, 468, 468f, 469f
in biomolecule stabilization, 6
Nucleoproteins, packing of, 318, 319t, 320f
Niacin, 482t, 490, 490f. See also
peptide conformation and, 20
Nucleosidases (nucleoside phosphorylases),
Nicotinamide; Nicotinic acid
Nonequilibrium reactions, 128-129
purine, deficiency of, 300
in citric acid cycle, 133
citric acid cycle regulation and, 135
Nucleoside diphosphate kinase, 85
deficiency of, 482t, 490
glycolysis regulation and, 140, 153-155
Nucleoside triphosphates
excess/toxicity of, 490
Nonesterified fatty acids. See Free fatty acids
group transfer potential of, 289-290,
Nick translation, 413
Nonfunctional plasma enzymes, 57. See also
289f, 290f, 290t
Nickel, 496t
Enzymes
nonhydrolyzable analogs of, 291, 292f
Nicks/nick-sealing, in DNA replication,
in diagnosis and prognosis, 57, 57t
in phosphorylation, 85
332, 332f
Nonheme iron, 92, 95f, 585
Nucleosides, 286-287, 288t
Nicotinamide, 482t, 490, 490f. See also
Nonhistone proteins, 314
Nucleosomes, 314, 315-316, 315f
Niacin
Non-insulin dependent diabetes mellitus
Nucleotide excision-repair of DNA, 336,
coenzymes derived from, 50-51
(NIDDM/type 2), 161
337, 338f
dehydrogenases and, 87, 89f
Nonoxidative phase, of pentose phosphate
Nucleotide sugars, in glycoprotein biosyn-
excess/toxicity of, 490
pathway, 163-166
thesis, 516-517, 516t, 520, 521t
Nicotinamide adenine dinucleotide
Nonrepetitive (unique-sequence) DNA,
Nucleotides, 286-292, 288t. See also
(NAD+), 87, 490, 490f
320, 320-321
Purine; Pyrimidines
absorption spectrum of, 56, 56f
Nonsense codons, 359, 361, 363
adenylyl kinase (myokinase) in
in citric acid cycle, 133
Nonsense mutations, 361
interconversion of, 84
as coenzyme, 87, 89f, 290t
Nonsteroidal anti-inflammatory drugs
as coenzymes, 290, 290t
Nicotinamide adenine dinucleotide
cyclooxygenase affected by, 193
DNA, deletion/insertion of, frameshift
phosphate (NADP+), 87, 490
prostaglandins affected by, 190
mutations and, 363, 364f
as coenzyme, 87, 89f, 290t
Norepinephrine, 439f, 447, 447f. See also
metabolism of, 293-302
in pentose phosphate pathway, 163,
Catecholamines
in mRNA, 358
164f, 165f
synthesis of, 267, 267f, 445-447, 447f
mutations caused by changes in,
Nicotinic acid, 482t, 490, 490f. See also
in thermogenesis, 217, 217f
361-363, 361f, 362f, 364f
Niacin
Northern blot transfer procedure, 305-306,
physiologic functions of, 289
as hypolipidemic drug, 229
403, 404f, 413
as polyfunctional acids, 290
NIDDM. See Non-insulin dependent
in gene isolation, 635t
polynucleotides, 291-292
diabetes mellitus
NPCs. See Nuclear pore complexes
synthetic analogs of, in chemotherapy,
Nidogen (entactin), in basal lamina, 540
NSF. See NEM-sensitive factor
290-291, 291f
Niemann-Pick disease, 203t
Nuclear export signals, 503
ultraviolet light absorbed by, 290
INDEX
/
677
Nucleus (cell), importins and exportins in
Oncogenes, 1
Overnutrition, 478-479
transport and, 501-503, 502f
cyclins and, 334
Oxaloacetate
Nutrition, 474-480. See also Diet
Oncoproteins, Rb protein and, 334
in amino acid carbon skeleton
biochemical research affecting, 2
Oncotic (osmotic) pressure, 580, 584
catabolism, 249, 250f
lipogenesis regulated by, 177-178
Oncoviruses, cyclins and, 334
in aspartate synthesis, 237-238, 238f
Nutritional deficiencies, 474
Open complex, 345
in citric acid cycle, 126, 127f, 130, 131f,
in AIDS and cancer, 479
Operator locus, 377-378, 377f, 378
133, 134f, 135
Nutritionally essential amino acids, 124,
Operon/operon hypothesis, 375, 376-378,
Oxalosis, 170
237t, 480. See also Amino acids
376f, 377f
Oxidases, 86, 86-87, 87f. See also specific
Nutritionally essential fatty acids, 190. See
Optical activity/isomer, 104
type
also Fatty acids
ORC. See Origin replication complex
ceruloplasmin as, 587
abnormal metabolism of, 195-196
ORE. See Origin replication element
copper in, 86
deficiency of, 191-192, 194-195
Ori (origin of replication), 326, 327f, 413
flavoproteins as, 86-87, 88f
Nutritionally nonessential amino acids,
Origin replication complex, 326
mixed-function, 89-90, 627. See also
124, 237, 237t, 480
Origin replication element, 326
Cytochrome P450 system
synthesis of, 237-241
Origin of replication (ori), 326, 327f,
Oxidation, 86-91
413
definition of, 86
Ornithine, 265, 266f
dehydrogenases in, 87-88, 88f, 89f
OR. See Right operator
catabolism of, 250, 251f
fatty acid, 180-189. See also Ketogenesis
O blood group substance, 618-619, 619f
in urea synthesis, 245, 246-247, 246f,
acetyl-CoA release and, 123-124,
O gene, 618-619
247
123f, 181-183, 181f, 182f
O-glycosidic linkage
Ornithine δ-aminotransferase, mutations
β, 181-183, 181f, 182f
of collagen, 537
in, 250
ketogenesis regulation and,
of proteoglycans, 542-543
Ornithine-citrulline antiporter, defective,
186-187, 187f, 188f
O-linked glycoproteins, 518, 518-520,
250
modified, 183, 183f
519f, 520f, 520t
Ornithine transcarbamoylase/L-Ornithine
clinical aspects of, 187-189
synthesis of, 520, 521t
transcarbamoylase
hypoglycemia caused by impairment
O-linked oligosaccharides, in mucins,
deficiency of, 247, 300
of, 187-188
519-520, 519f, 520f
in urea synthesis, 246-247, 246f
in mitochondria, 180-181, 181f
Obesity, 80, 205, 231, 474, 478
Orosomucoid (α1-acid glycoprotein),
hydroperoxidases in, 88-89
lipogenesis and, 173
583t
oxidases in, 86-87, 87f, 88f
Octamers, histone, 315, 315f
Orotate phosphoribosyltransferase, 296,
oxygen toxicity and, 90-91, 611-613,
Oculocerebrorenal syndrome, 512t
297, 298f
613t
1,25(OH)2-D3. See Calcitriol
Orotic aciduria, 300, 301
oxygenases in, 89-90, 90f
3β-OHSD. See 3β-Hydroxysteroid
Orotidine monophosphate (OMP), 296,
redox potential and, 86, 87t
dehydrogenase
298f
Oxidation-reduction (redox) potential, 86,
Okazaki fragments, 327, 330, 331f
Orotidinuria, 301
87t
Oleic acid, 112, 112f, 113, 113t, 114f, 190f
Orphan receptors, 436, 471
Oxidative decarboxylation, of
synthesis of, 191, 191f
Osmotic fragility test, 617
α-ketoglutarate, 131, 132f
Oligomers, import of by peroxisomes, 503
Osmotic lysis, complement in, 596
Oxidative phase, of pentose phosphate
Oligomycin, respiratory chain affected by,
Osmotic (oncotic) pressure, 580, 584
pathway, 163, 164f, 165f
95, 96f, 97f
Osteoarthritis, 535, 551t
Oxidative phosphorylation, 83, 92-101,
Oligonucleotide
proteoglycans in, 548
122. See also Phosphorylation;
definition of, 413
Osteoblasts, 549, 549f, 550
Respiratory chain
in primary structure determination, 26
Osteocalcin, 488, 496, 548t
chemiosmotic theory of, 92, 95-97, 97f
Oligosaccharide:protein transferase, 523
Osteoclasts, 549-550, 549f, 550f
clinical aspects of, 100-101
Oligosaccharide branches (antennae), 521
Osteocytes, 549, 549f
muscle generation of ATP by, 573,
Oligosaccharide chains
Osteogenesis imperfecta (brittle bones),
574-576, 575f, 575t
glycoprotein, 514, 515t, 581-582
551-552, 551t
poisons affecting, 92, 95, 96f
in N-glycosylation, 524, 525f
Osteoid, 549f, 550
Oxidative stress, 612
regulation of, 526
Osteomalacia, 482t, 484, 485, 551t
Oxidoreductases, 49, 86. See also specific type
sugars in, 515, 516t
Osteonectin, 548t
deficiency of, 100
glycosaminoglycans, 543
Osteopetrosis (marble bone disease), 552
Oxidosqualene:lanosterol cyclase, 220, 222f
Oligosaccharide processing, 521, 524, 525f
Osteoporosis, 485, 551t, 552
Oxygen
Golgi apparatus in, 509
Osteopontin, 548t
binding, 42, 42f. See also Oxygenation
regulation of, 526, 527f
Ouabain, 106
Bohr effect and, 44, 45f
Oligosaccharides, 102
Na+-K+ ATPase affected by, 428
histidines F8 and E7 in, 40, 41f
O-linked, in mucins, 519-520, 519f,
Outer mitochondrial membrane, 92, 93f
hemoglobin affinities (P50) for, 42-43,
520f
protein insertion in, 501
43f
OMP (orotidine monophosphate), 296,
Ovary, hormones produced by, 437,
myoglobin in storage of, 40, 41-42, 42f,
298f
442-445, 444f, 445f
573
678
/
INDEX
Oxygen (cont.)
storage/secretion of, 453, 454t
Periodic acid-Schiff reagent, in glycoprotein
reductive activation of, 627
synthesis of, 450, 451f
analysis, 515t
transport of, ferrous iron in, 40-41
Paroxysmal nocturnal hemoglobinuria,
Periodic hyperlysinemia, 258
Oxygen dissociation curve, for myoglobin
432t, 528, 530t, 531, 531f
Periodic paralysis
and hemoglobin, 41-42, 42f
Partition chromatography, for protein/
hyperkalemic, 569t
Oxygen radicals. See Free radicals
peptide purification, 21
hypokalemic, 569t
Oxygen toxicity, superoxide free radical
Passive diffusion/transport, 423, 423t, 424f
Peripheral proteins, 420-421, 421f
and, 90-91, 611-613, 613t. See
Pasteur effect, 157
Peripherin, 577t
also Free radicals
pBR322, 402, 402t, 403f
Permeability coefficients, of substances in
Oxygenases, 86, 89-90
PCR. See Polymerase chain reaction
lipid bilayer, 418, 419f
Oxygenation of hemoglobin
PDH. See Pyruvate dehydrogenase
Pernicious anemia, 482t, 492
conformational changes and, 42, 43f, 44f
PDI. See Protein disulfide isomerase
Peroxidases, 88, 192
apoprotein, 42
PECAM-1, 529, 529t
Peroxidation, lipid, free radicals produced
2,3-bisphosphoglycerate stabilizing,
Pedigree analysis, 409, 410f
by, 118-119, 120f
45, 45f
Pellagra, 482t, 490
Peroxins, 503
high altitude adaptation and, 46
Penicillamine, for Wilson disease, 589
Peroxisomal-matrix targeting sequences
mutant hemoglobins and, 46
Pentasaccharide, in N-linked glycoproteins,
(PTS), 503, 508t
Oxysterols, 119
521, 522f
Peroxisomes, 89, 503
Pentose phosphate pathway, 123, 163-166,
absence/abnormalities of, 503, 503t
164f, 165f, 167f
in Zellweger’s syndrome, 188, 503
Pi, in muscle contraction, 561, 561f
cytosol as location for reactions of, 163
biogenesis of, 503
P50, hemoglobin affinity for oxygen and,
enzymes of, 156t
in fatty acid oxidation, 182-183
42-43, 43f
erythrocyte hemolysis and, 169-170,
Pfeiffer syndrome, 551t
p53 protein/p53 gene, 339
613
PFK-1. See Phosphofructokinase
p160 coactivators, 472, 472t
impairment of, 169-170
PGHS. See Prostaglandin H synthase
p300 coactivator/CPB/p300, 461, 468,
NADPH produced by, 163, 164f,
PGIs. See Prostacyclins
469, 469f, 472, 472t
165f
PGs. See Prostaglandins
P450 cytochrome. See Cytochrome P450
for lipogenesis, 175f, 176, 176f
pH, 9-13. See also Acid-base balance
system
nonoxidative phase of, 163-166
amino acid net charge and, 16, 17f
P450scc (cytochrome P450 side chain
oxidative phase of, 163, 164f, 165f
buffering and, 11-12, 12f. See also
cleavage enzyme), 438, 440f, 442
ribose produced by, 163, 164f
Buffers
p/CIP coactivator, 472, 472t
Pentoses, 102, 102t
calculation of, 9-10
P component, in amyloidosis, 590
in glycoproteins, 109t
definition of, 9
P-selectin, 529t
physiologic importance of, 104-105,
enzyme-catalyzed reaction rate affected
PAC (P1-based) vector, 401-402, 402t, 413
105t
by, 64, 64f
PAF. See Platelet-activating factor
Pentosuria, essential, 163, 170
isoelectric, amino acid net charge and, 17
PAGE. See Polyacrylamide gel
PEPCK. See Phosphoenolpyruvate
Phage lambda, 378-383, 379f, 380f, 381f,
electrophoresis
carboxykinase
382f
Pain, prostaglandins in, 190
Pepsin, 477
Phages
Palindrome, 413
in acid-base catalysis, 52
for cloning in gene isolation, 635t
Palmitate, 173, 173-174
Pepsinogen, 477
in recombinant DNA technology, 401
Palmitic acid, 112t
Peptidases, in protein degradation, 242,
Phagocytic cells, respiratory burst of,
Palmitoleic acid, 113t, 190f
243f
622-623
Palmitoylation, in covalent modification,
Peptide bonds. See also Peptides
Phagocytosis, 429
mass increases and, 27t
formation of, 7, 368
Pharmacogenetics, 630, 631-632
Pancreatic insufficiency, in vitamin B12
partial double-bond character of, 19-20,
Pharmacogenomics, 632, 638
deficiency, 492
20f
Phasing, nucleosome, 315-316
Pancreatic islets, insulin produced by, 160
Peptides, 14-20, 439f. See also Amino
Phenobarbital, warfarin interaction and,
Pancreatic lipase, 475, 476f
acids; Proteins
cytochrome P450 induction
Panproteinase inhibitor, α2-macroglobulin
absorption of, 477
affecting, 628
as, 590
amino acids in, 14, 19, 19f
Phenylalanine, 16t
Pantothenic acid, 173, 482t, 495, 495i
formation of, L-α-amino acids in, 14
catabolism of, 255-258, 255f
in citric acid cycle, 133
as hormone precursors, 449-453
in phenylketonuria, 255, 255f
coenzymes derived from, 51
intracellular messengers used by,
requirements for, 480
deficiency of, 482t
457-468, 461t, 463t
in tyrosine synthesis, 239, 240f
Papain, immunoglobulin digestion by, 591
as polyelectrolytes, 19
Phenylalanine hydroxylase
PAPS. See Adenosine 3′-phosphate-
purification of, 21-24
defect in, 255
5′-phosphosulfate
Peptidyl prolyl isomerase, 508
localization of gene for, 407t
Parallel β sheet, 32, 33f
Peptidylglycine hydroxylase, vitamin C as
in tyrosine synthesis, 239, 240f
Parathyroid hormone (PTH), 438, 450,
coenzyme for, 496
Phenylethanolamine-N-methyltransferase
451f
Peptidyltransferase, 368, 370t
(PNMT), 447, 447f
INDEX
/
679
Phenylisothiocyanate (Edman reagent), in
in neutrophil activation, 621-622
Phospholipase Cβ, in platelet activation,
protein sequencing, 25, 26f
in platelet activation, 606-607, 606f
606, 606f
Phenylketonuria, 255-258
Phosphatidylinositol 3-kinase (PI-3 kinase)
Phospholipase D, 200, 201f
Phi (ϕ) angle, 31, 31f
in insulin signal transmission, 465, 466f
Phospholipases
Phosphagens, 83, 84f
in Jak/STAT pathway, 467
in glycoprotein analysis, 515t
Phosphatases
Phosphatidylserine, 115, 115f
in phosphoglycerol degradation and
acid, diagnostic significance of, 57t
in blood coagulation, 601
remodeling, 200-201, 201f
alkaline
membrane asymmetry and, 420
Phospholipids, 111, 205
in bone mineralization, 550
Phosphocreatine, in muscle, 556
digestion and absorption of, 475-477,
isozymes of, diagnostic significance of,
Phosphodiester, 291
476f
57t
Phosphodiesterases, 291
glycerol ether, synthesis of, 199, 200f
in recombinant DNA technology,
in calcium-dependent signal
in lipoprotein lipase activity, 207-208
400t
transduction, 463
in membranes, 114-116, 115f,
Phosphate transporter, 99, 99f
in cAMP-dependent signal transduction,
416-417, 417f, 419, 511
Phosphates/phosphorus, 496t
461, 462f
membrane asymmetry and, 420, 511
exchange transporters and, 99, 99f, 100,
cAMP hydrolyzed by, 147
in multiple sclerosis, 202
101f
Phosphoenolpyruvate, 156t
as second messenger precursors, 197
in extracellular and intracellular fluid,
free energy of hydrolysis of, 82t
synthesis of, 198f
416t
in gluconeogenesis, 133, 134f, 156t
Phosphoprotein phosphatases, in cAMP-
free energy of hydrolysis of, 82-83, 82t
Phosphoenolpyruvate carboxykinase
dependent signal transduction,
high-energy, 83. See also ATP
(PEPCK), 133, 134f
462, 462f
in energy capture and transfer, 82-83,
in gluconeogenesis regulation, 133, 134f,
Phosphoproteins, in cAMP-dependent
82f, 82t, 83f
153, 154f
signal transduction, 461, 462f
as “energy currency” of cell, 83-85,
Phosphofructokinase/
Phosphoric acid, pK/pKa value of, 12t
84f, 85f
phosphofructokinase-1, 156t
Phosphorus. See Phosphates
symbol designating, 83
in gluconeogenesis regulation, 157
Phosphorylase
transport of, creatine phosphate
in glycolysis, 137, 138f, 156t
in glycogen metabolism, 145-146,
shuttle in, 100, 101f
regulation and, 140
146f
low-energy, 83
muscle, deficiency of, 143, 152t
regulation of, 148-150, 150-151,
Phosphatidate, 198f, 199
Phosphofructokinase-2, 157, 158f
150f, 151f
in triacylglycerol synthesis, 197, 197f,
Phosphoglucomutase, in glycogen
liver, 147
198, 198f, 199
biosynthesis, 145, 146f
deficiency of, 152t
Phosphatidate phosphohydrolase, 198f, 199
6-Phosphogluconate dehydrogenase, 156t,
muscle, 147
Phosphatidic acid, 114, 115f, 416-417,
163, 164f, 165f
absence of, 152t
417f
3-Phosphoglycerate
activation of
Phosphatidic acid pathway, 476f, 477
in glycolysis, 137, 138f
calcium/muscle contraction and,
Phosphatidylcholines (lecithins), 114-115,
in serine synthesis, 238, 238f
148
115f
Phosphoglycerate kinase, in glycolysis, 137,
cAMP and, 147-148, 149f
in cytochrome P450 system, 617
138f
Phosphorylase a, 147, 149f
membrane asymmetry and, 420
in erythrocytes, 140, 140f
Phosphorylase b, 147, 149f
synthesis of, 197, 197f, 198f
Phosphoglycerate mutase, in glycolysis, 137,
Phosphorylase kinase
Phosphatidylethanolamine (cephalin), 115,
138f
calcium/calmodulin-sensitive, in
115f
Phosphoglycerides, in membranes,
glycogenolysis, 148
membrane asymmetry and, 420
416-417, 417f
deficiency of, 152t
synthesis of, 197, 197f
Phosphoglycerols
protein phosphatase-1 affecting, 147
Phosphatidylglycerol, 115, 115f
lysophospholipids in metabolism of, 116,
Phosphorylase kinase a, 148, 149f
Phosphatidylinositol/phosphatidylinositide,
116f
Phosphorylase kinase b, 148, 149f
115, 115f
synthesis of, 197f, 198f, 199
Phosphorylation
in blood coagulation, 601
Phosphohexoseisomerase, in glycolysis, 137,
in covalent modification, 76, 77-79, 78f,
GPI-linked glycoproteins and, 527.
138f
78t
See also Glycosylphosphatidyli-
Phosphoinositide-dependent kinase-1
mass increases and, 27t
nositol-anchored (GPI-
(PDK1), in insulin signal
multisite, in glycogen metabolism, 151
anchored/ GPI-linked)
transmission, 465
oxidative. See Oxidative phosphorylation
glycoproteins
Phospholipase A1, 200, 201f
in respiratory burst, 623
metabolism of, 464-465, 464f, 465f
Phospholipase A2, 200, 201f
Photolysis reaction, in vitamin D synthesis,
as second messenger/second messenger
in platelet activation, 606f, 607
445
precursor, 115, 115f, 437, 437t,
Phospholipase C, 200, 201f
Photosensitivity, in porphyria, 274
457, 463-465, 463t, 464f, 465f
in calcium-dependent signal transduction,
Phototherapy, cancer, porphyrins in, 273
synthesis of, 197, 197f, 198f
464-465, 464f, 465f
Phylloquinone, 482t, 486, 488f. See also
Phosphatidylinositol 4,5-bisphosphate, 115,
in Jak/STAT pathway, 467
Vitamin K
464-465, 465f
in respiratory burst, 623
Physical map, 633, 634f
680
/
INDEX
Physiologic (neonatal) jaundice, 282-283
Plasma thromboplastin antecedent
Polymerases
Phytanic acid, Refsum’s disease caused by
(PTA/factor XI), 599f, 600, 600t
DNA, 326, 327-328, 327f, 328, 328t
accumulation of, 188
deficiency of, 601
in recombinant DNA technology, 400t
Phytase, 477
Plasma thromboplastin component
RNA, DNA-dependent, in RNA
Phytic acid (inositol hexaphosphate), calcium
(PTC/factor IX), 599f, 600, 600t
synthesis, 342-343, 342f, 343t
absorption affected by, 477
coumarin drugs affecting, 604
Polymorphisms, 407
Pi, 589. See also α1-Antiproteinase
deficiency of, 604
acetyltransferase, 630
pI (isoelectric pH), amino acid net charge
Plasmalogens, 116, 117f, 199, 200f
cytochrome P450, 628, 630t
and, 17
Plasmids, 400-401, 401f, 402, 402t, 403f,
microsatellite, 322, 411, 413
PI-3 kinase
413
plasma protein, 582
in insulin signal transmission, 465, 466f
for cloning in gene isolation, 635t
restriction fragment length. See
in Jak/STAT pathway, 467
Plasmin, 604-605, 604f
Restriction fragment length
PIC. See Preinitiation complex
Plasminogen, 604
polymorphisms
PIG-A gene, mutations of in paroxysmal
activators of, 604-605, 604f, 605, 605f,
single nucleotide, 414
nocturnal hemoglobinuria, 531,
607t
Polynucleotide kinase, in recombinant
531f
Platelet-activating factor, 197, 621t
DNA technology, 400t
“Ping-Pong” mechanism, in facilitated
synthesis of, 198f, 199, 200f
Polynucleotides, 291-292
diffusion, 427, 427f
Platelets, activation/aggregation of, 598,
posttranslational modification of, 289
Ping-pong reactions, 69-70, 69f
605-607, 606f
Polyol (sorbitol) pathway, 172
Pinocytosis, 429-430
aspirin affecting, 607-608
Polypeptides
PIP2, in absorptive pinocytosis, 430
Pleckstrin, in platelet activation, 607
receptors for, 436
Pituitary hormones, 437. See also specific
PLP. See Pyridoxal phosphate
sequencing of
type
PNMT. See Phenylethanolamine-
cleavage in, 25, 26t
blood glucose affected by, 161
N-methyltransferase
Sanger’s determination of, 24-25
pK/pKa
pOH, in pH calculation, 9
Polyphosphoinositide pathway, platelet
of amino acids, 15-16t, 17, 17f, 18
Point mutations, 361
activation and, 605-607
environment affecting, 18, 18t
recombinant DNA technology in
Polyprenoids, 118, 119f
medium affecting, 13
detection of, 408-409, 408f, 409t
Polyribosomes (polysomes), 310, 370
of weak acids, 10-11, 11-12, 12t, 13, 17
Poisons, oxidative phosphorylation/
protein synthesis on, 498, 499f, 500f,
PKA. See Protein kinase A
respiratory chain affected
506
PKB. See Protein kinase B
by, 92, 95, 96f
plasma proteins, 581
PKC. See Protein kinase C
Pol II
signal hypothesis of binding of,
PKU. See Phenylketonuria
phosphorylation of, 350-351
503-505, 504t, 505f
Placenta, estriol synthesis by, 442
in preinitiation complex formation,
Polysaccharides, 102, 107-110, 108f, 109f.
Plaque hybridization, 403. See also
351-352
See also specific type
Hybridization
in transcription, 350-351
Polysomes. See Polyribosomes
Plasma, 580
Polarity
Polytene chromosomes, 318, 318f
Plasma cells, immunoglobulins synthesized
of DNA replication/synthesis, 330-331
Polyunsaturated fatty acids, 112, 113t. See
in, 591
of protein synthesis, 364
also Fatty acids; Unsaturated fatty
Plasma enzymes. See also Enzymes
of xenobiotics, metabolism and, 626
acids
diagnostic significance of, 57, 57t
Poly(A) tail, of mRNA, 309, 355-356
dietary, cholesterol levels affected by, 227
Plasma lipoproteins. See Lipoproteins
in initiation of protein synthesis, 365
eicosanoids formed from, 190, 192,
Plasma membrane, 415, 426-431, 426f.
Polyacrylamide gel electrophoresis, for
193f, 194f
See also Membranes
protein/peptide purification,
essential, 190, 190f
carbohydrates in, 110
24, 24f, 25f
synthesis of, 191, 191f, 192f
mutations in, diseases caused by, 431,
Polyadenylation sites, alternative, 394
POMC. See Pro-opiomelanocortin
432t
Polyamines, synthesis of, 265-266, 266f
(POMC) peptide family
Plasma proteins, 514, 580-591, 581f, 583t.
Polycistronic mRNA, 376
Pompe’s disease, 152t
See also specific type and
Polycythemia, 46
Porcine stress syndrome, 565
Glycoproteins
Polydystrophy, pseudo-Hurler, 532, 546t
Porphobilinogen, 270, 273f, 275f
in bone, 548t
Polyelectrolytes, peptides as, 19
Porphyrias, 274-278, 277f, 277t
concentration of, 580
Polyfunctional acids, nucleotides as, 290
Porphyrinogens, 272
electrophoresis for analysis of, 580, 582f
Polyisoprenoids, in cholesterol synthesis,
accumulation of in porphyria, 274-278
functions of, 583, 583t
220, 221f
Porphyrins, 270-278, 271f, 272f
half life of, 582
Polyisoprenol, in N-glycosylation, 521-522
absorption spectra of, 273-274, 277f
in inflammation, 621t
Polymerase chain reaction (PCR), 57,
heme synthesis and, 270-273, 273f,
polymorphism of, 582
405-406, 406f, 413, 414
274f, 275f, 276f
synthesis of
in gene isolation, 635t
reduced, 272
in liver, 125, 581
in microsatellite repeat sequence
spectrophotometry in detection of,
on polyribosomes, 581
detection, 322
273-274
transport, 454-455, 454t, 455t, 583t
in primary structure determination, 26
Positive nitrogen balance, 479
INDEX
/
681
Positive regulators, of gene expression, 374,
proteomics and, 28-29
Promoter recognition specificity, 343
375t, 378, 380
Sanger’s technique in determination of,
Promoters, in transcription, 342, 342f
Posttranslational processing, 30, 37-39,
24-25
alternative use of in regulation, 354-355,
38f, 371
Primary transcript, 342
355f, 393-394
of collagen, 537-538, 537t
Primases, DNA, 327, 327f, 328t
bacterial, 345-346, 345f
in membrane assembly, 511-512
Primosome, 328, 414
eukaryotic, 346-349, 347f, 348f, 349f,
Posttranslational translocation, 499
Prion diseases (transmissible spongiform
384
Potassium, 496t
encephalopathies), 37
Promotor site, in operon model, 377f, 378
in extracellular and intracellular fluid,
Prion-related protein (PrP), 37
Proofreading, DNA polymerase, 328
416, 416t
Prions, 37
Pro-opiomelanocortin (POMC) peptide
permeability coefficient of, 419f
Proaccelerin (factor V), 600t, 601, 602f
family, 452-453, 453f. See also
Power stroke, 561
Proaminopeptidase, 477
specific type
PPI. See Peptidyl prolyl isomerase
Probes, 402, 414. See also DNA probes
Pro-oxidants, 612. See also Free radicals
PPi. See Pyrophosphate, inorganic
for gene isolation, 635t
Proparathyroid hormone (proPTH), 450,
PR. See Progesterone, receptors for
Probucol, 229
450f
Pravastatin, 229
Procarcinogens, 626
Propionate
PRE. See Progestin response element
Processivity, DNA polymerase, 328
blood glucose and, 159
Pre-β-lipoproteins, 205, 206t, 210
Prochymotrypsin, activation of, 77, 77f
in gluconeogenesis, 154f, 155
Precursor proteins, amyloid, 590
Procollagen, 371, 496, 537
metabolism of, 155, 155f
Pregnancy
Procollagen aminoproteinase, 537
Propionic acid, 112t
estriol synthesis in, 442
Procollagen carboxyproteinase, 537
Propionyl-CoA
fatty liver of, 188
Procollagen N-proteinase, disease caused by
fatty acid oxidation yielding, 182
hypoglycemia during, 161
deficiency of, 538t
methionine in formation of, 259, 259f
iron needs during, 586
Proconvertin (factor VII), 599f, 600t, 601
Propionyl-CoA carboxylase, 155, 155f
Pregnancy toxemia of ewes (twin lamb
coumarin drugs affecting, 604
Proproteins, 37-38, 76, 371
disease)
Prodrugs, 626
Propyl gallate, as antioxidant/food
fatty liver and, 212
Proelastase, 477
preservative, 119
ketosis in, 188
Proenzymes, 76
Prostacyclins, 112
Pregnenolone, 440f
rapid response to physiologic demand
clinical significance of, 196
in adrenal steroidogenesis, 438-440,
and, 76
clotting/thrombosis affected by, 607, 607t
440f, 441f
Profiling, protein-transcript, 412
Prostaglandin E2, 112, 113f
in testicular steroidogenesis, 442, 443f
Progesterone, 439f, 440f
Prostaglandin H synthase, 192
Preinitiation complex, 343, 351-352
binding of, 455, 455t
Prostaglandins, 112, 113f, 190, 192
assembly of, 351-352
receptors for, 471
cyclooxygenase pathway in synthesis of,
in protein synthesis, 365, 366f
synthesis of, 438, 442, 445f
192, 192-194, 193f, 194f
Prekallikrein, 599f, 600
Progesterone (∆4) pathway, 442, 443f
Prostanoids, 112, 119
Premenstrual syndrome, vitamin B6 in
Progestin response element, 459t
clinical significance of, 196
management of, sensory
Progestins, binding of, 455
cyclooxygenase pathway in synthesis of,
neuropathy and, 491
Prohormones, 371
192, 192-194, 193f, 194f
Prenatal diagnosis, recombinant DNA
Proinsulin, 449, 450f
Prosthetic groups, 50
technology in, 409
Prokaryotic gene expression. See also Gene
in catalysis, 50-51, 51f
Preprocollagen, 537
expression
Protamine, 603
Preprohormone, insulin synthesized as, 449,
eukaryotic gene expression compared
Proteases/proteinases, 8, 477, 624t. See also
450f
with, 391-395, 392t
specific type
Preproparathyroid hormone (preproPTH),
as model for study, 375
α2-macroglobulin binding of, 590
450, 451f
unique features of, 375-376
in cartilage, 553
Preproprotein, albumin synthesized as, 583
Prolactin, 437
as catalytically inactive proenzymes,
Preproteins, 498, 581
localization of gene for, 407t
76-77
Presequence. See Signal peptide
receptor for, 436
mucin resistance to, 520
Preventive medicine, biochemical research
Proline, 16t
of neutrophils, 623-624, 624t
affecting, 2
accumulation of (hyperprolinemia),
in protein degradation, 242, 243f, 477
Primaquine-sensitive hemolytic anemia, 613
249-250
Staphylococcus aureus V8, for polypeptide
Primary structure, 21-29, 31. See also
catabolism of, 249-250, 251f
cleavage, 25, 26t
Protein sequencing
synthesis of, 238, 239f
Protein 4.1, in red cell membranes, 615f,
amino acid sequence determining, 18-19
Proline dehydrogenase, block of proline
616f, 616t, 617
Edman reaction in determination of, 25,
catabolism at, 249-250
Protein C, in blood coagulation, 600t, 603
26f
Proline hydroxylase, vitamin C as coenzyme
Protein disulfide isomerase, protein folding
genomics in analysis of, 28
for, 496
and, 37, 508
molecular biology in determination of,
Proline-cis,trans-isomerase, protein folding
Protein-DNA interactions, bacteriophage
25-26
and, 37, 37f
lambda as paradigm for,
of polynucleotides, 291-292
Prolyl hydroxylase reaction, 240, 240f, 535
378-383, 379f, 380f, 381f, 382f
682
/
INDEX
Protein folding, 36-37, 37f
importins and exportins in, 501-503,
loss of in trauma/infection, 480
chaperones and, 499, 507-508, 508t
502f
in membranes, 419, 420t, 514. See also
after denaturation, 36
KDEL amino acid sequence and,
Glycoproteins; Membrane
Protein kinase A (PKA), 460, 462f
506-507, 508t
proteins
Protein kinase B (PKB), in insulin signal
membrane assembly and, 511-513,
ratio of to lipids, 416, 416f
transmission, 465, 466f
512f, 512t
modular principals in construction of, 30
Protein kinase C (PKC)
mitochondria in, 499-501, 501f
monomeric, 34
in calcium-dependent signal
peroxisomes/peroxisome disorders and,
phosphorylation of, 76, 77-79, 78f, 78t.
transduction, 464, 464f
503, 503t
See also Phosphorylation
in platelet activation, 606f, 607
protein destination and, 507, 507f, 508t
posttranslational modification of, 30,
Protein kinase D1, in insulin signal
retrograde transport and, 507
37-39, 38f, 371
transmission, 466f, 467
signal hypothesis of polyribosome binding
purification of, 21-24
Protein kinase-phosphatase cascade, as
and, 503-505, 504t, 505f
receptors as, 431, 436
second messenger, 437, 437t
signal sequences and, 492f, 498-499,
soluble, 30
Protein kinases, 77
499f
structure of, 31-36
in cAMP-dependent signal transduction,
transport vesicles and, 508-511, 509t,
diseases associated with disorders of,
460-461, 462f
510f
37
in cGMP-dependent signal transduction,
Protein turnover, 74, 242
folding and, 36-37, 37f
463
membranes affecting, 511
higher orders of, 30-39
deficiency of, 151-152
rate of enzyme degradation and, 74
molecular modeling and, 36
DNA-dependent, in double-strand break
Proteinases. See Proteases/proteinases
nuclear magnetic resonance spec-
repair, 338
Proteins. See also specific type and Peptides
troscopy in analysis of, 35-36
in glycogen metabolism, 147-148, 149f,
β-turns in, 32, 34f
primary, 21-29, 31. See also Primary
151, 151f
acute phase, 583, 583t
structure
in hormonal regulation, 436, 465-468
negative, vitamin A as, 483-484
prion diseases associated with
of lipolysis, 215, 216f
L-α-amino acids in, 14
alteration of, 37
in initiation of protein synthesis, 365
asymmetry of, membrane assembly and,
quaternary, 33-35, 35f
in insulin signal transmission, 465-467,
511, 512f
secondary, 31, 31-33, 31f, 32f, 33f,
466f
binding, 454-455, 454t, 455t
34f
in Jak/STAT pathway, 467, 467f
catabolism of, 242-248
supersecondary motifs and, 33
in NF-κB pathway, 468, 468f
classification of, 30
tertiary, 33-35, 35f
in protein phosphorylation, 77, 78f
configuration of, 30
x-ray crystallography in analysis of, 35
Protein-lipid respiratory chain complexes,
conformation of, 30
synthesis of, 358-373. See also Protein
93
peptide bonds affecting, 20
sorting
Protein-losing gastroenteropathy, 582
core, 542, 543f
amino acids in, 124, 124f
Protein phosphatase-1, 147, 148, 149f, 151,
in glycosaminoglycan synthesis,
elongation in, 367-370, 368f
151f
542-543
environmental threats affecting, 370
Protein phosphatases, 77. See also
degradation of, to amino acids, 242, 243f
in fed state, 232
Phosphatases
denaturation of
genetic code/RNA and, 307-308,
Protein profiling, 412
protein refolding and, 36
309t, 358-363. See also Genetic
Protein-RNA complexes, in initiation,
temperature and, 63
code
365-367, 366f
dietary
inhibition of by antibiotics, 371-372,
Protein S, in blood coagulation, 600t, 603
digestion and absorption of, 477
372f
Protein sequencing
metabolism of, in fed state, 232
initiation of, 365-367, 366f, 367f
Edman reaction in, 25, 26f
requirements for, 479-480
by mitochondria, 499-501, 501t
genomics and, 28
dimeric, 34
modular principles in, 30
mass spectrometry in, 27, 27f, 27t
domains of, 33-34
polysomes in, 370, 498, 499f
molecular biology in, 25-26
encoding of by human genome, 636,
posttranslational processing and, 371
peptide purification for, 21-24
637t
in ribosomes, 126, 127f
polypeptide cleavage and, 25, 26t
in extracellular and intracellular fluid,
recognition and attachment (charging)
proteomics and, 28-29
416, 416t
in, 360, 360f
purification for, 21-24, 22f, 23f, 24f,
fibrous, 30
recombinant DNA techniques for,
25f
collagen as, 38
407
Sanger’s method of, 24-25
function of, bioinformatics in
reticulocytes in, 611
Protein sorting, 498-513
identification of, 28-29
termination of, 369f, 370
chaperones and, 507-508, 508t
fusion, in enzyme study, 58, 59f
translocation and, 368
cotranslational insertion and, 505-506,
globular, 30
viruses affecting, 370-371, 371f
506f
Golgi apparatus in glycosylation and
transmembrane
disorders due to mutations in genes
sorting of, 509
ion channels as, 423-424, 425f, 426t
encoding, 512t, 513
import of, by mitochondria, 499-501,
in red cells, 615-616, 615f, 616f,
Golgi apparatus in, 498, 500f, 507, 509
501t
616t
INDEX
/
683
transport, 454-455, 454t, 455t
“Puffs,” polytene chromosome, 318, 318f
oxidation of, 134, 135f, 140-142, 141f,
xenobiotic cell injury and, 631
Pulsed-field gel electrophoresis, for gene
142f, 143t. See also Acetyl-CoA;
Proteoglycans, 109, 535, 538, 542-549,
isolation, 635t
Glycolysis
542f. See also
Pumps, 415
clinical aspects of, 142-143
Glycosaminoglycans
in active transport, 427-428, 428f
enzymes in, 156t
in bone, 548t
Purification, protein/peptide, 21-24
gluconeogenesis and, 153, 154f
carbohydrates in, 542, 542f, 543f
Purine nucleoside phosphorylase deficiency,
Pyruvate carboxylase, 133, 134f, 156t
in cartilage, 551, 553
300
in gluconeogenesis regulation, 133, 134f,
disease associations and, 548-549
Purines/purine nucleotides, 286-290, 286f,
153, 156t
functions of, 547-549, 548t
289f
Pyruvate dehydrogenase, 134, 135f, 140,
galactose in synthesis of, 167-169, 170f
dietarily nonessential, 293
141f, 156t
link trisaccharide in, 518
metabolism of, 293-302
deficiency of, 143
Proteolysis
disorders of, 300
regulation of, 141-142, 142f
in covalent modification, 76, 76-77, 77f
gout as, 299
acyl-CoA in, 141-142, 142f, 178
in prochymotrypsin activation, 77, 77f
uric acid formation and, 299, 299f
thiamin diphosphate as coenzyme for,
Proteome/proteomics, 28-29, 414,
synthesis of, 293-294, 294f, 295f, 296f,
488
636-637, 637-638
297f
Pyruvate dehydrogenase complex, 140
Prothrombin (factor II), 600t, 601, 602f
catalysts in, 293, 294f
Pyruvate kinase, 156t
activation of, 601
pyrimidine synthesis coordinated with,
deficiency of, 143, 619
coumarin drugs affecting, 487, 604
299
gluconeogenesis regulation and, 157
in vitamin K deficiency, 487
“salvage” reactions in, 294, 295f, 297f
in glycolysis, 137-139, 138f, 156t
Prothrombinase complex, 601
ultraviolet light absorbed by, 290
regulation and, 140
Proton acceptors, bases as, 9
Puromycin, 372, 372f
Proton donors, acids as, 9
Putrescine, in polyamine synthesis, 266f
Proton pump, respiratory chain complexes
Pyranose ring structures, 103f, 104
Q (coenzyme Q/ubiquinone), 92, 95f
as, 96, 96f, 97f
Pyridoxal phosphate, 50, 491, 491f
Q10 (temperature coefficient), enzyme-
Proton-translocating transhydrogenase, as
in heme synthesis, 270
catalyzed reactions and, 63
source of intramitochondrial
in urea biosynthesis, 243
QT interval, congenitally long, 432t
NADPH, 99
Pyridoxine/pyridoxal/pyridoxamine
Quaternary structure, 33-35, 35f
Protons, transport of, by hemoglobin, 44,
(vitamin B6), 482t, 491, 491f
of hemoglobins, allosteric properties and,
45f
deficiency of, 482t, 491
42-46
Protoporphyrin, 270, 272f
xanthurenate excretion in, 258, 258f
stabilizing factors and, 35
incorporation of iron into, 271-272, 272f
excess/toxicity of, 491
Protoporphyrin III, 271, 276f
Pyrimethamine, 494
Protoporphyrinogen III, 271, 276f
Pyrimidine analogs, in pyrimidine
R groups, amino acid properties affected by,
Protoporphyrinogen oxidase, 271, 275f, 276f
nucleotide biosynthesis, 297
18, 18t
Provitamin A carotenoids, 482-483
Pyrimidines/pyrimidine nucleotides,
pK/pKa, 18
Proximal histidine (histidine F8)
286-290, 286f, 289f
R (relaxed) state, of hemoglobin,
in oxygen binding, 40, 41f
dietarily nonessential, 293
oxygenation and, 43, 43f, 44f
replacement of in hemoglobin M, 46
metabolism of, 293-302, 301f
Rab protein family, 511
Proximity, catalysis by, 51
diseases caused by catabolite
RAC3 coactivator, 472, 472t
PrP (prion-related protein), 37
overproduction and, 300-301
Radiation, nucleotide excision-repair of
PRPP
water-soluble metabolites and, 300,
DNA damage caused by, 337
in purine synthesis, 294, 295f
301f
Radiation hybrid mapping, 635t
in pyrimidine synthesis, 296, 298f, 299
precursors of, deficiency of, 300-301
Ran protein, 501, 502f, 503
PRPP glutamyl amidotransferase, 294, 295f
synthesis of, 296-299, 298f
Rancidity, peroxidation causing, 118
PRPP synthetase, defect in, gout caused by,
catalysts in, 296
Rapamycin, mammalian target of (mTOR),
299
purine synthesis coordinated with, 299
in insulin signal transmission,
Pseudo-Hurler polydystrophy, 532, 546t,
regulation of, 297-299, 298f
466f, 467
547
ultraviolet light absorbed by, 290
RAR. See Retinoic acid receptor
Pseudogenes, 325, 414
Pyrophosphatase, inorganic
RARE. See Retinoic acid response element
Psi (ψ) angle, 31, 31f
in fatty acid activation, 85, 180
Rate constant, 62
PstI, 399t
in glycogen biosynthesis, 145, 146f
Keq as ratio of, 62-63
PstI site, insertion of DNA at, 402, 403f
Pyrophosphate
Rate of degradation (kdeg), control of, 74
PTA. See Plasma thromboplastin antecedent
free energy of hydrolysis of, 82t
Rate-limiting reaction, metabolism egulated
PTC. See Plasma thromboplastin
inorganic, 85, 85f
by, 73
component
Pyrrole, 40, 41f
Rate of synthesis (ks), control of, 74
Pteroylglutamic acid. See Folic acid
Pyruvate, 123
Rb protein. See Retinoblastoma protein
PTH. See Parathyroid hormone
formation of, in amino acid carbon
Reactant concentration, chemical reaction
PTSs. See Peroxisomal-matrix targeting
skeleton catabolism, 250-255,
rate affected by, 62
sequences
252f, 253f
Reactive oxygen species. See Free radicals
684
/
INDEX
Rearrangements, DNA
Redox state, 184
collection and oxidation of reducing
in antibody diversity, 325-326, 393,
Reduced porphyrins, 272
equivalents and, 92-93, 93f, 94f,
593-594
Reducing equivalents
95f
recombinant DNA technology in
in citric acid cycle, 130-133, 132f
dehydrogenases in, 87
detection of, 409, 409t
in pentose phosphate pathway, 166
energy for metabolism provided by,
recA, 381, 382f
respiratory chain in collection and oxida-
93-95, 98f
Receptor-associated coactivator 3 (RAC3
tion of, 92-93, 93f, 94f, 95f
oxidative phosphorylation at level of, 94
coactivator), 472, 472t
5α-Reductase, 442, 444f
poisons affecting, 92, 95, 96f
Receptor-effector coupling, 435-436
Reduction, definition of, 86
as proton pump, 96, 96f, 97f
Receptor-mediated endocytosis, 429f,
Reductive activation, of molecular oxygen,
redox potential of components of,
430
627
92-93, 94f, 95f
Receptors, 431, 436. See also specific type
Refsum’s disease, 188, 503, 503t
substrates for, citric acid cycle providing,
activation of in signal generation,
Regional asymmetries, membrane, 420
131,131f
456-457, 458f
Regulated secretion, 498
Respiratory control, 81, 94-95, 97, 97t,
nuclear, 436, 469, 469-471, 471f, 472t
Regulatory proteins, binding of to DNA,
98f, 134-135
Recognition domains, on hormone
motifs for, 387-390, 388t, 389f,
Respiratory distress syndrome, surfactant
receptors, 435
390f, 391f
deficiency causing, 115, 202
Recombinant DNA/recombinant DNA
Regurgitation hyperbilirubinemia, 282
Restriction endonucleases/enzymes, 312,
technology, 396-414, 635t
Relaxation phase
397-399, 399t, 400f, 414
base pairing and, 396-397
of skeletal muscle contraction, 561, 564
in recombinant DNA technology,
blotting techniques in, 403, 404f
of smooth muscle contraction
399-400, 399t, 400f, 400t, 401f
chimeric molecules in, 397-406
calcium in, 571
Restriction enzymes. See Restriction
cloning in, 400-402, 401f, 402t, 403f
nitric oxide in, 571-573, 573f
endonucleases
definition of, 414
Relaxed (R) state, of hemoglobin,
Restriction fragment length polymorphisms
DNA ligase in, 399-400
oxygenation and, 43, 43f, 44f
(RFLPs), 57, 409-411, 411f
DNA sequencing in, 404, 405f
Releasing factors (RF1/RF3), in protein
in forensic medicine, 411
double helix structure and, 396, 397
synthesis termination, 369f, 370
Restriction map, 399
in enzyme study, 58, 59f
Remnant removal disease, 228t
Retention hyperbilirubinemia, 282
gene mapping and, 406-407, 407t
Renal glomerulus, laminin in basal lamina
Reticulocytes, in protein synthesis, 611
in genetic disease diagnosis, 407-412,
of, 540-542
Retina
408f, 409t, 410f, 411f
Renal threshold for glucose, 161
gyrate atrophy of, 250
hybridization techniques in, 403-404
Renaturation, DNA, base pair matching
retinaldehyde in, 483, 484f
libraries and, 402
and, 305-306
Retinal. See also Retinol
oligonucleotide synthesis in, 404-405
Renin, 451, 452f
Retinaldehyde, 482, 483f
organization of DNA into genes and,
Renin-angiotensin system, 451-452, 452f
Retinitis pigmentosa, essential fatty acid
397, 398f, 399t
Repeat sequences, 637
deficiency and, 192
polymerase chain reaction in, 405-406,
amino acid, 519, 520f
Retinoblastoma protein, 333
406f
short interspersed (SINEs), 321-322,
Retinoic acid, 482, 483f. See also Retinol
practical applications of, 406-412
414
functions of, 483
restriction enzymes and, 397-400, 399t,
Repetitive-sequence DNA, 320, 321-322
receptors for, 471, 483
400f, 400t, 401f
Replication/synthesis. See DNA,
Retinoic acid receptor (RAR), 471, 483
terminology used in, 413-414
replication/synthesis of;
Retinoic acid response element, 459t
transcription and, 397, 398f
RNA, synthesis of
Retinoid X receptor (RXR), 470, 470f, 471,
Recombinant erythropoietin (epoetin
Replication bubbles, 331-333, 331f, 332f,
483
alfa/EPO), 526, 610
333f
Retinoids, 482-484, 483f, 484f. See also
Recombinant fusion proteins, in enzyme
Replication fork, 327-328, 327f
Retinol
study, 58, 59f
Reporter genes, 385-386, 387f, 388f
Retinol, 482, 482t, 483f, 484f. See also
Recombination, chromosomal, 323-324,
Repression, enzyme
Vitamin A
323f, 324f
enzyme synthesis control and, 74
deficiency of, 482t
Recruitment hypothesis, of preinitiation
in gluconeogenesis regulation, 155-157
functions of, 482t, 483, 484f
complex formation, 352
Repressor protein/gene, lambda (cI),
Retinol-binding protein, 583t
Red blood cells, 609-610, 610-619. See
379-383, 380f, 381f, 382f
Retrograde transport, 505, 510
also Erythrocytes
Repressors, 348
from Golgi apparatus, 507
recombinant DNA technology in study
in gene expression, 374, 377, 378, 385
Retroposons/retrotransposons, 321, 637
of, 624
tissue-specific expression and, 385
Retroviruses, reverse transcriptases in, 308,
Red thrombus, 598
Reproduction, prostaglandins in, 190
332-333
Red (slow) twitch fibers, 574-576, 575t
Respiration, 86
Reverse cholesterol transport, 210, 211f,
Redox (oxidation-reduction) potential, 86,
Respiratory burst, 479, 622-623
219, 224
87t
Respiratory chain, 92-101. See also
Reverse transcriptase/reverse transcription,
of respiratory chain components, 92-93,
Oxidative phosphorylation
308, 333, 414
94f, 95f
clinical aspects of, 100-101
in recombinant DNA technology, 400t
INDEX
/
685
Reversed-phase high-pressure
classes/species of, 307-308, 309t, 341,
RNA polymerases, DNA-dependent, in
chromatography, for protein/
342t
RNA synthesis, 342-343, 342f,
peptide purification, 23-24
complementarity of, 306, 309f
343t
Reversible covalent modifications, 77-79,
heterogeneous nuclear (hnRNA), 310
RNA primer, in DNA synthesis, 328, 329f,
78f, 78t. See also Phosphorylation
gene regulation and, 354
330f
Reye’s syndrome, orotic aciduria in, 300
messenger (mRNA), 307, 309-310,
RNA probes, 402, 414
RFLPs. See Restriction fragment length
310f, 311f, 341, 342t, 359
RNAP. See RNA polymerases
polymorphisms
alternative splicing and, 354, 354f,
RNase. See Ribonucleases
RFs. See Releasing factors
393-394, 636
ROS (reactive oxygen species). See Free
Rheumatoid arthritis, glycosylation
codon assignments in, 358, 359t
radicals
alterations in, 533
editing of, 356
Rotor syndrome, 283
Rho-dependent termination signals, 344,
expression of, detection of in gene
Rough endoplasmic reticulum
346, 346f
isolation, 635t
glycosylation in, 524-525, 525f
Rhodopsin, 483, 484f
modification of, 355-356
in protein sorting, 498, 499f, 500f
Riboflavin (vitamin B2), 86, 482t, 489-490
nucleotide sequence of, 358
protein synthesis and, 370
in citric acid cycle, 133
mutations caused by changes in,
routes of protein insertion into,
coenzymes derived from, 50-51, 489, 490
361-363, 361f, 362f, 364f
505-507, 506f
deficiency of, 482t, 490
polycistronic, 376
signal hypothesis of polyribosome
dehydrogenases dependent on, 87
recombinant DNA technology and,
binding to, 503-505, 504t, 505f
Ribonucleases, 312
397
rRNA. See Ribosomal RNA
Ribonucleic acid. See RNA
relationship of to chromosomal DNA,
RT-PCR, 414
Ribonucleoside diphosphates (NDPs),
321f
RXR. See Retinoid X receptor
reduction of, 294, 297f
stability of, regulation of gene
Ryanodine, 563
Ribonucleosides, 286, 287f
expression and, 394-395, 394f
Ryanodine receptor, 563, 564f
Ribonucleotide reductase complex, 294,
transcription starting point and,
mutations in gene for, diseases caused by,
297f
342
564-565, 565f, 630t
Ribose, 102
variations in size/complexity of, 397,
RYR. See Ryanodine receptor
in nucleosides, 286, 287f
399t
pentose phosphate pathway in
modification of, 355-356
production of, 123, 163, 166
processing of, 352-355
S50, 67
D-Ribose, 104f, 105t, 286
alternative, in regulation of gene
S phase of cell cycle, DNA synthesis during,
Ribose phosphate, pentose phosphate path-
expression, 354, 355f, 393-394
333-335, 334f, 335t
way in production of, 163,
in protein synthesis, 307-308, 309t
Saccharopine, in lysine catabolism, 256f,
164f
ribosomal (rRNA), 307-308, 310-311,
258
Ribose 5-phosphate, in purine synthesis,
341, 342t
Salt (electrostatic) bonds (salt bridges/
293-294, 295f
as peptidyltransferase, 368, 370t
linkages), 7
Ribose 5-phosphate ketoisomerase, 163,
processing of, 355
oxygen binding rupturing, Bohr effect
165f
small nuclear (snRNA), 308, 309t, 311,
protons and, 44-45, 45f
Ribosomal dissociation, in protein
341, 342t, 414
“Salvage” reactions
synthesis, 365, 366f
small stable, 311
in purine synthesis, 294, 295f, 297f
Ribosomal RNA (rRNA), 307-308,
splicing, 352-354, 414
in pyrimidine synthesis, 296
310-311, 341, 342t. See also
alternative, in regulation of gene
Sanfilippo syndrome, 546t
RNA
expression, 354, 354f,
Sanger’s method
as peptidyltransferase, 368, 370t
393-394, 636
for DNA sequencing, 404, 405f
processing of, 355
recombinant DNA technology and,
for polypeptide sequencing, 24-25
Ribosomes, 310, 312t
397, 398f
Sanger’s reagent (1-fluoro-2,4-dinitro-
bacterial, 371-372
structure of, 306-312, 308f, 309f, 311f,
benzene), for polypeptide
protein synthesis in, 126, 127f
312f
sequencing, 25
dissociation and, 370
synthesis of, 341-352
Sarcolemma, 556
Ribozymes, 308, 311, 356
initiation/elongation/termination in,
Sarcomere, 556-557, 557f
D-Ribulose, 105t, 106f
342, 342f, 343-344, 344f
Sarcoplasm, 556
Ribulose 5-phosphate 3-epimerase, 163,
transfer (tRNA), 308, 310, 312f, 341,
of cardiac muscle, 566
165f
342t, 360-361, 361f
Sarcoplasmic reticulum, calcium level in
Richner-Hanart syndrome, 255
aminoacyl, in protein synthesis, 368
skeletal muscle and, 563-564,
Ricin, 372, 518t
anticodon region of, 359
563f, 564f
Rickets, 482t, 484, 551t
processing and modification of, 355,
Saturated fatty acids, 111, 112, 112t
Right operator, 379-383, 380f, 382f
356
in membranes, 417, 418f
Rigor mortis, 562, 564
suppressor, 363
Saturation kinetics, 64f, 66
RNA, 303, 306-312, 341-357
xenobiotic cell injury and, 631
sigmoid substrate, Hill equation in
as catalyst, 356
RNA editing, 356
evaluation of, 66-67, 67f
in chromatin, 314
RNA polymerase III, 343t
Scavenger receptor B1, 210, 211f
686
/
INDEX
Scheie syndrome, 546t
in glycine synthesis, 238, 239f
Signal hypothesis, of polyribosome binding,
Schindler disease, 532-533, 533t
phosphorylated, 264
503-505, 504t, 505f
Scrapie, 37
synthesis of, 238, 238f
Signal peptidase, 504, 505f
Scurvy, 482t, 496
tetrahydrofolate and, 492-494, 493f
Signal peptide, 498, 503-504, 508t
collagen affected in, 38-39, 496,
Serine 195, in covalent catalysis, 53-54, 54f
albumin, 583
538-539
Serine hydroxymethyltransferase, 250, 252f,
in protein sorting, 498-499, 499f, 500f,
SDS-PAGE. See Sodium dodecyl
493-494
503-504, 505, 505f
sulfate-polyacrylamide gel
Serine protease inhibitor, 589. See also
Signal recognition particle, 504
electrophoresis
α1-Antiproteinase
Signal sequence. See Signal peptide
Se gene, 618
Serine proteases. See also specific type
Signal transducers and activators of
Sec1 proteins, 511
conserved residues and, 54, 55t
transcription (STATs), 467, 467f
Sec61p complex, 504
in covalent catalysis, 53-54, 54f
Signal transduction, 456-473
Second messengers, 76, 436-437, 437t,
zymogens of, in blood coagulation, 600,
GPI-anchors in, 528
457-468, 461t, 463t. See also
600t, 601
hormone response to stimulus and, 456,
specific type
Serotonin, 266-267, 621t
457f
calcium as, 436-437, 437t, 457
Serpin, 589. See also α1-Antiproteinase
intracellular messengers in, 457-468,
cAMP as, 147, 436, 437t, 457, 458-462,
Serum prothrombin conversion accelerator
461t, 463t. See also specific type
460t, 462f
(SPCA/factor VII), 599f, 600t,
in platelet activation, 606, 606f
cGMP as, 290, 436, 437t, 457, 462-463
601
signal generation and, 456-457, 458f,
diacylglycerol as, 464, 465f
coumarin drugs affecting, 604
459f, 459t
inositol trisphosphate as, 464-465, 464f,
Sex (gender), xenobiotic-metabolizing
transcription modulation and, 468-473,
465f
enzymes affected by, 630
470f, 471f, 472t
precursors of
Sex hormone-binding globulin
Silencers, 348
phosphatidylinositol as, 115, 115f
(testosterone-estrogen-binding
recombinant DNA technology and, 397
phospholipids as, 197
globulin), 455, 455t, 583t
Silencing mediator for RXR and TR
Secondary structure, 31, 31-33, 32f, 33f,
SGLT 1 transporter protein, 475, 475f
(SMRT), 472t, 473
34f
SGOT. See Aspartate aminotransferase
Silent mutations, 361
peptide bonds affecting, 31, 31f
SGPT. See Alanine aminotransferase
Silicon, 496t
supersecondary motifs and, 33
SH2 domains. See Src homology 2 (SH2)
Simple diffusion, 423, 423t, 424f
Secretor (Se) gene, 618
domains
Simvastatin, 229
Secretory component, of IgA, 595f
SHBG. See Sex hormone-binding globulin
SINEs. See Short interspersed repeat
Secretory granules, protein entry into, 507,
Short interspersed repeat sequences
sequences
507f
(SINEs), 321-322, 414
Single displacement reactions, 69, 69f
Secretory (exocytotic) pathway, 498
Shoshin beriberi, 489
Single nucleotide polymorphism (SNP), 414
Secretory vesicles, 498, 500f
Shotgun sequencing, 634
Single-pass membrane proteins,
D-Sedoheptulose, 106f
SI nuclease, in recombinant DNA
glycophorins as, 615-616,
Selectins, 528-530, 529f, 529t, 530f
technology, 400t
615f, 616f, 616t
Selectivity/selective permeability,
Sialic acids, 110, 110f, 116, 169, 171f
Single-stranded DNA, replication from,
membrane, 415, 423-426,
in gangliosides, 171f, 201, 203f
326. See also DNA,
423t, 424f, 425f, 426t
in glycoproteins, 109t, 516t
replication/synthesis of
Selenium, 496t
Sialidosis, 532-533, 533t, 546, 546t
Single-stranded DNA-binding proteins
in glutathione peroxidase, 88, 166
Sialoprotein, bone, 548t, 550
(SSBs), 326, 327, 327f, 328t
Selenocysteine, synthesis of, 240, 240f
Sialyl-LewisX, selectins binding, 530, 530f
Sister chromatid exchanges, 325, 325f
Selenophosphate synthetase/synthase, 240,
Sialylated oligosaccharides, selectins
Sister chromatids, 318, 319f
240f
binding, 530
Site-directed mutagenesis, in enzyme study,
Self-assembly
Sickle cell disease, 363, 619
58
in collagen synthesis, 537
pedigree analysis of, 409, 410f
Site-specific DNA methylases, 398
of lipid bilayer, 418
recombinant DNA technology in
Site specific integration, 324
Self-association, hydrophobic interactions
detection of, 408-409
Sitosterol, for hypercholesterolemia, 229
and, 6-7
Side chain cleavage enzyme P450
Size exclusion chromatography, for
Sensory neuropathy, in vitamin B6 excess,
(P450scc), 438, 440f, 442
protein/peptide purification,
491
Side chains, in porphyrins, 270, 271f
21-22, 23f
Sepharose-lectin column chromatography,
Sigmoid substrate saturation kinetics, Hill
SK. See Streptokinase
in glycoprotein analysis, 515t
equation in evaluation of, 66-67,
Skeletal muscle, 556, 568t. See also Muscle;
Sequential displacement reactions, 69, 69f
67f
Muscle contraction
Serine, 15t
Signal. See also Signal peptide
glycogen stores in, 573
catabolism of, pyruvate formation and,
generation of, 456-457, 458f, 459f, 459t
metabolism in, 125, 125f
250, 252f
in recombinant DNA technology, 414
lactate production and, 139
conserved residues and, 54, 55t
transmission of. See also Signal
as protein reserve, 576
in cysteine and homoserine synthesis,
transduction
slow (red) and fast (white) twitch fibers
239, 239f
across membrane, 415, 431
in, 574-576, 575t
INDEX
/
687
Skin
Sodium-potassium pump (Na+-K+ ATPase),
SR-B1. See Scavenger receptor B1
essential fatty acid deficiency affecting,
427-428, 428f
SRC-1 coactivator, 472, 472t
194-195
in glucose transport, 428, 429f
Src homology 2 (SH2) domains
mutant keratins and, 578
Solubility point, of amino acids, 18
in insulin signal transmission, 465, 466f,
vitamin D3 synthesis in, 445, 446f, 484,
Soluble NSF attachment factor (SNAP)
467
485f
proteins, 509, 510f, 511
in Jak/STAT pathway, 467, 467f
Sleep, prostaglandins in, 190
Solutions, aqueous, Kw of, 9
SRP. See Signal recognition particle
Sliding filament cross-bridge model, of
Solvent, water as, 5, 6f
SRS-A. See Slow-reacting substance of
muscle contraction, 557-559,
Sorbitol, in diabetic cataract, 172
anaphylaxis
558f
Sorbitol dehydrogenase, 167, 169f
ssDNA. See Single-stranded DNA
Slow acetylators, 630
Sorbitol intolerance, 172
Staphylococcus aureus V8 protease, for
Slow-reacting substance of anaphylaxis, 196
Sorbitol (polyol) pathway, 172
polypeptide cleavage, 25, 26t
Slow (red) twitch fibers, 574-576, 575t
Soret band, 273
STAR. See Steroidogenic acute regulatory
Sly syndrome, 546t
Southern blot transfer procedure, 305-306,
protein
Small intestine
403, 404f, 414
Starch, 107, 108f
cytochrome P450 isoforms in, 627
Southwestern blot transfer procedure, 403,
glycemic index of, 474
monosaccharide digestion in, 475, 475f
414
hydrolysis of, 474
Small nuclear RNA (snRNA), 308, 309t,
SPARC (bone) protein, 548t
Starling forces, 580
311, 341, 342t, 414
Sparteine, CYP2D6 in metabolism of,
Starvation, 80
Small nucleoprotein complex (snurp), 353
628
clinical aspects of, 236
Small stable RNA, 311
SPCA. See Serum prothrombin conversion
fatty liver and, 212
Smoking
accelerator
ketosis in, 188
CYP2A6 metabolism of nicotine and,
Specific acid/base catalysis, 51-52
metabolic fuel mobilization in, 232-234,
628
Specificity, enzyme, 49, 50f
234f, 234t
cytochrome P450 induction and, 628
Spectrin, 615, 615f, 616f, 616t, 617
triacylglycerol redirection and, 208
nucleotide excision-repair of DNA
abnormalities of, 617
Statin drugs, 229
damage caused by, 337
Spectrometry
STATs (signal transducers and activators of
Smooth endoplasmic reticulum, cy-
covalent modifications detected by, 27,
transcription), 467, 467f
tochrome P450 isoforms in,
27f, 27t
Stearic acid, 112t
627
for glycoprotein analysis, 514, 515t
Steely hair disease (Menkes disease), 588
Smooth muscle, 556, 568t
Spectrophotometry
Stem cells, differentiation of to red blood
actin-myosin interactions in, 572t
for NAD(P)+-dependent dehydrogenases,
cells, erythropoietin in regulation
contraction of
56, 56f
of, 610, 611f
calcium in, 570-571, 571f
for porphyrins, 273-274
Stereochemical (-sn-) numbering system,
myosin-based regulation of, 570
Spectroscopy, nuclear magnetic resonance
114, 115f
myosin light chain phosphorylation in,
(NMR)
Stereoisomers. See also Isomerism
570
for glycoprotein analysis, 514, 515f
of steroids, 117, 118f
relaxation of
protein structure demonstrated by,
Steroid nucleus, 117, 117f, 118f
calcium in, 571
35-36
Steroid receptor coactivator 1 (SRC-1
nitric oxide in, 571-573, 573f
Spermidine, synthesis of, 265-266, 266f
coactivator), 472, 472t
SMRT, 472t, 473
Spermine, synthesis of, 265-266, 266f
Steroid sulfates, 201
SNAP (soluble NSF attachment factor)
Spherocytosis, hereditary, 432t, 617, 617f
Steroidogenesis. See Steroids, synthesis of
proteins, 509, 510f
Sphingolipidoses, 202-203, 203t
Steroidogenic acute regulatory protein
SNAP 25, 511
Sphingolipids, 197
(STAR), 442
SNARE proteins, 509, 510f, 511
metabolism of, 201-202, 202f, 203f
Steroids, 117-118, 117f, 118f, 119f. See
SNAREpins, 511
clinical aspects of, 202-203, 203t
also specific type
SNP. See Single nucleotide polymorphism
in multiple sclerosis, 202
adrenal. See also Glucocorticoids;
snRNA. See Small nuclear RNA
Sphingomyelins, 116, 116f, 201, 202f
Mineralocorticoids
Snurp (small nucleoprotein [snRNP]
in membranes, 417
synthesis of, 438-442, 440f, 441f
complex), 353
membrane asymmetry and, 420
calcitriol as, 484
Sodium, 496t
Sphingophospholipids, 111
receptors for, 436
in extracellular and intracellular fluid,
Sphingosine, 116, 116f
stereoisomers of, 117, 118f
416, 416t
Spina bifida, folic acid supplements in
storage/secretion of, 453, 454t
permeability coefficient of, 419f
prevention of, 494
synthesis of, 123f, 124, 438, 438-445,
Sodium-calcium exchanger, 463
Spliceosome, 353, 414
439t, 440f, 441f
Sodium dodecyl sulfate-polyacrylamide gel
Spongiform encephalopathies, transmissible
transport of, 454-455, 455t
electrophoresis
(prion diseases), 37
vitamin D as, 484
for protein/peptide purification, 24, 24f,
Squalene, synthesis of, in cholesterol
Sterol 27-hydroxylase, 226
25f
synthesis, 219, 221f, 222f
Sterols, 117
red cell membrane proteins determined
Squalene epoxidase, in cholesterol synthesis,
in membranes, 417
by, 614-615, 615f
220, 222f
Stickler syndrome, 553
688
/
INDEX
Sticky end ligation/sticky-ended DNA, 299,
Sucrose, 106-107, 107f, 107t
t-SNARE proteins, 509, 511
398, 400f, 401f, 414
glycemic index of, 474
T (taut) state, of hemoglobin
“Sticky foot,” 527
Sugars. See also Carbohydrates
2,3-bisphosphoglycerate stabilizing, 45,
“Sticky patch,” in hemoglobin S, 46, 46f
amino (hexosamines), 106, 106f
45f
Stoichiometry, 60
glucose as precursor of, 169, 171f
oxygenation and, 43, 43f, 44f
Stokes radius, in size exclusion
in glycosaminoglycans, 109, 169, 171f
T tubular system, in cardiac muscle, 566
chromatography, 21
in glycosphingolipids, 169, 171f
T-type calcium channel, 567
Stop codon, 369f, 370
interrelationships in metabolism of,
TAFs. See TBP-associated factors
Stop-transfer signal, 506
171f
Talin, 540, 541f
Strain, catalysis by, 52
classification of, 102, 102t
Tandem, 414
Streptokinase, 605, 605f, 606t
deoxy, 106, 106f
Tandem mass spectrometry, 27
Streptomycin, 106
“invert,” 107
Tangier disease, 228t
Striated muscle, 556, 557, 557f. See also
isomerism of, 102-104, 103f
TaqI, 399t
Cardiac muscle; Skeletal muscle
nucleotide, in glycoprotein biosynthesis,
Target cells, 434-435, 435t
actin-myosin interactions in, 572t
516-517, 516t
receptors for, 435, 436f
Stroke, with mitochondrial encephalopathy
“Suicide enzyme,” cyclooxygenase as, 194
Targeted gene disruption/knockout, 412
and lactic acidosis (MELAS),
Sulfate
Tarui’s disease, 152t
100-101
active (adenosine 3′-phosphate-5′-
TATA binding protein, 346, 349f, 350,
Strong acids, 9
phosphosulfate), 289, 289f, 629
351
Strong bases, 9
in glycoproteins, 515
TATA box, in transcription control, 345,
Structural proteins, 535
in mucins, 520
345f, 346, 347f, 348, 348f, 351t
Stuart-Prower factor (factor X), 599f, 600,
Sulfatide, 116
Taurochenodeoxycholic acid, synthesis of,
600t
Sulfation, of xenobiotics, 629
226f
activation of, 599-600, 599f
Sulfo(galacto)-glycerolipids, 201
Taut (T) state, of hemoglobin
coumarin drugs affecting, 604
Sulfogalactosylceramide, 201
2,3-bisphosphoglycerate stabilizing, 45,
Substrate analogs, competitive inhibition
accumulation of, 203
45f
by, 67-68, 67f
Sulfonamides, hemolytic anemia
oxygenation and, 43, 43f, 44f
Substrate level, phosphorylation at, 94
precipitated by, 613
Tay-Sachs disease, 203t
Substrate shuttles
Sulfonylurea drugs, 188
TBG. See Thyroxine-binding globulin
coenzymes as, 50
Sulfotransferases, in glycosaminoglycan
TBP. See TATA binding protein
in extramitochondrial NADH oxidation,
synthesis, 543
TBP-associated factors, 346, 350, 351
99, 100f
Sunlight. See Ultraviolet light
TΨC arm, of tRNA, 310, 312f, 360, 361f
Substrate specificity, of cytochrome P450
Supercoils, DNA, 306, 332, 333f
TEBG. See Testosterone-estrogen-binding
isoforms, 627
Superoxide anion free radical, 90-91,
globulin
Substrates, 49
611-613, 613t. See also Free
Telomerase, 318
competitive inhibitors resembling,
radicals
Telomeres, 318, 319f
67-68, 67f
production of in respiratory burst, 622
Temperature
concentration of, enzyme-catalyzed
Superoxide dismutase, 90-91, 119,
chemical reaction rate affected by, 62,
reaction rate affected by, 64,
611-613, 613t, 622
62f
64f, 65f
Supersecondary structures, 33
enzyme-catalyzed reaction rate affected
Hill model of, 66-67, 67f
Suppressor mutations, 363
by, 63
Michaelis-Menten model of, 65-66,
Suppressor tRNA, 363
in fluid mosaic model of membrane
66f
Surfactant, 115, 197
structure, 422
conformational changes in enzymes
deficiency of, 115, 202
Temperature coefficient (Q10), enzyme-
caused by, 52, 53f
SV40 viruses, cancer caused by
catalyzed reactions and, 63
multiple, 69-70
Swainsonine, 527, 527t
Template binding, in transcription, 342,
Succinate, 131-133, 132f
Symport systems, 426, 426f
342f
Succinate dehydrogenase, 87, 132f,
Syn conformers, 287, 287f
Template strand DNA, 304, 306, 307f
133
Synaptobrevin, 511
transcription of in RNA synthesis,
inhibition of, 67-68, 67f
Syntaxin, 511
341-343, 342f
Succinate semialdehyde, 267, 268f
Synthesis, rate of (ks), control of, 74
Tenase complex, 600-601
Succinate thiokinase (succinyl-CoA
Terminal transferase, 400t, 414
synthetase), 131, 132f
Termination
Succinic acid, pK/pKa value of, 12t
t1/2. See Half life
chain
Succinyl-CoA, in heme synthesis, 270-273,
T3. See Triiodothyronine
in glycosaminoglycan synthesis, 543
273f, 274f, 275f, 276f
T4. See Thyroxine
in transcription cycle, 342, 342f
Succinyl-CoA-acetoacetate-CoA transferase
Tm. See Melting temperature/transition
of protein synthesis, 369f, 370
(thiophorase), 133, 186, 186f
temperature
of RNA synthesis, 342, 342f, 344, 344f
Succinyl-CoA synthetase (succinate
T lymphocytes, 591
Termination signals, 359
thiokinase), 131, 132f
t-PA. See Tissue plasminogen activator
for bacterial transcription, 346, 346f
Sucrase-isomaltase complex, 475
TΨC arm, of tRNA, 310, 312f, 360, 361f
for eukaryotic transcription, 349-350
INDEX
/
689
Tertiary structure, 33-35, 35f
Thiamin triphosphate, 489
Thyroglobulin, 447, 449
stabilizing factors and, 35
Thick (myosin) filaments, 557, 558f
Thyroid-binding globulin, 454, 583t
Testes, hormones produced by, 437, 442,
Thin (actin) filaments, 557, 558f, 559f
Thyroid hormone receptor-associated
443f. See also specific type
Thioesterase, 173
proteins (TRAPs), 472t, 473
Testosterone, 439f, 440f
6-Thioguanine, 290, 291f
Thyroid hormone response element, 459t
binding of, 455, 455t
Thiokinase (acyl-CoA synthetase)
storage/secretion of, 453, 454t
metabolism of, 442, 444f
in fatty acid activation, 180, 181f
Thyroid hormones, 437, 438
synthesis of, 442, 443f
in triacylglycerol synthesis, 199, 214f,
in lipolysis, 215, 216f
Testosterone-estrogen-binding globulin (sex
215
receptors for, 436, 471
hormone-binding globulin), 455,
Thiol-dependent transglutaminase. See
synthesis of, 447-449, 448f
455t, 583t
Transglutaminase
transport of, 454, 454t
Tetracycline (tet) resistance genes, 402,
Thiol ester plasma protein family, 590
Thyroid-stimulating hormone (TSH), 437,
403f
Thiolase, 181, 182f, 184
438, 439f, 449
Tetrahedal transition state intermediate, in
in mevalonate synthesis, 219, 220f
Thyroperoxidase, 449
acid-base catalysis, 52, 53f
Thiophorase (succinyl-CoA-
Thyrotropin-releasing hormone (TRH),
Tetrahydrofolate, 492, 493-494, 493f
acetoacetate-CoA transferase),
438, 439f
Tetraiodothyronine (thyroxine/T4), 438,
133, 186, 186f
Thyroxine (T4), 438, 447
447
Thioredoxin, 294
storage/secretion of, 453, 454t
storage/secretion of, 453, 454t
Thioredoxin reductase, 294, 297f
synthesis of, 447-449, 448f
synthesis of, 447-449, 448f
Threonine, 15t
transport of, 454, 454t
transport of, 454, 454t
catabolism of, 253f, 255
Thyroxine-binding globulin, 454, 454t
Tetramers
phosphorylated, 264
TIF2 coactivator, 472, 472t
hemoglobin as, 42
requirements for, 480
Tiglyl-CoA, catabolism of, 261f
histone, 314-315, 315
Thrombin, 601, 602, 603f
TIM. See Translocase-of-the-inner
Tetroses, 102, 102t
antithrombin III affecting, 603-604
membrane
Tf. See Transferrin
circulating levels of, 602-603
Timnodonic acid, 113t
TFIIA, 350
conserved residues and, 55t
Tin, 496t
TFIIB, 350
formation of fibrin and, 601-602, 603f
Tissue differentiation, retinoic acid in, 483
TFIID, 346, 350, 351
in platelet activation, 606, 606f
Tissue factor complex, 601
in preinitiation complex formation, 352
Thrombolysis
Tissue factor (factor III), 599f, 600t, 601
TFIIE, 350
laboratory tests in evaluation of, 608
Tissue factor pathway inhibitor, 601
TFIIF, 350
t-PA and streptokinase in, 605, 605f,
Tissue plasminogen activator (alteplase/
TFIIH, 350
606t
t-PA), 604-605, 605, 605f,
TFPI. See Tissue factor pathway inhibitor
Thrombomodulin, in blood coagulation,
606t, 607t
TfR. See Transferrin receptor
600t, 603, 607, 607t
Tissue-specific gene expression, 385
Thalassemias, α and β, 47, 610t
Thrombosis, 598-608. See also Coagulation
Titin, 566t
recombinant DNA technology in
antithrombin III in prevention of,
TMP (thymidine monophosphate), 288f,
detection of, 408f, 409, 409t
603-604
288t
Thanatophoric dysplasia, 551t
circulating thrombin levels and, 602-603
Tocopherol, 482t, 486, 486f. See also
Theca cells, hormones produced by, 442
endothelial cell products in, 607, 607t
Vitamin E
Theobromine, 289
hyperhomocysteinemia and, folic acid
as antioxidant, 91, 119, 486, 487f
Theophylline, 289
supplements in prevention of, 494
deficiency of, 482t
hormonal regulation of lipolysis and, 215
phases of, 598
Tocotrienol, 486, 486f. See also Vitamin E
Thermodynamics
in protein C or protein S deficiency, 603
Tolbutamide, 188
biochemical (bioenergetics), 80-85. See
t-PA and streptokinase in management
TOM. See Translocase-of-the-outer
also ATP
of, 605, 605f, 606t
membrane
glycolysis reversal and, 153-155
types of thrombi and, 598
Topogenic sequences, 506
laws of, 80-81
Thromboxane A2, 113f
Topoisomerases, DNA, 306, 328t, 332,
hydrophobic interactions and, 7
in platelet activation, 606f, 607
332f
Thermogenesis, 217, 217f
Thromboxanes, 112, 113f, 190, 192
Total iron-binding capacity, 586
diet-induced, 217, 478
clinical significance of, 196
Toxemia of pregnancy of ewes, ketosis and,
Thermogenin, 217, 217f
cyclooxygenase pathway in formation of,
188
Thiamin (vitamin B1), 482t, 488-489, 489f
192, 193f
Toxic hyperbilirubinemia, 283
in citric acid cycle, 133
Thymidine, 288t
Toxopheroxyl free radical, 486
coenzymes derived from, 51
base pairing of in DNA, 303, 304, 305f
TpC. See Troponin C
deficiency of, 482t, 489
Thymidine monophosphate (TMP), 288t
TpI. See Troponin I
pyruvate metabolism affected by, 140,
Thymidine-pseudouridine-cytidine (TΨC)
TpT. See Troponin T
143, 489
arm, of tRNA, 310, 312f, 360,
TR activator molecule 1 (TRAM-1
Thiamin diphosphate, 140, 166, 488-489,
361f
coactivator), 472, 472t
489f
Thymidylate, 303
TRAM (translocating chain-associated
Thiamin pyrophosphate, 50
Thymine, 288t
membrane) protein, 504
690
/
INDEX
TRAM-1 coactivator, 472, 472t
Transferrin, 478, 583t, 584-586, 585f,
Transposition, 324-325
Trans fatty acids, 113-114, 192
585t
retroposons/retrotransposons and, 321,
Transaldolase, 166
Transferrin receptor, 586
637
Transaminases. See Aminotransferases
Transfusion, ABO blood group and, 618
Transthyretin, 583t, 590
Transamination, 124, 124f
Transgenic animals, 385, 411-412, 414
Transverse asymmetry, 511
in amino acid carbon skeleton catabo-
enhancers/regulatory elements identified
Transversion mutations, 361, 361f
lism, 249-250, 249f, 250f, 251f
in, 386
TRAPs, 472t, 473
citric acid cycle in, 133-134, 134f
Transglutaminase, in blood coagulation,
Trauma, protein loss and, 480
in urea biosynthesis, 243-244, 243f
600, 600t, 602, 603f
TRE. See Thyroid hormone response
Transcortin (corticosteroid-binding
Transhydrogenase, proton-translocating, as
element
globulin), 454-455, 455t
source of intramitochondrial
Trehalase, 475
Transcript profiling, 412
NADPH, 99
Trehalose, 107t
Transcription, 306, 350-352, 351t, 414
Transient insertion signal. See Signal
TRH. See Thyrotropin-releasing hormone
activators and coactivators in control of,
peptide
Triacylglycerols (triglycerides), 114, 115f,
351, 351t
Transition mutations, 361, 361f
205
bacterial promoters in, 345-346, 345f
Transition state intermediate, tetrahedal, in
digestion and absorption of, 475-477,
control of fidelity and frequency of,
acid-base catalysis, 52, 53f
476f
344-350
Transition states, 61
excess of. See Hypertriacylglycerolemia
eukaryotic promoters in, 346-349, 347f,
Transition temperature/melting
interconvertability of, 231
348f, 349f
temperature (Tm), 305, 422
in lipoprotein core, 205, 207f
in gene expression regulation, 383-387,
Transketolase, 163-166, 165f, 170
metabolism of, 123, 123f, 125-126,
391, 392t. See also Gene
erythrocyte, in thiamin nutritional status
126f
expression
assessment, 489
in adipose tissue, 214-215, 214f
hormonal regulation of, 457, 458f,
thiamin diphosphate in reactions
fatty liver and, 212, 213f
468-473, 470f, 471f, 472t
involving, 166, 170, 488-489
hepatic, 211-212, 213f
initiation of, 342-343, 342f
Translation, 358, 414
high-density lipoproteins in,
NF-κB in regulation of, 468, 469f
Translocase-of-the-inner membrane, 499
209-211, 211f
nuclear receptor coregulators in,
Translocase-of-the-outer membrane, 499
hydrolysis in, 197
471-473, 472t
Translocating chain-associated membrane
reduction of serum levels of, drugs for,
recombinant DNA technology and, 397,
(TRAM) protein, 504
229
398f
Translocation, protein, 499
synthesis of, 198f, 199
retinoic acid in regulation of, 483
Translocation complexes, 499
transport of, 207, 208f, 209f, 210f
reverse, 414
Translocon, 504
Tricarboxylate anions, transporter systems
in retroviruses, 308, 332-333
Transmembrane proteins, 419
for, 98-99
in RNA synthesis, 306, 307f, 341-343,
ion channels as, 423-424, 425f, 426t
lipogenesis regulation and, 178
342f
in red cells, 615-616, 615f, 616f, 616t
Tricarboxylic acid cycle. See Citric acid
Transcription complex, eukaryotic, 306,
Transmembrane signaling, 415, 431
cycle
350-352, 351t
in platelet activation, 606, 606f
Triglycerides. See Triacylglycerols
Transcription control elements, 351, 351t
Transmissible spongiform encephalopathies
Triiodothyronine (T3), 438, 447
Transcription domains, definition of,
(prion diseases), 37
storage/secretion of, 453, 454t
387
Transport proteins, 454-456, 454t, 455t,
synthesis of, 447-449, 448f
Transcription factors, 351, 351t
583t
transport of, 454, 454t
nuclear receptor superfamily, 469-471,
Transport systems/transporters. See also
Trimethoprim, 494
471f, 472t
specific type
Trinucleotide repeat expansions, 322
Transcription start sites, alternative,
active, 423, 423t, 424f, 426-427,
Triokinase, 167, 169f
393-394
427-428, 428f
Triose phosphates, acylation of, 123
Transcription unit, 342, 345f
ADP/ATP, 95, 98f
Trioses, 102, 102t
Transcriptional intermediary factor 2 (TIF2
ATP-binding cassette, 210, 211f
Triphosphates, nucleoside, 287, 287f
coactivator), 472, 472t
in cotranslational insertion, 506, 506f
Triple helix structure, of collagen, 38, 38f,
Transcriptome information, 412, 414
disorders associated with mutations in
535-539, 536f
Transfection, identification of
genes encoding, 512t, 513
Triplet code, genetic code as, 358, 359t
enhancers/regulatory elements
exchange, 98-100, 98f, 99f
tRNA. See Transfer RNA
and, 386
facilitated diffusion, 423, 423t, 424f,
Tropocollagen, 38, 38f
Transfer RNA (tRNA), 308, 310, 312f,
426-427, 427, 427f
Tropoelastin, 539
341, 342t, 360-361, 361f. See
glucose. See Glucose transporters
Tropomyosin, 557, 559f, 562
also RNA
in inner mitochondrial membrane,
in red cell membranes, 616t
aminoacyl, in protein synthesis, 368
98-100, 98f, 99f
as striated muscle inhibitor, 563
anticodon region of, 359
membrane, 426-431, 426f
Troponin/troponin complex, 557, 559f,
processing and modification of, 355, 356
for nucleotide sugars, 517
562
suppressor, 363
Transport vesicles, 498, 508-511, 509t,
as striated muscle inhibitor, 563
Transferases, 50
510f
Troponin C, 562
INDEX
/
691
Troponin I, 562
Ubiquinone (Q/coenzyme Q), 92, 95f, 118
Uridine diphosphate N-acetylgalactosamine
Troponin T, 562
in cholesterol synthesis, 220, 221f
(UDP-GalNAc), 516t
Trypsin, 477
UDP-glucose. See Uridine diphosphate
Uridine diphosphate N-acetylglucosamine
conserved residues and, 55t
glucose
(UDP-GlcNAc), 516t
in digestion, 477
UDPGal. See Uridine diphosphate galactose
Uridine diphosphate galactose (UDPGal),
for polypeptide cleavage, 25, 26t
UDPGlc. See Uridine diphosphate glucose
167, 516-517, 516t
Trypsinogen, 477
UFA (unesterified fatty acids). See Free fatty
Uridine diphosphate galactose (UDPGal)
Tryptophan, 16t, 266-267, 490
acids
4-epimerase, 167, 170f
catabolism of, 257f, 258, 258f
Ulcers, 474
inherited defects in, 172
deficiency of, 490
Ultraviolet light
Uridine diphosphate glucose
niacin synthesized from, 490
nucleotide absorption of, 290
(UDP/UDPGlc), 145, 147f,
permeability coefficient of, 419f
nucleotide excision-repair of DNA
516, 516t
requirements for, 480
damage caused by, 337
in glycogen biosynthesis, 145, 146f
Tryptophan oxygenase/L-tryptophan
vitamin D synthesis and, 484, 485f
Uridine diphosphate glucose dehydroge-
oxygenase (tryptophan
UMP (uridine monophosphate), 288f, 288t
nase, 166, 168f
pyrrolase), 89, 257f, 258
Uncouplers/uncoupling proteins
Uridine diphosphate glucose
TSEs. See Transmissible spongiform
in respiratory chain, 95, 96f
pyrophosphorylase, 166, 168f
encephalopathies
chemiosmotic theory of action of, 97
in glycogen biosynthesis, 145, 146f
TSH. See Thyroid-stimulating hormone
undernutrition and, 479
Uridine diphosphate-glucuronate/glu-
α-Tubulin, 577
Undernutrition, 474, 478-479
curonic acid, 166-167, 168f, 290
β-Tubulin, 577
Unequal crossover, 324, 324f
Uridine diphosphate xylose (UDP-Xyl),
γ-Tubulin, 577
Unesterified fatty acids. See Free fatty acids
516t
Tumor cells, migration of, hyaluronic acid
Uniport systems, 426, 426f
Uridine monophosphate (UMP), 288f,
and, 548
Unique-sequence (nonrepetitive) DNA,
288t
Tumor suppressor genes, p53, 339
320, 320-321
Uridine triphosphate (UTP), in glycogen
Tunicamycin, 527, 527t
Universal donor/universal recipient, 618
biosynthesis, 145, 146f
β-Turn, 32, 34f
Unsaturated fatty acids, 111, 112, 113t. See
Uridyl transferase deficiency, 172
Twin lamb disease. See Pregnancy toxemia
also Fatty acids
Urobilinogens
of ewes
cis double bonds in, 112-114, 114f
conjugated bilirubin reduced to, 281,
Twitch fibers, slow (red) and fast (white),
dietary, cholesterol levels affected by,
282f
574-576, 575t
227
in jaundice, 284, 284t
Two-dimensional electrophoresis, protein
eicosanoids formed from, 190, 192,
normal values for, 284t
expression and, 28
193f, 194f
Urocanic aciduria, 250
TXs. See Thromboxanes
essential, 190, 190f, 193
Urokinase, 605, 605f
Tyk-2, in Jak-STAT pathway, 467
abnormal metabolism of, 195-196
Uronic acid pathway, 163, 166-167, 168f
Type A response, in gene expression, 374,
deficiency of, 191-192, 194-195
disruption of, 170
375f
prostaglandin production and, 190
Uronic acids, 109
Type B response, in gene expression,
in membranes, 417, 418f
in heparin, 545, 545f
374-375, 375f
metabolism of, 190-192
Uroporphyrinogen I, 271, 274f, 275f
Type C response, in gene expression, 375,
oxidation of, 183
Uroporphyrinogen I synthase, in porphyria,
375f
structures of, 190f
277t
Tyrosine, 15t, 16t, 267, 267f
synthesis of, 191, 191f
Uroporphyrinogen III, 271, 274f, 275f
catabolism of, 254f, 255
Unwinding, DNA, 326, 326-327
Uroporphyrinogen decarboxylase, 271,
epinephrine and norepinephrine formed
RNA synthesis and, 344
275f
from, 267, 267f
Uracil, 288t
in porphyria, 277t
in hemoglobin M, 46
deoxyribonucleosides of, in pyrimidine
Uroporphyrins, 270, 271f, 272f
in hormone synthesis, 438, 439-449,
synthesis, 296-297, 298f
spectrophotometry in detection of,
439t
Urate, as antioxidant, 119
273-274
phosphorylated, 264
Urea
UTP, in phosphorylation, 85
requirements for, 480
amino acid metabolism and, 124, 124f
synthesis of, 239, 240f
nitrogen catabolism producing,
Tyrosine aminotransferase, defect in, in
242-243, 245-247, 246f
V8 protease, for polypeptide cleavage, for
tyrosinemia, 255
permeability coefficient of, 419f
polypeptide cleavage, 25, 26t
Tyrosine hydroxylase, catecholamine
synthesis of, 243-244, 243f, 244f
vi. See Initial velocity
biosynthesis and, 446, 447f
metabolic disorders associated with,
Vmax. See Maximal velocity
Tyrosine kinase
247-248
V region/segment. See Variable regions/
in insulin signal transmission, 465-467,
gene therapy for, 248
segments
466f
Uric acid, 289
v-SNARE proteins, 509, 511
in Jak/STAT pathway, 467, 467f
purine catabolism in formation of, 299,
Valeric acid, 112t
Tyrosinemia, 255
299f
Valine, 15t
Tyrosinosis, 255
Uridine, 287f, 288t
catabolism of, 259, 260f, 262f
692
/
INDEX
Valine (cont.)
Vitamin B complex. See also specific vitamin
in coagulation, 486-488, 488f
interconversion of, 240
in citric acid cycle, 133
coumarin anticoagulants affecting,
requirements for, 480
coenzymes derived from, 50-51, 51f
604
Valinomycin, 99
Vitamin B1 (thiamin), 482t, 488-489, 489f
deficiency of, 482t
Van der Waals forces, 7
in citric acid cycle, 133
Vitamin K hydroquinone, 487, 488f
Vanadium, 496t
coenzymes derived from, 51
Vitamins, 2, 481-496, 482t. See also spe-
Variable numbers of tandemly repeated
deficiency of, 482t, 489
cific vitamin
units (VNTRs), in forensic
pyruvate metabolism affected by, 140,
in citric acid cycle, 133
medicine, 411
143, 489
digestion and absorption of, 477-478
Variable regions/segments, 591-592, 594f
Vitamin B2 (riboflavin), 86, 482t, 489-490
lipid- (fat) soluble, 482-488
gene for, 593
in citric acid cycle, 133
absorption of, 475
DNA rearrangement and, 325-326,
coenzymes derived from, 50-51, 489,
water-soluble, 488-496
393, 593-594
490
VLA-1/VLA-5/VLA-6, 622t
immunoglobulin heavy chain, 591, 592f,
deficiency of, 482t, 490
VLDL. See Very low density lipoproteins
594f
dehydrogenases dependent on, 87
VNTRs. See Variable numbers of tandemly
immunoglobulin light chain, 325-326,
Vitamin B6 (pyridoxine/pyridoxal/
repeated units
393, 591, 592f, 594f
pyridoxamine), 482t, 491, 491f
Voltage-gated channels, 424, 568t
Vascular system, nitric oxide affecting,
deficiency of, 482t, 491
von Gierke’s disease, 152t, 300
571-573, 573f, 574t
xanthurenate excretion in, 258, 258f
Von Willebrand factor, in platelet
Vasodilators, 556
excess/toxicity of, 491
activation, 605
nitric oxide as, 571-573, 573f, 574t
Vitamin B12 (cobalamin), 482t, 491-492,
VDRE. See Vitamin D response element
492f
Vector, 414
absorption of, 491-492
Warfarin, 486, 604
cloning, 400-402, 401f, 402t, 403f, 414
intrinsic factor in, 477, 491-492
phenobarbital interaction and,
expression, 402
deficiency of, 482t, 492
cytochrome P450 induction
Vegetarian diet, vitamin B12 deficiency and,
functional folate deficiency and, 492,
affecting, 628
491
494
vitamin K affected by, 487
Velocity
in methylmalonic aciduria, 155
Water, 2, 5-9
initial, 64
Vitamin B12-dependent enzymes, 292f, 492
as biologic solvent, 5, 6f
inhibitors affecting, 68, 68f, 69f
Vitamin C (ascorbic acid), 163, 482t,
biomolecular structure and, 6-7, 6t
maximal (Vmax)
495-496, 496f
dissociation of, 8-9
allosteric effects on, 75-76
as antioxidant, 119
in hydrogen bonds, 5, 6f
inhibitors affecting, 68, 68f, 69f
in collagen synthesis, 38, 496, 535
as nucleophile, 7-9
Michaelis-Menten equation in
deficiency of, 482t, 496
permeability coefficient of, 419f
determination of, 65-66, 66f
collagen affected in, 38-39, 496,
structure of, 5, 6f
substrate concentration and, 64, 64f
538-539
Water solubility, of xenobiotics, metabolism
Very low density lipoprotein receptor, 208
iron absorption and, 478, 496
and, 626
Very low density lipoproteins, 125, 205,
supplemental, 496
Watson-Crick base pairing, 7, 303
206t, 207
Vitamin D, 482t, 484-486
Waxes, 111
hepatic secretion of, dietary and hormonal
in calcium absorption, 477, 484,
Weak acids, 9
status and, 211-212, 213f
484-485
buffering capacity of, 11-12, 12f
metabolism of, 125, 126f, 207-209, 210f
deficiency of, 482t, 484, 485
dissociation constants for, 10-11, 12
in triacylglycerol transport, 207, 208f,
ergosterol as precursor for, 118, 119f
Henderson-Hasselbalch equation
210f
excess/toxicity of, 485-486
describing behavior of, 11, 12f
Vesicles
metabolism of, 484-485, 485f
physiologic significance of, 10-11
coating, 509, 510f
receptor for, 471
pK/pKa values of, 10-13, 12t,
brefeldin A affecting, 510-511
Vitamin D2 (ergocalciferol), 484
Weak bases, 9
secretory, 498, 500f
Vitamin D3 (cholecalciferol)
Wernicke-Korsakoff syndrome, 482t
targeting, 509, 510f
synthesis of in skin, 445, 446f, 484, 485f
Wernicke’s encephalopathy, 489
transport, 498, 508-511, 509t, 510f
in vitamin D metabolism, 484, 485f
Western blot transfer procedure, 403, 404f,
Vimentins, 577t, 578
Vitamin D-binding protein, 445
414
Vinculin, 540, 541f
Vitamin D receptor-interacting proteins
White blood cells, 620-624. See also specific
Viral oncogenes. See Oncogenes
(DRIPs), 472t, 473
type
Viruses, host cell protein synthesis affected
Vitamin D response element, 459t
growth factors regulating production of,
by, 370-371, 371f
Vitamin E, 482t, 486, 486f
610
Vision, vitamin A in, 482t, 483, 484f
as antioxidant, 91, 119, 486, 487f
recombinant DNA technology in study
Vitamin A, 482-484, 482t, 483f, 484f
deficiency of, 482t, 486
of, 624
deficiency of, 482t, 483-484
Vitamin H. See Biotin
White thrombus, 598
excess/toxicity of, 484
Vitamin K, 482t, 486-488, 488f, 604
White (fast) twitch fibers, 574-576, 575t
functions of, 482t, 483
calcium-binding proteins and, 487-488,
Whole genome shotgun approach, 634
in vision, 482t, 483
488f
Williams syndrome, 539
INDEX
/
693
Wilson disease, 432t, 587-589
responses to, 630-631, 630t,
Z line, 556, 557f, 558f
ceruloplasmin levels in, 587
631t
Zellweger’s (cerebrohepatorenal) syndrome,
gene mutations in, 432t, 588-589
toxic, 631, 631f
188, 503, 503t
Wobble, 361
Xeroderma pigmentosum, 337
Zinc, 496t
Xerophthalmia, vitamin A deficiency in,
Zinc finger motif, 387, 388t, 390, 390f
482t, 483
in DNA-binding domain, 470
X-linked disorders, RFLPs in diagnosis of,
XP. See Xeroderma pigmentosum
Zona fasciculata, steroid synthesis in, 440
411
Xylose, in glycoproteins, 516t
Zona glomerulosa, mineralocorticoid
X-ray diffraction and crystallography,
D-Xylose, 104f, 105t
synthesis in, 438
protein structure
D-Xylulose, 106f
Zona pellucida, glycoproteins in, 528
demonstrated by, 35
L-Xylulose, 105t
Zona reticularis, steroid synthesis in,
Xanthine, 289
accumulation of in essential pentosuria,
440
Xanthine oxidase, 87
170
ZP. See Zona pellucida
deficiency of, hypouricemia and, 300
ZP1-3 proteins, 528
Xanthurenate, excretion of in vitamin B6
Zwitterions, 16
deficiency, 258, 258f
Zymogens, 76, 477
Xenobiotics, metabolism of, 626-632
YAC vector. See Yeast artificial chromosome
in blood coagulation, 600, 600t, 601
conjugation in, 626, 628-630
(YAC) vector
rapid response to physiologic demand
cytochrome P450 system/hydroxylation
Yeast artificial chromosome (YAC) vector,
and, 76
in, 626-628, 629t
401-402, 402t
ZZ genotype, α1-antiproteinase deficiency
factors affecting, 630
for cloning in gene isolation, 635t
and
pharmacogenetics in drug research and,
Yeast cells, mitochondrial protein import
in emphysema, 589
631-632
studied in, 499
in liver disease, 590