Ranges of Normal Values in Human Whole Blood (B), Plasma (P), or Serum (S)a Normal Value (Varies with Procedure Used)
Determination Traditional Units SI Units
Normal Value (Varies with Procedure Used) Determination
Acetoacetate plus acetone (S) Aldosterone (supine) (P)
Alpha-amino nitrogen (P) Aminotransferases
Alanine aminotransferase Aspartate aminotransferase Ammonia (B)
Amylase (S)
Ascorbic acid (B)
Bilirubin (S)
Calcium (S)
Carbon dioxide content (S) Carotenoids (S)
Ceruloplasmin (S)
Chloride (S)
Cholesterol (S)
Cholesteryl esters (S)
Copper (total) (S)
Cortisol (P) (AM, fasting) Creatinine (P)
Glucose, fasting (P)
Iron (S)
Lactic acid (B)
Lipase (S)
Lipids, total (S)
Magnesium (S)
Osmolality (S)
PCO2 (arterial) (B)
Pepsinogen (P)
Phenylalanine (S)
Phosphatase, acid (S)
Phosphatase, alkaline (S) Phospholipids (S)
Phosphorus, inorganic (S) PO2 (arterial) (B)
Potassium (S)
Protein
Total (S)
Albumin (S)
Globulin (S)
Pyruvic acid (P)
Sodium (S)
Urea nitrogen (S)
Uric acid (S)
Women
Men
Traditional Units
0.3–2.0 mg/dL
3.0–10 ng/dL
3.0–5.5 mg/dL
SI Units
3–20 mg/L
83–227 pmol/L 2.1–3.9 mmol/L
3–48 units/L
0–55 units/L
12–55 μmol/L
53–123 units/L
0.4–1.5 mg/dL (fasting)
Conjugated (direct): up to 0.4 mg/dL
Total (conjugated plus free): up to 1.0 mg/dL
8.5–10.5 mg/dL; 4.3–5.3 meq/L
24–30 meq/L
0.8–4.0 μg/mL
23–43 mg/dL
100–108 meq/L
< 200 mg/dL
60–70% of total cholesterol
70–155 μg/dL
5–25 μg/dL
0.6–1.5 mg/dL
70–110 mg/dL
50–150 μg/dL
0.5–2.2 meq/L
3–19 units/L
450–1000 mg/dL
1.4–2.0 meq/L
280–296 mosm/kg H
2
O
35–45 mm Hg
200–425 units/mL
0–2 mg/dL
Males: 0–0.8 sigma unit/mL
Females: 0.01–0.56 sigma unit/mL
13–39 units/L (adults)
9–16 mg/dL as lipid phosphorus
2.6–4.5 mg/dL (infants in first year: up to 6.0 mg/dL)
75–100 mm Hg
3.5–5.0 meq/L
12–55 μmol/L
884–2050 nmol s
–1
/L 23–85 μmol/L
Up to 7 μmol/L
Up to 17 μmol/L
2.1–2.6 mmol/L
24–30 mmol/L
1.5–7.4 μmol/L
240–430 mg/L
100–108 mmol/L < 5.17 mmol/L
11.0–24.4 μmol/L
0.14–0.69 μmol/L
53–133 μmol/L
3.9–6.1 mmol/L
9.0–26.9 μmol/L
0.5–2.2 mmol/L
4.5–10 g/L
0.7–1.0 mmol/L
280–296 mmol/kg H
2
O
4.7–6.0 kPa
pH (B) 7.35–7.45
0–120 μmol/L
0.22–0.65 μmol s
–1
/L
2.9–5.2 mmol/L
0.84–1.45 mmol/L
10.0–13.3 kPa
3.5–5.0 mmol/L
6.0–8.0 g/dL
3.1–4.3 g/dL
2.6–4.1 g/dL
0–0.11 meq/L
135–145 meq/L
8–25 mg/dL
60–80 g/L
31–43 g/L
26–41 g/L
0–110 μmol/L 135–145 mmol/L 2.9–8.9 mmol/L
2.3–6.6 mg/dL 3.6–8.5 mg/dL 137–393 μmol/L 214–506 μmol/L
a
Based in part on Kratz A, et al. Laboratory reference values. N Engl J Med 2004;351:1548. Ranges vary somewhat from one laboratory to another depending on the
details of the methods used, and specific values should be considered in the context of the range of values for th e laboratory that made the determination. a LANGE
medical book
Ganong’s
Review of
Medical Physiology
Twenty-Third Edition
Kim E. Barrett, PhD
Professor
Department of Medicine
Dean of Graduate Studies
University of California, San Diego La Jolla, California
Scott Boitano, PhD
Associate Professor, Physiology Arizona Respiratory Center
Bio5 Collaborative Research Institute University of Arizona
Tucson, Arizona
Susan M. Barman, PhD
Professor
Department of Pharmacology/Toxicology Michigan State University East Lansing, Mich igan
Heddwen L. Brooks, PhD
Associate Professor
Department of Physiology
College of Medicine
University of Arizona
Tucson, Arizona
New York Milan Chicago San Francisco New Delhi San Juan Lisbon London Seoul Si ngapore Madrid Mexico City Sydney Toronto
Copyright © 2010 by The McGraw-Hill Companies, Inc. All rights reserved. Except a s permitted under the United States Copyright Act
of 1976, no part of this publication may be reproduced or distributed in any form or b y any means, or stored in a database or retrieval
system, without the prior written permission of the publisher.
ISBN: 978-0-07-160568-7
MHID: 0-07-160568-1
The material in this eBook also appears in the print version of this title: ISBN: 978-0-07- 160567-0, MHID: 0-07-160567-3.
All trademarks are trademarks of their respective owners. Rather than put a trademark symbol after every occurrence of a trademarked
name, we use names in an editorial fashion only, and to the benefit of the trademark ow ner, with no intention of infringement of the
trademark. Where such designations appear in this book, they have been printed with i nitial caps.
McGraw-Hill eBooks are available at special quantity discounts to use as premiums and sales promotions, or for use in corporate training
programs. To contact a representative please e-mail us at bulksales@mcgraw-hill.com.
Medicine is an ever-changing science. As new research and clinical experience broade n our knowledge, changes in treatment and drug
therapy are required. The authors and the publisher of this work have checked with so urces 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 th e publisher nor any other party who has been
involved in the preparation or publication of this work warrants that the information con tained herein is in every respect accurate or
complete, and they disclaim all responsibility for any errors or omissions or for the resu lts obtained from use of the information
contained in this work. Readers are encouraged to confirm the information contained h erein with other sources. For example and in
particular, readers are advised to check the product information sheet included in the pa ckage 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 infrequently used drugs.
TERMS OF USE
This is a copyrighted work and The McGraw-Hill Companies, Inc. (“McGraw-Hill”) a nd its licensors reserve all rights in and to the
work. Use of this work is subject to these terms. Except as permitted under the Copyrig ht Act of 1976 and the right to store and retrieve
one copy of the work, you may not decompile, disassemble, reverse engineer, reprodu ce, modify, create derivative works based upon,
transmit, distribute, disseminate, sell, publish or sublicense the work or any part of it with out McGraw-Hill’s prior consent. You may use
the work for your own noncommercial and personal use; any other use of the work is strictly prohibited. Your right to use the work may
be terminated if you fail to comply with these terms.
THE WORK IS PROVIDED “AS IS.” McGRAW-HILL AND ITS LICENSORS MA KE NO GUARANTEES OR WARRANTIES AS
TO THE ACCURACY, ADEQUACY OR COMPLETENESS OF OR RESULTS TO B E OBTAINED FROM USING THE WORK,
INCLUDING ANY INFORMATION THAT CAN BE ACCESSED THROUGH TH E WORK VIA HYPERLINK OR OTHERWISE,
AND EXPRESSLY DISCLAIM ANY WARRANTY, EXPRESS OR IMPLIED, INC LUDING BUT NOT LIMITED TO IMPLIED
WARRANTIES OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PU RPOSE. McGraw-Hill and its licensors do not
warrant or guarantee that the functions contained in the work will meet your requireme nts or that its operation will be uninterrupted or
error free. Neither McGraw-Hill nor its licensors shall be liable to you or anyone else f or any inaccuracy, error or omission, regardless of
cause, in the work or for any damages resulting therefrom. McGraw-Hill has no respo nsibility for the content of any information
accessed through the work. Under no circumstances shall McGraw-Hill and/or its licens ors be liable for any indirect, incidental, special,
punitive, consequential or similar damages that result from the use of or inability to use th e work, even if any of them has been advised of
the possibility of such damages. This limitation of liability shall apply to any claim or caus e whatsoever whether such claim or cause
arises in contract, tort or otherwise.
Dedication to
WILLIAM FRANCIS GANONG
William Francis (“Fran”) Ganong was an outstanding scientist, educator, and writer. He was completely dedicated to the field of
physiology and medical education in general. Chairman of the Department of Physiolog y at the University of California, San Francisco,
for many years, he received numerous teaching awards and loved working with medic al students.
Over the course of 40 years and some 22 editions, he was the sole author of the best se lling Review of Medical Physiology, and a co-
author of 5 editions of Pathophysiology of Disease: An Introduction to Clinical Medicin e. He was one of the “deans” of the Lange group
of authors who produced concise medical text and review books that to this day remain extraordinarily popular in print and now in digital
formats. Dr. Ganong made a gigantic impact on the education of countless medical stud ents and clinicians.
A general physiologist par excellence and a neuroendocrine physiologist by subspecial ty, Fran developed and maintained a rare
understanding of the entire field of physiology. This allowed him to write each new edi tion (every 2 years!) of the Review of Medical
Physiology as a sole author, a feat remarked on and admired whenever the book came up for discussion among physiologists. He was an
excellent writer and far ahead of his time with his objective of distilling a complex subjec t into a concise presentation. Like his good
friend, Dr. Jack Lange, founder of the Lange series of books, Fran took great pride in the many different translations of the Review of
Medical Physiology and was always delighted to receive a copy of the new edition in an y language.
He was a model author, organized, dedicated, and enthusiastic. His book was his pride and joy and like other best-selling authors, he
would work on the next edition seemingly every day, updating references, rewriting as needed, and always ready and on time when the
next edition was due to the publisher. He did the same with his other book, Pathophysio logy of Disease: An Introduction to Clinical
Medicine, a book that he worked on meticulously in the years following his formal retir ement and appointment as an emeritus professor
at UCSF.
Fran Ganong will always have a seat at the head table of the greats of the art of medical science education and communication. He died
on December 23, 2007. All of us who knew him and worked with him miss him greatl y.
iii
Key Features of the 23rd Edition of
Ganong’s Review of Medical Physiology
Thoroughly updated to reflect the latest research and developments in imp ortant areas such as the cellular basis of neurophysiology
Incorporates examples from clinical medicine throughout the chapters to illustrate important physiologic concepts
Delivers more detailed, clinically-relevant, high-yield information per page than any similar text or review
NEW full-color illustrations—the authors have worked with an outstanding team of medical illustrators, photographers, educators,
and students to provide an unmatched collection of 600 illustrations and tables
NEW boxed clinical cases—featuring examples of diseases that illus trate important physiologic principles
NEW high-yield board review questions at the end of each chapter
NEW larger 8½ X 11” trim-size enhances the rich visual content
NEW companion online learning center (LangeTextbooks.com) offers a wealth of innovative learning tools and illustrations
NEW iPod-compatible
review—Medical PodClass
offers audio and text for
study on the go
Full-color illustrations
enrich the text
KEY FEATURES v
Clinical Cases illustrate essential
physiologic principles
Summary tables and charts
encapsulate important information
Chapters conclude with Chapter Summaries and review questions
About the Authors
KIM E. BARRETT
Kim Barrett received her PhD in biological chemistry from University College London in 1982. Following postdoctoral training at the
National Institutes of Health, she joined the faculty at the University of California, San D iego, School of Medicine in 1985, rising to her
current rank of Professor of Medicine in 1996. Since 2006, she has also served the U niversity as Dean of Graduate Studies. Her
American Physiological Society (APS) and recently served on its council. She has also s erved as Chair of the Central Nervous System
Section of APS as well as Chair of both the Women in Physiology and Section Advisory Committees of APS. In her spare time, she
enjoys daily walks, aerobic exercising, and mind-challenging activities like puzzles of va rious sorts.
SCOTT BOITANO
research interests focus on the physiology and pathophysiology of the intestinal epitheliu m, and how its function is altered by
commensal, probiotics, and pathogenic bacteria as well as in specific disease states, such as inflammatory bowel diseases. She has
published almost 200 articles, chapters, and reviews, and has received several honors f or her research accomplishments including the
Bowditch and Davenport Lectureships from the American Physiological Society and the degree of Doctor of Medical Sciences, honoris
causa, from Queens University, Belfast. She is also a dedicated and award-winning inst ructor of medical, pharmacy, and graduate
students, and has taught various topics in medical and systems physiology to these group s for more than 20 years. Her teaching
experiences led her to author a prior volume (Gastrointestinal Physiology, McGraw-Hi ll, 2005) and she is honored to have been invited to
take over the helm of Ganong.
Scott Boitano received his PhD in genetics and cell biology from Washington State Univ ersity in Pullman, Washington, where he
acquired an interest in cellular signaling. He fostered this interest at University of Califor nia, Los Angeles, where he focused his research
on second
messengers and cellular physiology of the lung epithelium. He continued to foster these research interests at the University of Wyoming
and at his current positions with the Department of Physiology and the Arizona Respira tory Center, both at the University of Arizona.
HEDDWEN L. BROOKS
SUSAN M. BARMAN
Susan Barman received her PhD in physiology from Loyola University School of Med icine in Maywood, Illinois. Afterward she went to
Michigan State University (MSU) where she is currently a Professor in the Department of Pharmacology/ Toxicology and the
Neuroscience Program. Dr Barman has had a career-long interest in neural control of cardiorespiratory function with an emphasis on the
characterization
Heddwen Brooks received her PhD from Imperial College, University of London and is an Associate Professor in the Department of
Physiology at the University of Arizona (UA). Dr Brooks is a renal physiologist and is best known for her development of microarray
technology to address in vivo signaling pathways involved in the hormonal regulation o f
and origin of the naturally occurring discharges of sympathetic and phrenic nerves. Sh e was a recipient of a prestigious National
Institutes of Health MERIT (Method to Extend Research in Time) Award. She is also a recipient of an Outstanding University Woman
Faculty Award from the MSU Faculty Professional Women's Association and an MSU C ollege of Human Medicine Distinguished
Faculty Award. She has been very active in the renal function. Dr Brooks’ many awar ds include the American Physiological Society
(APS) Lazaro J. Mandel Young Investigator Award, which is for an individual demon strating outstanding promise in epithelial or renal
physiology. She will receive the APS Renal Young Investigator Award at the 20 09 annual meeting of the Federation of American
Societies for Experimental Biology. Dr Brooks is a member of the APS Renal Stee ring Section and the APS Committee of Committees.
She is on the Editorial Board of the American Journal of Physiology-Renal Physiology (since 2001), and she has also served on study
sections of the National Institutes of Health and the American Heart Association.
Contents
Preface ix
SECTION I
CELLULAR & MOLECULAR BASIS FOR MEDICAL PHYSIOLOGY 1
1. General Principles & Energy
Production in Medical Physiology 1 2. Overview of Cellular Physiology
in Medical Physiology 31
3. Immunity, Infection, & Inflammation 63
SECTION II
PHYSIOLOGY OF NERVE
& MUSCLE CELLS 79
4. Excitable Tissue: Nerve 79
5. Excitable Tissue: Muscle 93
6. Synaptic & Junctional Transmission 115
7. Neurotransmitters & Neuromodulators 129
8. Properties of Sensory Receptors 149
9. Reflexes 157
SECTION III
CENTRAL & PERIPHERAL
NEUROPHYSIOLOGY 167
10. Pain & Temperature 167
11. Somatosensory Pathways 173
12. Vision 181
13. Hearing & Equilibrium 203
14. Smell & Taste 219
15. Electrical Activity of the Brain, Sleep–Wake States, & Circadian Rhythms 229
16. Control of Posture & Movement 241
17. The Autonomic Nervous System 261
18. Hypothalamic Regulation of
Hormonal Functions 273
19. Learning, Memory, Language,
& Speech 289
SECTION IV
ENDOCRINE & REPRODUCTIVE PHYSIOLOGY 301
20. The Thyroid Gland 301
21. Endocrine Functions of the
Pancreas & Regulation of
Carbohydrate Metabolism 315
22. The Adrenal Medulla &
Adrenal Cortex 337
23. Hormonal Control of Calcium
and Phosphate Metabolism &
the Physiology of Bone 363
24. The Pituitary Gland 377
25. The Gonads: Development & Function of the Reproductive System 391
SECTION V
GASTROINTESTINAL
PHYSIOLOGY 429
26. Overview of Gastrointestinal
Function & Regulation 429
vii viii CONTENTS
27. Digestion, Absorption, &
Nutritional Principles 451
28. Gastrointestinal Motility 469
29. Transport & Metabolic
Functions of the Liver 479
SECTION VI
CARDIOVASCULAR
PHYSIOLOGY 489
30. Origin of the Heartbeat & the
Electrical Activity of the Heart 489 31. The Heart as a Pump 507
32. Blood as a Circulatory Fluid & the
Dynamics of Blood & Lymph Flow 521 33. Cardiovascular Regulatory Mec hanisms 555 34. Circulation Through Special Regions
569
SECTION VII
RESPIRATORY PHYSIOLOGY 587
35. Pulmonary Function 587
36. Gas Transport & pH in the Lung 609
37. Regulation of Respiration 625
SECTION VIII
RENAL PHYSIOLOGY 639
38. Renal Function & Micturition 639
39. Regulation of Extracellular Fluid Composition & Volume 665
40. Acidification of the Urine &
Bicarbonate Excretion 679
Answers to Multiple Choice Questions 687 Index 689
Preface
From the Authors
We are very pleased to launch the 23rd edition of Ganong's Review of Medical P hysiology. The current authors have attempted to
maintain the highest standards of excellence, accuracy, and pedagogy developed by Fr an Ganong over the 46 years in which he educated
countless students worldwide with this textbook.
At the same time, we have been attuned to the evolving needs of both students and prof essors in medical physiology. Thus, in addition to
usual updates on the latest research and developments in areas such as the cellular basis of physiology and neurophysiology, this edition
has added both outstanding pedagogy and learning aids for students.
We are truly grateful for the many helpful insights, suggestions, and reviews from arou nd the world that we received from colleagues
and students. We hope you enjoy the new features and the 23rd edition!
This edition is a revision of the original works of Dr. Francis Ganong.
New 4 Color Illustrations
• We have worked with a large team of medical illustrators, photographers, educators, and students to build an accurate, up-to-date, and
visually appealing new illustration program. Full-color illustrations and tables are provide d throughout, which also include detailed
figure legends that tell a short story or describes the key point of the illustration.
New 8
1
/
2
x 11 Format
• Based on student and instructor focus groups, we have increased the trim size, which will provide additional white space and allow our
new art program to really show!
New Boxed Clinical Cases
• Highlighted in a shaded background, so students can recognize the boxed clinical case s, examples of diseases illustrating important
physiological principles are provided.
New End of Chapter Board
Review Questions
• New to this edition, chapters now conclude with board review questions.
New Media
• This new edition has focused on creating new student content that is built upon learnin g outcomes and assessing student performance.
Free with every student copy is an iPod Review Tutorial Product. Questions and art bas ed from each chapter tests students
comprehension and is easy to navigate with a simple click of the scroll bar!
• Online Learning Center will provide students and faculty with cases and art and board review questions on a dedicated website.
ix
This page intentionally left blank
SECTION I CELLULAR & MOLECULAR
BASIS OF MEDICAL PHYSIOLOGY
General Principles & Energy Production in Medical Physiology
CH APTER
1
OBJEC TIV ES
After studying this chapter, you should be able to:
Name the different fluid compartments in the human body.
Define moles, equivalents, and osmoles.
Define pH and buffering.
Understand electrolytes and define diffusion, osmosis, and tonicity.
Define and explain the resting membrane potential.
Understand in general terms the basic building blocks of the cell: nucleotides, amino acids, car bohydrates, and fatty acids.
Understand higher-order structures of the basic building blocks: DNA, RNA, proteins, and lipi ds.
Understand the basic contributions of these building blocks to cell structure, function, and energy balance.
INTRODUCTION
In unicellular organisms, all vital processes occur in a single cell. As the evolution of mu lticellular organisms has progressed, various cell
groups organized into tissues and organs have taken over particular functions. In hum ans and other vertebrate animals, the specialized
cell groups include a gastrointestinal system to digest and absorb food; a respiratory sys tem to take up O
2
and eliminate CO
2
; a urinary
system to remove wastes; a cardiovascular system to distribute nutrients, O
2
, and the products of metabolism; a reproductive system to
perpetuate the species; and nervous and endocrine systems to coordinate and integrate t he functions of the other systems. This book is
concerned with the way these systems function and the way each contributes to the fun ctions of the body as a whole.
In this section, general concepts and biophysical and biochemical principles that are basi c to the function of all the systems are presented.
In the first chapter, the focus is on review of basic biophysical and biochemical princip les and the introduction of the molecular building
blocks that contribute to cellular physiology. In the second chapter, a review of basic ce llular morphology and physiology is presented. In
the third chapter, the process of immunity and inflammation, and their link to physiology , are considered.
1
GENERAL PRINCIPLES
THE BODY AS AN
ORGANIZED “SOLUTION”
The cells that make up the bodies of all but the simplest multicellular animals, both aquatic and terrestrial, exist in an “internal sea” of
extracellular fluid (ECF) enclosed within the integument of the animal. From th is fluid, the cells take up O
2
and nutrients; into it, they
discharge metabolic waste products. The ECF is more dilute than present-day seawater, but its composition closely resembles that of the
primordial oceans in which, presumably, all life originated.
In animals with a closed vascular system, the ECF is divided into two components: the interstitial fluid and the circulating blood
plasma. The plasma and the cellular elements of the blood, principally red blood cells , fill the vascular system, and together they
constitute the total blood volume. The interstitial fluid is that part of the ECF that is o utside the vascular system, bathing the cells. The
special fluids considered together as transcellular fluids are discussed in the following te xt. About a third of the total body water is
extracellular; the remaining two thirds is intracellular (intracellular fluid). In the aver age young adult male, 18% of the body weight is
protein and related substances, 7% is mineral, and 15% is fat. The remaining 60% is w ater. The distribution of this water is shown in
Figure 1–1A.
The intracellular component of the body water accounts for about 40% of body weigh t and the extracellular component for about 20%.
Approximately 25% of the extracellular component is in the vascular system (plasma = 5% of body weight) and 75% outside the blood
vessels (interstitial fluid = 15% of body weight). The total blood volume is about 8% of body weight. Flow between these compartments
is tightly regulated.
UNITS FOR MEASURING
CONCENTRATION OF SOLUTES
In considering the effects of various physiologically important substances and the intera ctions between them, the number of molecules,
electric charges, or particles of a substance per unit volume of a particular body fluid a re often more meaningful than simply the weight
of the substance per unit volume. For this reason, physiological concentrations are freq uently expressed in moles, equivalents, or
osmoles.
Moles
A mole is the gram-molecular weight of a substance, ie, the molecular weight of the sub stance in grams. Each mole (mol) consists of 6 ×
10
23
molecules. The millimole (mmol) is 1/1000 of a mole, and the micromole (μmol) is 1/1,0 00,000 of a mole. Thus, 1 mol of NaCl =
23 g + 35.5 g = 58.5 g, and 1 mmol = 58.5 mg. The mole is the standard unit for expr essing the amount of substances in the SI unit
system.
The molecular weight of a substance is the ratio of the mass of one molecule of the subs tance to the mass of one twelfth the mass of an
atom of carbon-12. Because molecular weight is a ratio, it is dimensionless. The dalton ( Da) is a unit of mass equal to one twelfth the
mass of an atom of carbon-12. The kilodalton (kDa = 1000 Da) is a useful unit for ex pressing the molecular mass of proteins. Thus, for
example, one can speak of a 64-kDa protein or state that the molecular mass of the pro tein is 64,000 Da. However, because molecular
weight is a dimensionless ratio, it is incorrect to say that the molecular weight of the prote in is 64 kDa.
Equivalents
The concept of electrical equivalence is important in physiology because many of the so lutes in the body are in the form of charged
particles. One equivalent (eq) is 1 mol of an ionized substance divided by its valence. O ne mole of NaCl dissociates into 1 eq of Na
+
and
1 eq of Cl
. One equivalent of Na
+
= 23 g, but 1 eq of Ca
2+
= 40 g/2 = 20 g. The milliequivalent (meq) is 1/1000 of 1 eq.
Electrical equivalence is not necessarily the same as chemical equivalence. A gram equiv alent is the weight of a substance that is
chemically equivalent to 8.000 g of oxygen. The normality (N) of a solution is the num ber of gram equivalents in 1 liter. A 1 N solution
of hydrochloric acid contains both H
+
(1 g) and Cl
(35.5 g) equivalents, = (1 g + 35.5 g)/L = 36.5 g/L.
WATER, ELECTROLYTES, & ACID/BASE
The water molecule (H
2
O) is an ideal solvent for physiological reactions. H
2
O has a dipole moment where oxygen slightly pulls away
electrons from the hydrogen atoms and creates a charge separation that makes the mole cule polar. This allows water to dissolve a variety
of charged atoms and molecules. It also allows the H
2
O molecule to interact with other H
2
O molecules via hydrogen bonding. The
resultant hydrogen bond network in water allows for several key properties in physiolo gy: (1) water has a high surface tension, (2) water
has a high heat of vaporization and heat capacity, and (3) water has a high dielectric co nstant. In layman’s terms, H
2
O is an excellent
biological fluid that serves as a solute; it provides optimal heat transfer and conduction o f current.
Electrolytes (eg, NaCl) are molecules that dissociate in water to their cation (Na
+
) and anion (Cl
) equivalents. Because of the net
charge on water molecules, these electrolytes tend not to reassociate in water. There are many important electrolytes in physiology,
notably Na
+
, K
+
, Ca
2+
, Mg
2+
, Cl
, and HCO3–. It is important to note that electrolytes and other charged compounds (eg , proteins) are
unevenly distributed in the body fluids (Figure 1–1B). These separations play an impor tant role in physiology.
Stomach
Intestines
Lungs
Blood plasma:
Skin
5% body weight
Kidneys
Extra
cellular
fluid: Interstitial fluid:20% body
15% body weight
weight
Intracellular fluid: 40% body weight
A
200
Extracellular fluid Intracellular fluid
Plasma
Interstitial fluid Misc. 150 phosphates K
+
100
Na
+
Cl
Na
+
Cl
Na
+ 50
HCO
3
− Prot− K+ K+
HCO
3
HCO
3
Prot−
0
B
Cl
FIGURE 1–1 Organization of body fluids and electrolytes into compartments. A) Body fluids are divided into Intracellular and
extracellular fluid compartments (ICF and ECF, respectively). Their contribution to perc entage body weight (based on a healthy young
adult male; slight variations exist with age and gender) emphasizes the dominance of flui d makeup of the body. Transcellular fluids,
which constitute a very small percentage of total body fluids, are not shown. Arrows represent fluid movement between compartments.
B) Electrolytes and proteins are unequally distributed among the body fluids. This unev en distribution is crucial to physiology. Prot
,
protein, which tends to have a negative charge at physiologic pH.
pH AND BUFFERING
The maintenance of a stable hydrogen ion concentration ([H
+
]) in body fluids is essential to life. The pH of a solution is defined as the
logarithm to the base 10 of the reciprocal of the H
+
concentration ([H
+
]), ie, the negative logarithm of the [H
+
]. The pH of water at 25
°C, in which H
+
and OH
ions are present in equal numbers, is 7.0 (Figure 1–2). For each pH unit less than 7.0, the [H
+
] is increased
tenfold; for each pH unit above 7.0, it is decreased tenfold. In the plasma of healthy ind ividuals, pH is slightly alkaline, maintained in the
narrow range of 7.35 to 7.45. Conversely, gastric fluid pH can be quite acidic (on the order of 2.0) and pancreatic secretions can be quite
alkaline (on the order of 8.0). Enzymatic activity and protein structure are frequently se nsitive to pH; in any given body or cellular
compartment, pH is maintained to allow for maximal enzyme/protein efficiency.
Molecules that act as H
+
donors in solution are considered acids, while those that tend to remove H
+
from solutions are considered
bases. Strong acids (eg, HCl) or bases (eg, NaOH) dissociate completely in water and t hus can most change the [H
+
] in solution. In
physiological compounds, most acids or bases are considered “weak,” that is, they cont ribute relatively few H
+
or take away relatively
few H
+
from solution. Body pH is stabilized by the bufferi ng capacity of the body fluids. A buffer is a substance that has the ability to
bind or release H
+
in solution, thus keeping the pH of the solution relatively constant despite the addition of considerable quantities of
acid or base. Of course there are a number of buffers at work in biological fluids at an y given time. All buffer pairs in a homogenous
solution are in equilibrium with the same [H
+
]; this is known as the isohydric principle. One outcome of this principle is that by
assaying a single buffer system, we can understand a great deal about all of the biologic al buffers in that system.
H
+
concentration
(mol/L) pH
10
1
1
10
2
2
10
3
3
10
4
4
10
5
5
For pure water,
10−6 6
[H
+
] = 10
7
mol/L
10
7
7
10
8
8
10
−9
9
10
−10
10
10
−11
11
10
−12
12
10
−13
13
10
−14
14
FIGURE 1–2 Proton concentration and pH. Relative proton (H
+
) concentrations for solutions on a pH scale are shown. (Redraw n from
Alberts B et al: Molecular Biology of the Cell, 4th ed. Garland Science, 2002.)
When acids are placed into solution, there is a dissociation of some of the component ac id (HA) into its proton (H
+
) and free acid (A
).
This is frequently written as an equation:
HA
H
+
+ A
.
According to the laws of mass action, a relationship for the dissociation can be defined mathematically as:
K
a
= [H
+
] [A
] / [HA]
where K
a
is a constant, and the brackets represent concentrations of the individual species. In lay man’s terms, the product of the proton
concentration ([H
+
]) times the free acid concentration ([A
]) divided by the bound acid concentration ([HA]) is a defined constant (K).
This can be rearranged to read:
[H
+
] = K
a
[HA]/[A
]
If the logarithm of each side is taken:
log [H
+
] = logK
a
+ log[HA]/[A
]
Both sides can be multiplied by –1 to yield:
–log [H
+
] = –logK
a
+ log[A
]/[HA]
This can be written in a more conventional form known as the Henderson Hasselbach equation:
pH = pK
a
+ log [A
]/[HA]
This relatively simple equation is quite powerful. One thing that we can discern right aw ay is that the buffering capacity of a particular
weak acid is best when the pK
a
of that acid is equal to the pH of the solution, or when:
[A–] = [HA], pH = pK
a
Similar equations can be set up for weak bases. An important buffer in the body is carb onic acid. Carbonic acid is a weak acid, and thus
is only partly dissociated into H
+
and bicarbonate:
H
2
CO
3 →
H
+
+ HCO3–
If H
+
is added to a solution of carbonic acid, the equilibrium shifts to the left and most of the a dded H
+
is removed from solution. If OH
is added, H
+
and OH
combine, taking H
+
out of solution. However, the decrease is countered by more dissociation of H
2
CO
3
, and the
decline in H
+
concentration is minimized. A unique feature of bicarbonate is the linkage between its b uffering ability and the ability for
the lungs to remove carbon dioxide from the body. Other important biological buffers include phosphates and proteins.
DIFFUSION
Diffusion is the process by which a gas or a substance in a solution expands, because o f the motion of its particles, to fill all the available
volume. The particles (molecules or atoms) of a substance dissolved in a solvent are in c ontinuous random movement. A given particle is
equally likely to move into or out of an area in which it is present in high concentration. However, because there are more particles in the
area of high concentration, the total number of particles moving to areas of lower conc entration is greater; that is, there is a net flux of
solute particles from areas of high to areas of low concentration. The time required for equilibrium by diffusion is proportionate to the
square of the diffusion distance. The magnitude of the diffusing tendency from one re gion to another is directly proportionate to the
cross-sectional area across which diffusion is taking place and the concentration gradient, or chemical gradient, which is the
difference in concentration of the diffusing substance divided by the thickness of the b oundary (Fick’s law of diffusion). Thus,
J = –DA Δc
Δx
where J is the net rate of diffusion, D is the diffusion coefficient, A is the area, and Δc/Δ x is the concentration gradient. The minus sign
indicates the direction of diffusion. When considering movement of molecules from a h igher to a lower concentration, Δc/Δx is negative,
so multiplying by –DA gives a positive value. The permeabilities of the boundaries acro ss which diffusion occurs in the body vary, but
diffusion is still a major force affecting the distribution of water and solutes.
OSMOSIS
When a substance is dissolved in water, the concentration of water molecules in the solu tion is less than that in pure water, because the
addition of solute to water results in a solution that occupies a greater volume than does the water alone. If the solution is placed on one
side of a membrane that is permeable to water but not to the solute, and an equal volume of water is placed on the other, water molecules
diffuse down their concentration (chemical) gradient into the solution (Figure 1–3). Thi s process—the diffusion of solvent molecules
into a region in which there is a higher concentration of a solute to which the membrane is imp ermeable—is called osmosis. It is an
important factor in physiologic processes. The tendency for movement of solvent molec ules to a region of greater solute concentration
can be prevented by applying pressure to the more concentrated solution. The pressure necessary to prevent solvent migration is the
osmotic pressure of the solution.
Osmotic pressure—like vapor pressure lowering, freezingpoint depression, and boiling -point elevation—depends on the number rather
than the type of particles in a solution; that is, it is a fundamental colligative property of so lutions. In an ideal solution, osmotic pressure
(P) is related to temperature and volume in the same way as the pressure of a gas:
nRT
P = ---------
V
where n is the number of particles, R is the gas constant, T is the absolute temperature, a nd V is the volume. If T is held constant, it is
clear that the osmotic pressure is proportional to the number of particles in solution per u nit volume of solution.
Semipermeable
membrane
Pressure
FIGURE 1–3 Diagrammatic representation of osmosis. Water molecules are represented by sma ll open circles, solute molecules by
large solid circles. In the diagram on the left, water is placed on one side of a memb rane permeable to water but not to solute, and an
equal volume of a solution of the solute is placed on the other. Water molecules move down the ir concentration (chemical) gradient into
the solution, and, as shown in the diagram on the right, the volume of the solution increases . As indicated by the arrow on the right, the
osmotic pressure is the pressure that would have to be applied to prevent the movement of the water molecules.
For this reason, the concentration of osmotically active particles is usually expressed in osm oles. One osmole (Osm) equals the gram-
molecular weight of a substance divided by the number of freely moving particles that e ach molecule liberates in solution. For biological
solutions, the milliosmole (mOsm; 1/1000 of 1 Osm) is more commonly used.
If a solute is a nonionizing compound such as glucose, the osmotic pressure is a functio n of the number of glucose molecules present. If
the solute ionizes and forms an ideal solution, each ion is an osmotically active particle. F or example, NaCl would dissociate into Na
+
and Cl
ions, so that each mole in solution would supply 2 Osm. One mole of Na
2
SO
4
would dissociate into Na
+
, Na
+
, and SO42–
supplying 3 Osm. However, the body fluids are not ideal solutions, and although the di ssociation of strong electrolytes is complete, the
number of particles free to exert an osmotic effect is reduced owing to interactions betw een the ions. Thus, it is actually the effective
concentration (activity) in the body fluids rather than the number of equivalents of an electro lyte in solution that determines its osmotic
capacity. This is why, for example, 1 mmol of NaCl per liter in the body fluids contribu tes somewhat less than 2 mOsm of osmotically
active particles per liter. The more concentrated the solution, the greater the deviation fro m an ideal solution.
The osmolal concentration of a substance in a fluid is measured by the degree to which it depresses the freezing point, with 1 mol of an
ideal solution depressing the freezing point 1.86 °C. The number of milliosmoles per lite r in a solution equals the freezing point
depression divided by 0.00186. The osmolarity is the number of osmoles per liter of so lution (eg, plasma), whereas the osmolality is the
number of osmoles per kilogram of solvent. Therefore, osmolarity is affected by the vo lume of the various solutes in the solution and the
temperature, while the osmolality is not. Osmotically active substances in the body are dis solved in water, and the density of water is 1,
so osmolal concentrations can be expressed as osmoles per liter (Osm/L) of water. In th is book, osmolal (rather than osmolar)
concentrations are considered, and osmolality is expressed in milliosmoles per liter (of w ater).
Note that although a homogeneous solution contains osmotically active particles and can be said to have an osmotic pressure, it can exert
an osmotic pressure only when it is in contact with another solution across a membrane permeable to the solvent but not to the solute.
OSMOLAL CONCENTRATION
OF PLASMA: TONICITY
The freezing point of normal human plasma averages –0.54 °C, which corresponds to an osmolal concentration in plasma of 290
mOsm/L. This is equivalent to an osmotic pressure against pure water of 7.3 atm. The os molality might be expected to be higher than
this, because the sum of all the cation and anion equivalents in plasma is over 300. It is n ot this high because plasma is not an ideal
solution and ionic interactions reduce the number of particles free to exert an osmotic ef fect. Except when there has been insufficient
time after a sudden change in composition for equilibrium to occur, all fluid compartme nts of the body are in (or nearly in) osmotic
equilibrium. The term tonicity is used to describe the osmolality of a solu tion relative to plasma. Solutions that have the same osmolality
as plasma are said to be isotonic; those with greater osmolality are hypertonic; an d those with lesser osmolality are hypotonic. All
solutions that are initially isosmotic with plasma (ie, that have the same actual osmotic pres sure or freezing-point depression as plasma)
would remain isotonic if it were not for the fact that some solutes diffuse into cells and o thers are metabolized. Thus, a 0.9% saline
solution remains isotonic because there is no net movement of the osmotically active part icles in the solution into cells and the particles
are not metabolized. On the other hand, a 5% glucose solution is isotonic when initially i nfused intravenously, but glucose is
metabolized, so the net effect is that of infusing a hypotonic solution.
It is important to note the relative contributions of the various plasma components to the total osmolal concentration of plasma. All but
about 20 of the 290 mOsm in each liter of normal plasma are contributed by Na
+
and its accompanying anions, principally Cl
and
HCO3–. Other cations and anions make a relatively small contribution. Although the co ncentration of the plasma proteins is large when
expressed in grams per liter, they normally contribute less than 2 mOsm/L because of th eir very high molecular weights. The major
nonelectrolytes of plasma are glucose and urea, which in the steady state are in equilibri um with cells. Their contributions to osmolality
are normally about 5 mOsm/L each but can become quite large in hyperglycemia or ure mia. The total plasma osmolality is important in
assessing dehydration, overhydration, and other fluid and electrolyte abnormalities (Clin ical Box 1–1).
CLINICAL BOX 1–1 Plasma Osmolality & Disease
Unlike plant cells, which have rigid walls, animal cell membranes are flexible. Therefore , animal cells swell when exposed to
extracellular hypotonicity and shrink when exposed to extracellular hypertonicity. Cells contain ion channels and pumps that can be
activated to offset moderate changes in osmolality; however, these can be overwhelmed under certain pathologies. Hyperosmolality can
cause coma (hyperosmolar coma). Because of the predominant role of the major solute s and the deviation of plasma from an ideal
solution, one can ordinarily approximate the plasma osmolality within a few mosm/liter b y using the following formula, in which the
constants convert the clinical units to millimoles of solute per liter:
Osmolality (mOsm/L) = 2[Na
+
] (mEq/L) + 0.055[Glucose] (mg/dL) + 0.36[BUN] (mg/dL) BUN is the blood urea nitrogen. T he formula
is also useful in calling attention to abnormally high concentrations of other solutes. An o bserved plasma osmolality (measured by
freezing-point depression) that greatly exceeds the value predicted by this formula prob ably indicates the presence of a foreign substance
such as ethanol, mannitol (sometimes injected to shrink swollen cells osmotically), or pois ons such as
ethylene glycol or methanol (components of antifreeze).
NONIONIC DIFFUSION
Some weak acids and bases are quite soluble in cell membranes in the undissociated form , whereas they cannot cross membranes in the
charged (ie, dissociated) form. Consequently, if molecules of the undissociated substanc e diffuse from one side of the membrane to the
other and then dissociate, there is appreciable net movement of the undissociated substan ce from one side of the membrane to the other.
This phenomenon is called nonionic diffusion.
DONNAN EFFECT
When an ion on one side of a membrane cannot diffuse through the membrane, the di stribution of other ions to which the membrane is
permeable is affected in a predictable way. For example, the negative charge of a nond iffusible anion hinders diffusion of the diffusible
cations and favors diffusion of the diffusible anions. Consider the following situation,
X Y
m
K
+
K
+
Cl
Cl
Prot
in which the membrane (m) between compartments X and Y is impermeable to charged proteins (Prot
) but freely permeable to K
+
and
Cl
. Assume that the concentrations of the anions and of the cations on the two sides are in itially equal. Cl
diffuses down its
concentration gradient from Y to X, and some K
+
moves with the negatively charged Cl
because of its opposite charge. Therefore
[K+x] > [K+y]
Furthermore,
[K+x] + [Cl–x] + [Prot–x] > [K+y] + [Cl–y] that is, more osmotically active particles a re on side X than on side Y.
Donnan and Gibbs showed that in the presence of a nondiffusible ion, the diffusible io ns distribute themselves so that at equilibrium their
concentration ratios are equal:
[K+x
] = [Cl
–y]
[K+y] [Cl–x]
Cross-multiplying,
[K+x] + [Cl–x] = [K+y] + [Cl–y]
This is the Gibbs–Donnan equation. It holds for any pair of cations and anions of the same valence.
The Donnan effect on the distribution of ions has three effects in the body introduced h ere and discussed below. First, because of
charged proteins (Prot
) in cells, there are more osmotically active particles in cells than in interstitial fluid, and be cause animal cells
have flexible walls, osmosis would make them swell and eventually rupture if it were no t for Na, K ATPase pumping ions back out of
cells. Thus, normal cell volume and pressure depend on Na, K ATPase. Second, becau se at equilibrium the distribution of permeant ions
across the membrane (m in the example used here) is asymmetric, an electrical differenc e exists across the membrane whose magnitude
can be determined by the Nernst equation. In the example used here, side X will be negative relative to side Y. The charges line up
along the membrane, with the concentration gradient for Cl
exactly balanced by the oppositely directed electrical gradient, and the same
holds true for K
+
. Third, because there are more proteins in plasma than in interstitial fluid, there is a Don nan effect on ion movement
across the capillary wall.
FORCES ACTING ON IONS
The forces acting across the cell membrane on each ion can be analyzed mathematically . Chloride ions (Cl
) are present in higher
concentration in the ECF than in the cell interior, and they tend to diffuse along this concentration gradient into the cell. The interior of
the cell is negative relative to the exterior, and chloride ions are pushed out of the cell alo ng this electrical gradient. An equilibrium is
reached between Cl
influx and Cl
efflux. The membrane potential at which this equilibrium exists is the equilib rium potential. Its
magnitude can be calculated from the Nernst equation, as follows:
RT ln
[Clo–]
ECl =
FZ i–]
Cl
[Cl
where
E
Cl
= equilibrium potential for Cl
R = gas constant
T = absolute temperature
F = the faraday (number of coulombs per mole of charge) Z
Cl
= valence of Cl
(–1)
[Clo–] = Cl
concentration outside the cell
[Cli–] = Cl
concentration inside the cell
Converting from the natural log to the base 10 log and replacing some of the constants with numerical values, the equation becomes:
ECl
= 61.5 log [Cl
i–
] at 37 °C
[Clo–]
Note that in converting to the simplified expression the concentration ratio is reversed be cause the –1 valence of Cl
has been removed
from the expression.
The equilibrium potential for Cl
(E
Cl
), calculated from the standard values listed in Table 1–1, is –70 mV, a value identical to the
measured resting membrane potential of –70 mV. Therefore, no forces other than thos e represented by the chemical and electrical
gradients need be invoked to explain
the distribution of Cl– across the membrane. +
A similar equilibrium potential can be calculated for K (E
K
):
EK
= RT ln [K
o+
] = 61.5log [K
o+
] at 37 °C
FZ
K
[Ki+] [Ki+]
where
E
K
= equilibrium potential for K
+
Z
+
(+1)
K
= valence of K
[Ko+] = K
+
concentration outside the cell
[Ki+] = K
+
concentration inside the cell
R, T, and F as above
In this case, the concentration gradient is outward and the electrical gradient inward. In mammalian spinal motor neurons, E
K
is –90 mV
(Table 1–1). Because the resting membrane potential is –70 mV, there is somewhat mor e K
+
in the neurons than can be accounted for by
the electrical and chemical gradients.
The situation for Na
+
is quite different from that for K
+
and Cl
. The direction of the chemical gradient for Na
+
is inward, to the area
where it is in lesser concentration, and the electrical gradient is in the same direction. E
Na
is +60 mV (Table 1–1). Because neither E
K
nor E
Na
is equal to the membrane potential,
TABLE 1–1 Concentration of some ions inside and outside mammalian spinal motor neu rons.
Concentration (mmol/L of H
2
O)
Ion Inside Cell Outside Cell Equilibrium Potential (mV)
Na
+
15.0 150.0
K
+
150.0 5.5 +60
–90
Cl
9.0 125.0 –70 NH
2
Adenine N
N
CH
2
N
N
O
O
C
HH
C
RiboseO O O
H H
O PO PO P
HO OH
O O O
Resting membrane potential = –70 mV Adenosine 5'-monophosphate (AMP)
Adenosine 5'-diphosphate (ADP)
one would expect the cell to gradually gain Na
+
and lose K
+
if only passive electrical and chemical forces were acting across the
membrane. However, the intracellular concentration of Na
+
and K
+
remain constant because of the action of the Na, K ATPase that
actively transports Na
+
out of the cell and K
+
into the cell (against their respective electrochemical gradients).
Adenosine 5'-triphosphate (ATP)
FIGURE 1–4 Energy-rich adenosine derivatives. Ade nosine triphosphate is broken down into its backbone purine base and sugar (at
right) as well as its high energy phosphate derivatives (across bottom). (Reproduc ed, with permission, from Murray RK et al: Harper’s Biochemistry,
26th ed. McGraw-Hill, 2003.)
GENESIS OF THE MEMBRANE POTENTIAL
The distribution of ions across the cell membrane and the nature of this membrane prov ide the explanation for the membrane potential.
The concentration gradient for K
+
facilitates its movement out of the cell via K
+
channels, but its electrical gradient is in the opposite
(inward) direction. Consequently, an equilibrium is reached in which the tendency of K
+
to move out of the cell is balanced by its
tendency to move into the cell, and at that equilibrium there is a slight excess of cations o n the outside and anions on the inside. This
condition is maintained by Na, K ATPase, which uses the energy of ATP to pump K
+
back into the cell and keeps the intracellular
concentration of Na
+
low. Because the Na, K ATPase moves three Na
+
out of the cell for every two K
+
moved in, it also contributes to
the membrane potential, and thus is termed an electrogenic pump. It should b e emphasized that the number of ions responsible for the
membrane potential is a minute fraction of the total number present and that the total con centrations of positive and negative ions are
equal everywhere except along the membrane.
ENERGY PRODUCTION
ENERGY TRANSFER
Energy is stored in bonds between phosphoric acid residues and certain organic compo unds. Because the energy of bond formation in
some of these phosphates is particularly high, relatively large amounts of energy (10–1 2 kcal/mol) are released when the bond is
hydrolyzed. Compounds containing such bonds are called high-energy phosphate com pounds. Not all organic phosphates are of the
high-energy type. Many, like glucose 6-phosphate, are low-energy phosphates that on hydrolysis liberate 2–3 kcal/mol. Some of the
intermediates formed in carbohydrate metabolism are high-energy phosphates, but the m ost important high-energy phosphate compound
is adenosine triphosphate (ATP). This ubiquitous molecule (Fig ure 1–4) is the energy storehouse of the body. On hydrolysis to
adenosine diphosphate (ADP), it liberates energy directly to such processes as muscle c ontraction, active transport, and the synthesis of
many chemical compounds. Loss of another phosphate to form adenosine monophosp hate (AMP) releases more energy.
Another group of high-energy compounds are the thioesters, the acyl derivatives of m ercaptans. Coenzyme A (CoA) is a widely
distributed mercaptan-containing adenine, ribose, pantothenic acid, and thioethanolamin e (Figure 1–5). Reduced CoA (usually
abbreviated HS–CoA) reacts with acyl groups (R–CO–) to form R–CO–S–CoA deriv atives. A prime example is the reaction of HS-CoA
with acetic acid to form acetylcoenzyme A (acetyl-CoA), a compound of pivotal impor tance in intermediary metabolism. Because acetyl-
CoA has a much higher energy content than acetic acid, it combines readily with substan ces in reactions that would otherwise require
outside energy. Acetyl-CoA is therefore often called “active acetate.” From the point of view of energetics, formation of 1 mol of any
acyl-CoA compound is equivalent to the formation of 1 mol of ATP.
BIOLOGIC OXIDATIONS
Oxidation is the combination of a substance with O
2
, or loss of hydrogen, or loss of electrons. The corresponding reverse processes are
called reduction. Biologic oxidations are catalyzed by specific enzymes. Cofactors (s imple ions) or coenzymes (organic, nonprotein
substances) are accessory substances that
Pantothenic acid β-Alanine Thioethanolamine
H
3
C OH O
H
O
H
CH
2
C CH C N CH
2
CH
2
C N CH
2
CH
2
SH
O
NH 2
H
3
C
O P O
N
N
Pyrophosphate Adenine O
N
N
O
PO
CH
2
Coenzyme A
O
O
HH
H H
Ribose 3-phosphate
O OH
PO O O O
R C OH + HS CoA R CS CoA + HOH FIGURE 1–5 Coenzym e A (CoA) and its derivatives. Left: Formula of reduced
coenzyme A (HS-CoA) with its components highlighted. Right: Form ula for reaction of CoA with biologically important compounds to
form thioesters. R, remainder of molecule.
usually act as carriers for products of the reaction. Unlike the enzymes, the coenzymes may catalyze a variety of reactions.
A number of coenzymes serve as hydrogen acceptors. One common form of biologic oxidation is removal of hydrogen from an R–OH
group, forming R=O. In such dehydrogenation reactions, nicotinamide adenine dinucl eotide (NAD
+
) and dihydronicotinamide adenine
dinucleotide phosphate (NADP
+
) pick up hydrogen, forming dihydronicotinamide adenine dinucleotide (NADH) and
dihydronicotinamide adenine dinucleotide phosphate (NADPH) (Figure 1–6). The hyd rogen is then transferred to the flavoprotein–
cytochrome system, reoxidizing the NAD
+
and NADP
+
. Flavin adenine dinucleotide (FAD) is formed when riboflavin is
phosphorylated, forming flavin mononucleotide (FMN). FMN then combines with AM P, forming the dinucleotide. FAD can accept
hydrogens in a similar fashion, forming its hydro (FADH) and dihydro (FADH
2
) derivatives.
The flavoprotein–cytochrome system is a chain of enzymes that transfers hydrogen to oxygen, forming water. This process occurs in the
mitochondria. Each enzyme in the chain is reduced
NH
2
N
N
N OH
N
HH
POCH2O
OH* OH
H
O
H
O
CONH
2
O
P
OCH
+
2
N O
O
HH
HH OH OH
Adenine Ribose Diphosphate Ribose Nicotinamide
H
HH
CONH
2
CONH
2
+
+ R' N
+
+ R'H
2
+ H N
R R
Oxidized coenzyme Reduced coenzyme
FIGURE 1–6 Structures of molecules important in oxidation reduction reactio ns to produce energy. Top: Formula of the oxidized
form of nicotinamide adenine dinucleotide (NAD
+
). Nicotinamide adenine dinucleotide phosphate (NADP
+
) has an additional phosphate
group at the location marked by the asterisk. Bottom: Reaction by which NAD
+
and NADP
+
become reduced to form NADH and
NADPH. R, remainder of molecule; R’, hydrogen donor.
H
+ Outer lamella
I
nner lamella
ATP ADP
FIGURE 1–7 Simplified diagram of transport of protons across the inner a nd outer lamellas of the inner mitochondrial
membrane. The electron transport system (flavoprotein-cytochrome system) h elps create H
+
movement from the inner to the outer
lamella. Return movement of protons down the proton gradient generates ATP.
and then reoxidized as the hydrogen is passed down the line. Each of the enzymes is a protein with an attached nonprotein prosthetic
group. The final enzyme in the chain is cytochrome c oxidase, which transfers hydrog ens to O
2
, forming H
2
O. It contains two atoms of
Fe and three of Cu and has 13 subunits.
The principal process by which ATP is formed in the body is oxidative ph osphorylation. This process harnesses the energy from a
proton gradient across the mitochondrial membrane to produce the high-energy bond of ATP and is briefly outlined in Figure 1–7.
Ninety percent of the O
2
consumption in the basal state is mitochondrial, and 80% of this is coupled to ATP synth esis. About 27% of the
ATP is used for protein synthesis, and about 24% is used by Na, K ATPase, 9% by glu coneogenesis, 6% by Ca
2+
ATPase, 5% by
myosin ATPase, and 3% by ureagenesis.
MOLECULAR BUILDING BLOCKS
NUCLEOSIDES, NUCLEOTIDES,
& NUCLEIC ACIDS
Nucleosides contain a sugar linked to a nitrogen-containing base. The physiologically important bases, purines and pyrimidines, have
ring structures (Figure 1–8). These structures are bound to ribose or 2-deoxyribose to complete the nucleoside. When inorganic
phosphate is added to the nucleoside, a nucleotide is formed. Nucleosides and nucleotides for m the backbone for RNA and DNA, as
well as a variety of coenzymes and regulatory molecules (eg, NAD
+
, NADP
+
, and ATP) of physiological importance (Table 1–2).
Nucleic acids in the diet are digested and their constituent purines and pyrimidines absor bed, but most of the purines and pyrimidines are
synthesized from amino acids, principally in the liver. The nucleotides and RNA and DN A are then synthesized. RNA is in dynamic
equilibrium with the amino acid pool, but DNA, once formed, is metabolically stable thro ughout life. The purines and pyrimidines
released by the breakdown of nucleotides may be reused or catabolized. Minor amoun ts are excreted unchanged in the urine.
The pyrimidines are catabolized to the β-amino acids, β- alanine and β-a minoisobutyrate. These amino acids have their amino group on
β-carbon, rather than the α-carbon typical to physiologically active amino acids. Becaus e β-aminoisobutyrate is a product of thymine
degradation, it can serve as a measure of DNA turnover. The β-amino acids are furthe r degraded to CO
2
and NH
3
.
Uric acid is formed by the breakdown of purines and by
Uric acid is formed by the breakdown of purines and by PRPP) and glutamine (Figur e 1–9). In humans, uric acid is excreted in the urine,
but in other mammals, uric acid is further oxidized to allantoin before excretion. The no rmal blood uric acid level in humans is
approximately 4 mg/dL (0.24 mmol/L). In the kidney, uric acid is filtered, reabsorbed, and secreted. Normally, 98% of the filtered uric
acid is reabsorbed and the remaining 2% makes up approximately 20% of the amount excreted. The remaining 80% comes from the
tubular secretion. The uric acid excretion on a purine-free diet is about 0.5 g/24 h and on a regular diet about 1 g/24 h. Excess uric acid in
the blood or urine is a characteristic of gout (Clinical Box 1–2).
C
N
Adenine:
N
1
6
5
C 7
Guanine: 8CH
H C2 3 4 C 9
Hypoxanthine:
N N
6-Aminopurine
1-Amino
6-oxypurine
6-Oxypurine
2,6-Dioxypurine
H
Xanthine: Purine nucleus
H
Cytosine: C
N 3 4 5C H
Uracil: C
2
1
6
CHH Thymine: N
Pyrimidine nucleus
TABLE 1–2 Purine- and pyrimidinecontaining compounds.
Type of
Compound Components
Nucleoside Purine or pyrimidine plus ribose or 2-deoxyribose
4-Amino
2-oxypyrimidine 2,4-Dioxypyrimidine
5-Methyl
2,4-dioxypyrimidine Nucleotide
(mononucleotide) Nucleoside plus phosphoric acid residue
Nucleic acid Many nucleotides forming double-helical structures of two polynucleotide chains
Nucleoprotein
Contain ribose Nucleic acid plus one or more simple basic proteins
Ribonucleic acids (RNA)
FIGURE 1–8 Principal physiologically important purines and pyrimidines. Purine and pyrimidine structures are shown next to
representative molecules from each group. Oxypurines and oxypyrimidines may form enol d erivatives (hydroxypurines and
hydroxypyrimidines) by migration of hydrogen to the oxygen substituents.
Contain
2-deoxyribose Deoxyribonucleic acids (DNA) Adenosine Guanosine
CLINICAL BOX 1–2
Hypoxanthine
Xanthine oxidase
5-PRPP + Glutamine
Xanthine
O
Xanthine oxidase
C NH HN C
C O O C C
N
NH
H
Uric acid (excreted in humans)
O NH H
2
N C
C O O C C
N
H
NH
H
Allantoin (excreted in other mammals)
FIGURE 1–9 Synthesis and breakdown of uric acid. Adenosine is converted to hypoxa nthine, which is then converted to xanthine, and
xanthine is converted to uric acid. The latter two reactions are both catalyzed by xanthin e oxidase. Guanosine is converted directly to
xanthine, while 5-PRPP and glutamine can be converted to uric acid. An additional oxida tion of uric acid to allantoin occurs in some
mammals.
Gout
Gout is a disease characterized by recurrent attacks of arthritis; urate deposits in the joint s, kidneys, and other tissues; and elevated blood
and urine uric acid levels. The joint most commonly affected initially is the metatarsophal angeal joint of the great toe. There are two
forms of “primary” gout. In one, uric acid production is increased because of various enzyme abnormalities. In the other, there is a
selective deficit in renal tubular transport of uric acid. In “secondary” gout, the uric acid levels in the body fluids are elevated as a result
of decreased excretion or increased production secondary to some other disease proce ss. For example, excretion is decreased in patients
treated with thiazide diuretics and those with renal disease. Production is increased in leu kemia and pneumonia because of increased
breakdown of uric acid-rich white blood cells.
The treatment of gout is aimed at relieving the acute arthritis with drugs such as colchicin e or nonsteroidal anti-inflammatory agents and
decreasing the uric acid level in the blood. Colchicine does not affect uric acid metabolis m, and it apparently relieves gouty attacks by
inhibiting the phagocytosis of uric acid crystals by leukocytes, a process that in some wa y produces the joint symptoms. Phenylbutazone
and probenecid inhibit uric acid reabsorption in the renal tubules. Allopurinol, which di rectly inhibits xanthine oxidase in the purine
degradation pathway, is one of the drugs used to decrease uric acid production.
DNA
Deoxyribonucleic acid (DNA) is found in bacteria, in the nuclei of eukaryotic cells, an d in mitochondria. It is made up of two extremely
long nucleotide chains containing the bases adenine (A), guanine (G), thymine (T), and cytosine (C) (Figure 1–10). The chains are bound
together by hydrogen bonding between the bases, with adenine bonding to thymine an d guanine to cytosine. This stable association forms
a double-helical structure (Figure 1–11). The double helical structure of DNA is compa cted in the cell by association with histones, and
further compacted into chromosomes. A diploid human cell contains 46 chromosom es.
A fundamental unit of DNA, or a gene, can be defined as the sequence of DNA n ucleotides that contain the information for the
production of an ordered amino acid sequence for a single polypeptide chain. Interesti ngly, the protein encoded by a single gene may be
subsequently divided into several different physiologically active proteins. Information i s accumulating at an accelerating rate about the
structure of genes and their regulation. The basic structure of a typical eukaryotic gene is shown in diagrammatic form in Figure 1–12. It
is made up of a strand of DNA that includes coding and noncoding regions. In eukary otes, unlike prokaryotes, the portions of the genes
that dictate the formation of proteins are usually broken into several segments (exo ns) separated by segments that are not translated
(introns). Near the transcription start site of the gene is a promoter, which is the site at which RNA polymerase and its cofactors bind .
It often includes a thymidine–adenine–thymidine–adenine (TATA) sequence (TATA b ox), which ensures that transcription starts at the
proper point. Farther out in the 5' region are regulatory elements, which include enhancer and silencer sequences. It has been estimated
that each gene has an average of five regulatory sites. Regulatory sequences are somet imes found in the 3'-flanking region as well.
Gene mutations occur when the base sequence in the DNA is altered from its origina l sequence. Such alterations can affect protein
structure and be passed on to daughter cells after cell division. Point mutations are single base substitutions. A variety of chemical
modifications (eg, alkylating or intercalating agents, or ionizing radiation) can lead to ch anges in DNA sequences and mutations. The
collection of genes within the full expression of DNA from an organism is termed its genome. An indication of the complexity of DNA
in the human haploid genome (the total genetic message) is its size; it is made up of 3 × 1 0
9
base pairs that can code for approximately
30,000 genes. This genetic message is the blueprint for
NH
2
NH
2
Phosphate
N
Phosphate
N
O
Base (cytosine)
O Base (cytosine)
O P O CH
2
N
O –
O P O CH2 N
O
O
O–
O
O–
C
H H
C
Sugar (deoxyribose)
C
H H
C
Sugar (ribose)
H C C H H C C H
OH H
OH OH
A Typical deoxyribonucleotide Typical ribonucleotide
Phosphate
NH2
O N N
Adenine (DNA and RNA) OP O CH
2
N
N
O
– O
O
Sugar N HNO
Guanine (DNA and RNA) OP O CH
2
N
N NH2Nucleotide – OO
NH
2
O
N Cytosine (DNA and RNA) OP O CH
2
N O
O
O
O
CH
3
O NH
Thymine (DNA only) OP O CH
2
N O
O
– O
O
Uracil (RNA only) NH
O
OP O CH
2
N O
O
O
B
FIGURE 1–10 Basic structure of nucleotides and nucleic acids. A) At left, the nucleotide cytosine is shown with deoxyribose and at
right with ribose as the principal sugar. B) Purine bases adenine and guanine are bound to each other or to pyrimidine bases, cytosine,
thymine, or uracil via a phosphodiester backbone between 2'-deoxyribosyl moieties attach ed to the nucleobases by an N-glycosidic bond.
Note that the backbone has a polarity (ie, a 5' and a 3' direction). Thymine is only fo und in DNA, while the uracil is only found in RNA.
GC
T A
T A
C G A T
Minor groove
G C
3.4 nm C G
A T Major groove
T A
2.0 nm
FIGURE 1–11 Double-helical structure of DNA. The compact structure has an approximatel y 2.0 nm thickness and 3.4 nm between full
turns of the helix that contains both major and minor grooves. The structure is maintained in the double helix by hydrogen bonding
between purines and pyrimidines across individual strands of DNA. Adenine (A) is bound to thymine (T) and cytosine (C) to guanine
(G). (Reproduced with permission from Murray RK et al: Harper’s Biochemistry, 26th ed. McGraw-Hill, 2003.)
REPLICATION: MITOSIS & MEIOSIS
At the time of each somatic cell division (mitosis), the two DNA chains separ ate, each serving as a template for the synthesis of a new
complementary chain. DNA polymerase catalyzes this reaction. One of the double helic es thus formed goes to one daughter cell and one
goes to the other, so the amount of DNA in each daughter cell is the same as that in the parent cell. The life cycle of the cell that begins
after mitosis is highly regulated and is termed the cell cycle (Figure 1–13). The G
1
(or Gap 1) phase represents a period of cell growth
and divides the end of mitosis from the DNA synthesis (or S) phase. Following DNA sy nthesis, the cell enters another period of cell
growth, the G
2
(Gap 2) phase. The ending of this stage is marked by chromosome condensation and the beginning of mitosis (M stage).
In germ cells, reduction division (meiosis) takes place during maturation. The net result is that one of ea ch pair of chromosomes ends up
in each mature germ cell; consequently, each mature germ cell contains half the amount of chromosomal material found in somatic cells.
Therefore, when a sperm unites with an ovum, the resulting zygote has the full complem ent of DNA, half of which came from the father
and half from the mother. The term “ploidy” is sometimes used to refer to the number o f chromosomes in cells. Normal resting diploid
cells are euploid and become tetraploid just before division. Aneuploidy is the condition in which a cell contains other than the haploid
number of chromosomes or an exact multiple of it, and this condition is common in canc erous cells.
the heritable characteristics of the cell and its descendants. The proteins formed from the DNA blueprint include all the enzymes, and
these in turn control the metabolism of the cell.
Each nucleated somatic cell in the body contains the full genetic message, yet there is gre at differentiation and specialization in the
functions of the various types of adult cells. Only small parts of the message are normal ly transcribed. Thus, the genetic message is
normally maintained in a repressed state. However, genes are controlled both spatially a nd temporally. First, under physiological
conditions, the double helix requires highly regulated interaction by proteins to unravel for replication, transcription, or both.
RNA
The strands of the DNA double helix not only replicate themselves, but also serve as tem plates by lining up complementary bases for the
formation in the nucleus of ribonucleic acids (RNA). RNA differs from DNA in that it is single-st randed, has uracil in place of
thymine, and its sugar moiety is ribose rather than 2'-deoxyribose (Figure 1–13). The p roduction of RNA from DNA is called
transcription. Transcription can lead to several types of RNA including: messenger RN A (mRNA), transfer RNA (tRNA), ribosomal
RNA (rRNA), and other RNAs. Transcription is catalyzed by various forms of RNA polymerase.
Basal Regulatory promoter region region Transcription Poly(A)
start site addition site Exon Exon
DNA 5' CAAT TATA AATAAA 3'
5' Intron 3' Noncoding Noncoding region region
FIGURE 1–12 Diagram of the components of a typical eukaryotic gene. The region that produces intro ns and exons is flanked by
noncoding regions. The 5'-flanking region contains stretches of DNA that interact with proteins to facilitate or inhibit transcription. The
3'-flanking region contains the poly(A) addition site. (Modified from Murray RK et al: Harper’s Biochem istry, 26th ed. McGraw-Hill, 2003.)
Mitotic phase
elophase
Mitosis
G
2
G
2
Final growth and activity before mitosis
Cytokinesis
G
1
Centrioles replicate
S
DNA replication
Interphase FIGURE 1–13 Sequence of events during the cell cycle. Immediate ly following mitosis (M) the cell enters a gap phase
(G1) before a DNA synthesis phase (S) a second gap phase (G2) and back to mi tosis. Collectively G1, S, and G2 phases are referred to as
interphase (I).
Typical transcription of an mRNA is shown in Figure 1–14. When suitably activated, tra nscription of the gene into a premRNA starts at
the cap site and ends about 20 bases beyond the AATAAA sequence. The RN A transcript is capped in the nucleus by addition of 7-
methylguanosine triphosphate to the 5' end; this cap is necessary for proper binding to t he ribosome. A poly(A) tail of about 100 bases is
added to the untranslated segment at the 3' end to help maintain the stability of the mRNA . The pre-mRNA formed by capping and
addition of the poly(A) tail is then processed by elimination of the introns, and once this posttranscriptional modification is complete, the
mature mRNA moves to the cytoplasm. Posttranscriptional modification of the pre-mRN A is a regulated process where differential
splicing can occur to form more than one mRNA from a single pre-mRNA. The intron s of some genes are eliminated by spliceosomes,
complex units that are made up of small RNAs and proteins. Other introns are eliminated by selfsplicing by the RNA they contain.
Because of introns and splicing, more than one mRNA can be formed from the same g ene.
Most forms of RNA in the cell are involved in translation, or protein synthesis. A b rief outline of the transition from transcription to
translation is shown in Figure 1–15. In the cytoplasm, ribosomes provide a template for tRNA to deliver specific amino acids to a
growing polypeptide chain based on specific sequences in mRNA. The mRNA molecu les are smaller than the DNA molecules, and each
represents a
Flanking DNA Introns
Gene Exons
Transcription Cap
Pre
mRNA
Flanking DNA
Poly(A)
RNA processing Poly(A)
mRNA Poly(A) Translation
FIGURE 1–14 Transcription of a typical mRNA. Steps in transcription from a typical gene to a processed mRNA are shown. Cap, cap
site. (Modified from Baxter JD: Principles of endocrinology. In: Cecil Textbook of Medicine, 16th ed. Wyng aarden JB, Smith LH Jr (editors). Saunders, 1982.)
transcript of a small segment of the DNA chain. For comparison, the molecules of tRNA contain only 70–80 nitrogenous bases,
compared with hundreds in mRNA and 3 billion in DNA.
AMINO ACIDS & PROTEINS
AMINO ACIDS
Amino acids that form the basic building blocks for proteins are identified in Table 1–3. These amino acids are often referred to by their
corresponding three-letter, or single-letter abbreviations. Various other important amino acids such as ornithine, 5-hydroxytryptophan, L-
dopa, taurine, and thyroxine (T
4
) occur in the body but are not found in proteins. In higher animals, the L isomers of th e amino acids are
the only naturally occurring forms in proteins. The L isomers of hormones such as thyr oxine are much more active than the D isomers.
The amino acids are acidic, neutral, or basic in reaction, depending on the relative prop ortions of free acidic (–COOH) or basic (–NH
2
)
groups in the molecule. Some of the amino acids are nutritionally essential amino acids, that is, they must be obtained in the diet,
because they cannot be made in the body. Arginine and histidine must be provided thro ugh diet during times of rapid growth or recovery
from illness and are termed conditionally essential. All others are nonessentia l amino acids in the sense that they can be synthesized in
vivo in amounts sufficient to meet metabolic needs.
DNA
RNA strand formed on DNA strand
(transcription)
tRNA
Amino acid
adenylate
Chain separation
Posttranscriptional modification
Activating enzyme Messenger RNA Coding triplets for
A3
A
A
2
Translation Posttranslational modification
Translation Posttranslational modification
Ribosome
tRNA-amino acid-adenylate A
3
A
2
A
1
complex
A4
Peptide chain
FIGURE 1–15 Diagrammatic outline of transcription to translation. Fr om the DNA molecule, a messenger RNA is produced and
presented to the ribosome. It is at the ribosome where charged tRNA match up with their com plementary codons of mRNA to position
the amino acid for growth of the polypeptide chain. DNA and RNA are represented as lines with multiple short projections representing
the individual bases . Small boxes labeled A represent individual amino acids.
TABLE 1–3 Amino acids found in proteins.*
Amino acids with aliphatic side chains Amino acids with acidic side chains, or their amide s Alanine (Ala, A) Aspartic acid (Asp, D)
Valine (Val, V) Asparagine (Asn, N) Leucine (Leu, L) Glutamine (Gln, Q) Isoleucine (IIe, I) Glutamic acid (Glu, E) Hydroxy l-
substituted amino acids γ-Carboxyglutamic acid
b
(Gla) Serine (Ser, S) Threonine (Thr, T)
Amino acids with side chains containing basic groups Arginine
c
(Arg, R)
Sulfur-containing amino acids Cysteine (Cys, C) Methionine (Met, M)
a
Selenocysteine
Lysine (Lys, K)
Hydroxylysine
b
(Hyl) Histidine
c
(His, H)
Imino acids (contain imino group but no amino group) Amino acids with aromatic ring side chains Proline (Pro, P)
Phenylalanine (Phe, F) Tyrosine (Tyr, Y) 4-Hydroxyproline
b
(Hyp)
b
3-Hydroxyproline
Tryptophan (Trp, W)
*Those in bold type are the nutritionally essential amino acids. The generally accepted three-letter and on e-letter abbreviations for the amino acids are shown in
parentheses.
a
Selenocysteine is a rare amino acid in which the sulfur of cysteine is replaced by selenium. The codon UG A is usually a stop codon, but in certain
situations it codes for selenocysteine.
b
There are no tRNAs for these four amino acids; they are formed by post-translational modification of the corresponding
unmodified amino acid in peptide linkage. There are tRNAs for selenocysteine and the remaining 20 amino acids, and they are incorporated into peptides and proteins
under direct genetic control.
c
Arginine and histidine are sometimes called “conditionally essential”—they are not necessary for maintena nce of nitrogen balance, but are
needed for normal growth.
THE AMINO ACID POOL
Although small amounts of proteins are absorbed from the gastrointestinal tract and som e peptides are also absorbed, most ingested
proteins are digested and their constituent amino acids absorbed. The body’s own prote ins are being continuously hydrolyzed to amino
acids and resynthesized. The turnover rate of endogenous proteins averages 80–100 g /d, being highest in the intestinal mucosa and
practically nil in the extracellular structural protein, collagen. The amino acids formed by endogenous protein breakdown are identical to
those derived from ingested protein. Together they form a common amino acid po ol that supplies the needs of the body (Figure 1–16).
this text, amino acid chains containing 2–10 amino acid residues are called peptides, cha ins containing more than 10 but fewer than 100
amino acid residues are called polypeptides, and chains containing 100 or more amino a cid residues are called proteins.
Diet Body Inert protein protein (hair, etc)
Urinary excretion Amino acid pool
Transamination
CommonAmination metabolicDeamination pool
PROTEINS
Proteins are made up of large numbers of amino acids linked into chains by pepti de bonds joining the amino group of one amino acid to
the carboxyl group of the next (Figure 1–17). In addition, some proteins contain carbo hydrates (glycoproteins) and lipids (lipoproteins).
Smaller chains of amino acids are called peptides or polypeptides. The boundaries between peptides, p olypeptides, and proteins are not
well defined. For
Creatine Purines, Hormones, NH
4
+
pyrimidines neurotransmitters
Urea
FIGURE 1–16 Amino acids in the body. There is an extensive network of amino acid turnover in the body. Boxes represent large pools
of amino acids and some of the common interchanges are represented by arrows. Note that most amino acids come from the diet and end
up in protein, however, a large portion of amino acids are interconverted and can feed into and out of a common metabolic pool through
amination reactions.
H OR H O
HH H H NC C N C C OH H–N C C H
R O R
Amino acid Polypeptide chain
FIGURE 1–17 Amino acid structure and formation of peptide bonds. The dashed line shows where peptide bonds are formed
between two amino acids. The highlighted area is released as H
2
O. R, remainder of the amino acid. For example, in glycine, R = H; in
glutamate, R = —(CH
.
2
)
2
—COO
The order of the amino acids in the peptide chains is called the primary structure of a protein. The chains are twisted and folded in
complex ways, and the term secondary structure of a protein refers to the spatial arrange ment produced by the twisting and folding. A
common secondary structure is a regular coil with 3.7 amino acid residues per turn (α- helix). Another common secondary structure is a
β-sheet. An antiparallel β-sheet is formed when extended polypeptide chains fold back and forth on one another and hydrogen bonding
occurs between the peptide bonds on neighboring chains. Parallel β-sheets between pol ypeptide chains also occur. The tertiary
structure of a protein is the arrangement of the twisted chains into layers, crystals, or fib ers. Many protein molecules are made of several
proteins, or subunits (eg, hemoglobin), and the term quaternary structure is used to refer to the a rrangement of the subunits into a
functional structure.
PROTEIN SYNTHESIS
The process of protein synthesis, translation, is the conversion of information en coded in mRNA to a protein (Figure 1–15). As
described previously, when a definitive mRNA reaches a ribosome in the cytoplasm, it dictates the formation of a polypeptide chain.
Amino acids in the cytoplasm are activated by combination with an enzyme and adenosi ne monophosphate (adenylate), and each
activated amino acid then combines with a specific molecule of tRNA. There is at least one tRNA for each of the 20 unmodified amino
acids found in large quantities in the body proteins of animals, but some amino acids hav e more than one tRNA. The tRNA–amino acid–
adenylate complex is next attached to the mRNA template, a process that occurs in the ri bosomes. The tRNA “recognizes” the proper
spot to attach on the mRNA template because it has on its active end a set of three bases that are complementary to a set of three bases in
a particular spot on the mRNA chain. The genetic code is made up of such triplets (c odons), sequences of three purine, pyrimidine, or
purine and pyrimidine bases; each codon stands for a particular amino acid.
Translation typically starts in the ribosomes with an AUG (transcribed from ATG in the gene), which codes for methionine. The amino
terminal amino acid is then added, and the chain is lengthened one amino acid at a time. The mRNA attaches to the 40S subunit of the
ribosome during protein synthesis, the polypeptide chain being formed attaches to the 6 0S subunit, and the tRNA attaches to both. As the
amino acids are added in the order dictated by the codon, the ribosome moves along th e mRNA molecule like a bead on a string.
Translation stops at one of three stop, or nonsense, codons (UGA, UAA, or UAG), an d the polypeptide chain is released. The tRNA
molecules are used again. The mRNA molecules are typically reused approximately 10 times before being replaced. It is common to
have more than one ribosome on a given mRNA chain at a time. The mRNA chain plus its collection of ribosomes is visible under the
electron microscope as an aggregation of ribosomes called a polyribosome.
POSTTRANSLATIONAL MODIFICATION
After the polypeptide chain is formed, it “folds” into its biological form and can be furth er modified to the final protein by one or more
of a combination of reactions that include hydroxylation, carboxylation, glycosylation, o r phosphorylation of amino acid residues;
cleavage of peptide bonds that converts a larger polypeptide to a smaller form; and the further folding, packaging, or folding and
packaging of the protein into its ultimate, often complex configuration. Protein folding is a complex process that is dictated primarily by
the sequence of the amino acids in the polypeptide chain. In some instances, however, n ascent proteins associate with other proteins
called chaperones, which prevent inappropriate contacts with other proteins and ensure that the final “proper” conformation of the
nascent protein is reached.
Proteins also contain information that helps to direct them to individual cell compartments . Many proteins that are going to be secreted or
stored in organelles and most transmembrane proteins have at their amino terminal a signal peptide (leader sequence) that guides them
into the endoplasmic reticulum. The sequence is made up of 15 to 30 predominantly hy drophobic amino acid residues. The signal
peptide, once synthesized, binds to a signal recognition particle (SRP), a complex molecule made up of six polypeptides and 7S RNA,
one of the small RNAs. The SRP stops translation until it binds to a translocon, a pore in the endoplasmic reticulum that is a
heterotrimeric structure made up of Sec 61 proteins. The ribosome also binds, and the s ignal peptide leads the growing peptide chain
into the cavity of the endoplasmic reticulum (Figure 1–18). The signal
5' 3'
N
UAA
SRP
NN NN
C C C C N
N N
FIGURE 1–18 Translation of protein into endoplasmic reticulum according to the signal hypothesis. The ribosomes synthesizing a
protein move along the mRNA from the 5' to the 3' end. When the signal pep tide of a protein destined for secretion, the cell membrane,
or lysosomes emerges from the large unit of the ribosome, it binds to a signal recognition particle (SRP), and this arrests further
translation until it binds to the translocon on the endoplasmic reticulum. N, amino end of protein; C, carboxyl end of protein. (Reproduced,
with permission, from Perara E, Lingappa VR: Transport of proteins into and across the endoplasm ic reticulum membrane. In: Protein Transfer and Organelle
Biogenesis. Das RC, Robbins PW (editors). Academic Press, 1988.)
peptide is next cleaved from the rest of the peptide by a signal peptidase while the rest o f the peptide chain is still being synthesized.
SRPs are not the only signals that help to direct proteins to their proper place in or out o f the cell; other signal sequences,
posttranslational modifications, or both (eg, glycosylation) can serve this function.
PROTEIN DEGRADATION
Like protein synthesis, protein degradation is a carefully regulated, complex process. It has been estimated that overall, up to 30% of
newly produced proteins are abnormal, such as can occur during improper folding. A ged normal proteins also need to be removed as
they are replaced. Conjugation of proteins to the 74-amino-acid polypeptide ubiquit in marks them for degradation. This polypeptide is
highly conserved and is present in species ranging from bacteria to humans. The proce ss of binding ubiquitin is called ubiquitination,
and in some instances, multiple ubiquitin molecules bind (polyubiquitination). Ubiquitin ation of cytoplasmic proteins, including
integral proteins of the endoplasmic reticulum, marks the proteins for degradation in mu ltisubunit proteolytic particles, or proteasomes.
Ubiquitination of membrane proteins, such as the growth hormone receptors, also mark s them for degradation, however these can be
degraded in lysosomes as well as via the proteasomes.
There is an obvious balance between the rate of production of a protein and its destruc tion, so ubiquitin conjugation is of major
importance in cellular physiology. The rates at which individual proteins are metabolized vary, and the body has mechanisms by which
abnormal proteins are recognized and degraded more rapidly than normal body consti tuents. For example, abnormal hemoglobins are
metabolized rapidly in individuals with congenital hemoglobinopathies.
CATABOLISM OF AMINO ACIDS
The short-chain fragments produced by amino acid, carbohydrate, and fat catabolism a re very similar (see below). From this common
metabolic pool of intermediates, carbohydrates, proteins, and fats can be synthesized. These fragments can enter the citric acid cycle, a
final common pathway of catabolism, in which they are broken down to hydrogen ato ms and CO
2
. Interconversion of amino acids
involve transfer, removal, or formation of amino groups. Transamination reactions, conversion of one amino acid to the corresponding
keto acid with simultaneous conversion of another keto acid to an amino acid, occur in many tissues:
Alanine +
α
-Ketoglutarate
Pyruvate + Glutamate
The transaminases involved are also present in the circulation. When damage to many active cells occurs as a result of a pathologic
process, serum transaminase levels rise. An example is the rise in plasma aspartate aminotr ansferase (AST) following myocardial
infarction.
Oxidative deamination of amino acids occurs in the liver. An imino acid is formed by dehydrogenation, and this compound is
hydrolyzed to the corresponding keto acid, with production of NH4+:
Amino acid + NAD
+
→ Imino acid + NADH + H
+
Imino acid + H
2
O → Keto acid + NH4+
Interconversions between the amino acid pool and the common metabolic pool are sum marized in Figure 1–19. Leucine, isoleucine,
phenylalanine, and tyrosine are said to be ketogenic because they are converted to th e ketone body acetoacetate (see below). Alanine
and many other amino acids are glucogenic or gluconeogenic; that is, they give rise to compou nds that can readily be converted to
glucose.
UREA FORMATION
Most of the NH 4+ formed by deamination of amino acids in the liver is converted to u rea, and the urea is excreted in the urine. The
NH4+ forms carbamoyl phosphate, and in the mitochondria it is transferred to ornithine , forming citrulline. The enzyme involved is
ornithine carbamoyltransferase. Citrulline is converted to arginine, after which urea is s plit off and ornithine is regenerated (urea cycle;
Figure 1–20). The overall reaction in the urea cycle consumes 3 ATP (not shown) and thus requires significant energy. Most of the urea
is formed in the liver, and in severe liver disease the blood urea nitrogen (BUN) falls a nd blood NH
3
rises (see Chapter 29). Congenital
deficiency of ornithine carbamoyltransferase can also lead to NH
3
intoxication, even in individuals who are heterozygous for this
deficiency.
Hydroxyproline Serine
Cysteine
Threonine
Glycine
Tryptophan Lactate
Transaminase Alanine
Pyruvate Acetyl-CoA
Glucose Phosphoenolpyruvate
carboxykinase
Phosphoenolpyruvate Oxaloacetate
Tyrosine
Phenylalanine Fumarate Transaminase
Aspartate Citrate
Isoleucine Methionine Valine
Succinyl-CoA
CO
2
Propionate α-Ketoglutarate
Histidine Proline
Histidine Proline
Glutamine Arginine
CO
2
Transaminase
Glutamate
FIGURE 1–19 Involvement of the citric acid cycle in transamination and gluco neogenesis. The bold arrows indicate the main
pathway of gluconeogenesis. Note the many entry positions for groups of amino acids into the citric acid cycle. (Reproduced with permission
from Murray RK et al: Harper’s Biochemistry, 26th ed. McGraw-Hill, 2003.)
METABOLIC FUNCTIONS
OF AMINO ACIDS
In addition to providing the basic building blocks for proteins, amino acids also have me tabolic functions. Thyroid hormones,
catecholamines, histamine, serotonin, melatonin, and intermediates in the urea cycle are f ormed from specific amino acids. Methionine
and cysteine provide the sulfur contained in proteins, CoA, taurine, and other biologica lly important compounds. Methionine is
converted into S-adenosylmethionine, which is the active methylating agent in the synthe sis of compounds such as epinephrine.
CARBOHYDRATES
Carbohydrates are organic molecules made of equal amounts of carbon and H
2
O. The simple sugars, or monosaccharides, including
pentoses (5 carbons; eg, ribose) and hexoses (6 carbons; eg, glucose) perform both structura l (eg, as part of nucleotides discussed
previously) and functional roles (eg, inositol 1,4,5 trisphosphate acts as a cellular signali ng molecules) in the body. Monosaccharides can
be linked together to form disaccharides (eg, sucrose), or polysaccharides (eg, glycog en). The placement of sugar moieties onto proteins
(glycoproteins) aids in cellular targeting, and in the case of some
Argininosuccinate
Aspartate Fumarate
Cyto
H
2
N H
2
N
O C
NH
2
+
C
HN HN Citrulline
+
NO Arginine (CH
2
)
3
(CH
2
)
3
HC NH
3
+
HC NH
3
+
Ornithine
−COO− COOH3N+
Pi
Mito (CH
2
)
3 Urea
HC NH3+ NH2
Carbamoyl
phosphate
COO− C
O
NH
4
+
NH
3
NH
2
FIGURE 1–20 Urea cycle. The processing of NH
3
to urea for excretion contains several coordinative steps in both the cytoplasm (Cyto)
and the mitochondria (Mito). The production of carbamoyl phosphate and its c onversion to citrulline occurs in the mitochondria, whereas
other processes are in the cytoplasm.
H CO H CO CH
2
OH H COH H COH CO
the body because, except for the quantitatively unimportant production from glycerol, t here is no pathway for conversion.
HO CH HO CH HO CH H COH HO CH H COH H COH H COH H COH
CH
2
OH CH
2
OH CH
2
OH
D-Glucose D-Galactose D-Fructose
FIGURE 1–21 Structures of principal dietary hexoses. Glucose, galactose, a nd fructose are shown in their naturally occurring D
isomers.
receptors, recognition of signaling molecules. In this section we will discuss a major role for carbohydrates in physiology, the production
and storage of energy.
Dietary carbohydrates are for the most part polymers of hexoses, of which the most im portant are glucose, galactose, and fructose
(Figure 1–21). Most of the monosaccharides occurring in the body are the D isomers. The principal product of carbohydrate digestion
and the principal circulating sugar is glucose. The normal fasting level of plasma glucos e in peripheral venous blood is 70 to 110 mg/dL
(3.9–6.1 mmol/ L). In arterial blood, the plasma glucose level is 15 to 30 mg/ dL higher than in venous blood.
Once it enters the cells, glucose is normally phosphorylated to form glucose 6-phosphat e. The enzyme that catalyzes this reaction is
hexokinase. In the liver, there is an additional enzyme called glucokina se, which has greater specificity for glucose and which, unlike
hexokinase, is increased by insulin and decreased in starvation and diabetes. The glucos e 6-phosphate is either polymerized into
glycogen or catabolized. The process of glycogen formation is called glycogenesis, and glycogen breakd own is called glycogenolysis.
Glycogen, the storage form of glucose, is present in most body tissues, but the major su pplies are in the liver and skeletal muscle. The
breakdown of glucose to pyruvate or lactate (or both) is called glycolysis. Glucos e catabolism proceeds via cleavage through fructose to
trioses or via oxidation and decarboxylation to pentoses. The pathway to pyruvate throu gh the trioses is the Embden–Meyerhof
pathway, and that through 6-phosphogluconate and the pentoses is the direct oxidative pathway (hexose monophosphate shunt).
Pyruvate is converted to acetyl-CoA. Interconversions between carbohydrate, fat, and protein include conversion of the glycerol from fats
to dihydroxyacetone phosphate and conversion of a number of amino acids with carbo n skeletons resembling intermediates in the
Embden–Meyerhof pathway and citric acid cycle to these intermediates by deamination. In this way, and by conversion of lactate to
glucose, nonglucose molecules can be converted to glucose (gluconeogenesis). Glucose c an be converted to fats through acetyl-CoA, but
because the conversion of pyruvate to acetyl-CoA, unlike most reactions in glycolysis, is irreversible, fats are not converted to glucose
via this pathway. There is therefore very little net conversion of fats to carbohydrates in
CITRIC ACID CYCLE
The citric acid cycle (Krebs cycle, tricarboxylic acid cycle) is a sequence of reactions in which ac etyl-CoA is metabolized to CO
2
and H
atoms. Acetyl-CoA is first condensed with the anion of a four-carbon acid, oxaloacetat e, to form citrate and HS-CoA. In a series of seven
subsequent reactions, 2CO
2
molecules are split off, regenerating oxaloacetate (Figure 1–22). Four pairs of H atom s are transferred to the
flavoprotein– cytochrome chain, producing 12ATP and 4H
2
O, of which 2H
2
O is used in the cycle. The citric acid cycle is the common
pathway for oxidation to CO
2
and H
2
O of carbohydrate, fat, and some amino acids. The major entry into it is through acetylC oA, but a
number of amino acids can be converted to citric acid cycle intermediates by deaminatio n. The citric acid cycle requires O
2
and does not
function under anaerobic conditions.
ENERGY PRODUCTION
The net production of energy-rich phosphate compounds during the metabolism of glu cose and glycogen to pyruvate depends on
whether metabolism occurs via the Embden– Meyerhof pathway or the hexose monop hosphate shunt. By oxidation at the substrate level,
the conversion of 1 mol of phosphoglyceraldehyde to phosphoglycerate generates 1 m ol of ATP, and the conversion of 1 mol of
phosphoenolpyruvate of ATP, and the conversion of 1 mol of phosphoenolpyruvate phosp hate produces, via the Embden–Meyerhof
pathway, 2 mol of phosphoglyceraldehyde, 4 mol of ATP is generated per mole of glu cose metabolized to pyruvate. All these reactions
occur in the absence of O
2
and consequently represent anaerobic production of energy. However, 1 mol of ATP is used in forming
fructose 1,6-diphosphate from fructose 6-phosphate and 1 mol in phosphorylating glu cose when it enters the cell. Consequently, when
pyruvate is formed anaerobically from glycogen, there is a net production of 3 mol of ATP per mole of glucose 6-phosphate; however,
when pyruvate is formed from 1 mol of blood glucose, the net gain is only 2 mol of A TP.
A supply of NAD
+
is necessary for the conversion of phosphoglyceraldehyde to phosphoglycerate. Und er anaerobic conditions
(anaerobic glycolysis), a block of glycolysis at the phosphoglyceraldehyde conversion step might be expected to develop as soon as the
available NAD
+
is converted to NADH. However, pyruvate can accept hydrogen from NADH, form ing NAD
+
and lactate:
Pyruvate + NADH
Lactate + NAD
+
In this way, glucose metabolism and energy production can continue for a while withou t O
2
. The lactate that accumulates is converted
back to pyruvate when the O
2
supply is restored, with NADH transferring its hydrogen to the flavoprotein– cytochro me chain.
Pyruvate 3C
NAD
+
NADH + H
+ CO2
Acetyl-CoA 2C
Oxaloacetate 4C
NADH + H+
Citrate 6C NAD
+
Malate 4C
Isocitrate 6C
Fumarate 4C
NAD+
FADH
2 CO2
NADH + H
+
FAD
-Ketoglutarate 5C Succinate 4C
α
P CO
2 GTP
NAD
+
Succinyl-CoA 4C NADH
+
H+
GDP
FIGURE 1–22 Citric acid cycle. The numbers (6C, 5C, etc) indicate the number o f carbon atoms in each of the intermediates. The
conversion of pyruvate to acetyl-CoA and each turn of the cycle provide four NADH an d one FADH
2
for oxidation via the flavoprotein-
cytochrome chain plus formation of one GTP that is readily converted to ATP.
During aerobic glycolysis, the net production of ATP is 19 times greater than the two A TPs formed under anaerobic conditions. Six
ATPs are formed by oxidation via the flavoprotein–cytochrome chain of the two NAD Hs produced when 2 mol of
phosphoglyceraldehyde is converted to phosphoglycerate (Figure 1–22), six ATPs are formed from the two NADHs produced when 2
mol of pyruvate is converted to acetyl-CoA, and 24 ATPs are formed during the subse quent two turns of the citric acid cycle. Of these,
18 are formed by oxidation of six NADHs, 4 by oxidation of two FADH
2
s, and 2 by oxidation at the substrate level when succinyl-CoA
is converted to succinate. This reaction actually produces GTP, but the GTP is converted to ATP. Thus, the net production of ATP per
mol of blood glucose metabolized aerobically via the Embden–Meyerhof pathway and citric acid cycle is 2 + [2 × 3] + [2 × 3] + [2 × 12]
= 38.
Glucose oxidation via the hexose monophosphate shunt generates large amounts of NA DPH. A supply of this reduced coenzyme is
essential for many metabolic processes. The pentoses formed in the process are building blocks for nucleotides (see below). The amount
of ATP generated depends on the amount of NADPH converted to NADH and then o xidized.
“DIRECTIONAL-FLOW VALVES”
Metabolism is regulated by a variety of hormones and other factors. To bring about an y net change in a particular metabolic process,
regulatory factors obviously must drive a chemical reaction in one direction. Most of th e reactions in intermediary metabolism are freely
reversible, but there are a number of “directional-flow valves,” ie, reactions that procee d in one direction under the influence of one
enzyme or transport mechanism and in the opposite direction under the influence of an other. Five examples in the intermediary
metabolism of carbohydrate are shown in Figure 1–23. The different pathways for fat ty acid synthesis and catabolism (see below) are
another example. Regulatory factors exert their influence on metabolism by acting direc tly or indirectly at these directional-flow valves.
GLYCOGEN SYNTHESIS & BREAKDOWN
Glycogen is a branched glucose polymer with two types of glycoside linkages: 1:4α and 1:6α (Figure 1–24). It is synthesized on
glycogenin, a protein primer, from glucose 1-phosphate via uridine diphosphogluco se (UDPG). The enzyme glycogen synthase
catalyses the final synthetic step. The availability of
1. Glucose entry into cells and glucose exit from cells
Hexokinase
2. Glucose Glucose 6-phosphate Glucose 6-phosphatase
glycogenin is one of the factors determining the amount of glycogen synthesized. The b reakdown of glycogen in 1:4α linkage is
catalyzed by phosphorylase, whereas another enzyme catalyzes the breakdown of gly cogen in 1:6α linkage.
Glycogen synthase 3. Glucose 1-phosphate Glycogen Phosphorylase
Phospho
fructokinase
4. Fructose 6-phosphate
5. Phosphoenolpyruvate 5. Phosphoenolpyruvate 5. Phosphoenolpyr uvate
biphosphate
biphosphatase
ADP ATP
Pyruvate kinase
Pyruvate
Phosphoenolpyruvate carboxykinase Oxaloacetate
Pyruvate
Oxaloacetate
Malate Malate
FIGURE 1–23 Directional flow valves in energy production reactions. In carbohydrate met abolism there are several reactions that
proceed in one direction by one mechanism and in the other direction by a diff erent mechanism, termed “directional-flow valves.” Five
examples of these reactions are illustrated (numbered at left). The double line in example 5 repr esents the mitochondrial membrane.
Pyruvate is converted to malate in mitochondria, and the malate diffuses out of the mitochondria to the cytosol, where it is converted to
phosphoenolpyruvate.
FACTORS DETERMINING THE
PLASMA GLUCOSE LEVEL
The plasma glucose level at any given time is determined by the balance between the am ount of glucose entering the bloodstream and
the amount leaving it. The principal determinants are therefore the dietary intake; the rate of entry into the cells of muscle, adipose
tissue, and other organs; and the glucostatic activity of the liver (Figure 1–25). Five perc ent of ingested glucose is promptly converted
into glycogen in the liver, and 30–40% is converted into fat. The remainder is metaboliz ed in muscle and other tissues. During fasting,
liver glycogen is broken down and the liver adds glucose to the bloodstream. With mor e prolonged fasting, glycogen is depleted and
there is increased gluconeogenesis from amino acids and glycerol in the liver. Plasma gl ucose declines modestly to about 60 mg/dL
during prolonged starvation in normal individuals, but symptoms of hypoglycemia do n ot occur because gluconeogenesis prevents any
further fall.
CH
2
OH CH
2
OH O O
O
O
1:6α linkage
CH
2
OH CH
2
OH CH
2
OH Glycogen
O O O
CH
2
OH CH
2
OH CH
2
O O
O O O O O Glycogen synthase 1:4α linkage
Uridine
Phosphorylase a
diphospho
glucose
O−
CH
2
OH CH
2
O PO O
O−
O
O−
O PO
O
Glucose Glucose
1-phosphate 6-phosphate
FIGURE 1–24 Glycogen formation and breakdown. Glycogen is the main storage for glu cose in the cell. It is cycled: built up from
glucose
6-phosphate when energy is stored and broken down to glucose 6-phosphate when e nergy is required. Note the intermediate glucose
1-phosphate and enzymatic control by phosphorylase a and glycogen kinase.
Diet
Amino Glycerol acids
Intestine Liver Lactate
Plasma glucose 70 mg/dL
(3.9 mmol/L)
Kidney Brain Fat Muscle and other tissues
Urine (when plasma glucose > 180 mg/dL)
the intestines and liver, so its value in replenishing carbohydrate elsewhere in the body i s limited.
Fructose 6-phosphate can also be phosphorylated in the 2 position, forming fructose 2, 6-diphosphate. This compound is an important
regulator of hepatic gluconeogenesis. When the fructose 2,6-diphosphate level is high, conversion of fructose 6-phosphate to fructose
1,6-diphosphate is facilitated, and thus breakdown of glucose to pyruvate is increased. A decreased level of fructose 2,6-diphosphate
facilitates the reverse reaction and consequently aids gluconeogenesis.
FIGURE 1–25 Plasma glucose homeostasis. Notice the glucostat ic function of the liver, as well as the loss of glucose in the urine when
the renal threshold is exceeded (dashed arrows).
METABOLISM OF HEXOSES
OTHER THAN GLUCOSE
Other hexoses that are absorbed from the intestine include galactose, which is liberated b y the digestion of lactose and converted to
glucose in the body; and fructose, part of which is ingested and part produced by hydr olysis of sucrose. After phosphorylation, galactose
reacts with uridine diphosphoglucose (UDPG) to form uridine diphosphogalactose. Th e uridine diphosphogalactose is converted back to
UDPG, and the UDPG functions in glycogen synthesis. This reaction is reversible, and conversion of UDPG to uridine
diphosphogalactose provides the galactose necessary for formation of glycolipids and m ucoproteins when dietary galactose intake is
inadequate. The utilization of galactose, like that of glucose, depends on insulin. In the in born error of metabolism known as
galactosemia, there is a congenital deficiency of galactose 1-phosphate uridyl transfe rase, the enzyme responsible for the reaction
between galactose 1-phosphate and UDPG, so that ingested galactose accumulates in the circulation. Serious disturbances of growth and
development result. Treatment with galactose-free diets improves this condition without l eading to galactose deficiency, because the
enzyme necessary for the formation of uridine diphosphogalactose from UDPG is pre sent.
Fructose is converted in part to fructose 6-phosphate and then metabolized via fructose 1,6-diphosphate. The enzyme catalyzing the
formation of fructose 6-phosphate is hexokinase, the same enzyme that catalyzes the co nversion of glucose to glucose 6-phosphate.
However, much more fructose is converted to fructose 1-phosphate in a reaction cataly zed by fructokinase. Most of the fructose 1-
phosphate is then split into dihydroxyacetone phosphate and glyceraldehyde. The glyce raldehyde is phosphorylated, and it and the
dihydroxyacetone phosphate enter the pathways for glucose metabolism. Because the r eactions proceeding through phosphorylation of
fructose in the 1 position can occur at a normal rate in the absence of insulin, it has been recommended that fructose be given to
diabetics to replenish their carbohydrate stores. However, most of the fructose is metab olized in
FATTY ACIDS & LIPIDS
The biologically important lipids are the fatty acids and their derivatives, the neutral fats ( triglycerides), the phospholipids and related
compounds, and the sterols. The triglycerides are made up of three fatty acids bound to glycerol (Table 1–4). Naturally occurring fatty
acids contain an even number of carbon atoms. They may be saturated (no double bon ds) or unsaturated (dehydrogenated, with various
numbers of double bonds). The phospholipids are constituents of cell membranes and p rovide structural components of the cell
membrane, as well as an important source of intra- and intercellular signaling molecules . Fatty acids also are an important source of
energy in the body.
FATTY ACID OXIDATION & SYNTHESIS
In the body, fatty acids are broken down to acetyl-CoA, which enters the citric acid cy cle. The main breakdown occurs in the
mitochondria by β-oxidation. Fatty acid oxidation begins with activation (formation of th e CoA derivative) of the fatty acid, a reaction
that occurs both inside and outside the mitochondria. Medium- and short-chain fatty aci ds can enter the mitochondria without difficulty,
but long-chain fatty acids must be bound to carnitine in ester linkage before they can cross the inner mitochondrial membrane. Carnitine
is β-hydroxyγ-trimethylammonium butyrate, and it is synthesized in the body from lysin e and methionine. A translocase moves the fatty
acid–carnitine ester into the matrix space. The ester is hydrolyzed, and the carnitine recy cles. β-oxidation proceeds by serial removal of
two carbon fragments from the fatty acid (Figure 1–26). The energy yield of this proc ess is large. For example, catabolism of 1 mol of a
six-carbon fatty acid through the citric acid cycle to CO
2
and H
2
O generates 44 mol of ATP, compared with the 38 mol generated by
catabolism of 1 mol of the six-carbon carbohydrate glucose.
KETONE BODIES
In many tissues, acetyl-CoA units condense to form acetoacetylCoA (Figure 1–27). In the liver, which (unlike other tissues) contains a
deacylase, free acetoacetate is formed. This β-keto acid is converted to β-hydroxybutyr ate and acetone, and because these compounds are
metabolized with difficulty in
TABLE 1–4. Lipids.
Typical fatty acids:
O
Palmitic acid: CH
5
(CH
2
)
14
—C—OH O
Stearic acid: CH
5
(CH
2
)
16
—C—OH O
Oleic acid: CH
5
(CH
2
)
7
CH=CH(CH
2
)
7
—C—OH (Unsaturated)
Triglycerides (triacylglycerols): Esters of glycerol and three fatty acids.
O CH
2
—O—C—R CH
2
OH
O O
CH
2
—O—C—R + 3H
2
O CHOH + 3HO—C—R O
CH
2
—O—C—R CH
2
OH Triglyceride Glycerol
R = Aliphatic chain of various lengths and degrees of saturation.
Phospholipids:
A. Esters of glycerol, two fatty acids, and
1. Phosphate = phosphatidic acid
2. Phosphate plus inositol = phosphatidylinositol
3. Phosphate plus choline = phosphatidylcholine (lecithin)
4. Phosphate plus ethanolamine = phosphatidyl-ethanolamine (cephalin)
5. Phosphate plus serine = phosphatidylserine
CELLULAR LIPIDS
The lipids in cells are of two main types: structural lipids, which are an inherent part of the membranes and other parts of cells; and
neutral fat, stored in the adipose cells of the fat depots. Neutral fat is mobi lized during starvation, but structural lipid is preserved. The
fat depots obviously vary in size, but in nonobese individuals they make up about 15% of body weight in men and 21% in women. They
are not the inert structures they were once thought to be but, rather, active dynamic tissu es undergoing continuous breakdown and
resynthesis. In the depots, glucose is metabolized to fatty acids, and neutral fats are synth esized. Neutral fat is also broken down, and
free fatty acids are released into the circulation.
A third, special type of lipid is brown fat, which makes up a small percentage of total bo dy fat. Brown fat, which is somewhat more
abundant in infants but is present in adults as well, is located between the scapulas, at the nape of the neck, along the great vessels in the
thorax and abdomen, and in other scattered locations in the body. In brown fat depots, the fat cells as well as the blood vessels have an
extensive sympathetic innervation. This is in contrast to white fat depots, in which some f at cells may be innervated but the principal
sympathetic innervation is solely on blood vessels. In addition, ordinary lipocytes have o nly a single large droplet of white fat, whereas
brown fat cells contain several small droplets of fat. Brown fat cells also contain many m itochondria. In these mitochondria, an inward
proton conductance that generates ATP takes places as usual, but in addition there is a se cond proton conductance that does not generate
ATP. This “shortcircuit” conductance depends on a 32-kDa uncoupling protein (UCP1 ). It causes uncoupling of metabolism and
generation of ATP, so that more heat is produced.
B. Other phosphate-containing derivatives of glycerol
C. Sphingomyelins: Esters of fatty acid, phosphate, choline, and the amino alcohol sph ingosine.
Cerebrosides: Compounds containing galactose, fatty acid, and sphingosine.
Sterols: Cholesterol and its derivatives, including steroid hormones, bile acids, and various vitam ins.
the liver, they diffuse into the circulation. Acetoacetate is also formed in the liver via the formation of 3-hydroxy-3-methylglutaryl-CoA,
and this pathway is quantitatively more important than deacylation. Acetoacetate, β-hydr oxybutyrate, and acetone are called ketone
bodies. Tissues other than liver transfer CoA from succinyl-CoA to acetoaceta te and metabolize the “active” acetoacetate to CO
2
and
H
2
O via the citric acid cycle. Ketone bodies are also metabolized via other pathways. Aceto ne is discharged in the urine and expired air.
An imbalance of ketone bodies can lead to serious health problems (Clinical Box 1–3).
PLASMA LIPIDS & LIPID TRANSPORT
The major lipids are relatively insoluble in aqueous solutions and do not circulate in the f ree form. Free fatty acids (FFAs) are bound to
albumin, whereas cholesterol, triglycerides, and phospholipids are transported in the fo rm of lipoprotein complexes. The complexes
greatly increase the solubility of the lipids. The six families of lipoproteins (Table 1–5) ar e graded in size and lipid content. The density
of these lipoproteins is inversely proportionate to their lipid content. In general, the lipop roteins consist of a hydrophobic core of
triglycerides and cholesteryl esters surrounded by phospholipids and protein. These lipo proteins can be transported from the intestine to
the liver via an exogenous pathway, and between other tissues via an endogenous pathway.
Dietary lipids are processed by several pancreatic lipases in the intestine to form mixed m icelles of predominantly FFA, 2-
monoglycerols, and cholesterol derivatives (see Chapter 27). These micelles ad ditionally can contain important water-insoluble
molecules such as vitamins A, D, E, and K. These mixed micelles a re taken up into cells of the intestinal
Fatty acid "Active" fatty acid
O O Mg
2+
R CH
2
CH
2
C OH + HS-CoA
ATP ADP
H
2
O + R CH
2
CH
2
C S CoA
Oxidized
flavoprotein
Reduced
flavoprotein
OH O O R C CH
CH C S CoA
2
C S CoA H
2
O + R CH
H β-Hydroxy fatty acid–CoA α,β-Unsaturated fatty acid–CoA NAD
+
NADH + H
+
O O O O R C CH
2
CS CoA + HS-CoA R C S CoA + CH
3
CS CoA
β-Keto fatty acid–CoA "Active" fatty acid + Acetyl–CoA
R = Rest of fatty acid chain. FIGURE 1–26 Fatty acid oxidation. This process, splitting off two ca rbon fragments at a time, is repeated
to the end of the chain.
O O
β-Ketothiolase
O O
CH
3
C S CoA + CH
3
C S CoA CH
3
C CH
2
C S CoA + HS-CoA 2 Acetyl-CoA Acetoacetyl-CoA
CH O O O O
Deacylase
CH
3
C CH
2
C O
+ H
+
+ HS-CoA
3
C CH
2
C S CoA + H
2
O
(liver only)
Acetoacetyl-CoA Acetoacetate
OH O
Acetyl-CoA + Acetoacetyl-CoA CH
3
C CH
2
C S CoA + H
+
CH
2
COO
3-Hydroxy-3-methylglutaryl-CoA (HMG-CoA)
HMG-CoA Acetoacetate + H
+
+ Acetyl-CoA
Acetoacetate
O O
CH
3
C
CH
2
C
O
+ H
+
Tissues except liver CO
2
+ ATP
–CO
2
+2H –2H
O
CH
3
C CH
3 O
Acetone
CH
3
CHOH CH
2
C O
+ H
+
β-Hydroxybutyrate FIGURE 1–27 Formation and metabolism of ketone bodies. No te the two pathways
for the formation of acetoacetate.
CLINICAL BOX 1–3 Diseases Associated with Imbalance of β-oxidation of F atty Acids
Ketoacidosis
The normal blood ketone level in humans is low (about 1 mg/dL) and less than 1 mg is excreted per 24 h, because the ketones are
normally metabolized as rapidly as they are formed. However, if the entry of acetyl-Co A into the citric acid cycle is depressed because
of a decreased supply of the products of glucose metabolism, or if the entry does not in crease when the supply of acetyl-CoA increases,
acetyl-CoA accumulates, the rate of condensation to acetoacetyl-CoA increases, and mo re acetoacetate is formed in the liver. The ability
of the tissues to oxidize the ketones is soon exceeded, and they accumulate in the bloods tream (ketosis). Two of the three ketone bodies,
acetoacetate and β-hydroxybutyrate, are anions of the moderately strong acids acetoace tic acid and β-hydroxybutyric acid. Many of their
protons are buffered, reducing the decline in pH that would otherwise occur. Howeve r, the buffering capacity can be exceeded, and the
metabolic acidosis that develops in conditions such as diabetic ketosis can be severe
mucosa where large lipoprotein complexes, chylomicrons, are formed. The chylomicrons and their remn ants constitute a transport
system for ingested exogenous lipids (exogenous pathway). Chylomicrons can enter th e circulation via the lymphatic ducts. The
chylomicrons are cleared from the circulation by the action of lipoprotein lipase , which is located on the surface of the endothelium of
the capillaries. The enzyme catalyzes the breakdown of the triglyceride in the chylomicr ons to FFA and glycerol, which then enter
adipose
and even fatal. Three conditions lead to deficient intracellular glucose supplies, and henc e to ketoacidosis: starvation; diabetes mellitus;
and a high-fat, low-carbohydrate diet. The acetone odor on the breath of children who have been vomiting is due to the ketosis of
starvation. Parenteral administration of relatively small amounts of glucose abolishes the k etosis, and it is for this reason that
carbohydrate is said to be antiketogenic.
Carnitine Deficiency
Deficient β-oxidation of fatty acids can be produced by carnitine deficiency or genetic defects in the translocase or other enzymes
involved in the transfer of long-chain fatty acids into the mitochondria. This causes card iomyopathy. In addition, it causes
hypoketonemic hypoglycemia with coma, a serious and often fatal condition triggered by fasting, in which glucose stores are used up
because of the lack of fatty acid oxidation to provide energy. Ketone bodies are not fo rmed in normal amounts because of the lack of
adequate CoA in the liver.
cells and are reesterified. Alternatively, the FFA can remain in the circulation bound to a lbumin. Lipoprotein lipase, which requires
heparin as a cofactor, also removes triglycerides from circulating very low density lipop roteins (VLDL). Chylomicrons depleted of
their triglyceride remain in the circulation as cholesterol-rich lipoproteins called chylom icron remnants, which are 30 to 80 nm in
diameter. The remnants are carried to the liver, where they are internalized and degrad ed.
TABLE 1–5 The principal lipoproteins.*
Composition (%)
Lipoprotein Size (nm) Protein Free Cholesterol
Cholesteryl Esters Triglyceride Phospholipid Origin
Chylomicrons 75–1000 2 2 3 90 3 Intestine
Chylomicron remnants 30–80 … … … … … Capillaries
Very low density lipoproteins 30–80 (VLDL)
17 Liver and intestine 8 4 16 55
Intermediate-density lipo25–40 proteins (IDL)
10 5 25 40 20 VLDL
Low-density lipoproteins 20 (LDL)
20 7 46 6 21 IDL
High-density lipoproteins 7.5–10 (HDL)
50 4 16 5 25 Liver and intestine
*The plasma lipids include these components plus free fatty acids from adipose tissue, which circulate bou nd to albumin.
The endogenous system, made up of VLDL, intermediate-density lipoproteins (IDL), low -density lipoproteins (LDL), and high-
density lipoproteins (HDL), also transports triglycerides and cholesterol throughout t he body. VLDL are formed in the liver and
transport triglycerides formed from fatty acids and carbohydrates in the liver to extrahe patic tissues. After their triglyceride is largely
removed by the action of lipoprotein lipase, they become IDL. The IDL give up phosp holipids and, through the action of the plasma
enzyme lecithin-cholesterol acyltransferase (LCAT), pick up cholester yl esters formed from cholesterol in the HDL. Some IDL are
taken up by the liver. The remaining IDL then lose more triglyceride and protein, prob ably in the sinusoids of the liver, and become
LDL. LDL provide cholesterol to the tissues. The cholesterol is an essential constituent in cell membranes and is used by gland cells to
make steroid hormones.
FREE FATTY ACID METABOLISM
In addition to the exogenous and endogenous pathways described above, FFA are also synthesized in the fat depots in which they are
stored. They can circulate as lipoproteins bound to albumin and are a major source of e nergy for many organs. They are used extensively
in the heart, but probably all tissues can oxidize FFA to CO
2
and H
2
O.
The supply of FFA to the tissues is regulated by two lipases. As noted above, lipoprotein lipase on the surface of the endothelium of the
capillaries hydrolyzes the triglycerides in chylomicrons and VLDL, providing FFA and glycerol, which are reassembled into new
triglycerides in the fat cells. The intracellular hormone-sensitive lipase of adip ose tissue catalyzes the breakdown of stored triglycerides
into glycerol and fatty acids, with the latter entering the circulation. Hormone-sensitive lip ase is increased by fasting and stress and
decreased by feeding and insulin. Conversely, feeding increases and fasting and stress decrease the activity of lipoprotein lipase.
CHOLESTEROL METABOLISM
Cholesterol is the precursor of the steroid hormones and bile acids and is an essential cons tituent of cell membranes. It is found only in
animals. Related sterols occur in plants, but plant sterols are not normally absorbed from the gastrointestinal tract. Most of the dietary
cholesterol is contained in egg yolks and animal fat.
Cholesterol is absorbed from the intestine and incorporated into the chylomicrons forme d in the intestinal mucosa. After the
chylomicrons discharge their triglyceride in adipose tissue, the chylomicron remnants br ing cholesterol to the liver. The liver and other
tissues also synthesize cholesterol. Some of the cholesterol in the liver is excreted in the b ile, both in the free form and as bile acids.
Some of the biliary cholesterol is reabsorbed from the intestine. Most of the cholesterol i n the liver is incorporated into VLDL and
circulates in lipoprotein complexes.
The biosynthesis of cholesterol from acetate is summarized in Figure 1–28. Cholesterol feeds back to inhibit its own synthesis by
inhibiting HMG-CoA reductase, the enzyme that converts 3-hydroxy-3-methylglutaryl-coen zyme A (HMG-CoA) to mevalonic acid.
Thus, when dietary cholesterol intake is high, hepatic cholesterol synthesis is decreased, and vice versa. However, the feedback
compensation is incomplete, because a diet that is low in cholesterol and saturated fat lead s to only a modest decline in circulating
plasma cholesterol. The most effective and most commonly used cholesterol-lowering d rugs are lovastatin and other statins, which
reduce cholesterol synthesis by inhibiting HMG-CoA. The relationship between choleste rol and vascular disease is discussed in Clinical
Box 1–4.
ESSENTIAL FATTY ACIDS
Animals fed a fat-free diet fail to grow, develop skin and kidney lesions, and become in fertile. Adding linolenic, linoleic, and
arachidonic acids to the diet cures all the deficiency symptoms. These three acids are po lyunsaturated fatty acids and because of their
action are called essential fatty acids. Similar deficiency symptoms h ave not been unequivocally demonstrated in humans, but there is
reason to believe that some unsaturated fats are essential dietary constituents, especially f or children.
Acetyl-CoA
Acetoacetyl-CoA
Acetoacetyl-CoA methylglutaryl-CoA HMG-CoA reductase
Mevalonic acid
Acetoacetate Acetoacetate Squalene
Cholesterol
CH
3
HOOC CH
2
CH
2
COH Squalene
2
OH
(C
30
H
50
) HO Mevalonic acid Cholesterol (C
27
H
46
O)
FIGURE 1–28 Biosynthesis of cholesterol. Six mevalonic acid molecules condense to form squal ene, which is then hydroxylated to
cholesterol. The dashed arrow indicates feedback inhibition by cholesterol of HMG-CoA reductase, the e nzyme that catalyzes mevalonic
acid formation.
CLINICAL BOX 1–4 CLINICAL BOX 1–5 Cholesterol & Athero sclerosis
The interest in cholesterol-lowering drugs stems from the role of cholesterol in the etiolo gy and course of atherosclerosis. This
extremely widespread disease predisposes to myocardial infarction, cerebral thrombosis , ischemic gangrene of the extremities, and other
serious illnesses. It is characterized by infiltration of cholesterol and oxidized cholesterol into macrophages, converting them into foam
cells in lesions of the arterial walls. This is followed by a complex sequence of changes involving platelets, macrophages, smooth muscle
cells, growth factors, and inflammatory mediators that produces proliferative lesions wh ich eventually ulcerate and may calcify. The
lesions distort the vessels and make them rigid. In individuals with elevated plasma choles terol levels, the incidence of atherosclerosis
and its complications is increased. The normal range for plasma cholesterol is said to be 120 to 200 mg/dL, but in men, there is a clear,
tight, positive correlation between the death rate from ischemic heart disease and plasma cholesterol levels above 180 mg/dL.
Furthermore, it is now clear that lowering plasma cholesterol by diet and drugs slows an d may even reverse the progression of
atherosclerotic lesions and the complications they cause.
In evaluating plasma cholesterol levels in relation to atherosclerosis, it is important to anal yze the LDL and HDL levels as well. LDL
delivers cholesterol to peripheral tissues, including atheromatous lesions, and the LDL pl asma concentration correlates positively with
myocardial infarctions and ischemic strokes. On the other hand, HDL picks up choleste rol from peripheral tissues and transports it to the
liver, thus lowering plasma cholesterol. It is interesting that women, who have a lower in cidence of myocardial infarction than men, have
higher HDL levels. In addition, HDL levels are increased in individuals who exercise an d those who drink one or two alcoholic drinks
per day, whereas they are decreased in individuals who smoke, are obese, or live sede ntary lives. Moderate drinking decreases the
incidence of myocardial infarction, and obesity and smoking are risk factors that increa se it. Plasma cholesterol and the incidence of
cardiovascular diseases are increased in familial hypercholesterolemia, due to vari ous loss-of-function mutations in the genes for LDL
receptors.
Dehydrogenation of fats is known to occur in the body, but there does not appear to b e any synthesis of carbon chains with the
arrangement of double bonds found in the essential fatty acids.
EICOSANOIDS
One of the reasons that essential fatty acids are necessary for health is that they are the p recursors of prostaglandins, prostacyclin,
thromboxanes, lipoxins, leukotrienes, and related compounds. These substances are cal led eicosanoids, reflecting
Pharmacology of Prostaglandins
Because prostaglandins play a prominent role in the genesis of pain, inflammation, and fever, pharmacologists have long sought drugs to
inhibit their synthesis. Glucocorticoids inhibit phospholipase A
2
and thus inhibit the formation of all eicosanoids. A variety of
nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit both cyclooxygenases, inhibiting the production of PGH
2
and its derivatives.
Aspirin is the bestknown of these, but ibuprofen, indomethacin, and others are also use d. However, there is evidence that prostaglandins
synthesized by COX2 are more involved in the production of pain and inflammation, a nd prostaglandins synthesized by COX1 are more
involved in protecting the gastrointestinal mucosa from ulceration. Drugs such as celeco xib and rofecoxib that selectively inhibit COX2
have been developed, and in clinical use they relieve pain and inflammation, possibly w ith a significantly lower incidence of
gastrointestinal ulceration and its complications than is seen with nonspecific NSAIDs. H owever, rofecoxib has been withdrawn from the
market in the United States because of a reported increase of strokes and heart attacks in individuals using it. More research is underway
to better understand all the effects of the COX enzymes, their products, and their inhibi tors.
their origin from the 20-carbon (eicosa-) polyunsaturated fatty acid arachidonic acid (arachidonate) and the 20-carbon derivatives of
linoleic and linolenic acids.
The prostaglandins are a series of 20-carbon unsaturated fatty acids containing a cyclopen tane ring. They were first isolated from semen
but are now known to be synthesized in most and possibly in all organs in the body. Pr ostaglandin H
2
(PGH
2
) is the precursor for
various other prostaglandins, thromboxanes, and prostacyclin. Arachidonic acid is form ed from tissue phospholipids by phospholipase
A
2
. It is converted to prostaglandin H
2
(PGH
2
) by prostaglandin G/H synthases 1 and 2. These are bifunctional enzymes that have both
cyclooxygenase and peroxidase activity, but they are more commonly known by the n ames cyclooxygenase 1 (COX1) and
cyclooxygenase 2 (COX2). Their structures are very similar, but COX1 is constitutive where as COX2 is induced by growth factors,
cytokines, and tumor promoters. PGH
2
is converted to prostacyclin, thromboxanes, and prostaglandins by various tissue isome rases. The
effects of prostaglandins are multitudinous and varied. They are particularly important i n the female reproductive cycle, in parturition, in
the cardiovascular system, in inflammatory responses, and in the causation of pain. Dru gs that target production of prostaglandins are
among the most common over the counter drugs available (Clinical Box 1–5).
Arachidonic acid also serves as a substrate for the production of several physiologically important leukotrienes and lipoxins. The
leukotrienes, thromboxanes, lipoxins, and prostaglandins have been called local hormon es. They have short half-lives and are inactivated
in many different tissues. They undoubtedly act mainly in the tissues at sites in which the y are produced. The leukotrienes are mediators
of allergic responses and inflammation. Their release is provoked when specific allerge ns combine with IgE antibodies on the surfaces of
mast cells (see Chapter 3). They produce bronchoconstriction, constrict arterioles, incre ase vascular permeability, and attract neutrophils
and eosinophils to inflammatory sites. Diseases in which they may be involved include a sthma, psoriasis, adult respiratory distress
syndrome, allergic rhinitis, rheumatoid arthritis, Crohn’s disease, and ulcerative colitis.
CHAPTER SUMMARY
Cells contain approximately one third of the body fluids, while the remaining extracellular fluid is found between cells (interstitial
fluid) or in the circulating blood plasma.
The number of molecules, electrical charges, and particles of substances in sol ution are important in physiology.
The high surface tension, high heat capacity, and high electrical capacity allow H
2
O to function as an ideal solvent in physiology.
Biological buffers including bicarbonate, proteins, and phosphates can bind or rel ease protons in solution to help maintain pH.
Biological buffering capacity of a weak acid or base is greatest when pK
a
= pH.
Fluid and electrolyte balance in the body is related to plasma osmolality. Isotonic sol utions have the same osmolality as blood plasma,
hypertonic have higher osmolality, while hypotonic have lower osmolality.
Although the osmolality of solutions can be similar across a plasma membrane, the distrib ution of individual molecules and
distribution of charge across the plasma membrane can be quite different. These are affect ed by the Gibbs-Donnan equilibrium and can
be calculated using the Nernst potential equation.
There is a distinct difference in concentration of ions in the extracellular and intracellular fluids (concentration gradient). The
separation of concentrations of charged species sets up an electrical gradient at the plas ma membrane (inside negative). The
electrochemical gradient is in large part maintained by the Na, K ATPase.
Cellular energy can be stored in high-energy phosphate compounds, including ad enosine triphosphate (ATP). Coordinated oxidation-
reduction reactions allow for production of a proton gradient at the inner mitocho ndrial membrane that ultimately yields to the
production of ATP in the cell.
Nucleotides made from purine or pyrimidine bases linked to ribose or 2-deoxyrib ose sugars with inorganic phosphates are the basic
building blocks for nucleic acids, DNA, and RNA.
DNA is a double-stranded structure that contains the fundamental information for an organism. During cell division, DNA is faithfully
replicated and a full copy of DNA is in every cell. The fundamental unit of DNA is the ge ne, which encodes information to make
proteins in the cell. Genes are transcribed into messenger RNA, and with the help of r ibosomal RNA and transfer RNAs, translated into
proteins.
Amino acids are the basic building blocks for proteins in the cell and can also serve as sour ces for several biologically active
molecules. They exist in an “amino acid pool” that is derived from the diet, protein degradat ion, and de novo and resynthesis.
Translation is the process of protein synthesis. After synthesis, proteins ca n undergo a variety of posttranslational modifications prior
to obtaining their fully functional cell state.
Carbohydrates are organic molecules that contain equal amounts of C and H
2
O. Carbohydrates can be attached to proteins
(glycoproteins) or fatty acids (glycolipids) and are critically important for the production and storage of cellular and body energy, with
major supplies in the form of glycogen in the liver and skeletal muscle. The breakdown of glucose to generate energy, or glycolysis, can
occur in the presence or absence of O
2
(aerobic or anaerobically). The net production of ATP during aerobic glycolysis is 19 tim es
higher than anaerobic glycolysis.
Fatty acids are carboxylic acids with extended hydrocarbon chains. They are an important energy source for cells and their derivatives,
including triglycerides, phospholipids and sterols, and have additional important cellula r applications. Free fatty acids can be bound to
albumin and transported throughout the body. Triglycerides, phospholipids, and cholestero l are transported as lipoprotein complexes.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. The membra ne potential of a particular cell is at the K
+
equilibrium. The intracellular concentration for K
+
is at 150 mmol/L
and the extracellular concentration for K
+
is at 5.5 mmol/L. What is the resting potential?
A) –70 mv
B) –90 mv
C) +70 mv
D) +90 mv
2. The difference in concentration of H
+
in a solution of pH 2.0 compared with one of pH 7.0 is
A) 5-fold.
B) 1/5 as much.
C) 10
5
fold.
D) 10
–5
as much.
3. Transcription refers to
A) the process where an mRNA is used as a template for protein production.
B) the process where a DNA sequence is copied into RNA for the purpose of gene expression .
C) the process where DNA wraps around histones to form a nucleosome.
D) the process of replication of DNA prior to cell division. 4. The primary structure of a protein refers to
A) the twist, folds, or twist and folds of the amino acid sequence into stabilized structure s within the protein (ie, α-helices and β-sheets).
B) the arrangement of subunits to form a functional structure.
C) the amino acid sequence of the protein.
D) the arrangement of twisted chains and folds within a protein into a stable structure.
5. Fill in the blanks: Glycogen is a storage form of glucose. _______ refers to the process of making glycogen and _______ refers to the
process of breakdown of glycogen.
A) Glycogenolysis, glycogenesis
B) Glycolysis, glycogenolysis
C) Glycogenesis, glycogenolysis
D) Glycogenolysis, glycolysis
6. The major lipoprotein source of the cholesterol used in cells is A) chylomicrons.
B) intermediate-density lipoproteins (IDLs).
C) albumin-bound free fatty acids.
D) LDL.
E) HDL.
7. Which of the following produces the most high-energy phosphate compounds?
A) aerobic metabolism of 1 mol of glucose
B) anaerobic metabolism of 1 mol of glucose
C) metabolism of 1 mol of galactose
D) metabolism of 1 mol of amino acid
E) metabolism of 1 mol of long-chain fatty acid
8. When LDL enters cells by receptor-mediated endocytosis, which of the follo wing does not occur?
A) Decrease in the formation of cholesterol from mevalonic acid.
B) Increase in the intracellular concentration of cholesteryl esters.
C) Increase in the transfer of cholesterol from the cell to HDL. D) Decrease in the rate of synthesis of LDL receptors. E) Decrease in the
cholesterol in endosomes.
CHAPTER RESOURCES
Alberts B, et al: Molecular Biology of the Cell, 5th ed. Garland Science, 2007.
Hille B: Ionic Channels of Excitable Membranes, 3rd ed. Sinauer Associates, 2001.
Kandel ER, Schwartz JH, Jessell TM: Principles of Neural Science, 4th ed. McGraw- Hill, 2000.
Macdonald RG, Chaney WG: USMLE Road Map, Biochemistry. McGraw- Hill, 2007 .
Murray RK, et al: Harper’s Biochemistry, 26th ed. McGraw-Hill, 2003.
Pollard TD, Earnshaw WC: Cell Biology, 2nd ed. Saunders, Elsevier, 2008.
Sack GH, Jr. USMLE Road Map, Genetics. McGraw Hill, 2008.
Scriver CR, et al (editors): The Metabolic and Molecular Bases of Inherited Disease, 8th ed. McGraw-Hill, 2001.
Sperelakis N (editor): Cell Physiology Sourcebook, 3rd ed. Academic Press, 2001 .
Overview of Cellular Physiology in Medical Physiology
CH APTER
2
OBJEC TIV ES
After studying this chapter, you should be able to:
Name the prominent cellular organelles and state their functions in cells.
Name the building blocks of the cellular cytoskeleton and state their contributions to cell structure and function.
Name the intercellular and cellular to extracellular connections.
Define the processes of exocytosis and endocytosis, and describe the contribution of each to normal cell function.
Define proteins that contribute to membrane permeability and transport.
Describe specialized transport and filtration across the capillary wall.
Recognize various forms of intercellular communication and describe ways in which chemical messengers (including second
messengers) affect cellular physiology.
Define cellular homeostasis.
INTRODUCTION
The cell is the fundamental working unit of all organisms. In humans, cells can be highl y specialized in both structure and function;
alternatively, cells from different organs can share features and function. In the previo us chapter, we examined some basic principles of
biophysics and the catabolism and metabolism of building blocks found in the cell. In so me of those discussions, we examined how the
building blocks could contribute to basic cellular physiology (eg, DNA replication, trans cription, and translation). In this chapter, we will
briefly review more of the fundamental aspects of cellular and molecular physiology. A dditional aspects that concern specialization of
cellular and molecular physiology are considered in the next chapter concerning immun e function and in the relevant chapters on the
various organs.
FUNCTIONAL MORPHOLOGY OF THE CELL
A basic knowledge of cell biology is essential to an understanding of the organ systems in the body and the way they function. A key
tool for examining cellular constituents is the microscope. A light microscope can resolv e structures as close as 0.2 μm, while an electron
microscope can resolve structures as close as 0.002 μm. Although cell dimensions are q uite variable, this resolution can give us a good
look at the inner workings of the cell. The advent of common access to fluorescent, con focal, and other microscopy along with
specialized probes for both static and dynamic cellular structures further expanded the examination of cell structure and function.
Equally revolutionary advances in the modern biophysical, biochemical, and molecular biology techniques have also greatly contributed
to our knowledge of the cell.
The specialization of the cells in the various organs is considerable, and no cell can be c alled “typical” of all cells in the body. However,
a number of structures (organelles) are common to most cells. These structures are shown in F igure 2–1. Many of them can be isolated
by ultracentrifugation combined with
31
Secretory granules
Golgi
apparatus
Centrioles
Rough endoplasmic reticulum Smooth
endoplasmic
reticulum
Lysosomes Nuclear envelope
Lipid
droplets Mitochondrion
Nucleolus Globular heads
FIGURE 2–1 Diagram showing a hypothetical cell in the center as seen with th e light microscope. Individual organelles are
expanded for closer examination. (Adapted from Bloom and Fawcett. Reproduced with permission fro m Junqueira LC, Carneiro J, Kelley RO: Basic Histology,
9th ed. McGraw-Hill, 1998.)
other techniques. When cells are homogenized and the resulting suspension is centrifug ed, the nuclei sediment first, followed by the
mitochondria. High-speed centrifugation that generates forces of 100,000 times gravity or more causes a fraction made up of granules
called the microsomes to sediment. This fraction includes organelles such as the ribosomes and peroxisomes.
CELL MEMBRANES
The membrane that surrounds the cell is a remarkable structure. It is made up of lipids a nd proteins and is semipermeable, allowing
some substances to pass through it and excluding others. However, its permeability can also be varied because it contains numerous
regulated ion channels and other transport proteins that can change the amounts of sub stances moving across it. It is generally referred to
as the plasma membrane. The nucleus and other organelles in the cell are bound by similar membranous structures.
Although the chemical structures of membranes and their properties vary considerably from one location to another, they have certain
common features. They are generally about 7.5 nm (75 Å) thick. The major lipids are phospholipids such as phosphatidylcholine and
phosphatidylethanolamine. The shape of the phospholipid molecule reflects its solubility properties: the head end of the molecule
contains the phosphate portion and is relatively soluble in water (polar, hydrophilic) and the tail s are relatively insoluble (nonpolar,
hydrophobic). The possession of both hydrophilic and hydrophobic properties make the lipid an amphipathic molecule. In the
membrane, the hydrophilic ends of the molecules are exposed to the aqueous environm ent that bathes the exterior of the cells and the
aqueous cytoplasm; the hydrophobic ends meet in the water-poor interior of the membr ane (Figure 2–2). In prokaryotes (ie, bacteria in
which there is no nucleus), the membranes are relatively simple, but in eukaryotes (cells containing nuclei), cell membranes contain
various glycosphingolipids, sphingomyelin, and cholesterol in addition to phospholipids and phosphatidylcholine.
Many different proteins are embedded in the membrane. They exist as separate globula r units and many pass through the membrane
(integral proteins), whereas others (peripheral proteins) stud the inside and outside of the membrane (Fi gure 2–2). The amount of
protein varies significantly with the function of the membrane but makes up on average 50% of the mass of the membrane; that is, there
is about one protein molecule per 50 of the much smaller phospholipid molecules. The p roteins in the membranes carry out many
functions. Some are cell adhesion molecules that anchor cells to their neighbors or to basal laminas. Some proteins function as pumps,
actively
Extracellular fluid
Carbohydrate portion of
glycoprotein Transmembrane
Phospholipidsproteins
Channel
Intregral
proteins
Peripheral protein
Polar regions
Nonpolar regions
Intracellular fluid
FIGURE 2–2 Organization of the phospholipid bilayer and associated protein s in a biological membrane. The phospholipid
molecules each have two fatty acid chains (wavy lines) attached to a phosphate head (o pen circle). Proteins are shown as irregular
colored globules. Many are integral proteins, which extend into the membrane, but periph eral proteins are attached to the inside or
outside (not shown) of the membrane. Specific protein attachments and cholesterol co mmonly found in the bilayer are omitted for clarity.
(Reproduced with permission from Widmaier EP, Raff H, Strang K: Vander’s Human Physiology: The Mechanisms of Body Function, 11th ed. McGraw-Hill, 2008.)
transporting ions across the membrane. Other proteins function as carriers, transporting substances down electrochemical gradients by
facilitated diffusion. Still others are ion channels, which, when activated, permit the passage of ions into or out of the cell. The role of
the pumps, carriers, and ion channels in transport across the cell membrane is discussed below. Proteins in another group function as
receptors that bind ligands or messenger molecules, initiating physiologic changes inside the cell. Proteins also function as enzymes,
catalyzing reactions at the surfaces of the membrane. Examples from each of these gro ups are discussed later in this chapter.
The uncharged, hydrophobic portions of the proteins are usually located in the interior of the membrane, whereas the charged,
hydrophilic portions are located on the surfaces. Peripheral proteins are attached to the surfaces of the membrane in various ways. One
common way is attachment to glycosylated forms of phosphatidylinositol. Proteins held b y these glycosylphosphatidylinositol (GPI)
anchors (Figure 2–3) include enzymes such as alkaline phosphatase, various antigens, a nu mber of cell adhesion molecules, and three
proteins that combat cell lysis by complement. Over 45 GPIlinked cell surface proteins h ave now been described in humans. Other
proteins are lipidated, that is, they have specific lipids attached to them (Figure 2–3). Proteins may be myristolated, palmitoylated, or
prenylated (ie, attached to geranylgeranyl or farnesyl groups).
The protein structure—and particularly the enzyme content—of biologic membranes va ries not only from cell to cell, but also within the
same cell. For example, some of the enzymes embedded in cell membranes are differen t from those in mitochondrial membranes. In
epithelial cells, the enzymes in the cell membrane on the mucosal surface differ from tho se in the
Lipid membrane
N-Myristoyl
Cytoplasmic or external face of membrane O
N Gly Protein COOH H
S-Cys Protein NH
2
S-Palmitoyl O
S-Cys Protein NH
2
Geranylgeranyl
S-Cys Protein NH
2
Farnesyl O C CCH
2
C C CH O O GPI anchor O C OO OC Protein (Glycosylphosphatidylinositol)
H2 O
Hydrophobic domain Hydrophilic domain
FIGURE 2–3 Protein linkages to membrane lipids. Some are linked by their am ino terminals, others by their carboxyl terminals. Many
are attached via glycosylated forms of phosphatidylinositol (GPI anchors). (R eproduced with permission from Fuller GM, Shields D: Molecular Basis
of Medical Cell Biology. McGraw-Hill, 1998.)
Intramemb space
H+ H+ H+ H+
Inner mito
CoQ Cyt c
membrane
Matrix space ADP
AS
ATP
Complex I II III IV V
Subunits from 70 1 3 2
mDNA
Subunits from 39 4 10 10 14
nDNA
FIGURE 2–4 Components involved in oxidative phosphorylation in mitochondr ia and their origins. As enzyme complexes I
through IV convert 2-carbon metabolic fragments to CO
2
and H
2
O, protons (H
+
) are pumped into the intermembrane space. The
proteins diffuse back to the matrix space via complex V, ATP synthase (AS), in which ADP is converted to ATP. The enzyme
complexes are made up of subunits coded by mitochondrial DNA (mDNA) and nuclear DNA (nDNA), and the figures document the
contribution of each DNA to the complexes.
cell membrane on the basal and lateral margins of the cells; that is, the cells are polarized. Such polarization makes transport acro ss
epithelia possible. The membranes are dynamic structures, and their constituents are bein g constantly renewed at different rates. Some
proteins are anchored to the cytoskeleton, but others move laterally in the membrane.
Underlying most cells is a thin, “fuzzy” layer plus some fibrils that collectively make up t he basement membrane o r, more properly,
the basal lamina. The basal lamina and, more generally, the extracellu lar matrix are made up of many proteins that hold cells together,
regulate their development, and determine their growth. These include collagens, laminin s, fibronectin, tenascin, and various
proteoglycans.
MITOCHONDRIA
Over a billion years ago, aerobic bacteria were engulfed by eukaryotic cells and evolv ed into mitochondria, providing the eukaryotic
Over a billion years ago, aerobic bacteria were engulfed by eukaryotic cells and evolv ed into mitochondria, providing the eukaryotic
cells with the ability to form the energy-rich compound ATP by oxidative pho sphorylation. Mitochondria perform other functions,
including a role in the regulation of apoptosis (programmed cell death), but oxidative phosphory lation is the most crucial. Each
eukaryotic cell can have hundreds to thousands of mitochondria. In mammals, they are generally depicted as sausage-shaped organelles
(Figure 2–1), but their shape can be quite dynamic. Each has an outer membrane, an in termembrane space, an inner membrane, which is
folded to form shelves (cristae), and a central matrix space. The enzyme complexes responsib le for oxidative phosphorylation are lined
up on the cristae (Figure 2–4).
Consistent with their origin from aerobic bacteria, the mitochondria have their own gen ome. There is much less DNA in the
mitochondrial genome than in the nuclear genome, and 99% of the proteins in the mitoc hondria are the products of nuclear genes, but
mitochondrial DNA is responsible for certain key components of the pathway for oxid ative phosphorylation. Specifically, human
mitochondrial DNA is a double-stranded circular molecule containing approximately 16 ,500 base pairs (compared with over a billion in
nuclear DNA). It codes for 13 protein subunits that are associated with proteins encode d by nuclear genes to form four enzyme
complexes plus two ribosomal and 22 transfer RNAs that are needed for protein produ ction by the intramitochondrial ribosomes.
The enzyme complexes responsible for oxidative phosphorylation illustrate the interactio ns between the products of the mitochondrial
genome and the nuclear genome. For example, complex I, reduced nicotinamide adeni ne dinucleotide dehydrogenase (NADH), is made
up of 7 protein subunits coded by mitochondrial DNA and 39 subunits coded by nucle ar DNA. The origin of the subunits in the other
complexes is shown in Figure 2–4. Complex II, succinate dehydrogenase-ubiquinone oxidoreductase; complex III,
ubiquinonecytochrome c oxidoreductase; and complex IV, cytochrome c oxidase, act w ith complex I, coenzyme Q, and cytochrome c to
convert metabolites to CO
2
and water. Complexes I, III, and IV pump protons (H
+
) into the intermembrane space during this electron
transfer. The protons then flow down their electrochemical gradient through complex V , ATP synthase, which harnesses this energy to
generate ATP.
As zygote mitochondria are derived from the ovum, their inheritance is maternal. This m aternal inheritance has been used as a tool to
track evolutionary descent. Mitochondria have an ineffective DNA repair system, and the mutation rate for mitochondrial DNA is over
10 times the rate for nuclear DNA. A large number of relatively rare diseases have now been traced to mutations in mitochondrial DNA.
These include for the most part disorders of tissues with high metabolic rates in which en ergy production is defective as a result of
abnormalities in the production of ATP.
LYSOSOMES
In the cytoplasm of the cell there are large, somewhat irregular structures surrounded b y membrane. The interior of these structures,
which are called lysosomes, is more acidic than the
TABLE 2–1 Some of the enzymes found in lysosomes and the cell component s that are their substrates.
CLINICAL BOX 2–1
Enzyme Substrate
Ribonuclease RNA
Deoxyribonuclease DNA
Phosphatase Phosphate esters
Glycosidases Complex carbohydrates; glycosides and polysaccharides
Arylsulfatases Sulfate esters
Collagenase Collagens
Cathepsins Proteins
Lysosomal Diseases
When a lysosomal enzyme is congenitally absent, the lysosomes become engorged with the material the enzyme normally degrades.
This eventually leads to one of the lysosomal storage diseases. For example, α-gala ctosidase A deficiency causes Fabry disease, and β-
galactocerebrosidase deficiency causes Gaucher disease. These diseases are rare, but th ey are serious and can be fatal. Another example
is the lysosomal storage disease called Tay–Sachs disease, which causes mental retardatio n and blindness. Tay–Sachs is caused by the
loss of hexosaminidase A, a lysosomal enzyme that catalyzes the biodegradation of gang liosides (fatty acid derivatives).
rest of the cytoplasm, and external material such as endocytosed bacteria, as well as wor n-out cell components, are digested in them. The
interior is kept acidic by the action of a proton pump, or H
+
, ATPase. This integral membrane protein uses the energy of ATP to move
protons from the cytosol up their electrochemical gradient and keep the lysosome relativ ely acidic, near pH 5.0. Lysosomes can contain
over 40 types of hydrolytic enzymes, some of which are listed in Table 2–1. Not surpr isingly, these enzymes are all acid hydrolases, in
that they function best at the acidic pH of the lysosomal compartment. This can be a safe ty feature for the cell; if the lysosome were to
break open and release its contents, the enzymes would not be efficient at the near neut ral cytosolic pH (7.2), and thus would be unable
to digest cytosolic enzymes they may encounter. Diseases associated with lysosomal dysf unction are discussed in Clinical Box 2–1.
PEROXISOMES
Peroxisomes are 0.5 μm in diameter, are surrounded by a membrane, and contain enzymes that can either produce H
2
O
2
(oxidases) or
break it down (catalases). Proteins are directed to the peroxisome by a unique signal s equence with the help of protein chaperones,
peroxins. The peroxisome membrane contains a number of peroxisome-specifi c proteins that are concerned with transport of substances
into and out of the matrix of the peroxisome. The matrix contains more than 40 enzyme s, which operate in concert with enzymes outside
the peroxisome to catalyze a variety of anabolic and catabolic reactions (eg, breakdown of lipids). Peroxisomes can form by budding of
endoplasmic reticulum, or by division. A number of synthetic compounds were found to cause proliferation of peroxisomes by acting on
receptors in the nuclei of cells. These peroxisome proliferation activated receptors (PPARs) are members of the nuclear receptor
superfamily. When activated, they bind to DNA, producing changes in the production of mRNAs. The known effects for PPARs are
extensive and can affect most tissues and organs.
CYTOSKELETON
All cells have a cytoskeleton, a system of fibers that not only maintains the structure of the cell b ut also permits it to change shape and
move. The cytoskeleton is made up primarily of microtubules, intermediate filaments, a nd microfilaments (Figure 2–5), along with
proteins that anchor them and tie them together. In addition, proteins and organelles mo ve along microtubules and microfilaments from
one part of the cell to another, propelled by molecular motors.
Microtubules (Figures 2–5 and 2–6) are long, hollow structures with 5-nm walls surrounding a cavity 15 nm in diameter. They are made
up of two globular protein subunits: α- and β-tubulin. A third subunit, γ-tubulin, is assoc iated with the production of microtubules by the
centrosomes. The α and β subunits form heterodimers, which aggregate to form long tu bes made up of stacked rings, with each ring
usually containing 13 subunits. The tubules interact with GTP to facilitate their formation. Although microtubule subunits can be added
to either end, microtubules are polar with assembly predominating at the “+” end and di sassembly predominating at the “–” end. Both
processes occur simultaneously in vitro. The growth of microtubules is temperature sen sitive (disassembly is favored under cold
conditions) as well as under the control of a variety of cellular factors that can directly i nteract with microtubules in the cell.
Because of their constant assembly and disassembly, microtubules are a dynamic portion of the cell skeleton. They provide the tracks
along which several different molecular motors move transport vesicles, organelles such as secretory granules, and mitochondria, from
one part of the cell to another. They also form the spindle, which moves the chromosom es in mitosis. Cargo can be transported in either
direction on microtubules.
There are several drugs available that disrupt cellular function through interaction with m icrotubules. Microtubule assembly is prevented
by colchicine and vinblastine. The anticancer drug paclitaxel (Taxol) bi nds to microtubules and
Cytoskeletal filaments Diameter (nm) Protein subunit
Microfilament 7 Actin
Intermediate filament 10 Several proteins
Microtubule 25 Tubulin
FIGURE 2–5 Cytoskeletal elements of the cell. Artistic impressions that depict the major cytoskeletal elements are shown on the left,
with basic properties of these elements on the right. (Reproduced with permission from Widm aier EP, Raff H, Strang KT: Vander’s Human Physiology: The
Mechanisms of Body Function, 11th ed. McGraw-Hill, 2008.)
makes them so stable that organelles cannot move. Mitotic spindles cannot form, and the cells die.
Intermediate filaments (Figures 2–5 and 2–6) are 8 to 14 nm in diameter and are made up of various subunits. Some of these filaments
connect the nuclear membrane to the cell membrane. They form a flexible scaffolding f or the cell and help it resist external pressure. In
their absence, cells rupture more easily, and when they are abnormal in humans, blister ing of the skin is common. The proteins that make
up intermediate filaments are celltype specific, and are thus frequently used as cellular m arkers. For example, vimentin is a major
intermediate filament in fibroblasts, whereas cytokeratin is expressed in epithelial cells.
Microfilaments (Figures 2–5 and 2–6) are long solid fibers with a 4 to 6 nm diam eter that are made up of actin. Although actin is most
often associated with muscle contraction, it is present in all types of cells. It is the most ab undant protein in mammalian cells, sometimes
accounting for as much as 15% of the total protein in the cell. Its structure is highly con served; for example, 88% of the amino acid
sequences in yeast and rabbit actin are identical. Actin filaments polymerize and depolym erize in vivo, and it is not uncommon to find
polymerization occurring at one end of the filament while depolymerization is occurring at the other end. Filamentous (F) actin refers to
intact microfilaments and globular (G) actin refers to the unpolymerized protein actin s ubunits. F-actin fibers attach to various parts of
the cytoskeleton and can interact directly or indirectly with membrane-bound proteins. T hey reach to the tips of the microvilli on the
epithelial cells of the intestinal mucosa. They are also abundant in the lamellipodia that cel ls put out when they crawl along surfaces. The
actin filaments interact with integrin
FIGURE 2–6 Microfilaments and microtubules. Electron microg raph (Left) of the cytoplasm of a fibroblast, displaying actin
microfilaments (MF) and microtubules (MT). (Reproduced, with permission, from Junque ira LC, Carneiro J: Basic Histology, 10th ed. McGraw-Hill, 2003.)
Fluorescent micrographs of airway epithelial cells displaying actin microfilaments stained with phalloidin (Middle) and microtubules
visualized with an antibody to β-tubulin (Right). Both fluorescent microgr aphs are counterstained with Hoechst dye (blue) to visualize
nuclei. Note the distinct differences in cytoskeletal structure.
receptors and form focal adhesion complexes, which serve as points of traction with the su rface over which the cell pulls itself. In
addition, some molecular motors use microfilaments as tracks. they perform functions a s diverse as contraction of muscle and cell
migration.
MOLECULAR MOTORS
The molecular motors that move proteins, organelles, and other cell parts (collectively re ferred to as “cargo”) to all parts of the cell are
100 to 500 kDa ATPases. They attach to their cargo at one end of the molecule and to microtubules or actin polymers with the other end,
sometimes referred to as the “head.” They convert the energy of ATP into movement a long the cytoskeleton, taking their cargo with
them. There are three super families of molecular motors: kinesin, dynein, and myosin. Examples of individual proteins from each
superfamily are shown in Figure 2–7. It is important to note that there is extensive variat ion among superfamily members, allowing for
specialization of function (eg, choice of cargo, cytoskeletal filament type, and/or directio n of movement).
The conventional form of kinesin is a doubleheaded molecule that tends to move it s cargo toward the “+” ends of microtubules. One
head binds to the microtubule and then bends its neck while the other head swings forw ard and binds, producing almost continuous
movement. Some kinesins are associated with mitosis and meiosis. Other kinesins perform different functions, including, in some
instances, moving cargo to the “–” end of microtubules. Dyneins have two heads, with the ir neck pieces embedded in a complex of
proteins. Cytoplasmic dyneins have a function like that of conventional kinesin, except they tend to move particles and membranes to
the “–” end of the microtubules. The multiple forms of myosin in the body are divid ed into 18 classes. The heads of myosin molecules
bind to actin and produce motion by bending their neck regions (myosin II) or walking along microfilaments, one head after the other
(myosin V). In these ways,
CENTROSOMES
Near the nucleus in the cytoplasm of eukaryotic animal cells is a centrosome . The centrosome is made up of two centrioles and
surrounding amorphous pericentriolar material. The centrioles are short cylinders arra nged so that they are at right angles to each
other. Microtubules in groups of three run longitudinally in the walls of each centriole ( Figure 2–1). Nine of these triplets are spaced at
regular intervals around the circumference.
The centrosomes are microtubule-organizing centers (MTOCs) that con tain γ-tubulin. The microtubules grow out of this γ-tubulin in
the pericentriolar material. When a cell divides, the centrosomes duplicate themselves, an d the pairs move apart to the poles of the
mitotic spindle, where they monitor the steps in cell division. In multinucleate cells, a centr osome is near each nucleus.
CILIA
Cilia are specialized cellular projections that are used by unicellular organisms to prop el themselves through liquid and by multicellular
organisms to propel mucus and other substances over the surface of various epithelia. C ilia are functionally indistinct from the eukaryotic
flagella of sperm cells. Within the cilium there is an axoneme that comprises a unique arrangemen t of nine outer microtubule doublets
and two inner microtubules (“9+2” arrangement). Along this cytoskeleton is axon emal dynein. Coordinated dynein-microtubule
interactions within the axoneme are the basis of ciliary and sperm movement. At the bas e of the axoneme and just inside lies the basal
body. It has nine circumferential triplet microtubules, like a centriole, and there is ev idence that basal bodies and centrioles are
interconvertible.
Cargo
Conventional kinesin Light
chains
4 nm
80 nm
Cytoplasmic dynein
Cargo-binding domain
Head 1 Head 2 Head 2 Head 1 ADP ADP ATP
Actin Myosin V
FIGURE 2–7 Three examples of molecular motors. Conventional kinesin is shown attached to cargo, in this case a membrane-bound
organelle. The way that myosin V “walks” along a microtubule is also shown. Note that the heads of the motors hydrolyze ATP and use
the energy to produce motion.
CELL ADHESION MOLECULES
Cells are attached to the basal lamina and to each other by cell adhesion molecu les (CAMs) that are prominent parts of the intercellular
connections described below. These adhesion proteins have attracted great attention in r ecent years because of their unique structural and
signaling functions found to be important in embryonic development and formation of the nervous system and other tissues, in holding
tissues together in adults, in inflammation and wound healing, and in the metastasis of tum ors. Many CAMs pass through the cell
membrane and are anchored to the cytoskeleton inside the cell. Some bind to like molecu les on other cells (homophilic binding), whereas
others bind to nonself molecules (heterophilic binding). Many bind to laminins, a family of large crossshaped molecules with multiple
receptor domains in the extracellular matrix.
Nomenclature in the CAM field is somewhat chaotic, partly because the field is growing so rapidly and partly because of the extensive
use of acronyms, as in other areas of modern biology. However, the CAMs can be div ided into four broad families: (1) integrins,
heterodimers that bind to various receptors; (2) adhesion molecules of the IgG superfamily of immunoglobulins; (3) cadherins, Ca
2+
-
dependent molecules that mediate cell-to-cell adhesion by homophilic reactions; and (4) selectins, which have lectin-like domains that
bind carbohydrates. Specific functions of some of these molecules are addressed in late r chapters.
The CAMs not only fasten cells to their neighbors, but they also transmit signals into and out of the cell. For example, cells that lose their
contact with the extracellular matrix via integrins have a higher rate of apoptosis than an chored cells, and interactions between integrins
and the cytoskeleton are involved in cell movement.
INTERCELLULAR CONNECTIONS
Intercellular junctions that form between the cells in tissues can be broadly split into two groups: junctions that fasten the cells to one
another and to surrounding tissues, and junctions that permit transfer of ions and other molecules from one cell to another. The types of
junctions that tie cells together and endow tissues with strength and stability include tight jun ctions, which are also known as the
zonula occludens (Figure 2–8). The desmosome and zonula adherens also help to hold cells together, and the hemidesmosome and
focal adhesions attach cells to their basal laminas. The gap junction forms a cytoplas mic “tunnel” for diffusion of small molecules (<
1000 Da) between two neighboring cells.
Tight junctions characteristically surround the apical margins of the cells in epithelia such as the intestinal mucosa, the walls of the renal
tubules, and the choroid plexus. They are also important to endothelial barrier function. They are made up of ridges—half from one cell
and half from the other—which adhere so strongly at cell junctions that they
Tight
junction
(zonula
occludens)
Zonula
adherens
Desmosomes
Gap
junctions
Hemidesmosome
FIGURE 2–8 Intercellular junctions in the mucosa of the small intestine. Tight jun ctions (zonula occludens), adherens junctions
(zonula adherens), desmosomes, gap junctions, and hemidesmosomes are all shown in relative positions in a polarized epithelial cell.
almost obliterate the space between the cells. There are three main families of transmemb rane proteins that contribute to tight junctions:
occludin, junctional adhesion molecules (JAMs), and claudins; and several more p roteins that interact from the cytosolic side. Tight
junctions permit the passage of some ions and solute in between adjacent cells (parac ellular pathway) and the degree of this
“leakiness” varies, depending in part on the protein makeup of the tight junction. Extrac ellular fluxes of ions and solute across epithelia
at these junctions are a significant part of overall ion and solute flux. In addition, tight ju nctions prevent the movement of proteins in the
plane of the membrane, helping to maintain the different distribution of transporters and channels in the apical and basolateral cell
membranes that make transport across epithelia possible.
In epithelial cells, each zonula adherens is usually a continuous structure on the basal sid e of the zonula occludens, and it is a major site
of attachment for intracellular microfilaments. It contains cadherins.
Desmosomes are patches characterized by apposed thickenings of the membranes of tw o adjacent cells. Attached to the thickened area in
each cell are intermediate filaments, some running parallel to the membrane and others r adiating away from it. Between the two
membrane thickenings the intercellular space contains filamentous material that includes c adherins and the extracellular portions of
several other transmembrane proteins.
Hemidesmosomes look like half-desmosomes that attach cells to the underlying basal lam ina and are connected
A
Presynaptic cytoplasm
3.5 nm 20 nm
Postsynaptic cytoplasm Normal
extracellular
space
Channel formed by pores in
each membrane
B 6 connexin subunits = 1 connexon (hemichannel) Each of the 6 connexins has 4 membrane-s panning regions
Presynaptic cytoplasm Cytoplasmic loops for regulation
Extracellular space
Extracellular loops for
homophilic interactions FIGURE 2–9 Gap junction connecting the cytoplasm of two cells. A) A gap junction plaque, or collection of
individual gap junctions, is shown to form multiple pores between cells that allow for the tran sfer of small molecules. Inset is electron
micrograph from rat liver (N. Gilula). B) Topographical depiction of individual connexon and corresponding 6 connexin proteins that
traverse the membrane. Note that each connexin traverses the membrane four times. (Reproduced with permission from Kandel ER, Schwartz JH,
Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
intracellularly to intermediate filaments. However, they contain integrins rather than cadh erins. Focal adhesions also attach cells to their
basal laminas. As noted previously, they are labile structures associated with actin filamen ts inside the cell, and they play an important
role in cell movement.
GAP JUNCTIONS
At gap junctions, the intercellular space narrows from 25 nm to 3 nm, and units called conne xons in the membrane of each cell are lined
up with one another (Figure 2–9). Each connexon is made up of six protein subunits c alled connexins. They surround a channel that,
when lined up with the channel in the corresponding connexon in the adjacent cell, per mits substances to pass between the cells without
entering the ECF. The diameter of the channel is normally about 2 nm, which permits th e passage of ions, sugars, amino acids, and other
solutes with molecular weights up to about 1000. Gap junctions thus permit the rapid pro pagation of electrical activity from cell to cell,
as well as the exchange of various chemical messengers. However, the gap junction ch annels are not simply passive, nonspecific
conduits. At least 20 different genes code for connexins in humans, and mutations in th ese genes can lead to diseases that are highly
selective in terms of the tissues involved and the type of communication between cells pr oduced. For instance, X-linked Charcot–
Marie–Tooth disease is a peripheral neuropathy associated with mutation of o ne particular connexin gene. Experiments in mice in
which particular connexins are deleted by gene manipulation or replaced with different connexins confirm that the particular connexin
subunits that make up connexons determine their permeability and selectivity. Recently it has been shown that connexons can be used as
channels to release small molecules from the cytosol into the ECF.
NUCLEUS & RELATED STRUCTURES
A nucleus is present in all eukaryotic cells that divide. If a cell is cut in half, the anucleate portion eventually dies without dividing. The
nucleus is made up in large part of the chromosomes, the structures in the nucleus that carry a co mplete blueprint for all the heritable
species and individual characteristics of the animal. Except in germ cells, the chromosom es occur in pairs, one originally from each
parent. Each chromosome is made up of a giant molecule of DNA. The DNA stra nd is about 2 m long, but it can fit in the nucleus
because at intervals it is wrapped around a core of histone proteins to form a nucleosome. There are about 25 million nucleosomes in
each nucleus. Thus, the structure of the chromosomes has been likened to a string of be ads. The beads are the nucleosomes, and the
linker DNA between them is the string. The whole complex of DNA and proteins is cal led chromatin. During cell division, the coiling
around histones is loosened, probably by acetylation of the histones, and pairs of chrom osomes become visible, but between cell
divisions only clumps of chromatin can be discerned in the nucleus. The ultimate units o f heredity are the genes on the chromosomes).
As discussed in Chapter 1, each gene is a portion of the DNA molecule.
The nucleus of most cells contains a nucleolus (Figure 2–1), a patchwork of gran ules rich in RNA. In some cells, the nucleus contains
several of these structures. Nucleoli are most prominent and numerous in growing cells . They are the site of synthesis of ribosomes, the
structures in the cytoplasm in which proteins are synthesized.
The interior of the nucleus has a skeleton of fine filaments that are attached to the nuclear membrane , or envelope (Figure 2–1), which
surrounds the nucleus. This membrane is a double membrane, and spaces between the two folds are called perinuclear cisterns. The
membrane is permeable only to small molecules. However, it contains nuclear pore complexes. Each complex has eightfold symmetry
and is made up of about 100 proteins organized to form a tunnel through which transp ort of proteins and mRNA occurs. There are many
transport pathways, and proteins called importins and exportins have been isola ted and characterized. Much current research is focused
on transport into and out of the nucleus, and a more detailed understanding of these pr ocesses should emerge in the near future.
ENDOPLASMIC RETICULUM
The endoplasmic reticulum is a complex series of tubules in the cytoplasm of the cell (Figure 2–1). The inner limb of its membrane is
continuous with a segment of the nuclear membrane, so in effect this part of the nuclear membrane is a cistern of the endoplasmic
reticulum. The tubule walls are made up of membrane. In rough, or granular, endoplasmic r eticulum, ribosomes are attached to the
cytoplasmic side of the membrane, whereas in smooth, or agranular, endoplasmic reticulum , ribosomes are absent. Free ribosomes are
also found in the cytoplasm. The granular endoplasmic reticulum is concerned with pro tein synthesis and the initial folding of
polypeptide chains with the formation of disulfide bonds. The agranular endoplasmic re ticulum is the site of steroid synthesis in steroid-
secreting cells and the site of detoxification processes in other cells. A modified endopla smic reticulum, the sarcoplasmic reticulum,
plays an important role in skeletal and cardiac muscle. In particular, the endoplasmic or sarcoplasmic reticulum can sequester Ca
2+
ions
and allow for their release as signaling molecules in the cytosol.
RIBOSOMES
The ribosomes in eukaryotes measure approximately 22 × 32 nm. Each is made up of a large and a small subunit called, on the basis of
their rates of sedimentation in the ultracentrifuge, the 60S and 40S subunits. The ribosom es are complex structures, containing many
different proteins and at least three ribosomal RNAs. They are the sites of protein synth esis. The ribosomes that become attached to the
endoplasmic reticulum synthesize all transmembrane proteins, most secreted proteins, an d most proteins that are stored in the Golgi
apparatus, lysosomes, and endosomes. These proteins typically have a hydrophobic signal peptide at one end (Figure 2–10). The
polypeptide chains that form these proteins are extruded into the endoplasmic reticulum. The free ribosomes synthesize cytoplasmic
proteins such as hemoglobin and the proteins found in peroxisomes and mitochondria.
GOLGI APPARATUS
& VESICULAR TRAFFIC
The Golgi apparatus is a collection of membrane-enclosed sacs (cisterns) that are stacke d like dinner plates (Figure 2–1). There are
usually about six sacs in each apparatus, but there may be more. One or more Golgi ap parati are present in all eukaryotic cells, usually
near the nucleus. Much of the organization of the Golgi is directed at proper glycosylat ion of proteins and lipids. There are more than
200 enzymes that function to add, remove, or modify sugars from proteins and lipids in the Golgi apparatus.
mRNA from Gene A
Cytoplasm mRNA from Gene B
Free ribosome
Signal sequence
Rough
endoplasmic reticulum
Carbohydrate group
Growing
polypeptide chain
Cleaved signal sequences
Vesicle
Secretory vesicle
Golgi apparatus
Lysosome
Exocytosis
Secreted protein
from Gene A Extracellular fluid
Digestive protein
from
Gene B
Plasma membrane
FIGURE 2–10 Rough endoplasmic reticulum and protein translation. Messenger RNA and ribosomes meet up in the cytosol for
translation. Proteins that have appropriate signal peptides begin translation, then associate with the endoplasmic reticulum (ER) to
complete translation. The association of ribosomes is what gives the ER its “rough” appea rance. (Reproduced with permission from Widmaier EP,
Raff H, Strang KT: Vander’s Human Physiology: The Mechanisms of Body Function, 11th ed. McGraw -Hill, 2008.)
ER Golgi apparatus Secretory granules Regulated secretion
Constitutive secretion
Recycling
Endocytosis
Nucleus Lysosome Late endosome Early endosome FIGURE 2–11 Cellular st ructures involved in protein processing. See text for
details.
The Golgi apparatus is a polarized structure, with cis and trans sides (Figure 2–11). Me mbranous vesicles containing newly synthesized
proteins bud off from the granular endoplasmic reticulum and fuse with the cistern on t he cis side of the apparatus. The proteins are then
passed via other vesicles to the middle cisterns and finally to the cistern on the trans side, from which vesicles branch off into the
cytoplasm. From the trans Golgi, vesicles shuttle to the lysosomes and to the cell exterior via constitutive and nonconstitutive pathways,
both involving exocytosis. Conversely, vesicles are pinched off from the cell membrane b y endocytosis and pass to endosomes. From
there, they are recycled.
Vesicular traffic in the Golgi, and between other membranous compartments in the cell, is regulated by a combination of common
mechanisms along with special mechanisms that determine where inside the cell they will go. One prominent feature is the involvement
of a series of regulatory proteins controlled by GTP or GDP binding (small G proteins) associated with vesicle assembly and delivery. A
second prominent feature is the presence of proteins called SNAREs (for soluble N-eth ylmaleimide-sensitive factor attachment receptor).
The v- (for vesicle) SNAREs on vesicle membranes interact in a lock-and-key fashion with t- (for target) SNAREs. Individual vesicles
also contain structural protein or lipids in their membrane that help to target them for spe cific membrane compartments (eg, Golgi sacs,
cell membranes).
QUALITY CONTROL
The processes involved in protein synthesis, folding, and migration to the various parts of the cell are so complex that it is remarkable
that more errors and abnormalities do not occur. The fact that these processes work as well as they do is because of mechanisms at each
level that are responsible for “quality control.” Damaged DNA is detected and repaired or bypassed. The various RNAs are also checked
during the translation process. Finally, when the protein chains are in the endoplasmic re ticulum and Golgi apparatus, defective
structures are detected and the abnormal proteins are degraded in lysosomes and protea somes. The net result is a remarkable accuracy in
the production of the proteins needed for normal body function.
APOPTOSIS
In addition to dividing and growing under genetic control, cells can die and be absorbe d under genetic control. This process is called
programmed cell death, or apoptosis (Gr. apo “away” + ptosis “fall”). It can be called “cell suicide” in the sense that the cell’s own
genes play an active role in its demise. It should be distinguished from necrosis (“cell mu rder”), in which healthy cells are destroyed by
external processes such as inflammation.
Apoptosis is a very common process during development and in adulthood. In the cent ral nervous system, large numbers of neurons are
produced and then die during the remodeling that occurs during development and syna pse formation. In the immune system, apoptosis
gets rid of inappropriate clones of immunocytes and is responsible for the lytic effects o f glucocorticoids on lymphocytes. Apoptosis is
also an important factor in processes such as removal of the webs between the fingers i n fetal life and regression of duct systems in the
course of sexual development in the fetus. In adults, it participates in the cyclic breakdow n of the endometrium that leads to
menstruation. In epithelia, cells that lose their connections to the basal lamina and neighbo ring cells undergo apoptosis. This is
responsible for the death of the enterocytes sloughed off the tips of intestinal villi. Abno rmal apoptosis probably occurs in autoimmune
diseases, neurodegenerative diseases, and cancer. It is interesting that apoptosis occurs i n invertebrates, including nematodes and insects.
However, its molecular mechanism is much more complex than that in vertebrates.
One final common pathway bringing about apoptosis is activation of caspases, a group of cysteine proteases. Many of these have been
characterized to date in mammals; 11 have been found in humans. They exist in cells as inactive proenzymes until activated by the
cellular machinery. The net result is DNA fragmentation, cytoplasmic and chromatin con densation, and eventually membrane bleb
formation, with cell breakup and removal of the debris by phagocytes (see Clinical Box 2–2).
TRANSPORT ACROSS
CELL MEMBRANES
There are several mechanisms of transport across cellular membranes. Primary pathway s include exocytosis, endocytosis, movement
through ion channels, and primary and secondary active transport. Each of these are d iscussed below.
EXOCYTOSIS
Vesicles containing material for export are targeted to the cell membrane (Figure 2–11) , where they bond in a similar manner to that
discussed in vesicular traffic between Golgi stacks, via the v-SNARE/t-SNARE arrange ment. The area of fusion then breaks down,
leaving the contents of the vesicle outside and the cell membrane intact. This is the Ca
2+
-dependent process of exocytosis (Figure 2–12).
Note that secretion from the cell occurs via two pathways (Figure 2–11). In the nonconstitutive pathway, proteins from the Golgi
apparatus initially enter secretory granules, where processing of prohormones to the m ature hormones occurs before exocytosis. The
other pathway, the constitutive pathway, involves the prompt transpor t of proteins to the cell membrane in vesicles, with little or no
processing or storage. The nonconstitutive pathway is sometimes called the regulated pathw ay, but this term is misleading because the
output of proteins by the constitutive pathway is also regulated.
ENDOCYTOSIS
Endocytosis is the reverse of exocytosis. There are various types of endocytosis named for the size of particles being ingested as well as
the regulatory requirements for the particular process. These include phagocytosis, pinocytos is, clathrinmediated endocytosis,
caveolae-dependent uptake, and nonclathrin/noncaveolae endocytosis.
Phagocytosis (“cell eating”) is the process by which bacteria, dead tissue, or other bits of microscopic material are engulfed by cells such
as the polymorphonuclear leukocytes of the blood. The material makes contact with the cell membrane, which then invaginates. The
invagination is pinched off, leaving the engulfed material in the membrane-enclosed vac uole and the cell membrane intact. Pinocytosis
(“cell drinking”) is a similar process with the vesicles much smaller in size and the substa nces ingested are in solution. The small size
membrane that is ingested should not be misconstrued; cells undergoing active pinocytos is (eg, macrophages) can ingest the equivalent
of their entire cell membrane in just 1 hour.
CLINICAL BOX 2–2 Molecular Medicine
Fundamental research on molecular aspects of genetics, regulation of gene expression, and protein synthesis has been paying off in
clinical medicine at a rapidly accelerating rate.
One early dividend was an understanding of the mechanisms by which antibiotics exert their effects. Almost all act by inhibiting protein
synthesis at one or another of the steps described previously. Antiviral drugs act in a sim ilar way; for example, acyclovir and ganciclovir
act by inhibiting DNA polymerase. Some of these drugs have this effect primarily in ba cteria, but others inhibit protein synthesis in the
cells of other animals, including mammals. This fact makes antibiotics of great value for research as well as for treatment of infections.
Single genetic abnormalities that cause over 600 human diseases have now been identifi ed. Many of the diseases are rare, but others are
more common and some cause conditions that are severe and eventually fatal. Example s include the defectively regulated Cl
channel in
cystic fibrosis and the unstable trinucleotide repeats in various parts of the genome th at cause Huntington’s disease, the fragile X
syndrome, and several other neurologic diseases. Abnormalities in mitochondrial DNA can also cause human diseases such as Leber’s
hereditary optic neuropathy and some forms of cardiomyopathy. Not surprisingly, gen etic aspects of cancer are probably receiving the
greatest current attention. Some cancers are caused by oncogenes, genes that are carried in the gen omes of cancer cells and are
responsible for producing their malignant properties. These genes are derived by soma tic mutation from closely related proto-
oncogenes, which are normal genes that control growth. Over 100 oncogenes have been de scribed. Another group of genes produce
proteins that suppress tumors, and more than 10 of these tumor suppressor gen es have been described. The most studied of these is the
p53 gene on human chromosome 17. The p53 protein produced by this gene triggers apoptosis. It is also a nuclear transcription factor
that appears to increase production of a 21-kDa protein that blocks two cell cycle enzym es, slowing the cycle and permitting repair of
mutations and other defects in DNA. The p53 gene is mutated in up to 50% of human cancers, with the production of p53 proteins that
fail to slow the cell cycle and permit other mutations in DNA to persist. The accumulated mutations eventually cause cancer.
Clathrin-mediated endocytosis occurs at membrane indentations where the protein cla thrin accumulates. Clathrin molecules have the
shape of triskelions, with three “legs” radiating from a central hub (Figure 2–13). As e ndocytosis progresses,
Exocytosis
Cytoplasm
Endocytosis
FIGURE 2–12 Exocytosis and endocytosis. Note that in exocytosis the cytoplasm ic sides of two membranes fuse, whereas in
endocytosis two noncytoplasmic sides fuse. (Reproduced with permission from A lberts B et al: Molecular Biology of the Cell, 4th ed. Garland Science, 2002.)
the clathrin molecules form a geometric array that surrounds the endocytotic vesicle. At the neck of the vesicle, the GTP binding protein
dynamin is involved, either directly or indirectly, in pinching off the vesicle. Once the complete v esicle is formed, the clathrin falls off
and the three-legged proteins recycle to form another vesicle. The vesicle fuses with an d dumps its contents into an early endosome
(Figure 2–11). From the early endosome, a new vesicle can bud off and return to the cell membrane. Alternatively, the early endosome
can become a late endosome and fuse with a lysosome (Figure 2–11) in w hich the contents are digested by the lysosomal proteases.
Clathrin-mediated endocytosis is responsible for the internal
FIGURE 2–13 Clathrin molecule on the surface of an endocytotic vesicle. Note the characteristic triskelion shape and the fact that
with other clathrin molecules it forms a net supporting the vesicle.
ization of many receptors and the ligands bound to them— including, for example, ner ve growth factor and low-density lipoproteins. It
also plays a major role in synaptic function.
It is apparent that exocytosis adds to the total amount of membrane surrounding the cell, and if membrane were not removed elsewhere at
an equivalent rate, the cell would enlarge. However, removal of cell membrane occurs by endocytosis, and such exocytosis–endocytosis
coupling maintains the surface area of the cell at its normal size.
RAFTS & CAVEOLAE
Some areas of the cell membrane are especially rich in cholesterol and sphingolipids and have been called rafts. These rafts are probably
the precursors of flask-shaped membrane depressions called caveolae (little caves) when their walls become infiltrated with a protein
called caveolin that resembles clathrin. There is considerable debate about the functions of rafts and caveolae, with evidence that they
are involved in cholesterol regulation and transcytosis. It is clear, however, that choleste rol can interact directly with caveolin, effectively
limiting the protein’s ability to move around in the membrane. Internalization via caveola e involves binding of cargo to caveolin and
regulation by dynamin. Caveolae are prominent in endothelial cells, where they help in the uptake of nutrients from the blood.
COATS & VESICLE TRANSPORT
It now appears that all vesicles involved in transport have protein coats. In humans, 53 coat complex subunits have been identified.
Vesicles that transport proteins from the trans Golgi to lysosomes have assembly pro tein 1 (AP-1) clathrin coats, and endocytotic
vesicles that transport to endosomes have AP-2 clathrin coats. Vesicles that transport bet ween the endoplasmic reticulum and the Golgi
have coat proteins I and II (COPI and COPII). Certain amino acid sequences or attach ed groups on the transported proteins target the
proteins for particular locations. For example, the amino acid sequence Asn–Pro–any a mino acid–Tyr targets transport from the cell
surface to the endosomes, and mannose-6-phosphate groups target transfer from the G olgi to mannose-6-phosphate receptors (MPR) on
the lysosomes.
Various small G proteins of the Rab family are especially important in vesicular traffic. T hey appear to guide and facilitate orderly
attachments of these vesicles. To illustrate the complexity of directing vesicular traffic, hu mans have 60 Rab proteins and 35 SNARE
proteins.
MEMBRANE PERMEABILITY &
MEMBRANE TRANSPORT PROTEINS
An important technique that has permitted major advances in our knowledge about tran sport proteins is patch clamping. A micropipette
is placed on the membrane of a cell and forms a tight seal to the membrane. The patch o f membrane under the pipette tip usually contains
only a few transport proteins, allowing for their detailed biophysical study (Figure 2–14 ). The cell can be left intact (cell-attached patch
clamp). Alternatively, the patch can be pulled loose from the cell, forming an inside-out patch. A third alternative is to suck out the
patch with the micropipette still attached to the rest of the cell membrane, providing direc t access to the interior of the cell (whole cell
recording).
Small, nonpolar molecules (including O
2
and N
2
) and small uncharged polar molecules such as CO
2
diffuse across the lipid membranes
of cells. However, the membranes have very limited permeability to other substances. In stead, they cross the membranes by endocytosis
and exocytosis and by passage through highly specific transport proteins, tr ansmembrane proteins that form channels for ions or
transport substances such as glucose, urea, and amino acids. The limited permeability ap plies even to water, with simple diffusion being
supplemented throughout the body with various water channels (aquaporins). For reference, the sizes of ions and other biologically
important substances are summarized in Table 2–2.
Some transport proteins are simple aqueous ion channels, though many of these have speci al features that make them selective for a
given substance such as Ca
2+
or, in the case of aquaporins, for water. These membrane-spanning proteins (or colle ctions of proteins)
have tightly regulated pores that can be gated opened or closed in response to local changes
Inside-out patch
Electrode
Pipette
Cell
membrane
Closed
ms Open
FIGURE 2–14 Patch clamp to investigate transport. In a patch clamp exper iment, a small pipette is carefully maneuvered to seal off a
portion of a cell membrane. The pipette has an electrode bathed in an appropriat e solution that allows for recording of electrical changes
through any pore in the membrane (shown below). The illustrated setup is termed an “inside-out patch” because of the orientation of the
membrane with reference to the electrode. Other configurations include cell attache d, whole cell, and outside-out patches. (Modified from
Ackerman MJ, Clapham DE: Ion channels: Basic science and clinical disease. N Engl J Med 1997;336:1575.)
(Figure 2–15). Some are gated by alterations in membrane potential (voltage-gated), whereas others are opened or closed in response to
a ligand (ligand-gated). The ligand is
TABLE 2–2 Size of hydrated ions and other substances of biologic interest.
Substance Atomic or Molecular Weight Radius (nm)
Cl
35 0.12
K
+ 39 0.12
H
2
O 18 0.12
Ca
2+
40 0.15
Na
+
23 0.18
Urea 60 0.23
Li
+
7 0.24
Glucose 180 0.38
Sucrose 342 0.48
Inulin 5000 0.75
Albumin 69,000 7.50 Data from Moore EW: Physiology of Intestinal Water and Electrolyte Abso rption. American Gastroenterological Association, 1976.
Closed Open A Ligand-gated
Bind ligand
B Phosphorylation-gated
Phosphorylate
Dephosphorylate Pi P C Voltage-gated
Change
membrane
potential
+++ + – – – –
– – – – +++ +
D Stretch or pressure-gated
the bound molecule from one side of the cell membrane to the other. Molecules move f rom areas of high concentration to areas of low
concentration (down their chemical gradient), and cations move to negatively charged areas whereas anions move to positively charged
areas (down their electrical gradient). When carrier proteins move substances in th e direction of their chemical or electrical gradients,
no energy input is required and the process is called facilitated diffusion. A typical example is glu cose transport by the glucose
transporter, which moves glucose down its concentration gradient from the ECF to the cytoplasm of the cell. Other carriers transport
substances against their electrical and chemical gradients. This form of transport require s energy and is called active transport. In
animal cells, the energy is provided almost exclusively by hydrolysis of ATP. Not surpr isingly, therefore, many carrier molecules are
ATPases, enzymes that catalyze the hydrolysis of ATP. One of these ATPases is sodium– potassium adenosine triphosphatase (Na, K
ATPase), which is also known as the Na, K pump. Th ere are also H, K ATPases in the gastric mucosa and the renal tubules.
Ca
2+
ATPase pumps Ca
2+
out of cells. Proton ATPases acidify many intracellular organelles, including parts of the Golgi complex and
lysosomes.
Some of the transport proteins are called uniports because they transport only one substa nce. Others are called symports because
transport requires the binding of more than one substance to the transport protein and t he substances are transported across the
membrane together. An example is the symport in the intestinal mucosa that is responsibl e for the cotransport by facilitated diffusion of
Na
+
and glucose from the intestinal lumen into mucosal cells. Other transporters are called anti ports because they exchange one
substance for another.
Stretch
Cytoskeleton
FIGURE 2–15 Regulation of gating in ion channels. Several types of gating are shown for ion channels. A) Ligand-gated channels
open in response to ligand binding. B) Protein phosphorylation or dephosphoryla tion regulate opening and closing of some ion channels.
C) Changes in membrane potential alter channel openings. D) Mechanical stretch of the membrane results in channel opening.
(Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural S cience, 4th ed. McGraw-Hill, 2000.)
often external (eg, a neurotransmitter or a hormone). However, it can also be internal; intracellular Ca
2+
, cAMP, lipids, or one of the G
proteins produced in cells can bind directly to channels and activate them. Some channe ls are also opened by mechanical stretch, and
these mechanosensitive channels play an important role in cell movement.
Other transport proteins are carriers that bind ions and other molecules and then change their c onfiguration, moving
ION CHANNELS
There are ion channels specific for K
+
, Na
+
, Ca
2+
, and Cl
, as well as channels that are nonselective for cations or anions. Each type of
channel exists in multiple forms with diverse properties. Most are made up of identical o r very similar subunits. Figure 2–16 shows the
multiunit structure of various channels in diagrammatic cross-section.
Most K
+
channels are tetramers, with each of the four subunits forming part of the pore throug h which K
+
ions pass. Structural analysis
of a bacterial voltage-gated K
+
channel indicates that each of the four subunits have a paddle-like extension containing four charges.
When the channel is closed, these extensions are near the negatively charged interior of the cell. When the membrane potential is
reduced, the paddles containing the charges bend through the membrane to its exterior surface, causing the channel to open. The
bacterial K
+
channel is very similar to the voltage-gated K
+
channels in a wide variety of species, including mammals. In the
acetylcholine ion channel and other ligand-gated cation or anion channels, five subunits make up the pore. Members of the ClC family of
Cl
channels are dimers, but they have two pores, one in each subunit. Finally, aquaporins are tetramers
AB C D
FIGURE 2–16 Different ways in which ion channels form pores. Many K
+
channels are tetramers (A), with each protein subunit
forming part of the channel. In ligand-gated cation and anion channels (B) such as the ace tylcholine receptor, five identical or very
similar subunits form the channel. Cl
channels from the ClC family are dimers (C), with an intracellular pore in each subunit.
Aquaporin water channels (D) are tetramers with an intracellular channel in each subun it. (Reproduced with permission from Jentsch TJ: Chloride
channels are different. Nature 2002;415:276.)
with a water pore in each of the subunits. Recently, a number of ion channels with intri nsic enzyme activity have been cloned. More than
30 different voltage-gated or cyclic nucleotide-gated Na
+
and Ca
2+
channels of this type have been described. Representative Na
+
, Ca
2+
,
and K
+
channels are shown in extended diagrammatic form in Figure 2–17.
Another family of Na
+
channels with a different structure has been found in the apical membranes of epithelial cells in the kidneys, colon,
lungs, and brain. The epithelial sodium channels (ENaCs) are made up of three subunits encoded by three different genes. Each of the
subunits probably spans the membrane twice, and the amino terminal and carboxyl term inal are located inside the cell. The α subunit
transports Na
+
, whereas the β and γ subunits do not. However, the addition of the β and γ subunits in creases Na
+
transport through the α
subunit. ENaCs are inhibited by the diuretic amiloride, which binds to the α subunit, and they used to be called amilorideinhibitable
Na
+
channels. The ENaCs in the kidney play an important role in the regulation of ECF volume by aldosterone. ENaC knockout mice
are born alive but promptly die because they cannot move Na
+
, and hence water, out of their lungs.
Humans have several types of Cl
channels. The ClC dimeric channels are found in plants, bacteria, and animals, and ther e are nine
different ClC genes in humans. Other Cl
channels have the same pentameric form as the acetylcholine receptor; examples includ e the γ-
aminobutyric acid A (GABA
A
) and glycine receptors in the central nervous system (CNS). The cystic fibrosis transme mbrane
conductance regulator (CFTR) that is mutated in cystic fibrosis is also a Cl
channel. Ion channel mutations cause a variety of
channelopathies—diseases that mostly affect muscle and brain tissue and produce episodic paralyses or convulsions.
Na, K ATPase
As noted previously, Na, K ATPase catalyzes the hydrolysis of ATP to adenosine dipho sphate (ADP) and uses the energy to extrude
three Na
+
from the cell and take two K
+
into the cell for each molecule of ATP hydrolyzed. It is an electro genic pump in that it moves
three positive charges out of the cell for each two that it moves in, and it is therefore said to have a coupling ratio of 3:2. It is found in
all parts of the body. Its activity is inhibited by ouabain and related digitalis glycosides us ed in the treatment of heart failure. It is a
heterodimer made up of an α subunit with a molecular weight of approximately 100,00 0 and a β subunit with a molecular weight of
approximately 55,000. Both extend through the cell membrane (Figure 2–18). Separat ion of the subunits eliminates activity. The β
subunit is a glycoprotein, whereas Na
+
and K
+
transport occur through the α subunit. The β subunit has a single membrane-spanning
domain and three extracellular glycosylation sites, all of which appear to have attached c arbohydrate residues. These residues account for
one third of its molecular weight. The α subunit probably spans the cell membrane 10 tim es, with the amino and carboxyl terminals both
located intracellularly. This subunit has intracellular Na
+
- and ATP-binding sites and a phosphorylation site; it also has extracellular
binding sites for K
+
and ouabain. The endogenous ligand of the ouabain-binding site is unsettled. When N a
+
binds to the α subunit, ATP
also binds and is converted to ADP, with a phosphate being transferred to Asp 376, the phosphorylation site. This causes a change in the
configuration of the protein, extruding Na
+
into the ECF. K
+
then binds extracellularly, dephosphorylating the α subunit, which returns to
its previous conformation, releasing K
+
into the cytoplasm.
The α and β subunits are heterogeneous, with α
1
, α
2
, and α
3
subunits and β
1
, β
2
, and β
3
subunits described so far. The α
1
isoform is found in the membranes of most cells, whereas α
2
is present in muscle, heart, adipose tissue, and brain, and α
3
is present in
heart and brain. The β
1
subunit is widely distributed but is absent in certain astrocytes, vestibular cells of the inne r ear, and glycolytic
fast-twitch muscles. The fast-twitch muscles contain only β
2
subunits. The different α and β subunit structures of Na, K ATPase in
various tissues probably represent specialization for specific tissue functions.
REGULATION OF Na, K ATPase ACTIVITY
The amount of Na
+
normally found in cells is not enough to saturate the pump, so if the Na
+
increases, more is pumped out. Pump
The amount of Na
+
normally found in cells is not enough to saturate the pump, so if the Na
+
increases, more is pumped out. Pump
activity is affected by second messenger molecules (eg, cAMP and diacylglycerol [DAG ]). The magnitude and direction of the altered
pump effects vary with the experimental conditions. Thyroid hormones increase pump activity by a genomic action to increase the
formation of Na, K ATPase molecules. Aldosterone also increases the number of pump s, although this effect is probably secondary.
Dopamine in the kidney inhibits the pump by phosphorylating it, causing a natriuresis. I nsulin increases pump activity, probably by a
variety of different mechanisms.
Na
+
channel I II III IV Extracellular side
123 5
P
6 123 5
P
6 123 5
P
6 123 5
P
6
Cytoplasmic side
NH
2 COOH
Ca
2+
channel
123 5
P
6 123 5
P
6 123 5
P
6 123 5
P
6
COOH NH
2
K
+
channel
123 5
P
6
COOH NH
2
FIGURE 2–17 Diagrammatic representation of the pore-forming subunits of three ion channels. The
α
subunit of the Na
+
and Ca
2+
channels traverse the membrane 24 times in four repeats of six membrane-spanning un its. Each repeat has a “P” loop between membrane
spans 5 and 6 that does not traverse the membrane. These P loops are though t to form the pore. Note that span 4 of each repeat is colored
in red, representing its net “+” charge. The K
+
channel has only a single repeat of the six spanning regions and P loop. Four K
+
subunits
are assembled for a functional K
+
channel. (Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Princi ples of Neural Science,
4th ed. McGraw-Hill, 2000.)
SECONDARY ACTIVE TRANSPORT
In many situations, the active transport of Na
+
is coupled to the transport of other substances (secondary active transport). For
example, the luminal membranes of mucosal cells in the small intestine contain a symport that transports glucose into the cell only if Na
+
binds to the protein and is transported into the cell at the same time. From the cells, the glu cose enters the blood. The electrochemical
gradient for Na
+
is maintained by the active transport of Na
+
out of the mucosal cell into ECF. Other examples are shown in Figure 2–
19. In the heart, Na,K ATPase indirectly affects Ca
2+
transport. An antiport in the membranes of cardiac muscle cells normally
exchanges intracellular Ca
2+
for extracellular Na
+
.
Active transport of Na
+
and K
+
is one of the major energyusing processes in the body. On the average, it accounts for
2K
+
Ouabain β
ECF
3
2
Active transport 2K
+
Ouabain
3Na
+
3Na
+
Na
+ ATP
ADP + Pi
Cl− Na
+ Ca
2+
Na
+
Cytoplasm
1 α
4
5
3Na
+
FIGURE 2–18 Na
+
–K
+
ATPase. The intracellular portion of the
α
subunit has a Na
+
-binding site (1), a phosphorylation site (4), and an
ATP-binding site (5). The extracellular portion has a K
+
-binding site (2) and an ouabain-binding site (3). (From Horisberger J-D et al: Structure–
function relationship of Na–K-ATPase. Annu Rev Physiol 1991;53:565. Reproduced with permission fro m the Annual Review of Physiology, vol. 53. Copyright © 1991
by Annual Reviews)
about 24% of the energy utilized by cells, and in neurons it accounts for 70%. Thus, it accounts for a large part of the basal metabolism.
A major payoff for this energy use is the establishment of the electrochemical gradient i n cells.
Na
+
15 meq/L
K+, 2Cl−
K
+
150 − Na
+
H+ Cl− 7 − SugarsK+
or amino
K
+
acids Cl−
H
+
−− − − Vm = −70 mV + + + +
Na
+
Na
+
140 meq/L K
+
4 −
Cl− 105 −
FIGURE 2–19 Composite diagram of main secondary effects of active transport of Na
+
and K
+
. Na,K ATPase converts the
chemical energy of ATP hydrolysis into maintenance of an inward gradient for Na
+
and an outward gradient for K
+
. The energy of the
gradients is used for countertransport, cotransport, and maintenance of the membrane poten tial. Some examples of cotransport and
countertransport that use these gradients are shown. (Reproduced with permission from Sk ou JC: The Na–K pump. News Physiol Sci 1992;7:95.)
TRANSPORT ACROSS EPITHELIA
In the gastrointestinal tract, the pulmonary airways, the renal tubules, and other structure s, substances enter one side of a cell and exit
another, producing movement of the substance from one side of the epithelium to the o ther. For transepithelial transport to occur, the
cells need to be bound by tight junctions and, obviously, have different ion channels an d transport proteins in different parts of their
membranes. Most of the instances of secondary active transport cited in the preceding p aragraph involve transepithelial movement of
ions and other molecules.
THE CAPILLARY WALL
FILTRATION
The capillary wall separating plasma from interstitial fluid is different from the cell memb ranes separating interstitial fluid from
intracellular fluid because the pressure difference across it makes filtration a significant factor in pr oducing movement of water and
solute. By definition, filtration is the process by which fluid is forced through a membra ne or other barrier because of a difference in
pressure on the two sides.
ONCOTIC PRESSURE
The structure of the capillary wall varies from one vascular bed to another. However, in skeletal muscle and many other organs, water
and relatively small solutes are the only substances that cross the wall with ease. The aper tures in the junctions between the endothelial
cells are too small to permit plasma proteins and other colloids to pass through in signific ant quantities. The colloids have a high
molecular weight but are present in large amounts. Small amounts cross the capillary wa ll by vesicular transport, but their effect is slight.
Therefore, the capillary wall behaves like a membrane impermeable to colloids, and thes e exert an osmotic pressure of about 25 mm Hg.
The colloid osmotic pressure due to the plasma colloids is called the oncotic pressure. F iltration across the capillary membrane as a
result of the hydrostatic pressure head in the vascular system is opposed by the oncotic pressure. The way the balance between the
hydrostatic and oncotic pressures controls exchanges across the capillary wall is conside red in detail in Chapter 32.
TRANSCYTOSIS
Vesicles are present in the cytoplasm of endothelial cells, and tagged protein molecules in jected into the bloodstream have been found in
the vesicles and in the interstitium. This indicates that small amounts of protein are transpo rted out of capillaries across endothelial cells
by endocytosis on the capillary side followed by exocytosis on the interstitial side of the cells. The transport mechanism makes use of
coated vesicles that appear to be coated with caveolin and is called transcytosis, vesicular transport, or cytopempsis.
GAP JUNCTIONS SYNAPTIC PARACRINE AND AUTOCRINE ENDOCRINE
A
P
Message transmission Directly from cell
to cell
Across synaptic cleft
By diffusion in interstitial fluid By circulating body fluids
Local or general Local Local Locally diffuse General
Specificity depends on Anatomic location Anatomic location and receptors
Receptors
FIGURE 2–20 Intercellular communication by chemical mediators. A, autocrine; P, para crine.
INTERCELLULAR COMMUNICATION
Cells communicate with one another via chemical messengers. Within a given tissue, som e messengers move from cell to cell via gap
junctions without entering the ECF. In addition, cells are affected by chemical messenge rs secreted into the ECF, or by direct cell–cell
contacts. Chemical messengers typically bind to protein receptors on the surface of the c ell or, in some instances, in the cytoplasm or the
nucleus, triggering sequences of intracellular changes that produce their physiologic eff ects. Three general types of intercellular
communication are mediated by messengers in the ECF: (1) neural communicat ion, in which neurotransmitters are released at synaptic
junctions from nerve cells and act across a narrow synaptic cleft on a postsynaptic cell; (2) endocrine communication, in which
hormones and growth factors reach cells via the circulating blood or the lymph; and (3 ) paracrine communication, in which the
products of cells diffuse in the ECF to affect neighboring cells that may be some distanc e away (Figure 2–20). In addition, cells secrete
chemical messengers that in some situations bind to receptors on the same cell, that is, the cell that secreted the messenger (autocrine
communication). The chemical messengers include amines, amino acids, steroids, polyp eptides, and in some instances, lipids, purine
nucleotides, and pyrimidine nucleotides. It is worth noting that in various parts of the bo dy, the same chemical messenger can function as
a neurotransmitter, a paracrine mediator, a hormone secreted by neurons into the blood (neural hormone), and a hormone secreted by
gland cells into the blood.
An additional form of intercellular communication is called juxtacrine communication. Some cells express multiple repeats of growth
factors such as transforming growth factor alpha (TGFα) extracellularly on transmembrane pro teins that provide an anchor to the cell.
Other cells have TGFα receptors. Consequently, TGFα anchored to a cell can bind to a TGFα receptor on another cell, linking the two.
This could be important in producing local foci of growth in tissues.
RECEPTORS FOR CHEMICAL MESSENGERS
The recognition of chemical messengers by cells typically begins by interaction with a re ceptor at that cell. There have been over 20
families of receptors for chemical messengers characterized. These proteins are not stati c components of the cell, but their numbers
increase and decrease in response to various stimuli, and their properties change with ch anges in physiological conditions. When a
hormone or neurotransmitter is present in excess, the number of active receptors gener ally decreases (down-regulation), whereas in the
presence of a deficiency of the chemical messenger, there is an increase in the number of active receptors (up-regulation). In its actions
on the adrenal cortex, angiotensin II is an exception; it increases rather than decreases th e number of its receptors in the adrenal. In the
case of receptors in the membrane, receptor-mediated endocytosis is responsible for do wn-regulation in some instances; ligands bind to
their receptors, and the ligand– receptor complexes move laterally in the membrane to c oated pits, where they are taken into the cell by
endocytosis (internalization). This decreases the number of receptors in the membrane. Some receptors are recycled after
internalization, whereas others are replaced by de novo synthesis in the cell. Another ty pe of down-regulation is desensitization, in
which receptors are chemically modified in ways that make them less responsive.
Receptors
MECHANISMS BY WHICH
CHEMICAL MESSENGERS ACT
Receptor–ligand interaction is usually just the beginning of the cell response. This event is transduced into secondary responses within
the cell that can be divided into four broad categories: (1) ion channel activation, (2) G-protein activation, (3) activation of enzyme
activity within the cell, or (4) direct activation of transcription. Within each of these grou ps, responses can be quite varied. Some of the
common mechanisms by which
TABLE 2–3 Common mechanisms by which chemical messengers in the ECF bring about changes in cell function.
Mechanism Examples
Open or close ion channels in cell membrane
Acetylcholine on nicotinic cholinergic receptor; norepinephrine on K
+
channel in the heart
Act via cytoplasmic or nuclear receptors to increase transcription of selected mRNAs
Thyroid hormones, retinoic acid, steroid hormones
Activate phospholipase C with intracellular production of DAG, IP
3
, and other inositol phosphates Angiotensin II, norepinephrine via
α
1
-adrenergic receptor, vasopressin via V
1
receptor
Activate or inhibit adenylyl cyclase, causing increased or decreased intracellular
production of cAMP
Norepinephrine via
β1
-adrenergic receptor (increased cAMP); norepinephrine via
α2
-adrenergic receptor (decreased cAMP)
Increase cGMP in cell Atrial natriuretic peptide; nitric oxide
Increase tyrosine kinase activity of cytoplasmic portions of transmembrane rec eptors
Insulin, epidermal growth factor (EGF), platelet-derived growth factor (PDGF), monocyte colo nystimulating factor (M-CSF)
pathways. Cellular phosphorylation is under the control of two groups of proteins: k inases, enzymes that catalyze the phosphorylation of
tyrosine or serine and threonine residues in proteins (or in some cases, in lipids); and phosph atases, proteins that remove phosphates
from proteins (or lipids). Some of the larger receptor families are themselves kinases. T yrosine kinase receptors initiate phosphorylation
on tyrosine residues on complementary receptors following ligand binding. Serine/threo nine kinase receptors initiate phosphorylation on
serines or threonines in complementary receptors following ligand binding. Cytokine re ceptors are directly associated with a group of
protein kinases that are activated following cytokine binding. Alternatively, second mess engers changes can lead to phosphorylation
further downstream in the signaling pathway. More than 300 protein kinases have been described. Some of the principal ones that are
important in mammalian cell signaling are summarized in Table 2–4. In general, addition of phosphate groups changes the conformation
of the proteins, altering their functions and consequently the functions of the cell. The c lose relationship between phosphorylation and
dephosphorylation of cellular proteins allows for a temporal control of activation of cel l signaling pathways. This is sometimes referred
to as a “phosphate timer.”
Increase serine or threonine kinase activity
TGFβ, activin, inhibin
STIMULATION OF TRANSCRIPTION
The activation of transcription, and subsequent translation, is a common outcome of cell ular signaling. There are three chemical
messengers exert their intracellular effects are summarized in Table 2–3. Ligands such a s acetylcholine bind directly to ion channels in
the cell membrane, changing their
conductance. Thyroid and steroid hormones, 1,25-dihydroxycholecalciferol, and retin oids enter cells and act on one or another member
of a family of structurally related cytoplasmic or
nuclear receptors. The activated receptor binds to DNA and increases transcription of s elected mRNAs. Many other ligands in
the ECF bind to receptors on the surface of cells and trigger the
release of intracellular mediators such as cAMP, IP
3
, and DAG
that initiate changes in cell function. Consequently, the extracellular ligands are called “first messengers” and the intracellular mediators
are called “second messengers.” Second
messengers bring about many short-term changes in cell function by altering enzyme fu nction, triggering exocytosis, and so
on, but they also can lead to the alteration of transcription of
various genes. A variety of enzymatic changes, protein–protein
interactions or second messenger changes can be activated
within a cell in an orderly fashion following receptor recognition of the primary messen ger. The resulting cell signaling
pathway provides amplification of the primary signal and distribution of the sign al to appropriate targets within the cell. Extensive cell
signaling pathways also provide opportunities for
feedback and regulation that can fine tune the signal for the correct physiological respo nse by the cell.
The most predominant posttranslation modification of proteins, phosphorylation, is a com mon theme in cell signaling
TABLE 2–4 Sample protein kinases.
Phosphorylate serine or threonine residues, or both
Calmodulin-dependent
Myosin light-chain kinase
Phosphorylase kinase
Ca
2+
/calmodulin kinase I
Ca
2+
/calmodulin kinase II
Ca
2+
/calmodulin kinase III
Calcium-phospholipid-dependent
Protein kinase C (seven subspecies)
Cyclic nucleotide-dependent
cAMP-dependent kinase (protein kinase A; two subspecies)
cGMP-dependent kinase
Phosphorylate tyrosine residues
Insulin receptor, EGF receptor, PDGF receptor, and
M-CSF receptor
distinct pathways for primary messengers to alter transcription of cells. First, as is the cas e with steroid or thyroid hormones, the primary
messenger is able to cross the cell membrane and bind to a nuclear receptor, which then can directly interact with DNA to alter gene
expression. A second pathway to gene transcription is the activation of cytoplasmic pro tein kinases that can move to the nucleus to
phosphorylate a latent transcription factor for activation. This pathway is a common end point of signals that go through the mitogen
activated protein (MAP) kinase cascade. MAP kinases can be activated foll owing a variety of receptor ligand interactions through
second messenger signaling. They comprise a series of three kinases that coordinate a s tepwise phosphorylation to activate each protein
in series in the cytosol. Phosphorylation of the last MAP kinase in series allows it to migra te to the nucleus where it phosphorylates a
latent transcription factor. A third common pathway is the activation of a latent transcript ion factor in the cytosol, which then migrates to
the nucleus and alters transcription. This pathway is shared by a diverse set of transcrip tion factors that include nuclear factor kappa B
(NFκB; activated following tumor necrosis family receptor binding and others), and signal transducers of activated trans cription
(STATs; activated following cytokine receptor binding). In all cases the binding of the activated tran scription factor to DNA increases
(or in some cases, decreases) the transcription of mRNAs encoded by the gene to whic h it binds. The mRNAs are translated in the
ribosomes, with the production of increased quantities of proteins that alter cell function .
INTRACELLULAR Ca
2+
AS A SECOND MESSENGER
Ca
2+
regulates a very large number of physiological processes that are as diverse as prolifer ation, neural signaling, learning, contraction,
secretion, and fertilization, so regulation of intracellular Ca
2+
is of great importance. The free Ca
2+
concentration in the cytoplasm at rest
is maintained at about 100 nmol/ L. The Ca
2+
concentration in the interstitial fluid is about 12,000 times the cytoplasmic concentration
(ie, 1,200,000 nmol/L), so there is a marked inwardly directed concentration gradient a s well as an inwardly directed electrical gradient.
Much of the intracellular Ca
2+
is stored at relatively high concentrations in the endoplasmic reticulum and other organe lles (Figure 2–
21), and these organelles provide a store from which Ca
2+
can be mobilized via ligand-gated channels to increase the concentration of
free Ca
2+
in the cytoplasm. Increased cytoplasmic Ca
2+
binds to and activates calcium-binding proteins. These proteins can have direct
effects in cellular physiology, or can activate other proteins, commonly protein kinases, to further cell signaling pathways.
Ca
2+
can enter the cell from the extracellular fluid, down its electrochemical gradient, throug h many different Ca
2+
channels. Some of
these are ligand-gated and others are voltagegated. Stretch-activated channels exist in so me cells as well.
Ca 2+ CaBP Effects
(volt)
2H+
ATP
Ca2+ Ca2+(lig) Ca2+
Ca2+
3Na
+
(SOCC)
2+Ca
Mitochondrion Endoplasmic reticulum
2+
handling in mammalian cells. Ca
2+
is FIGURE 2–21 Ca
stored in the endoplasmic reticulum and, to a lesser extent, mitochondria and can be released from them to replenish cytoplasmic Ca
2+
.
Calciumbinding proteins (CaBP) bind cytoplasmic Ca
2+
and, when activated in this fashion, bring about a variety of physiologic effects.
Ca
2+
enters the cells via voltage-gated (volt) and ligand-gated (lig) Ca
2+
channels and store-operated calcium channels ( SOCCs). It is
transported out of the cell by Ca, Mg ATPases (not shown), Ca, H ATPase and an Na, Ca an tiport. It is also transported into the ER by
Ca ATPases.
Many second messengers act by increasing the cytoplasmic Ca
2+
concentration. The increase is produced by releasing Ca
2+
from
intracellular stores—primarily the endoplasmic reticulum—or by increasing the entry of Ca
2+
into cells, or by both mechanisms. IP
3
is
the major second messenger that causes Ca
2+
release from the endoplasmic reticulum through the direct activation of a ligand-gated
channel, the IP
3
receptor. In effect, the generation of one second messenger (IP
3
) can lead to the release of another second messenger
(Ca
2+
). In many tissues, transient release of Ca
2+
from internal stores into the cytoplasm triggers opening of a population of Ca
2+
channels in the cell membrane (store-operated Ca
2+
channels; SOCCs). The resulting Ca
2+
influx replenishes the total intracellular
Ca
2+
supply and refills the endoplasmic reticulum. The exact identity of the SOCCs is still unk nown, and there is debate about the signal
from the endoplasmic reticulum that opens them.
As with other second messenger molecules, the increase in Ca
2+
within the cytosol is rapid, and is followed by a rapid decrease. Because
the movement of Ca
2+
outside of the cytosol (ie, across the plasma membrane or the membrane of the internal store) requires that it
move up its electrochemical gradient, it requires energy. Ca
2+
movement out of the cell is facilitated by the plasma membrane Ca
2+
ATPase. Alternatively, it can be transported by an antiport that exchanges three Na
+
for each Ca
2+
driven by the energy stored in the Na
+
electrochemical gradient. Ca
2+
movement into the internal stores is through the action of the sarcoplasmic or endoplasmic reticulum
Ca
2+
ATPase, also known as the SERCA pump.
CALCIUM-BINDING PROTEINS
Many different Ca
2+
-binding proteins have been described, including troponin, calmodulin, and calbindin. Troponin is the
70 90
L F
R V
F
F A
K D E
P
F
T
M M A R K
R E D
GD M I N Ca E LCa I K E G R
V D
A
T D E
N G
T
S E
Y
I S
A A H
E
D G
D
V
N 60 80
100
M I 50 A E N E G L N
T
(Me)3
110M D Q L E KT
P
L
40
G Q N T D
120
R
S
L E
E V D E
M
M E
G K
30 G K
V G D G IT T T I T
COOH
V D 130 R
L
E
Ca N F E A
N
Ca I
E
G K
10 A
T Y N A E
I
M
E E
140
20 E K
D
F
L
S
F A M Q
V
F
Q
E
E
T
L
Q
D
A
NH Ac
FIGURE 2–22 Structure of calmodulin from bovine brain. Single-letter abbreviations are used fo r the amino acid residues. Note the
four calcium domains (purple residues) flanked on either side by stretches of α helix. (R eproduced with permission from Cheung WY: Calmodulin:
An overview. Fed Proc 1982;41:2253.)
Ca
2+
-binding protein involved in contraction of skeletal muscle (Chapter 5). Calmodulin con tains 148 amino acid residues (Figure 2–22)
and has four Ca
2+
-binding domains. It is unique in that amino acid residue 115 is trimethylated, and it is ex tensively conserved, being
found in plants as well as animals. When calmodulin binds Ca
2+
, it is capable of activating five different calmodulin-dependent kinases
(CaMKs; Table 2–4), among other proteins. One of the kinases is myosin light-chain kinase, whi ch phosphorylates myosin. This brings
about contraction in smooth muscle. CaMKI and CaMKII are concerned with synaptic function, and CaMKIII is concerned with protein
synthesis. Another calmodulin-activated protein is calcineurin, a phosphatase that inactivates Ca
2+
channels by dephosphorylating them.
It also plays a prominent role in activating T cells and is inhibited by some immunosuppr essants.
MECHANISMS OF DIVERSITY
OF Ca
2+
ACTIONS
It may seem difficult to understand how intracellular Ca
2+
can have so many varied effects as a second messenger. Part of the
explanation is that Ca
2+
may have different effects at low and at high concentrations. The ion may be at high c oncentration at the site of
its release from an organelle or a channel (Ca
2+
sparks) and at a subsequent lower concentration after it diffuses throughout the cell.
Some of the changes it produces can outlast the rise in intracellular Ca
2+
concentration because of the way it binds to some of the Ca
2+
-
binding proteins. In addition, once released, intracellular Ca
2+
concentrations frequently oscillate at regular intervals, and there is
evidence that the frequency and, to a lesser extent, the amplitude of those oscillations co des information for effector mechanisms.
Finally, increases in intracellular Ca
2+
concentration can spread from cell to cell in waves, producing coordinated events such as the
rhythmic beating of cilia in airway epithelial cells.
G PROTEINS
A common way to translate a signal to a biologic effect inside cells is by way of nucleoti de regulatory proteins that are activated after
binding GTP (G proteins). When an activating signal reaches a G protein, the protein exc hanges GDP for GTP. The GTP–protein
complex brings about the activating effect of the G protein. The inherent GTPase activit y of the protein then converts GTP to GDP,
restoring the G protein to an inactive resting state. G proteins can be divided into two pr incipal groups involved in cell signaling: small
G proteins and heterotrimeric G proteins. Other groups that have similar regulation and are also important to cel l physiology include
elongation factors, dynamin, and translocation GTPases.
There are six different families of small G proteins (or small GTPases) that are all highly regulate d. GTPase activating proteins
(GAPs) tend to inactivate small G proteins by encouraging hydrolysis of GTP to GDP in the central binding site. Guanine exchange
factors (GEFs) tend to activate small G proteins by encouraging exchange of GDP for GTP in the active site. Some of the small G
proteins contain lipid modifications that help to anchor them to membranes, while others are free to diffuse throughout the cytosol. Small
G proteins are involved in many cellular functions. Members of the Rab family regulate the rate of vesicle traffic between the
endoplasmic reticulum, the Golgi apparatus, lysosomes, endosomes, and the cell membra ne. Another family of small GTPbinding
proteins, the Rho/Rac family, mediates interactions between the cytoskeleton and cell me mbrane; and a third family, the Ras family,
regulates growth by transmitting signals from the cell membrane to the nucleus.
Another family of G proteins, the larger heterotrimeric G proteins, couple cell surface receptors to catalytic u nits that catalyze the
intracellular formation of second messengers or couple the receptors directly to ion cha nnels. Despite the knowledge of the small G
proteins described above, the heteromeric G proteins are frequently referred to in the s hortened “G protein” form because they were the
first to be identified. Heterotrimeric G proteins are made up of three subunits designated α, β, and γ (Figure 2–23). Both the α and the γ
subunits have lipid modifications that anchor these proteins to plasma membrane. The α subunit is bound to GDP. When a ligand binds to
a G protein-coupled receptor (GPCR), this GDP is exchanged for GTP and the α subu nit separates from the combined β and γ subunits.
The separated α subunit brings about many biologic effects. The β and γ subunits are tig htly bound in the cell and together form a
signaling molecule that can also activate a variety of effectors. The intrinsic
Nucleotide exchange
TABLE 2–5 Some of the ligands for receptors coupled to heterotrimeric G proteins.
Input
GDP
GTP Output Class Ligand GTPase activity Neurotransmitters Epinephrine
Norepinephrine
Dopamine
5-Hydroxytryptamine
α
β γ
α
β γ
Histamine
Acetylcholine Effectors Adenosine
FIGURE 2–23 Heterotrimeric G proteins. Top Summary of overall reaction that occurs in the Gα subunit. Bottom: When the ligand
(square) binds to the G protein-coupled receptor in the cell membrane, GTP replaces GDP on the α subunit. GTPα separates from the βγ
subunit and GTPα and βγ both activate various effectors, producing physiologic effects . The intrinsic GTPase activity of GTPα then
converts GTP to GDP, and the α, β, and γ subunits reassociate. Opioids
Tachykinins Substance P
Neurokinin A
Neuropeptide K
Other peptides Angiotensin II
GTPase activity of the α subunit then converts GTP to GDP, and this leads to reassociation of the α with the βγ subunit and termination
of effector activation. The GTPase activity of the α subunit can be accelerated by a fam ily of regulators of G protein signaling (RGS).
Heterotrimeric G proteins relay signals from over 1000 GPCRs, and their effectors in t he cells include ion channels and enzymes (Table
2–5). There are 20 α, 6 β, and 12 γ genes, which allow for over 1400 α, β, and γ com binations. Not all combinations occur in the cell, but
over 20 different heterotrimeric G proteins have been well documented in cell signaling . They can be divided into five families, each
with a relatively characteristic set of effectors.
Arginine vasopressin
Oxytocin
VIP, GRP, TRH, PTH
Glycoprotein hormones TSH, FSH, LH, hCG
Arachidonic acid derivatives Thromboxane A
2
Other Odorants
Tastants
Endothelins
Platelet-activating factor
Cannabinoids
G PROTEIN-COUPLED RECEPTORS
All the heterotrimeric G protein-coupled receptors (GPCRs) that have been characterized to d ate are proteins that span the cell
membrane seven times. Because of this structure they are alternatively referred to as seven-helix receptors or serpentine receptors. A
very large number have been cloned, and their functions are multiple and diverse. The topological structures of two of them are shown in
Figure 2–24. These receptors further assemble into a barrel-like structure. Upon ligand binding, a conformational change activates a
resting heterotrimeric G protein associated with the cytoplasmic leaf of the plasma memb rane. Activation of a single receptor can result
in 1, 10, or more active heterotrimeric G proteins, providing amplification as well as tran sduction of the first messenger. Bound receptors
can be inactivated to limit the amount of cellular signaling. This frequently occurs throug h phosphorylation of the cytoplasmic side of the
receptor.
Light
INOSITOL TRISPHOSPHATE
& DIACYLGLYCEROL AS
SECOND MESSENGERS
The link between membrane binding of a ligand that acts via Ca
2+
and the prompt increase in the cytoplasmic Ca
2+
concentration is
often inositol trisphosphate (inositol 1,4,5-trisphosphate; IP
3
). When one of these ligands binds to its receptor, activation of the
receptor produces activation of phospholipase C (PLC) on the inner surface of the me mbrane. Ligands bound to G protein-coupled
receptor can do this through the G
q
heterotrimeric G proteins, while ligands bound to tyrosine kinase receptors can do this through other
cell signaling pathways. PLC has at least eight isoforms; PLC
β
H D
P V H S G N T T L L F D S D N G P P G M NH
2
D
V
2-Adrenergic receptor
T
E
C Y H K E TC
D
surface
IR
Extracellular
D
E
K
D
A
Q
A
C
E
MW N F G N
F H
F
W T F
D N L
I
VVG M A
K W
A
T
M C R
N Q
L I N E F W Y W H Q A Y I V H
P K
E V Y
I L M S B A G F T S I D M Q I P A I A S V I N V I L L N V I V P V V V L C L F S S I V I F F W L G L A I V A L G M V T A S T L Q S S F Y V P L W C Y V N S F G N V L D I E T V I W P L V L T F A F N
V L V I A C A L L C V I V M L I V M V F T G M I P L I Y
T A I S T I A V D V M R V YSL C R S P D F R
I A F Q F N I K V KK Y
R Y A R
T
A F
L
E
N F A K H E K L C
F
L
E R L Q T V T
A K
I T
Q V A K R Q
L Q K K L
Cytoplasmic
T L I S C
S P F K Y Q S L D
S
H F R G E S K S L
surface
P
R
R
R R
N L G Q V E Q D G R S G H G
L
S S
K S
S
A E G M Y D T K G N G N S S Y G N G Y A G
C
Q L G Q E K E S E R L C E D P P G T E S F V N C
Q HOOC L P S D N T S C N R G Q S D L S L S P V T
G
P S
R V V C T K N S F P V Y F N P G E T G N M NH
2
F
E
A Intradiskal S C G I D Y
Rhodopsin
P
surface
C
YQ
Y
F
G P
M
Q
T
Y
V
T
G
G
P
L
F C E
H
A
Y
N
P
E
E G L I E
G S
DH E Y T QP
WQ F
L S
G
R
N H F
F S N T
S M T Y L F A T W G V E S F F I Y
G P
I F M
L A A Y T T T F L G G E L P P A V I Y M F A V Q T I P A M F L Q G F I A L A C A F V V A Y P F F A L I M L V M F L W S L A L A M V H F I I L W C I K T S A G F P D A V V L A W T F P L I L F A V
Y N I N F L A L N L
I
E R Y A V G M V I F F I V M I
P V I
Y T L Y L I Y V V V I A H C Y G I V M I M M
V
N C
N Q
R
T
L
K
E
L
T N K Q
F R N C M
V
P
V
PM S N F R FG
V
E K
E A
K Q V
Q H K K
L R T
F T V
T T
K E
A A A Q QQ E S
A
L
Cytoplasmic surface
T T
C C G
HOOC A P A V Q S T E T K S V T T S A E D D G L P N K
FIGURE 2–24 Structures of two G protein-coupled receptors. The individual amino acid residue s are identified by their single-letter
codes, and the orange residues are sites of phosphorylation. The Y-shaped symbols identify gly cosylation sites. Note the extracellular
amino terminal, the intracellular carboxyl terminal, and the seven membrane-spann ing portions of each protein. (Reproduced with permission
from Benovic JL et al: Lightdependent phosphorylation of rhodopsin by
β
-adrenergic receptor kinase. Reprinted by permission from Nature 1986;321:869. Copyright ©
1986 by Macmillan Magazines)
is activated by heterotrimeric G proteins, while PLC γ forms are activated through tyros ine kinase receptors. PLC isoforms can catalyze
the hydrolysis of the membrane lipid phosphatidylinositol 4,5-diphosphate (PIP
2
) to form IP
3
and diacylglycerol (DAG) (Figure 2–25).
The IP
3
diffuses to the endoplasmic reticulum, where it triggers the release of Ca
2+
into the cytoplasm by binding the IP
3
receptor, a
ligand-gated Ca
2+
channel (Figure 2–26). DAG is also a second messenger; it stays in the cell membrane, where it activates one of
several isoforms of protein kinase C.
Phosphatidylinositol PIP (PI)
PIP
2
Diacylglycerol
Phospholipase C
P P 1 1
4 4
P
Inositol IP +
CDP-diacylglycerol
P
1 P
IP
3
4
5
1
P
P
54 PP
IP
2
Phosphatidic acid
FIGURE 2–25 Metabolism of phosphatidylinositol in cell membranes. Phosphatidy linositol is successively phosphorylated to form
phosphatidylinositol 4-phosphate (PIP), then phosphatidylinositol 4,5-bisphosphate (PIP
2
). Phospholipase C
β
and phospholipase Cγ
catalyze the breakdown of PIP
2
to inositol 1,4,5-trisphosphate (IP
3
) and diacylglycerol. Other inositol phosphates and
phosphatidylinositol derivatives can also be formed. IP
3
is dephosphorylated to inositol, and diacylglycerol is metabolized to cytosine
diphosphate (CDP)-diacylglycerol. CDP-diacylglycerol and inositol then combine to form phosphatidylinositol, completing the cycle.
(Modified from Berridge MJ: Inositol triphosphate and diacylglycerol as second messengers. Biochem J 198 4;220:345.)
Stimulatory receptor ISF
PIP2
PLC
β γ
α
Tyrosine Gq, etc
kinase
Cytoplasm DAG
O O O HO P OO CH
2
Adenine
O O O HO P OO CH
2
Adenine
OH OH OH
O
PKC ATP
H
HH
H
IP
3
Phosphoproteins OH OH
Adenylyl cyclase
CaBP Ca
2+
Physiologic effects ER
Physiologic effects PP
FIGURE 2–26 Diagrammatic representation of release of inositol tripho sphate (IP
3
) and diacylglycerol (DAG) as second
messengers. Binding of ligand to G protein-coupled receptor activates phospholipase C (PL C)
β
. Alternatively, activation of receptors
with intracellular tyrosine kinase domains can activate PLCγ. The resulting hydrolysis of phosphatidylinositol 4,5-diphosphate (PIP
2
)
produces IP
3
, which releases Ca
2+
from the endoplasmic reticulum (ER), and DAG, which activates protein kinase C (PKC). CaBP,
Ca
2+
-binding proteins. ISF, interstitial fluid.
O CH
2
Adenine
O
cAMP
H
HH
H
H2O
O P O OH OH
Phosphodiesterase
CYCLIC AMP
Another important second messenger is cyclic adenosine 3',5'monophosphate (cyclic AMP or cAMP; Figure 2–27). Cyclic AM P is
formed from ATP by the action of the enzyme adenylyl cyclase and conv erted to physiologically inactive 5'AMP by the action of the
enzyme phosphodiesterase. Some of the phosphodiesterase isoforms that break down cAMP are inhibited by methylxanthines such as
caffeine and theophylline. Consequently, these compounds can augment hormonal and transmitter effects mediated via cAMP. Cyclic
AMP activates one of the cyclic nucleotide-dependent protein kinases (protein k inase A, PKA) that, like protein kinase C, catalyzes the
phosphorylation of proteins, changing their conformation and altering their activity. In addition, the active catalytic subunit of PKA
moves to the nucleus and phosphorylates the cAMP-responsive element-binding prote in (CREB). This transcription factor then binds
to DNA and alters transcription of a number of genes.
PRODUCTION OF cAMP
BY ADENLYL CYCLASE
Adenylyl cyclase is a transmembrane protein, and it crosses the membrane 12 times. Ten isoforms of this enzyme have been described
and each can have distinct regulatory properties, permitting the cAMP pathway to be cu stomized to specific tissue needs. Notably,
stimulatory heterotrimeric G proteins (G
s
) activate, while inhibitory heterotrimeric G proteins (G
i
) inactivate adenylyl cyclase (Figure 2–
28). When the appropriate ligand binds to a stimulatory receptor, a G
s
α subunit activates one of the adenylyl cyclases. Conversely, when
the
AMP
O
HO P O CH
2
Adenine
O
OH
H
HH
H
OH OH
FIGURE 2–27 Formation and metabolism of cAMP. The second me ssenger cAMP is a made from ATP by adenylyl cyclase and
broken down into cAMP by phosphodiesterase.
appropriate ligand binds to an inhibitory receptor, a G
i
α subunit inhibits adenylyl cyclase. The receptors are specific, responding at low
threshold to only one or a select group of related ligands. However, heterotrimeric G p roteins mediate the stimulatory and inhibitory
effects produced by many different ligands. In addition, cross-talk occurs between the phospholipase C system and the adenylyl cyclase
system, as several of the isoforms of adenylyl cyclase are stimulated by calmodulin. Fina lly, the effects of protein kinase A and protein
kinase C are very widespread and can also affect directly, or indirectly, the activity at ad enylyl cyclase. The close relationship between
activation of G proteins and adenylyl cyclases also allows for spatial regulation of cAM P production. All of these events, and others,
allow for fine-tuning the cAMP response for a particular physiological outcome in the c ell.
Two bacterial toxins have important effects on adenylyl cyclase that are mediated by G proteins. The A subunit of cholera toxin
catalyzes the transfer of ADP ribose to an arginine residue in the middle of the α subun it of G
s
. This inhibits
Stimulatory
receptor
ISF
Adenylyl cyclase Inhibitory receptor ANP
NH
2
NH
2
NH
2
ST
α
β γ
G
S
α
Cytoplasm
ATP
CAMP
β γ
G
i
PDE
ISF
M
C
NH
2
PTK
NH2
5' AMP
Protein kinase A
Phosphoproteins
cyc cyc cyc COOH
COOH COOH
EGF
PDGF
PTK PTP
NH
2
PTK
PTPPTP
COOH COOH
COOHCOOH
Physiologic effects Guanylyl cyclases Tyrosine kinases Tyrosine
phosphatases FIGURE 2–28 The cAMP system. Activation of adenylyl cy clase catalyzes the conversion of ATP to cAMP. Cyclic AMP
activates protein kinase A, which phosphorylates proteins, producing physiologic effects. St imulatory ligands bind to stimulatory
receptors and activate adenylyl cyclase via G
s
. Inhibitory ligands inhibit adenylyl cyclase via inhibitory receptors and G
i
. ISF, interstitial
fluid.
its GTPase activity, producing prolonged stimulation of adenylyl cyclase. Pertussis toxin catalyzes ADP-ribosylation of a cysteine
residue near the carboxyl terminal of the α subunit of G
i
. This inhibits the function of G
i
. In addition to the implications of these
alterations in disease, both toxins are used for fundamental research on G protein funct ion. The drug forskolin also stimulates adenylyl
cyclase activity by a direct action on the enzyme.
GUANYLYL CYCLASE
Another cyclic nucleotide of physiologic importance is cyclic guanosine monoph osphate (cyclic GMP or cGMP). Cyclic GMP is
important in vision in both rod and cone cells. In addition, there are cGMP-regulated io n channels, and cGMP activates cGMP-dependent
kinase, producing a number of physiologic effects.
Guanylyl cyclases are a family of enzymes that catalyze the formation of cGMP. They e xist in two forms (Figure 2–29). One form has an
extracellular amino terminal domain that is a receptor, a single transmembrane domain, a nd a cytoplasmic portion with guanylyl cyclase
catalytic activity. Three such guanylyl cyclases have been characterized. Two are recep tors for atrial natriuretic peptide (ANP; also
known as atrial natriuretic factor), and a third binds an Escherichia coli enterotoxin and the gastrointestinal polypeptide guanylin. The
other form of guanylyl cyclase is soluble, contains heme, and is not bound to the memb rane. There appear to be several isoforms of the
intracellular enzyme. They are activated by nitric oxide (NO) and NO-containing comp ounds.
FIGURE 2–29 Diagrammatic representation of guanylyl cyclases, tyrosine kin ases, and tyrosine phosphatases. ANP, atrial
natriuretic peptide; C, cytoplasm; cyc, guanylyl cyclase domain; EGF, epidermal gro wth factor; ISF, interstitial fluid; M, cell membrane;
PDGF, platelet-derived growth factor; PTK, tyrosine kinase domain; PTP, tyrosine ph osphatase domain; ST, E. coli enterotoxin. (Modified
from Koesling D, Böhme E, Schultz G: Guanylyl cyclases, a growing family of signal transducing e nzymes. FASEB J 1991;5:2785.)
GROWTH FACTORS
Growth factors have become increasingly important in many different aspects of physi ology. They are polypeptides and proteins that are
conveniently divided into three groups. One group is made up of agents that foster the multiplication or development of various types of
cells; nerve growth factor (NGF), insulin-like growth factor I (IGF-I), activins and inh ibins, and epidermal growth factor (EGF) are
examples. More than 20 have been described. The cytokines are a second group. The se factors are produced by macrophages and
lymphocytes, as well as other cells, and are important in regulation of the immune system (see Chapter 3). Again, more than 20 have
been described. The third group is made up of the colony-stimulating factors that regula te proliferation and maturation of red and white
blood cells.
Receptors for EGF, platelet-derived growth factor (PDGF), and many of the other fac tors that foster cell multiplication and growth have
a single membrane-spanning domain with an intracellular tyrosine kinase domain (Figur e 2–29). When ligand binds to a tyrosine kinase
receptor, it first causes a dimerization of two similar receptors. The dimerization results in partial activation of the intracellular tyrosine
kinase domains and a cross-phosphorylation to fully activate each other. One of the pa thways activated by phosphorylation leads,
through the small G protein Ras, to MAP kinases, and eventually to the production of tr anscription factors in the nucleus that alter gene
expression (Figure 2–30).
Receptors for cytokines and colony-stimulating factors differ from the other growth fa ctors in that most of them do not have tyrosine
kinase domains in their cytoplasmic portions and
Growth factor Ligand Receptor
Cell membrane
A
Receptor ISF
Inactive
Ras Ras
Active Ras Ras Cytoplasm
T
GDP GTP
K
Grb2
SOS Raf
STAT STAT MAP KK
MAP K
B Ligand
TF P P P P Nucleus STAT STAT
Altered gene activity
FIGURE 2–30 One of the direct pathways by which growth factors alter gene activity. TK, tyrosine kinase domain; Grb2, Ras
activator controller; Sos, Ras activator; Ras, product of the ras gene; MAP K, mitogen-activ ated protein kinase; MAP KK, MAP kinase
kinase; TF, transcription factors. There is cross-talk between this pathway and the cAMP pathway, as well as cross-talk with the IP
3
DAG pathway.
C
Ligand
P P P P
STAT
P P
STAT
some have little or no cytoplasmic tail. However, they initiate tyrosine kinase activity in th e cytoplasm. In particular, they activate the
so-called Janus tyrosine kinases (JAKs) in the cytoplasm (Figure 2–31). These i n turn phosphorylate STAT proteins. The
phosphorylated STATs form homo- and heterodimers and move to the nucleus, where they act as transcription factors. There are four
known mammalian JAKs and seven known STATs. Interestingly, the JAK–STAT path way can also be activated by growth hormone and
is another important direct path from the cell surface to the nucleus. However, it should be emphasized that both the Ras and the JAK–
STAT pathways are complex and there is cross-talk between them and other signaling p athways discussed previously.
Finally, note that the whole subject of second messengers and intracellular signaling has become immensely complex, with multiple
pathways and interactions. It is only possible in a book such as this to list highlights and p resent general themes that will aid the reader in
understanding the rest of physiology (see Clinical Box 2–3).
D
Ligand
P P P P
P
STAT Nucleus
DNA
FIGURE 2–31 Signal transduction via the JAK–STAT pathway. A) Ligan d binding leads to dimerization of receptor. B) Activation
and tyrosine phosphorylation of JAKs. C) JAKs phosphorylate STATs. D) STATs dimerize and move to nucleus, where they bind to
response elements on DNA. (Modified from Takeda K, Kishimoto T, Akira S: STAT6: Its role in interleukin 4-m ediated biological functions. J Mol Med
1997;75:317.)
HOMEOSTASIS
The actual environment of the cells of the body is the interstitial component of the ECF. Because normal cell function
CLINICAL BOX 2–3 Receptor & G Protein Diseases
Many diseases are being traced to mutations in the genes for receptors. For example, lo ss-of-function receptor mutafor receptors. For
example, loss-of-function receptor muta dihydroxycholecalciferol receptor and the ins ulin receptor. Certain other diseases are caused by
production of antibodies against receptors. Thus, antibodies against thyroidstimulating ho rmone (TSH) receptors cause Graves’ disease,
and antibodies against nicotinic acetylcholine receptors cause myasthenia gravis.
An example of loss of function of a receptor is the type of nephrogenic diabetes insipidus that is due to loss o f the ability of mutated V
2
vasopressin receptors to mediate concentration of the urine. Mutant receptors can gain as well as lose function. A gain-of-function
mutation of the Ca
2+
receptor causes excess inhibition of parathyroid hormone secretion and familial hypercalciuric hypocalcemia. G
proteins can also undergo loss-of-function or gain-of-function mutations that cause dis ease (Table 2–6), In one form of
pseudohypoparathyroidism, a mutated G
s
α fails to respond to parathyroid hormone, producing the symptoms of hypoparathyro idism
without any decline in circulating parathyroid hormone. Testotoxicosis is an interesting disease tha t combines gain and loss of function.
In this condition, an activating mutation of G
s
α causes excess testosterone secretion and prepubertal sexual maturation. However, this
mutation is temperature-sensitive and is active only at the relatively low temperature of th e testes (33 °C). At 37 °C, the normal
temperature of the rest of the body, it is replaced by loss of function, with the productio n of hypoparathyroidism and decreased
responsiveness to TSH. A different activating mutation in G
s
α is associated with the rough-bordered areas of skin pigmentation and
hypercortisolism of the McCune–Albright syndrome. This mutation occurs during fetal development, creating a mosaic of normal and
abnormal cells. A third mutation in G
s
α reduces its intrinsic GTPase activity. As a result, it is much more active than normal, an d excess
cAMP is produced. This causes hyperplasia and eventually neoplasia in somatotrope ce lls of the anterior pituitary. Forty percent of
somatotrope tumors causing acromegaly have cells containing a somatic mutation of this type.
depends on the constancy of this fluid, it is not surprising that in multicellular animals, an immense number of regulatory mechanisms
have evolved to maintain it. To describe “the various physiologic arrangements which s erve to restore the normal state, once it has been
disturbed,” W.B. Cannon coined the term homeostasis. The buffering p roperties of the body fluids and the renal and respiratory
adjustments to the presence of excess acid or alkali are examples of homeostatic
TABLE 2–6 Examples of abnormalities caused by loss- or gain-of-function mutations of he terotrimeric G protein-coupled
receptors and G proteins.
Site Type of
Mutation Disease
Receptor
Cone opsins Loss Color blindness
Rhodopsin Loss Congenital night blindness; two forms of retinitis pigmentosa
V
2
vasopressin Loss X-linked nephrogenic diabetes insipidus
ACTH Loss Familial glucocorticoid deficiency
LH Gain Familial male precocious puberty
TSH Gain Familial nonautoimmune hyperthyroidism
TSH Loss Familial hypothyroidism
Ca
2+
Gain Familial hypercalciuric hypocalcemia
Thromboxane A
2
Loss Congenital bleeding
Endothelin B Loss Hirschsprung disease
G protein
G
s
α Loss Pseudohypothyroidism type 1a
G
s
α Gain/loss Testotoxicosis
G
s
α Gain
(mosaic) McCune–Albright syndrome
G
s α Gain Somatotroph adenomas with acromegaly
G
i
α Gain Ovarian and adrenocortical tumors
Modified from Lem J: Diseases of G-protein-coupled signal transduction pathways: Th e mammalian visual system as a model. Semin
Neurosci 1998;9:232.
mechanisms. There are countless other examples, and a large part of physiology is con cerned with regulatory mechanisms that act to
maintain the constancy of the internal environment. Many of these regulatory mechanis ms operate on the principle of negative feedback;
deviations from a given normal set point are detected by a sensor, and signals from the sensor trigger compensatory changes that
continue until the set point is again reached.
CHAPTER SUMMARY
The cell and the intracellular organelles are surrounded by a semipermeable membr ane. Biological membranes have a lipid bilayer with
a hydrophobic core and hydrophilic outer regions that provide a barrier between inside and outside compartments as well as a template
for biochemical reactions. The membrane is populated by structural and functional p roteins that can be integrated into the membrane or
be associated with one side of the lipid bilayer. These proteins contribute greatly to the semipermeable properties of biological
membrane.
Mitochondria are organelles that allow for oxidative phosphorylation in eukaryotic cells. They contain their own DNA, however,
proteins in the mitochondria are encoded by both mitochondrial and cellular DNA. Mito chondria also are important in specialized
cellular signaling.
Lysosomes and peroxisomes are membrane-bound organelles that contribute to protein and lipid processing. They do this in part by
creating acidic (lysosomes) or oxidative (peroxisomes) contents relative to the cell cytosol.
The cytoskeleton is a network of three types of filaments that provide struc tural integrity to the cell as well as a means for trafficking of
organelles and other structures. Actin is the fundamental building block for thin filamen ts and represents as much as 15% of cellular
protein. Actin filaments are important in cellular contraction, migration, and signaling. A ctin filaments also provide the backbone for
muscle contraction. Intermediate filaments are primarily structural. Proteins that make up intermediate filaments are cell-type specific.
Microtubules are made up of tubulin subunits. Microtubules provide a dynamic structure in cells tha t allows for movement of cellular
components around the cell.
There are three superfamilies of molecular motor proteins in the cell that use th e energy of ATP to generate force, movement, or both.
Myosin is the force generator for muscle cell contraction. There are also cellular myosins th at interact with the cytoskeleton (primarily
thin filaments) to participate in contraction as well as movement of cell contents. Kinesins and cellular dyneins are motor proteins that
primarily interact with microtubules to move cargo around the cells.
Cellular adhesion molecules aid in tethering cells to each other or to the extrace llular matrix as well as providing for initiation of
cellular signaling. There are four main families of these proteins: integrins, immunoglobu lins, cadherins, and selectins.
Cells contain distinct protein complexes that serve as cellular connections to other cells or the extracellular matrix. Tight junctions
provide intercellular connections that link cells into a regulated tissue barrier. T ight junctions also provide a barrier to movement of
proteins in the cell membrane and thus, are important to cellular polarization. Gap junctio ns provide contacts between cells that allow for
direct passage of small molecules between two cells. Desmosomes and adherens junction s are specialized structures that hold cells
together. Hemidesmosomes and focal adhesions attach cells to their basal lamina.
The nucleus is an organelle that contains the cellular DNA and is the site of transcription. T here are several organelles that emanate
from the nucleus, including the endoplasmic reticulum and the Golgi apparatus. These two organe lles are important in protein processing
and the targeting of proteins to correct compartments within the cell.
Exocytosis and endocytosis are vesicular fusion events that allow for movement of proteins and lipids between the cell interior, the
plasma membrane, and the cell exterior. Exocytosis can be constitutive or non constitutive; both are regulated processes that require
specialized proteins for vesicular fusion. Endocytosis is the formation of vesicles at the p lasma membrane to take material from the
extracellular space into the cell interior. Some endocytoses are defined in part by the size of the vesicles formed whereas others are
defined by membrane structures that contribute to the endocytosis. All are tightly regula ted processes.
Membranes contain a variety of proteins and protein complexes that allow for transport of small molecules. Aqueous ion channels are
membrane-spanning proteins that can be gated open to allow for selective diffusion of ions acros s membranes and down their
electrochemical gradient. Carrier proteins bind to small molecules and undergo confo rmational changes to deliver small molecules across
the membrane. This facilitated transport can be passive or active. Active transport requires ener gy for transport and is typically provided
by ATP hydrolysis.
Cells can communicate with one another via chemical messengers. Individual mess engers (or ligands) typically bind to a plasma
membrane receptor to initiate intracellular changes that lead to physiologic changes. Plas ma membrane receptor families include ion
channels, G protein-coupled receptors, or a variety of enzyme-linked receptors (eg, tyrosine kin ase receptors). There are additional
cytosolic receptors (eg, steroid receptors) that can bind membrane-permeant compounds. Act ivation of receptors lead to cellular changes
that include changes in membrane potential, activation of heterotrimeric G proteins, incre ase in second messenger molecules, or initiation
of transcription.
Second messengers are molecules that undergo a rapid concentration changes in th e cell following primary messenger recognition.
Common second messenger molecules include Ca
2+
, cyclic adenosine monophosphate (cAMP), cyclic guanine monophosphate (c GMP),
inositol trisphosphate (IP
3
) and nitric oxide (NO).
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. The electrog enic Na, K ATPase plays a critical role in
cellular physiology by
A) using the energy in ATP to extrude 3 Na
+
out of the cell in
exchange for taking two K
+
into the cell.
B) using the energy in ATP to extrude 3 K
+
out of the cell in exchange for taking two Na
+
into the cell.
C) using the energy in moving Na
+
into the cell or K
+
outside
the cell to make ATP. +
outside of the cell or K
+
D) using the energy in moving Na
inside the cell to make ATP.
2. Cell membranes
A) contain relatively few protein molecules.
B) contain many carbohydrate molecules.
C) are freely permeable to electrolytes but not to proteins. D) have variable protein and lipid contents depending on their
location in the cell.
E) have a stable composition throughout the life of the cell. 3. Second messengers
A) are substances that interact with first messengers outside cells.
B) are substances that bind to first messengers in the cell membrane.
C) are hormones secreted by cells in response to stimulation by another hormone.
D) mediate the intracellular responses to many different hormones and neurotransmitter s.
E) are not formed in the brain.
4. The Golgi complex
A) is an organelle that participates in the breakdown of proteins and lipids.
B) is an organelle that participates in posttranslational processing of proteins.
C) is an organelle that participates in energy production.
D) is an organelle that participates in transcription and translation.
E) is a subcellular compartment that stores proteins for trafficking to the nucleus.
5. Endocytosis
A) includes phagocytosis and pinocytosis, but not clathrinmediated or caveolae-depend ent uptake of extracellular contents.
B) refers to the merging of an intracellular vesicle with the plasma membrane to deliver intracellular contents to the extracellular milieu.
C) refers to the invagination of the plasma membrane to uptake extracellular contents in to the cell.
D) refers to vesicular trafficking between Golgi stacks. 6. G protein-coupled receptors
A) are intracellular membrane proteins that help to regulate movement within the cell.
B) are plasma membrane proteins that couple the extracellular binding of primary s ignaling molecules to activation of small G proteins.
C) are plasma membrane proteins that couple the extracellular binding of primary s ignaling molecules to the activation of heterotrimeric
G proteins.
D) are intracellular proteins that couple the binding of primary messenger molecules with transcription.
7. Gap junctions are intercellular connections that
A) primarily serve to keep cells separated and allow for transport across a tissue barrier .
B) serve as a regulated cytoplasmic bridge for sharing of small molecules between cells.
C) serve as a barrier to prevent protein movement within the cellular membrane.
D) are cellular components for constitutive exocytosis that occurs between adjacent cells .
CHAPTER RESOURCES
Alberts B et al: Molecular Biology of the Cell, 5th ed. Garland Science, 2007.
Cannon WB: The Wisdom of the Body. Norton, 1932.
Junqueira LC, Carneiro J, Kelley RO: Basic Histology, 9th ed. McGraw-Hill, 1998.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural Science, 4th ed. Mc Graw-Hill, 2000.
Pollard TD, Earnshaw WC: Cell Biology, 2nd ed. Saunders, Elsevier, 2008.
Sperelakis N (editor): Cell Physiology Sourcebook, 3rd ed. Academic Press, 2001 .
This page intentionally left blank
Immunity, Infection, & Inflammation
CH APTER
3
OBJEC TIV ES
After studying this chapter, you should be able to:
Understand the significance of immunity, particularly with respect to defending
the body against microbial invaders.
Define the circulating and tissue cell types that contribute to immune and inflammatory r esponses.
Describe how phagocytes are able to kill internalized bacteria.
Identify the functions of hematopoietic growth factors, cytokines, and chemokines.
Delineate the roles and mechanisms of innate, acquired, humoral, and cellular immunity .
Understand the basis of inflammatory responses and wound healing.
INTRODUCTION
As an open system, the body is continuously called upon to defend itself from potentiall y harmful invaders such as bacteria, viruses, and
other microbes. This is accomplished by the immune system, which is subdivided into inn ate and adaptive (or acquired) branches. The
immune system is composed of specialized effector cells that sense and respond to forei gn antigens and other molecular patterns not
found in human tissues. Likewise, the immune system clears the body’s own cells that ha ve become senescent or abnormal, such as
cancer cells. Finally, occasionally, normal host tissues become the subject of inappropria te immune attack, such as in autoimmune
diseases or in settings where normal cells are harmed as innocent bystanders when the i mmune system mounts an inflammatory response
to an invader. It is beyond the scope of this volume to provide a full treatment of all asp ects of modern immunology. Nevertheless, the
student of physiology should have a working knowledge of immune functions and the ir regulation, due to a growing appreciation for the
ways in which the immune system can contribute to normal physiological regulation in a variety of tissues, as well as contributions of
immune effectors to pathophysiology.
IMMUNE EFFECTOR CELLS
Many immune effector cells circulate in the blood as the white blood cells. In addition, th e blood is the conduit for the precursor cells
that eventually develop into the immune cells of the tissues. The circulating immunologic cells include granulocytes
(polymorphonuclear leukocytes, PMNs), comprising neutrophils, eosinophils, and basop hils; lymphocytes; and
monocytes. Immune responses in the tissues are further amplified by these cells follow ing their extravascular migration, as well as tissue
macrophages (derived from monocytes) and mast cells (related to basophils). Act ing together, these cells provide the body with
powerful defenses against tumors and viral, bacterial, and parasitic infections.
63
GRANULOCYTES
All granulocytes have cytoplasmic granules that contain biologically active substances inv olved in inflammatory and allergic reactions.
The average half-life of a neutrophil in the circulation is 6 hours. To maintain the norm al circulating blood level, it is therefore necessary
to produce over 100 billion neutrophils per day. Many neutrophils enter the tissues, par ticularly if triggered to do so by an infection or by
inflammatory cytokines. They are attracted to the endothelial surface by cell adhesion m olecules known as selectins, and they roll along
it. They then bind firmly to neutrophil adhesion molecules of the integrin family. They n ext insinuate themselves through the walls of
the capillaries between endothelial cells by a process called diapedesis. Many of those that le ave the circulation enter the gastrointestinal
tract and are eventually lost from the body.
Invasion of the body by bacteria triggers the inflammatory response. The bo ne marrow is stimulated to produce and release large
numbers of neutrophils. Bacterial products interact with plasma factors and cells to prod uce agents that attract neutrophils to the infected
area (chemotaxis). The chemotactic agents, which are part of a large and expa nding family of chemokines (see following text), include
a component of the complement system (C5a); leukotrienes; and polypeptides from lym phocytes, mast cells, and basophils. Other plasma
factors act on the bacteria to make them “tasty” to the phagocytes (opsonizatio n). The principal opsonins that coat the bacteria are
immunoglobulins of a particular class (IgG) and complement proteins (see following tex t). The coated bacteria then bind to receptors on
the neutrophil cell membrane. This triggers, via heterotrimeric G protein-mediated respo nses, increased motor activity of the cell,
exocytosis, and the socalled respiratory burst. The increased motor activity leads to prom pt ingestion of the bacteria by endocytosis
(phagocytosis). By exocytosis, neutrophil granules discharge their contents into th e phagocytic vacuoles containing the bacteria and also
into the interstitial space (degranulation). The granules contain various pr oteases plus antimicrobial proteins called defensins. In
addition, the cell membrane-bound enzyme NADPH oxidase is act ivated, with the production of toxic oxygen metabolites. The
combination of the toxic oxygen metabolites and the proteolytic enzymes from the gran ules makes the neutrophil a very effective killing
machine.
Activation of NADPH oxidase is associated with a sharp increase in O
2
uptake and metabolism in the neutrophil (the respiratory burst)
and generation of O2– by the following reaction:
NADPH + H
+
+ 2O
2
+ → NADP
+
+ 2H
+
+ 2O2– O2– is a free radical formed by the addition of one electron to O
2
. Two O2– react
with two H
+
to form H
2
O
2
in a reaction catalyzed by the cytoplasmic form of superoxide dismutase (SOD-1):
O 2–+ O2– + H
+
+ H
+ SOD-1
→ H
2
O
2
+ O
2
O2– and H
2
O
2
are both oxidants that are effective bactericidal agents, but H
2
O
2
is
converted to H
2
O and O
2
by the enzyme catalase. The cytoplasmic form of SOD contains both Zn a nd Cu. It is found in many parts of
the body. It is defective as a result of genetic mutation in a familial form of amyotrophic lateral sclerosis (ALS; see Chapter 19).
Therefore, it may be that O2– accumulates in motor neurons and kills them in at least on e form of this progressive, fatal disease. Two
other forms of SOD encoded by at least one different gene are also found in humans.
Neutrophils also discharge the enzyme myeloperoxidase, which catalyzes the conversion of Cl
, Br
, I
, and SCN
to the corresponding
acids (HOCl, HOBr, etc). These acids are also potent oxidants. Because Cl
is present in greatest abundance in body fluids, the principal
product is HOCl.
In addition to myeloperoxidase and defensins, neutrophil granules contain an elastase, t wo metalloproteinases that attack collagen, and a
variety of other proteases that help destroy invading organisms. These enzymes act in a cooperative fashion with the O2–, H
2
O
2
, and
HOCl formed by the action of the NADPH oxidase and myeloperoxidase to produce a killing zone around the activated neutrophil. This
zone is effective in killing invading organisms, but in certain diseases (eg, rheumatoid ar thritis) the neutrophils may also cause local
destruction of host tissue.
The movements of the cell in phagocytosis, as well as migration to the site of infection, in volve microtubules and microfilaments (see
Chapter 1). Proper function of the microfilaments involves the interaction of the actin th ey contain with myosin-1 on the inside of the
cell membrane (see Chapter 1).
Like neutrophils, eosinophils have a short half-life in the circulation, are attracted to the surface of endothelial cells by selectins, bind to
integrins that attach them to the vessel wall, and enter the tissues by diapedesis. Like neutr ophils, they release proteins, cytokines, and
chemokines that produce inflammation but are capable of killing invading organisms. H owever, eosinophils have some selectivity in the
way in which they respond and in the killing molecules they secrete. Their maturation an d activation in tissues is particularly stimulated
by IL-3, IL-5, and GM-CSF (see below). They are especially abundant in the mucosa of the gastrointestinal tract, where they defend
against parasites, and in the mucosa of the respiratory and urinary tracts. Circulating eo sinophils are increased in allergic diseases such as
asthma and in various other respiratory and gastrointestinal diseases.
Basophils also enter tissues and release proteins and cytokines. They resemble but a re not identical to mast cells, and like mast cells they
contain histamine (see below). They release histamine and other inflammatory mediators when activated by binding of specific antigens
to cell-fixed IgE molecules, and are essential for immediate-type hypersensitivity reaction s. These range from mild urticaria and rhinitis
to severe anaphylactic shock. The antigens that trigger IgE formation and basophil (and mast cell) activation are innocuous to most
individuals, and are referred to as allergens.
MAST CELLS
Mast cells are heavily granulated cells of the connective tissue that are abundant in tiss ues that come into contact with the external
environment, such as beneath epithelial surfaces. Their granules contain proteoglycans, histamine, and many proteases. Like basophils,
they degranulate when allergens bind to IgE molecules directed against them that previo usly coat the mast cell surface. They are
involved in inflammatory responses initiated by immunoglobulins IgE and IgG (see belo w). The inflammation combats invading
parasites. In addition to this involvement in acquired immunity, they release TNFα in resp onse to bacterial products by an antibody-
independent mechanism, thus participating in the nonspecific innate immunity that combats infections prior to the development of an
adaptive immune response (see following text). Marked mast cell degranulation produc es clinical manifestations of allergy up to and
including anaphylaxis.
MONOCYTES
Monocytes enter the blood from the bone marrow and circulate for about 72 hours. T hey then enter the tissues and become tissue
macrophages (Figure 3–1). Their life span in the tissues is unknown, but bone marrow transplantation data in humans suggest that they
persist for about 3 months. It appears that they do not reenter the circulation. Some of th em end up as the multinucleated giant cells seen
in chronic inflammatory diseases such as tuberculosis. The tissue macrophages include t he Kupffer cells of the liver, pulmonary alveolar
macrophages (see Chapter 35), and microglia in the brain, all of which come from the circulation. In the past, they have been called the
reticuloendothelial system, but the general term tissue macrophage system seems more appropriate.
Macrophages are activated by cytokines released from T lymphocytes, among others. A ctivated macrophages migrate in response to
chemotactic stimuli and engulf and kill bacte
Macrophages
Pseudopods
Bacteria
FIGURE 3–1 Macrophages contacting bacteria and preparing to engulf them. Fig ure is a colorized version of a scanning electron
micrograph.
ria by processes generally similar to those occurring in neutrophils. They play a key ro le in immunity (see below). They also secrete up to
100 different substances, including factors that affect lymphocytes and other cells, pros taglandins of the E series, and clot-promoting
factors.
GRANULOCYTE & MACROPHAGE COLONY-STIMULATING FACTORS
The production of white blood cells is regulated with great precision in healthy individua ls, and the production of granulocytes is rapidly
and dramatically increased in infections. The proliferation and self-renewal of hematop oietic stem cells (HSCs) depends on stem cell
factor (SCF). Other factors specify particular lineages. The proliferation and ma turation of the cells that enter the blood from the
marrow are regulated by glycoprotein growth factors or hormones that cause cells in o ne or more of the committed cell lines to
proliferate and mature (Table 3–1). The regulation of erythrocyte production by erythropo ietin is discussed in Chapter 39. Three
additional factors are called colony-stimulating factors (CSFs), because they cause appropr iate single stem cells to proliferate in soft
agar, forming colonies in this culture medium. The factors stimulating the production of committed stem cells include granulocyte–
macrophage CSF (GM-CSF), granulocyte CSF (G-CSF), and macrophage CSF (M -CSF). Interleukins IL-1 and IL-6 followed by
IL-3 (Table 3–1) act in sequence to convert pluripotential uncommitted stem cells to committe d progenitor cells. IL-3 is also known as
multi-CSF. Each of the CSFs has a predominant action, but all the CSFs and interleukin s also have other overlapping actions. In
addition, they activate and sustain mature blood cells. It is interesting in this regard that th e genes for many of these factors are located
together on the long arm of chromosome 5 and may have originated by duplication of an ancestral gene. It is also interesting that basal
hematopoiesis is normal in mice in which the GMCSF gene is knocked out, indicating tha t loss of one factor can be compensated for by
others. On the other hand, the absence of GM-CSF causes accumulation of surfactant i n the lungs (see Chapter 35).
As noted in Chapter 39, erythropoietin is produced in part by kidney cells and is a circu lating hormone. The other factors are produced
by macrophages, activated T cells, fibroblasts, and endothelial cells. For the most part, th e factors act locally in the bone marrow
(Clinical Box 3–1).
LYMPHOCYTES
Lymphocytes are key elements in the production of immunity (see below). After birth, some lymphocytes are formed in the bone
marrow. However, most are formed in the lymph nodes (Figure 3–2), thymus, and sp leen from precursor cells that originally came from
the bone marrow and were processed in the thymus or bursal equivalent (see below). Lymphocytes enter the bloodstream for the most
part via the lymphatics. At
TABLE 3–1 Hematopoietic growth factors.
Cytokine IL-1
IL-3
IL-4
IL-5
IL-6
IL-11
Erythropoietin
Cell Lines
Stimulated
Erythrocyte
Granulocyte Megakaryocyte Monocyte
Erythrocyte
Granulocyte Megakaryocyte Monocyte
Basophil
Eosinophil
Erythrocyte
Granulocyte Megakaryocyte Monocyte
Erythrocyte
Granulocyte Megakaryocyte Erythrocyte
SCF
G-CSF Erythrocyte
Granulocyte Megakaryocyte Monocyte
Granulocyte
GM-CSF Erythrocyte
M-CSF Granulocyte Megakaryocyte Monocyte
Thrombopoietin Megakaryocyte
Cytokine Source Multiple cell types
T lymphocytes
T lymphocytes T lymphocytes Endothelial cells
Fibroblasts Macrophages Fibroblasts Osteoblasts
Kidney
Kupffer cells of liver Multiple cell types
Endothelial cells Fibroblasts
Monocytes
Endothelial cells Fibroblasts
Monocytes
T lymphocytes Endothelial cells Fibroblasts
Monocytes
Liver, kidney
any given time, only about 2% of the body lymphocytes are in the peripheral blood. M ost of the rest are in the lymphoid organs. It has
been calculated that in humans, 3.5 × 10
10
lymphocytes per day enter the circulation via the thoracic duct alone; however, this cou nt
includes cells that reenter the lymphatics and thus traverse the thoracic duct more than on ce. The effects of adrenocortical hormones on
the lymphoid organs, the circulating lymphocytes, and the granulocytes are discussed in Chapter 22.
Cortical follicles, B cells
Key: IL = interleukin; CSF = colony stimulating factor; G = granulocyte; M = macrophage; SCF = stem cel l factor.
Reproduced with permission from McPhee SJ, Lingappa VR, Ganong WF (editors): Pathophysio logy of Disease, 4th ed. McGraw-Hill, 2003.
CLINICAL BOX 3–1 Disorders of Phagocytic Function
More than 15 primary defects in neutrophil function have been described, along with a t least 30 other conditions in which there is a
secondary depression of the function of neutrophils. Patients with these diseases are pro ne to infections that are relatively mild when only
the neutrophil system is involved, but which can be severe when the monocyte-tissue m acrophage system is also involved. In one
syndrome (neutrophil hypomotility), actin in the neutrophils does not polymerize norma lly, and the neutrophils move slowly. In another,
there is a congenital deficiency of leukocyte integrins. In a more serious disease (chron ic granulomatous disease), there is a failure to
generate O2– in both neutrophils and monocytes and consequent inability to kill m any phagocytosed bacteria. In severe congenital
glucose 6-phosphate dehydrogenase deficiency, there are multiple infections because o f failure to generate the NADPH necessary for
O2– production. In congenital myeloperoxidase deficiency, microbial killing p ower is reduced because hypochlorous acid is not formed.
Paracortex, T cells
Medullary cords, plasma cells
FIGURE 3–2 Anatomy of a normal lymph node. (After Chandrasoma. Rep roduced with permission from McPhee SJ, Lingappa VR, Ganong WF [editors]:
Pathophysiology of Disease, 4th ed. McGraw-Hill, 2003.)
IMMUNITY
OVERVIEW
Insects and other invertebrates have only innate immunity. T his system is triggered by receptors that bind sequences of sugars, fats, or
amino acids in common bacteria and activate various defense mechanisms. The receptor s are coded in the germ line, and their
fundamental structure is not modified by exposure to antigen. The activated defenses in clude, in various species, release of interferons,
phagocytosis, production of antibacterial peptides, activation of the complement system, and several proteolytic cascades. Even plants
release antibacterial peptides in response to infection. In vertebrates, innate immunity is a lso present, but is complemented by adaptive
or acquired immunity, a system in which T and B lymphocytes are activated by very specific antigens. In both innate and acquired
immunity, the receptors involved recognize the shape of antigens, not their specific chem ical composition. In acquired immunity,
activated B lymphocytes form clones that produce more antibodies which attack foreign proteins. After the invasion is repelled, small
numbers persist as memory cells so that a second exposure to the same antigen provoke s a prompt and magnified immune attack. The
genetic event that led to acquired immunity occurred 450 million years ago in the ancest ors of jawed vertebrates and was probably
insertion of a transposon into the genome in a way that made possible the generation of the immense repertoire of T cell receptors that
are present in the body.
In vertebrates, including humans, innate immunity provides the first line of defense aga inst infections, but it also triggers the slower but
more specific acquired immune response (Figure 3–3). In vertebrates, natural and acq uired immune mechanisms also attack tumors and
tissue transplanted from other animals.
Once activated, immune cells communicate by means of cytokines and chemokines. The y kill viruses, bacteria, and other foreign cells by
secreting other cytokines and activating the complement system.
CYTOKINES
Cytokines are hormonelike molecules that act—generally in a paracrine fashion—to reg ulate immune responses. They are secreted not
only by lymphocytes and macrophages but by endothelial cells, neurons, glial cells, and other types of cells. Most of the cytokines were
initially named for their actions, for example, B cell-differentiating factor, B cell-stimulat ing factor 2. However, the nomenclature has
since been rationalized by international agreement to that of the interleukins. For exa mple, the name of B cell-differentiating factor was
changed to interleukin-4. A number of cytokines selected for their biological and clinica l relevance are listed in Table 3–2, but it would
be beyond the scope of this text to list all cytokines, which now number more than 100.
Many of the receptors for cytokines and hematopoietic growth factors (see above), as well as the receptors for prolactin
Plasma cell
B
γδ
T cell Chemokines T
H
2
N M IL-4
Bacteria Viruses Tumors Naive T cell
APC
T
H
1 Cytotoxic
lymphocyte
FIGURE 3–3 How bacteria, viruses, and tumors trigger innate immunity and initiate the acquired immune response. Arrows
indicate mediators/cytokines that act on the target cell shown and/or pathways o f differentiation. APC, antigen-presenting cell; M,
monocyte; N, neutrophil; T
H
1 and T
H
2, helper T cells type 1 and type 2, respectively.
TABLE 3–2 Examples of cytokines and their clinical relevance.
Cytokine Cellular Sources Major Activities Clinical Relevance
Interleukin-1 Macrophages
Activation of T cells and macrophages; promotion of inflammation
Implicated in the pathogenesis of septic shock, rheumatoid arthritis, and atherosclero sis
Interleukin-2 Type 1 (TH1) helper T cells
Activation of lymphocytes, natural killer cells, and macrophages
Used to induce lymphokine-activated killer cells; used in the treatment of metastatic r enal-cell carcinoma, melanoma, and various other
tumors
Interleukin-4 Type 2 (TH2) helper T cells,
mast cells, basophils, and eosinophils
Activation of lymphocytes, monocytes, and IgE class switching
As a result of its ability to stimulate IgE production, plays a part in mast-cell sensitization an d thus in allergy and in defense against
nematode infections
Interleukin-5 Type 2 (TH2) helper T cells, mast cells, and eosinophils Differentiation of eosinophils
Monoclonal antibody against interleukin-5 used to inhibit the antigen-induced late-ph ase eosinophilia in animal models of allergy
Interleukin-6 Type 2 (TH2) helper T cells
and macrophages
Activation of lymphocytes; differentiation of B cells; stimulation of the production of acu te-phase proteins
Overproduced in Castleman’s disease; acts as an autocrine growth factor in myelom a and in mesangial proliferative glomerulonephritis
Interleukin-8 T cells and macrophages
Chemotaxis of neutrophils, basophils, and T cells
Levels are increased in diseases accompanied by neutrophilia, making it a potent ially useful marker of disease activity
Interleukin-11 Bone marrow stromal cells
Stimulation of the production of acutephase proteins
Used to reduce chemotherapy-induced thrombocytopenia in patients with cancer
Interleukin-12 Macrophages and B cells
Stimulation of the production of interferon γ by type 1 (TH1) helper T cells and by natural killer cells ; induction of type 1 (TH1) helper T
cells
May be useful as an adjuvant for vaccines
Tumor necrosis factor α
Macrophages, natural killer cells, T cells, B cells, and mast cells
Promotion of inflammation Treatment with antibodies against tumor necrosis facto r α beneficial in rheumatoid arthritis
Lymphotoxin (tumor necrosis factor β) Type 1 (TH1) helper T cells and B cells
Promotion of inflammation Implicated in the pathogenesis of multiple sclerosis and insulin -dependent diabetes mellitus
Transforming growth factor β
T cells, macrophages, B cells, and mast cells
Immunosuppression May be useful therapeutic agent in multiple sclerosis and myasthen ia gravis
Granulocyte
macrophage colonystimulating factor T cells, macrophages, natural killer cells, and B ce lls Promotion of the growth of granulocytes and
monocytes
Used to reduce neutropenia after chemotherapy for tumors and in ganciclovir-treated patien ts with AIDS; used to stimulate cell
production after bone marrow transplantation
Interferonα Virally infected cells
Induction of resistance of cells to viral infection
Used to treat AIDS-related Kaposi sarcoma, melanoma, chronic hepatitis B infection, an d chronic hepatitis C infection
Interferonβ Virally infected cells
Induction of resistance of cells to viral infection
Used to reduce the frequency and severity of relapses in multiple sclerosis
Interferonγ Type 1 (TH1) helper T cells
and natural killer cells Activation of macrophages; inhibition of type 2 (TH2) helper T cells
Used to enhance the killing of phagocytosed bacteria in chronic granulomatous disease
Reproduced with permission from Delves PJ, Roitt IM: The immune system. First of two parts. N Engl J Med 2000;343:37.
(see Chapter 25), and growth hormone (see Chapter 24) are members of a cytokine-r eceptor superfamily that has three subfamilies
(Figure 3–4). The members of subfamily 1, which includes the receptors for IL-4 and IL-7, are homodimers. The members of subfamily
2, which includes the receptors for IL-3, IL-5, and IL-6, are heterodimers. The recep tor for IL-2 and several other cytokines is unique in
that it consists of a heterodimer plus an unrelated protein, the so-called Tac antigen. The other members of subfamily 3 have the same γ
chain as IL2R. The extracellular domain of the homodimer and heterodimer subunits al l contain four conserved cysteine residues plus a
conserved Trp-Ser-X-Trp-Ser domain, and although the
Erythropoietin G-CSF
IL-4
IL-7
Growth hormone PRL
IL-3
Sharedβ
IL-2 GM-CSF
subunit
IL-4 IL-5 IL-7 IL-6
IL-9
IL-11
Shared gp130
IL-15 LIF
subunit
OSM
CNTF
ECF
α
Cytoplasm
α
γ
Subfamily 1 β β Subfamily 2 Subfamily 3
FIGURE 3–4 Members of one of the cytokine receptor superfamilies, showing shared structural elements. Note that all the subunits
except the
α
subunit in subfamily 3 have four conserved cysteine residues (open boxes at top) and a Trp-Ser-X-Trp-Ser motif (pink).
Many subunits also contain a critical regulatory domain in their cytoplasmic port ions (green). CNTF, ciliary neurotrophic factor; LIF,
leukemia inhibitory factor; OSM, oncostatin M; PRL, prolactin. (Modified from D’Andrea AD: Cytokine rec eptors in congenital hematopoietic disease.
N Engl J Med 1994;330:839.)
intracellular portions do not contain tyrosine kinase catalytic domains, they activate cytop lasmic tyrosine kinases when ligand binds to
the receptors.
The effects of the principal cytokines are listed in Table 3–2. Some of them have system ic as well as local paracrine effects. For example,
IL-1, IL-6, and tumor necrosis factor α cause fever, and IL-1 increases slow-wave sle ep and reduces appetite.
Another superfamily of cytokines is the chemokine family. Chemokines are substances that attract neutrophils (see previous text) and
other white blood cells to areas of inflammation or immune response. Over 40 have no w been identified, and it is clear that they also play
a role in the regulation of cell growth and angiogenesis. The chemokine receptors are G protein-coupled receptors that cause, among
other things, extension of pseudopodia with migration of the cell toward the source of t he chemokine.
THE COMPLEMENT SYSTEM
The cell-killing effects of innate and acquired immunity are mediated in part by a system of more than 30 plasma proteins originally
named the complement system because they “complemented” the effects of antibodies. Three different pathways or enzyme cascades
activate the system: the classic pathway, triggered by immune complexes; the mannose-binding lectin pathway, triggered when this
lectin binds mannose groups in bacteria; and the alternative or properdin pathw ay, triggered by contact with various viruses, bacteria,
fungi, and tumor cells. The proteins that are produced have three functions: They help kill invading organisms by opsonization,
chemotaxis, and eventual lysis of the cells; they serve in part as a bridge from innate to a cquired immunity by activating B cells and
aiding immune memory; and they help dispose of waste products after apoptosis. Cell ly sis, one of the principal ways the complement
system kills cells, is brought about by inserting proteins called perforins into their cell membranes. These create holes, which permit
free flow of ions and thus disruption of membrane polarity.
INNATE IMMUNITY
The cells that mediate innate immunity include neutrophils, macrophages, and natural killer (NK) cells, large lymphocytes that are not
T cells but are cytotoxic. All these cells respond to lipid and carbohydrate sequences un ique to bacterial cell walls and to other substances
characteristic of tumor and transplant cells. Many cells that are not professional immuno cytes may nevertheless also contribute to innate
immune responses, such as endothelial and epithelial cells. The activated cells produce th eir effects via the release of cytokines, as well
as, in some cases, complement and other systems.
An important link in innate immunity in Drosophila is a receptor protein name d toll, which binds fungal antigens and triggers activation
of genes coding for antifungal proteins. An expanding list of toll-like receptors (TLRs) have now been identified in humans. One of
these, TLR4, binds bacterial lipopolysaccharide and a protein called CD14, and this initia tes a cascade of intracellular events that activate
transcription of genes for a variety of proteins involved in innate immune responses. Th is is important because bacterial
lipopolysaccharide produced by gram-negative organisms is the cause of septic shock. TLR2 mediates the response to microbial
lipoproteins, TLR6 cooperates with TLR2 in recognizing certain peptidoglycans, and TL R9 recognizes the DNA of certain bacteria.
ACQUIRED IMMUNITY
As noted previously, the key to acquired immunity is the ability of lymphocytes to produ ce antibodies (in the case of B cells) or cell-
surface receptors (in the case of T cells) that are specific for one of the many millions o f foreign agents that may invade the body. The
antigens stimulating production of T cell receptors or antibodies are usually proteins and polypeptides, but antibodies can also be formed
against nucleic acids and lipids if these are presented as nucleoproteins and lipoproteins, and antibodies to smaller molecules can be
produced experimentally if the molecules are bound to protein. Acquired immunity has two components: humoral immunity and cellular
immunity. Humoral immunity is mediated by circulating immunoglobulin antibodies in th e γ-globulin fraction of the plasma proteins.
Immunoglobulins are produced by differentiated forms of B lymphocytes known as plasma cells, and they activate the complement
system and attack and neutralize antigens. Humoral immunity is a major defense against bacterial infections. Cellular immunity is
mediated by T lymphocytes. It is responsible for delayed allergic reactions and rejection of transplants of foreign tissue. Cytotoxic T
cells attack and destroy cells that have the antigen which activated them. They kill by inse rting perforins (see above) and by initiating
apoptosis. Cellular immunity constitutes a major defense against infections due to viruses , fungi, and a few bacteria such as the tubercle
bacillus. It also helps defend against tumors.
DEVELOPMENT OF THE IMMUNE SYSTEM
During fetal development, and to a much lesser extent during adult life, lymphocyte pre cursors come from the bone marrow. Those that
populate the thymus (Figure 3–5) become transformed by the environment in this orga n into T lymphocytes. In birds, the precursors that
populate the bursa of Fabricius, a lymphoid structure near the cloaca, become transform ed into B lymphocytes. There is no bursa in
mammals, and the transformation to B lymphocytes occurs in bursal equivalents, t hat is, the fetal liver and, after birth, the bone
marrow. After residence in the thymus or liver, many of the T and B lymphocytes migr ate to the lymph nodes.
T and B lymphocytes are morphologically indistinguishable but can be identified by mar kers on their cell membranes. B cells
differentiate into plasma cells and memory B cells. There are three major types of T ce lls: cytotoxic T cells, helper T cells, and
memory T cells. There are two subtypes of helper T cells: T helper 1 (TH1) cells secrete IL-2 and γ- interferon and are concerned
primarily with cellular immunity; T helper 2 (TH2) cells secrete IL-4 and IL-5 and inter act primarily with B cells in relation to humoral
immunity. Cytotoxic T cells destroy transplanted and other foreign cells, with their devel opment aided and directed by helper T cells.
Markers on the surface of lymphocytes are assigned CD (clusters of differentiation) n umbers on the basis of their reactions to a
Memory T cells
Thymus T lymphocytes
Cytotoxic T cells
(mostly CD8 T cells) Cellular immunity
Bone marrow lymphocyte precursors Helper T cells (CD4 T cells)
B lymphocytes Bursal equivalent (liver, bone marrow) Plasma cells
IgG
IgA HumoralIgM
immunity
IgD
IgE
Memory B cells
FIGURE 3–5 Development of the system mediating acquired immunity.
panel of monoclonal antibodies. Most cytotoxic T cells display the glycoprotein CD8, an d helper T cells display the glycoprotein CD4.
These proteins are closely associated with the T cell receptors and may function as core ceptors. On the basis of differences in their
receptors and functions, cytotoxic T cells are divided into αβ and γδ types (see below). Natural killer cells (see above) are also cytotoxic
lymphocytes, though they are not T cells. Thus, there are three main types of cytotoxic lymphocytes in the body: αβ T cells, γδ T cells,
and NK cells.
MEMORY B CELLS & T CELLS
After exposure to a given antigen, a small number of activated B and T cells persist as m emory B and T cells. These cells are readily
converted to effector cells by a later encounter with the same antigen. This ability to pro duce an accelerated response to a second
exposure to an antigen is a key characteristic of acquired immunity. The ability persists f or long periods of time, and in some instances
(eg, immunity to measles) it can be lifelong.
After activation in lymph nodes, lymphocytes disperse widely throughout the body and are especially plentiful in areas where invading
organisms enter the body, for example, the mucosa of the respiratory and gastrointestin al tracts. This puts memory cells close to sites of
reinfection and may account in part for the rapidity and strength of their response. Che mokines are involved in guiding activated
lymphocytes to these locations.
Langerhans dendritic cells in the skin. Macrophages and B cells themselves, and likely m any other cell types, can also function as APCs.
In APCs, polypeptide products of antigen digestion are coupled to protein products of the major histocompatibility complex (MHC)
genes and presented on the surface of the cell. The products of the MHC genes are ca lled human leukocyte antigens (HLA).
The genes of the MHC, which are located on the short arm of human chromosome 6, encode glycoproteins and are divided into two
classes on the basis of structure and function. Class I antigens are composed of a 45-kD a heavy chain associated noncovalently with β
2
-
microglobulin encoded by a gene outside the MHC (Figure 3–6). They are found on a ll nucleated cells. Class II antigens are
heterodimers made up of a 29- to 34-kDa αα to 28-kDa β chain. They are present in a ntigen-presenting cells, including B cells, and in
activated T cells.
The class I MHC proteins (MHC-I proteins) are coupled primarily to peptide fragment s generated from proteins synthesized within cells.
The peptides to which the host is not tolerant (eg, those from mutant or viral proteins) ar e recognized by T cells. The digestion of these
proteins occurs in
α
1
α
2
ANTIGEN RECOGNITION
The number of different antigens recognized by lymphocytes in the body is extremely large. The repertoire develops initially without
exposure to the antigen. Stem cells differentiate into many million different T and B lymp hocytes, each with the ability to respond to a
particular antigen. When the antigen first enters the body, it can bind directly to the appr opriate receptors on B cells. However, a full
antibody response requires that the B cells contact helper T cells. In the case of T cells, t he antigen is taken up by an antigen-presenting
cell and partially digested. A peptide fragment of it is presented to the appropriate recep tors on T cells. In either case, the cells are
stimulated to divide, forming clones of cells that respond to this antigen (clonal selection). Ef fector cells are also subject to negative
selection, during which lymphocyte precursors that are reactive with self antige ns are normally deleted. This results in immune
tolerance. It is this latter process that presumably goes awry in autoimmune diseases, wh ere the body reacts to and destroys cells
expressing normal proteins, with accompanying inflammation that may lead to tissue des truction.
ANTIGEN PRESENTATION
Antigen-presenting cells (APCs) include specialized cells called dendritic cells in the lymph node s and spleen and the
N
N
C C
β
2
m
α
3
proteasomes, complexes of proteolytic enzymes that may be produced by genes in the MH C group, and the peptide fragments appear
to bind to MHC proteins in the endoplasmic reticulum. The class II MHC proteins (MH C-II proteins) are concerned primarily with
peptide products of extracellular antigens, such as bacteria, that enter the cell by endocy tosis and are digested in the late endosomes.
FIGURE 3–6 Structure of human histocompatibility antigen HLA-A2. T he antigen-binding pocket is at the top and is formed by the
α1
and
α2
parts of the molecule. The
α3
portion and the associated
β2
- microglobulin (
β2
m) are close to the membrane. The extension of
the C terminal from
α3
that provides the transmembrane domain and the small cytoplasmic portion of the molecule hav e been omitted.
(Reproduced with permission from Bjorkman PJ et al: Structure of the human histocompatibility antigen HLA-A2. Nature 1987;329:506.)
CD4
Class II Class I MHC
CD8
MHC
T CELL RECEPTORS
The MHC protein–peptide complexes on the surface of the antigen-presenting cells bin d to appropriate T cells. Therefore, receptors on
the T cells must recognize a very wide variety of complexes. Most of the receptors on c irculating T cells are made up of two polypeptide
units designated α and β. They form heterodimers that recognize the MHC proteins and the antigen fragments with which they are
combined (Figure 3–7). These cells are called αβ T cells. About 10% of the circulating T cells have two different polypeptides designated
γ and δ in their receptors, and they are called γδ T cells. These T cells are prominent in the mucosa of the gastrointestinal tract, and there
is evidence that they form a link between the innate and acquired immune systems by w ay of the cytokines they secrete (Figure 3–3).
CD8 occurs on the surface of cytotoxic T cells that bind MHC-I proteins, and CD4 oc curs on the surface of helper T cells that bind
MHC-II proteins (Figure 3–8). The CD8 and CD4 proteins facilitate the binding of the MHC proteins to the T cell receptors, and they
also foster lymphocyte development, but how they produce these effects is unsettled. Th e
TCR TCR
FIGURE 3–8 Diagrammatic summary of the structure of CD4 and CD8, and their re lation to MHC-I and MHC-II proteins. Note
that CD4 is a single protein, whereas CD8 is a heterodimer.
activated CD8 cytotoxic T cells kill their targets directly, whereas the activated CD4 helpe r T cells secrete cytokines that activate other
lymphocytes.
The T cell receptors are surrounded by adhesion molecules and proteins that bind to co mplementary proteins in the antigen-presenting
cell when the two cells transiently join to form the “immunologic synapse” that permits T cell activation to occur. It is now generally
accepted that two signals are necessary to produce activation. One is produced by the b inding of the digested antigen to the T cell
receptor. The other is produced by the joining of the surrounding proteins in the “syna pse.” If the first signal occurs but the second does
not, the T cell is inactivated and becomes unresponsive.
Antigen-presenting
Cytoplasm
cell membrane
ECF β
2
m α
3
MHC molecular complex α
1
/
α
2
Antigen fragment
Constant regions
Variable regions
S–S ECF
+ +
Cytoplasm β α
T cell receptor heterodimer (α:β) T cell
membrane
FIGURE 3–7 Interaction between antigen-presenting cell (top) and
αβ
T lymphocyte (bottom). The MHC proteins (in this case,
MHC-I) and their peptide antigen fragment bind to the
α
and
β
units that combine to form the T cell receptor.
B CELLS
As noted above, B cells can bind antigens directly, but they must contact helper T cells to produce full activation and antibody formation.
It is the TH2 subtype that is mainly involved. Helper T cells develop along the TH2 linea ge in response to IL-4 (see below). On the other
hand, IL-12 promotes the TH1 phenotype. IL-2 acts in an autocrine fashion to cause a ctivated T cells to proliferate. The role of various
cytokines in B cell and T cell activation is summarized in Figure 3–9.
The activated B cells proliferate and transform into memory B cells (see above) and plasma cells. The plasma cells secrete large
quantities of antibodies into the general circulation. The antibodies circulate in the globuli n fraction of the plasma and, like antibodies
elsewhere, are called immunoglobulins. The immunoglobulins are actually the se creted form of antigen-binding receptors on the B cell
membrane.
MHC class II
1
Macrophage (antigen
presenting
cell)
V
H
Antigenbinding site
V
H
JH
D
2 IL-1 Fab
CH1
V
L
CD4 TCR C
SS SS
L
SS
V
L
J
L
Cytokine
induced IL-2R activation
3
IL-2 Complement
CH2binding
Fc Macrophage
binding
C
H
3 Hinge
Activated B cell
4 CD4
Activated
4
T cell
4
Inflammation
IL-2R
Cytotoxic and delayed T cell hypersensitivity
CD8
Antibodyproducing cell MHC class I
Cell death
FIGURE 3–10 Typical immunoglobulin G molecule. Fab, portion of the molecule tha t is concerned with antigen binding; Fc, effector
portion of the molecule. The constant regions are pink and purple, and the variable regions are orange. The constant segment of the
heavy chain is subdivided into C
H
1, C
H
2, and C
H
3. SS lines indicate intersegmental disulfide bonds. On the right side, the C labels are
omitted to show regions J
H
, D, and J
L
.
FIGURE 3–9 Summary of acquired immunity. (1) An antigenpresent ing cell ingests and partially digests an antigen, then presents part
of the antigen along with MHC peptides (in this case, MHC II peptides on the cell surfa ce). (2) An “immune synapse” forms with a naive
CD4 T cell, which is activated to produce IL-2. (3) IL-2 acts in an autocrine fashion to cause the cell to multiply, forming a clone. (4)
The activated CD4 cell may promote B cell activation and production of plasma cells or it m ay activate a cytotoxic CD8 cell. The CD8
cell can also be activated by forming a synapse with an MCH I antigen-presenting cell. (Reprodu ced with permission from McPhee SJ, Lingappa
VR, Ganong WF [editors]: Pathophysiology of Disease, 4th ed. McGraw-Hill, 2003.)
IMMUNOGLOBULINS
Circulating antibodies protect their host by binding to and neutralizing some protein toxin s, by blocking the attachment of some viruses
and bacteria to cells, by opsonizing bacteria (see above), and by activating complement. Five general types of immunoglobulin
antibodies are produced by the lymphocyte–plasma cell system. The basic component o f each is a symmetric unit containing four
polypeptide chains (Figure 3–10). The two long chains are called heavy chain s, whereas the two short chains are called light chains.
There are two types of light chains, k and λ, and eight types of heavy chains. The chain s are joined by disulfide bridges that permit
mobility, and there are intrachain disulfide bridges as well. In addition, the heavy chains are flexible in a region called the hinge. Each
heavy chain has a variable (V) segment in which the amino acid sequence is highly var iable, a diversity (D) segment in which the amino
acid segment is also highly variable, a joining (J) segment in which the sequence is mode rately variable, and a constant (C) segment in
which the sequence is constant. Each light chain has a V, a J, and a C segment. The V s egments form part of the antigen-binding sites
(Fab portion of the molecule [Figure 3–10]). The Fc portion of the molecule is the effe ctor portion, which mediates the reactions
initiated by antibodies.
Two of the classes of immunoglobulins contain additional polypeptide components (Tab le 3–3). In IgMs, five of the basic
immunoglobulin units join around a polypeptide called the J chain to form a pentamer. I n IgAs, the secretory immunoglobulins, the
immunoglobulin units form dimers and trimers around a J chain and a polypeptide that c omes from epithelial cells, the secretory
component (SC).
In the intestine, bacterial and viral antigens are taken up by M cells (see Chapter 27) an d passed on to underlying aggregates of lymphoid
tissue (Peyer’s patches), where they activate naive T cells. These lymphocytes then form B cells that infiltrate mucosa of the
gastrointestinal, respiratory, genitourinary, and female reproductive tracts and the breas t. There they secrete large amounts of IgAs when
exposed again to the antigen that initially stimulated them. The epithelial cells produce the SC, which acts as a receptor for and binds the
IgA. The resulting secretory immunoglobulin passes through the epithelial cell and is sec reted by exocytosis. This system of secretory
immunity is an important and effective defense mechanism.
GENETIC BASIS OF DIVERSITY
IN THE IMMUNE SYSTEM
The genetic mechanism for the production of the immensely large number of different configurations of immunoglobulins produced by
human B cells is a fascinating biologic problem.
TABLE 3–3 Human immunoglobulins.
a
Immunoglobulin Function Heavy Chain Additional Chain
Plasma Concentration
Structure (mg/dL)
IgG Complement activation γ1
,
γ2
,
γ3
,
γ4
Monomer 1000
IgA Localized protection in external secretions (tears, intestinal secretions, etc)
α
1
,
α
2
J, SC Monomer; dimer with J or SC 200 chain; trimer with J chain
IgM Complement activation μ J Pentamer with J chain 120
IgD Antigen recognition by B cells δ Monomer 3
IgE Reagin activity; releases histamine from basophils and mast cells
a
In all instances, the light chains are k or γ
ε Monomer 0.05
Diversity is brought about in part by the fact that in immune globulin molecules there are two kinds of light chains and eight kinds of
heavy chains. As noted previously, there are areas of great variability (hypervariable reg ions) in each chain. The variable portion of the
heavy chains consists of the V, D, and J segments. In the gene family responsible for th is region, there are several hundred different
coding regions for the V segment, about 20 for the D segment, and 4 for the J segmen t. During B cell development, one V coding
region, one D coding region, and one J coding region are selected at random and reco mbined to form the gene that produces that
particular variable portion. A similar variable recombination takes place in the coding re gions responsible for the two variable segments
(V and J) in the light chain. In addition, the J segments are variable because the gene se gments join in an imprecise and variable fashion
(junctional site diversity) and nucleotides are sometimes added (junctional insertion diver sity). It has been calculated that these
mechanisms permit the production of about 10
15
different immunoglobulin molecules. Additional variability is added by somatic
mutation.
Similar gene rearrangement and joining mechanisms operate to produce the diversity in T cell receptors. In humans, the α subunit has a
V region encoded by 1 of about 50 different genes and a J region encoded by 1 of a nother 50 different genes. The β subunits have a V
region encoded by 1 of about 50 genes, a D region encoded by 1 of 2 genes, and a J region encoded by 1 of 13 genes. These variable
regions permit the generation of up to an estimated 10
15
different T cell receptors (Clinical Box 3–2 and Clinical Box 3–3).
A variety of immunodeficiency states can arise from defects in these various stages of B and T lymphocyte maturation. These are
summarized in Figure 3–12.
PLATELETS
Platelets are circulating cells that are important mediators of hemostasis. While not immun e cells, per se, they often participate in the
response to tissue injury in cooperation with inflammatory cell types (see below). They have a ring of microtubules around their
periphery and an extensively invaginated membrane with an intricate canalicular system in contact with the ECF. Their membranes
contain receptors for collagen, ADP, vessel wall von Willebrand factor (see below), an d fibrinogen. Their cytoplasm contains actin,
myosin, glycogen, lysosomes, and two types of granules: (1) dense granules, which co ntain the nonprotein substances that are secreted in
response to platelet activation, including serotonin, ADP, and other adenine nucleotides; and (2) α-granules, which contain secreted
proteins other than the hydrolases in lysosomes. These proteins include clotting factors a nd plateletderived growth factor (PDGF).
PDGF is also produced by macrophages and endothelial cells. It is a dimer made up of A and B subunit polypeptides. Homodimers (AA
and BB), as well as the heterodimer (AB), are produced. PDGF stimulates wound heali ng and is a potent mitogen for vascular smooth
muscle. Blood vessel walls as well as platelets contain von Willebrand factor, which, in a ddition to its role in adhesion, regulates
circulating levels of factor VIII (see below).
When a blood vessel wall is injured, platelets adhere to the exposed collagen and von Willebrand facto r in the wall via receptors on the
platelet membrane. Von Willebrand factor is a very large circulating molecule that is pro duced by endothelial cells. Binding produces
platelet activations which release the contents of their granules. The released ADP acts o n the ADP receptors in the platelet membranes
to produce further accumulation of more platelets (platelet aggregation). Humans have a t least three different types of platelet ADP
receptors: P2Y
1
, P2Y
2
, and P2X
1
. These are obviously attractive targets for drug development, and several new inhibitor s have shown
promise in the prevention of heart attacks and strokes. Aggregation is also fostered by platelet-activating factor (PAF), a cytokine
secreted by neutrophils and monocytes as well as platelets. This compound also has infla mmacytes as well as platelets. This compound
also has inflamma acetylglyceryl-3-phosphorylcholine, which is produced from membr ane lipids. It acts via a G protein-coupled receptor
to
CLINICAL BOX 3–2 CLINICAL BOX 3–3 Autoimmunity
Sometimes the processes that eliminate antibodies against self antigens fail and a variety o f different autoimmune diseases are
produced. These can be B cell- or T cell-mediated and can be organ-specific or system ic. They include type 1 diabetes mellitus
(antibodies against pancreatic islet B cells), myasthenia gravis (antibodies against nicotinic cholinergic receptors), and multiple sclerosis
(antibodies against myelin basic protein and several other components of myelin). In so me instances, the antibodies are against receptors
and are capable of activating those receptors; for example, antibodies against TSH recep tors increase thyroid activity and cause Graves’
disease (see Chapter 20). Other conditions are due to the production of antibodies agai nst invading organisms that cross-react with
normal body constituents (molecular mimicry). An example is rheumatic f ever following a streptococcal infection; a portion of cardiac
myosin resembles a portion of the streptococcal M protein, and antibodies induced by th e latter attack the former and damage the heart.
Some conditions may be due to bystander effects, in which inflammation sensitizes T cells in the neighborhood , causing them to
become activated when otherwise they would not respond. However, much is still unce rtain about the pathogenesis of autoimmune
disease.
increase the production of arachidonic acid derivatives, including thromboxane A
2
. The role of this compound in the balance between
clotting and anticlotting activity at the site of vascular injury is discussed in Chapter 32.
Platelet production is regulated by the colony-stimulating factors that control the product ion of megakaryocytes, plus thrombopoietin, a
circulating protein factor. This factor, which facilitates megakaryocyte maturation, is pro duced constitutively by the liver and kidneys,
and there are thrombopoietin receptors on platelets. Consequently, when the number of platelets is low, less is bound and more is
available to stimulate production of platelets. Conversely, when the number of platelets is high, more is bound and less is available,
producing a form of feedback control of platelet production. The amino terminal portio n of the thrombopoietin molecule has the platelet-
stimulating activity, whereas the carboxyl terminal portion contains many carbohydrate r esidues and is concerned with the bioavailability
of the molecule.
When the platelet count is low, clot retraction is deficient and there is poor constriction o f ruptured vessels. The resulting clinical
syndrome (thrombocytopenic purpura) is characterized by easy bruisability and m ultiple subcutaneous hemorrhages. Purpura may also
occur when the platelet count is normal, and in some of these cases, the circulating plate lets are abnormal (thrombasthenic purpura).
Individuals with thrombocytosis are predisposed to thrombotic events.
Tissue Transplantation
The T lymphocyte system is responsible for the rejection of transplanted tissue. When tis sues such as skin and kidneys are transplanted
from a donor to a recipient of the same species, the transplants “take” and function for a while but then become necrotic and are
“rejected” because the recipient develops an immune response to the transplanted tissue. This is generally true even if the donor and
recipient are close relatives, and the only transplants that are never rejected are those fro m an identical twin. A number of treatments
have been developed to overcome the rejection of transplanted organs in humans. The goal of treatment is to stop rejection without
leaving the patient vulnerable to massive infections. One approach is to kill T lymphocyt es by killing all rapidly dividing cells with drugs
such as azathioprine, a purine antimetabolite, but this makes patients susceptible to infectio ns and cancer. Another is to administer
glucocorticoids, which inhibit cytotoxic T cell proliferation by inhibiting production of IL -2, but these cause osteoporosis, mental
changes, and the other facets of Cushing syndrome (see Chapter 22). More recently, i mmunosuppressive drugs such as cyclosporine or
tacrolimus (FK-506) have found favor. Activation of the T cell receptor normally increases intracellular Ca
2+
, which acts via
calmodulin to activate calcineurin (Figure 3-11). Calcineurin dephosphorylates the trans cription factor NF-AT, which moves to the
nucleus and increases the activity of genes coding for IL-2 and related stimulatory cyto kines. Cyclosporine and tacrolimus prevent the
dephosphorylation of NF-AT. However, these drugs inhibit all T cell-mediated immune responses, and cyclosporine causes kidney
damage and cancer. A new and promising approach to transplant rejection is the produ ction of T cell unresponsiveness by using drugs
that block the costimulation that is required for normal activation (see text). Clinically eff ective drugs that act in this fashion could be of
great value to transplant surgeons.
INFLAMMATION &
WOUND HEALING
LOCAL INJURY
Inflammation is a complex localized response to foreign substances such as bacteria or in some instances to internally produced
substances. It includes a sequence of reactions initially involving cytokines, neutrophils, adhesion molecules, complement, and IgG.
PAF, an agent with potent inflammatory effects, also plays a role. Later, monocytes and lymphocytes are involved. Arterioles in the
inflamed area dilate, and capillary permeability is increased (see Chapters 33 and 34). W hen the inflammation occurs in or just under the
skin (Figure 3–13), it
T cell receptor
Ca
2+
NF
κ
B by increasing the production of I
κ
B
α
, and this is probably the main basis of their anti-inflammatory action (see Chapter 22).
CAM
Calcineurin
TCLBP
CsABP
NF-AT
P
IL-2 gene activation Nucleus
FIGURE 3–11 Action of cyclosporine (CsA) and tacrolimus (TCL) in lymphocytes. BP, bi nding protein; CAM, calmodulin.
SYSTEMIC RESPONSE TO INJURY
Cytokines produced in response to inflammation and other injuries also produce system ic responses. These include alterations in plasma
acute phase proteins, defined as proteins whose concentration is increased or decreased by at le ast 25% following injury. Many of the
proteins are of hepatic origin. A number of them are shown in Figure 3–14. The cause s of the changes in concentration are incompletely
understood, but it can be said that many of the changes make homeostatic sense. Thus, for example, an increase in C-reactive protein
activates monocytes and causes further production of cytokines. Other changes that oc cur in response to injury include somnolence,
negative nitrogen balance, and fever.
is characterized by redness, swelling, tenderness, and pain. Elsewhere, it is a key compo nent of asthma, ulcerative colitis, and many other
diseases.
Evidence is accumulating that a transcription factor, nuclear factorκB, plays a key role in the inflammatory response. NF
κ
B is a
heterodimer that normally exists in the cytoplasm of cells bound to I
κ
B
α
, which renders it inactive. Stimuli such as cytokines, viruses,
and oxidants separate NF
κ
B from I
κ
B
α
, which is then degraded. NF
κ
B moves to the nucleus, where it binds to the DNA of the genes for
numerous inflammatory mediators, resulting in their increased production and secretion . Glucocorticoids inhibit the activation of
WOUND HEALING
When tissue is damaged, platelets adhere to exposed matrix via integrins that bind to coll agen and laminin (Figure 3–13). Blood
coagulation produces thrombin, which promotes platelet aggregation and granule releas e. The platelet granules generate an inflammatory
response. White blood cells are attracted by selectins and bind to integrins on endothelia l cells, leading to their extravasation through the
blood vessel walls. Cytokines released by the white blood cells and platelets up-regulate integrins on macrophages, which migrate to the
area of injury, and on fibroblasts and epithelial cells, which mediate wound healing
Pluripotent stem cell Autosomal
recessive SCID
BONE MARROW Lymphoid progenitor
THYMUS X-linked SCID pre-B cell
X-linked agammaglobulinemia Immature T cell
B cell MHC class I deficiency MHC class II deficiency
Hyper-IgM syndrome CD8
cell CD4 cell
IgM IgG IgA IgE FIGURE 3–12 Sites of congenital blockade of B and T lymphocyte maturation in various immunodeficiency
states. SCID, severe combined immune deficiency. (Modified from Rosen FS, Cooper MD, Wedgwood RJP : The primary immunodeficiencies. N Engl J
Med 1995;333:431.)
Fibrin clot
Neutrophil
Macrophage
TGFβ1 Platelet TGF
αplug FGF
VEGF PDGF BB
IGFTGFβ1
PDGF AB
Blood vessel
VEGF
Neutrophil
FGF-2
FGF-2
Fibroblast
FIGURE 3–13 Cutaneous wound 3 days after injury, showing the mult iple cytokines and growth factors affecting the repair
process. VEGF, vascular endothelial growth factor. For other abbreviations, se e Appendix. Note the epidermis growing down under the
fibrin clot, restoring skin continuity. (Modified from Singer AJ, Clark RAF: Cutaneous wo und healing. N Engl J Med 1999;341:738.)
and scar formation. Plasmin aids healing by removing excess fibrin. This aids the migra tion of keratinocytes into the wound to restore
the epithelium under the scab. Collagen proliferates, producing the scar. Wounds gain 20% of their ultimate strength in 3 weeks and later
gain more strength, but they never reach more than about 70% of the strength of norm al skin.
30,100
30,000
700
600
C-reactive protein
500
Serum amyloid A 400
300
Haptoglobin
200
Fibrinogen
100
C3
0
Albumin
Transferrin 0 714 21
Time after inflammatory stimulus (d) FIGURE 3–14 Time course of changes in some major acute phase proteins. C3, C3 component
of complement. (Modified and reproduced with permission from Gitlin JD, Colten HR: Molecular biology of ac ute phase plasma proteins. In Pick F, et al [editors]:
Lymphokines, vol 14, pages 123–153. Academic Press, 1987.)
CHAPTER SUMMARY
Immune and inflammatory responses are mediated by several different cell types— granulocytes, lymphocytes, monocytes, mast cells,
tissue macrophages, and antigen presenting cells— that arise predominantly from the bo ne marrow and may circulate or reside in
connective tissues.
Granulocytes mount phagocytic responses that engulf and destroy bacteria. These are accompanied by the release of reactive oxygen
species and other mediators into adjacent tissues that may cause tissue injury.
Mast cells and basophils underpin allergic reactions to substances that would be trea ted as innocuous by nonallergic individuals.
A variety of soluble mediators orchestrate the development of immunologic effector cells and their subsequent immune and
inflammatory reactions.
Innate immunity represents an evolutionarily conserved, primitive response to stere otypical microbial components.
Acquired immunity is slower to develop than innate immunity, but long-lasting and more effective.
Genetic rearrangements endow B and T lymphocytes with a vast array of receptors capab le of recognizing billions of foreign antigens.
Self-reactive lymphocytes are normally deleted; a failure of this process leads to autoimmune disease. Disease can also result from
abnormal function or development of granulocytes and lymphocytes. In these latt er cases, deficient immune responses to microbial
threats usually result.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. In normal hu man blood
A) the eosinophil is the most common type of white blood cell. B) there are more lymph ocytes than neutrophils.
C) the iron is mostly in hemoglobin.
D) there are more white cells than red cells.
E) there are more platelets than red cells.
2. Lymphocytes
A) all originate from the bone marrow after birth. B) are unaffected by hormones.
C) convert to monocytes in response to antigens.
D) interact with eosinophils to produce platelets.
E) are part of the body’s defense against cancer.
3. The ability of the blood to phagocytose pathogens and mount a respiratory burst is increas ed by
A) interleukin-2 (IL-2).
B) granulocyte colony-stimulating factor (G-CSF). C) erythropoietin.
D) interleukin-4 (IL-4).
E) interleukin-5 (IL-5).
4. Cells responsible for innate immunity are activated most commonly by
A) glucocorticoids.
B) pollen.
C) carbohydrate sequences in bacterial cell walls.
D) eosinophils.
E) cytoplasmic proteins of bacteria.
CHAPTER RESOURCES
Delibro G: The Robin Hood of antigen presentation. Science 2004;302:485.
Delves PJ, Roitt IM: The immune system. (Two parts.) N Engl J Med 2000;343:37,108.
Dhainaut J-K, Thijs LG, Park G (editors): Septic Shock. WB Saunders, 2000.
Ganz T: Defensins and host defense. Science 1999;286:420.
Samstein B, Emond JC: Liver transplant from living related donors. Annu Rev Med 2 001;52:147.
Singer AJ, Clark RAF: Cutaneous wound healing. N Engl J Med 1999;341:738
Tedder TF, et al: The selectins: Vascular adhesion molecules. FASEB J 1995;9:866.
Tilney NL: Transplant: From Myth to Reality. Yale University Press, 2003.
Walport MJ: Complement. (Two parts) N Engl J Med 2001;344:1058, 1140.
SECTION II PHYSIOLOGY OF NERVE & MUSCLE CELLS CH APTER
Excitable Tissue: Nerve 4
OBJEC TIV ES
After studying this chapter, you should be able to:
After studying this chapter, you should be able to:
Name the parts of a neuron and their functions.
Name the various types of glia and their functions.
Describe the chemical nature of myelin, and summarize the differences in the ways in which u nmyelinated and myelinated neurons
conduct impulses.
Define orthograde and retrograde axonal transport and the molecular motors involved in each.
Describe the changes in ionic channels that underlie electrotonic potentials, the ac tion potential, and repolarization.
List the various nerve fiber types found in the mammalian nervous system.
Describe the function of neurotrophins.
INTRODUCTION
The human central nervous system (CNS) contains about 10
11
(100 billion) neurons. It also contains 10–50 times this number of glial
cells. The CNS is a complex organ; it has been calculated that 40% of the human genes partic ipate, at least to a degree, in its formation.
The neurons, the basic building blocks of the nervous system, have evolved from prim itive neuroeffector cells that respond to various
stimuli by contracting. In more complex animals, contraction has become the specialized function of muscle cells, whereas integration
and transmission of nerve impulses have become the specialized functions of neurons. This chapter describes the cellular components of
the CNS and the excitability of neurons, which involves the genesis of electrical signals that enable neu rons to integrate and transmit
impulses (action potentials, receptor potentials, and synaptic potentials).
79
CELLULAR ELEMENTS IN THE CNS
GLIAL CELLS
For many years following their discovery, glial cells (or glia) were viewed as CNS con nective tissue. In fact, the word glia is Greek for
glue. However, today theses cells are recognized for their role in communication within the CNS in partnership with neurons. Unlike
neurons, glial cells continue to undergo cell division in adulthood and their ability to pro liferate is particularly noticeable after brain
injury (eg, stroke).
There are two major types of glial cells in the vertebrate nervous system: microglia and m acroglia. Microglia are scavenger cells that
resemble tissue macrophages and remove debris resulting from injury, infection, and d isease (eg, multiple sclerosis, AIDS-related
dementia, Parkinson disease, and Alzheimer disease). Microglia arise from macrophage s outside of the nervous system and are
physiologically and embryologically unrelated to other neural cell types.
There are three types of macroglia: oligodendrocytes, Schwann cells, and astrocytes (F igure 4–1). Oligodendrocytes and Schwann cells
are involved in myelin formation around axons in the CNS and peripheral nervous sys tem, respectively. Astrocytes, which are found
throughout the brain, are of two subtypes. Fibrous astrocytes, which contain many intermediate filaments, ar e found primarily in white
matter. Protoplasmic astrocytes are found in gray matter and have a granular cy toplasm. Both types send processes to blood vessels,
where they induce capillaries to form the tight junctions making up the blood–brain barrier. They als o send processes that
NEURONS
Neurons in the mammalian central nervous system come in many different shapes and s izes. Most have the same parts as the typical
spinal motor neuron illustrated in Figure 4–2. The cell body (soma) contains the nucleus and is the metabolic center of the neuron.
Neurons have several processes called dendrites that extend outward from the cell bod y and arborize extensively. Particularly in the
cerebral and cerebellar cortex, the dendrites have small knobby projections called dendritic spines. A typical neuron also has a long
fibrous axon that originates from a somewhat thickened area of the cell body, the axon hillock. The first portion of the axon is called the
initial segment. The axon divides into presynaptic terminals, each ending in a n umber of synaptic knobs which are also called
terminal buttons or boutons. They contain granules or vesicles in which the syn aptic transmitters secreted by the nerves are stored.
Based on the number of processes that emanate from their cell body, neurons can be c lassified as unipolar, bipolar, and multipolar
(Figure 4–3).
A Oligodendrocyte B Schwann cell C Astrocyte
Oligodendrocyte Perineural in white matter
oligodendrocytes Nodes of Ranvier Capillary
End-foot
Neuron Layers
of myelin
Axons
envelop synapses and the surface of nerve cells. Protoplasmic astrocytes have a membr ane potential that varies with the external K
+
concentration but do not generate propagated potentials. They produce substances that are tropic to neurons, and they help maintain the
appropriate concentration of ions and neurotransmitters by taking up K
+
and the neurotransmitters glutamate and γ-aminobutyrate
(GABA).
Schwann
End-foot
cell
cell
Fibrous astrocyte
Nucleus
Inner
tongue
Axon
Neuron
FIGURE 4–1 The principal types of glial cells in the nervous system. A) Oligodendrocytes are small with relatively few processes.
Those in the white matter provide myelin, and those in the gray matter support neurons. B) Sch wann cells provide myelin to the
peripheral nervous system. Each cell forms a segment of myelin sheath about 1 mm lon g; the sheath assumes its form as the inner tongue
of the Schwann cell turns around the axon several times, wrapping in concen tric layers. Intervals between segments of myelin are the
nodes of Ranvier. C) Astrocytes are the most common glia in the CNS and are characte rized by their starlike shape. They contact both
capillaries and neurons and are thought to have a nutritive function. They are also involved in forming the blood–brain barrier. (From
Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 20 00.)
Cell body (soma)
Initial segment
of axon Node of Ranvier Schwann cell
Axon hillock
Nucleus
Terminal buttons
Dendrites
FIGURE 4–2 Motor neuron with a myelinated axon. A m otor neuron is comprised of a cell body (soma) with a nucleus, several
processes called dendrites, and a long fibrous axon that originates from the axon hillock. The first portion of the axon is called the initial
segment. A myelin sheath forms from Schwann cells and surrounds the axon except at its en ding and at the nodes of Ranvier. Terminal
buttons (boutons) are located at the terminal endings.
A Unipolar cell B Bipolar cell C Pseudo-unipolar cell
Dendrite Dendrites Peripheral axon to skin and
muscle
Cell body
Axon
Cell body Cell body Single bifurcated process
Axon Central axon
Invertebrate neuron Bipolar cell of retina Axon terminals Ganglion cell of dorsal root
D Three types of multipolar cells
Dendrites Apical
dendrite Cell body Cell body
Basal dendrite Axon
Dendrites
Cell body
Axon Axon
Motor neuron of spinal cord
Pyramidal cell of hippocampus Purkinje cell of cerebellum
FIGURE 4–3 Some of the types of neurons in the mammalian nervous system . A) Unipolar neurons have one process, with different
segments serving as receptive surfaces and releasing terminals. B) Bipolar neurons have t wo specialized processes: a dendrite that carries
information to the cell and an axon that transmits information from the cell. C) Some sensory neu rons are in a subclass of bipolar cells
called pseudo-unipolar cells. As the cell develops, a single process splits into two, both of which function as axons—one going to skin or
muscle and another to the spinal cord. D) Multipolar cells have one axon and many dendri tes. Examples include motor neurons,
hippocampal pyramidal cells with dendrites in the apex and base, and cerebellar Purkinje cells with an extensive dendritic tree in a single
plane. (From Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed . McGraw-Hill, 2000.)
The conventional terminology used for the parts of a neuron works well enough for s pinal motor neurons and interneurons, but there are
problems in terms of “dendrites” and “axons” when it is applied to other types of neuro ns found in the nervous system. From a
functional point of view, neurons generally have four important zones: (1) a receptor, or dendritic zone, where multiple local potential
changes generated by synaptic connections are integrated; (2) a site where propagated action potentials are generated (the initial segment
in spinal motor neurons, the initial node of Ranvier in cutaneous sensory neurons); (3) an axonal process that transmits propagated
impulses to the nerve endings; and (4) the nerve endings, where action potentials cause the release of synaptic transmitters. The cell body
is often located at the dendritic zone end of the axon, but it can be within the axon (eg, auditory neurons) or attached to the side of the
axon (eg, cutaneous neurons). Its location makes no difference as far as the receptor f unction of the dendritic zone and the transmission
function of the axon are concerned.
The axons of many neurons are myelinated, that is, they acquire a sheath of myelin, a protein–lipid complex that is wrapped around the
axon (Figure 4–2). In the peripheral nervous system, myelin forms when a Schwann c ell wraps its membrane around an axon up to 100
times (Figure 4–1). The myelin is then compacted when the extracellular portions of a m embrane protein called protein zero (P
0
) lock to
the extracellular portions of P
0
in the apposing membrane. Various mutations in the gene for P
0
cause peripheral neuropathies; 29
different mutations have been described that cause symptoms ranging from mild to seve re. The myelin sheath envelops the axon except
at its ending and at the nodes of Ranvier, periodic periodic μm constrictions that are about 1 mm apart (Figure 4 –2). The insulating
function of myelin is discussed later in this chapter. Not all neurons are myelinated; some are unmyelinated, that is, simply surrounded
by Schwann cells without the wrapping of the Schwann cell membrane that produces m yelin around the axon.
In the CNS of mammals, most neurons are myelinated, but the cells that form the myelin are oligodendrocytes rather than Schwann cells
(Figure 4–1). Unlike the Schwann cell, which forms the myelin between two nodes of Ranvier on a single neuron, oligodendrocytes emit
multiple processes that form myelin on many neighboring axons. In multiple sclerosis, a crippling autoimmune disease, patchy
destruction of myelin occurs in the CNS (see Clinical Box 4–1). The loss of myelin is as sociated with delayed or blocked conduction in
the demyelinated axons.
CLINICAL BOX 4–1 Demyelinating Diseases
Normal conduction of action potentials relies on the insulating properties of myelin. Thus, defects in myelin can have major adverse
neurological consequences. One example is multiple sclerosis (MS), an autoim mune disease that affects over 3 million people
worldwide, usually striking between the ages of 20 and 50 and affecting women about twice as often as men. The cause of MS appears to
include both genetic and environmental factors. It is most common among Caucasians li ving in countries with temperate climates,
including Europe, southern Canada, northern United States, and southeastern Australia. Environmental triggers include early exposure to
viruses such as Epstein-Barr virus and those that cause measles, herpes, chicken pox, o r influenza. In MS, antibodies and white blood
cells in the immune system attack myelin, causing inflammation and injury to the sheath a nd eventually the nerves that it surrounds. Loss
of myelin leads to leakage of K
+
through voltage-gated channels, hyperpolarization, and failure to conduct action poten tials. Typical
physiological deficits range from muscle weakness, fatigue, diminished coordination, slu rred speech, blurred or hazy vision, bladder
dysfunction, and sensory disturbances. Symptoms are often exasperated by increased b ody temperature or ambient temperature.
Progression of the disease is quite variable. In the most common form, transient episode s appear suddenly, last a few weeks or months,
and then gradually disappear. Subsequent episodes can appear years later, and eventua lly full recovery does not occur. Others have a
progressive form of the disease in which there are no periods of remission. Diagnosing MS is very difficult and generally is delayed until
multiple episodes occur with deficits separated in time and space. Nerve conduction tests can detect slowed conduction in motor and
sensory pathways. Cerebral spinal fluid analysis can detect the presence of oligoclonal b ands indicative of an abnormal immune reaction
against myelin. The most definitive assessment is magnetic resonance imaging (MRI) to visualize multiple scarred (sclerotic) areas in
the brain. Although there is no cure for MS, some drugs (eg, β-interferon) that suppre ss the immune response reduce the severity and
slow the progression of the disease.
AXONAL TRANSPORT
Neurons are secretory cells, but they differ from other secretory cells in that the secreto ry zone is generally at the end of the axon, far
removed from the cell body. The apparatus for protein synthesis is located for the most part in the cell body, with transport of proteins
and polypeptides to the axonal ending by axoplasmic flow. Thus, the cell body maintains the function al and anatomic integrity of the
axon; if the axon is cut, the part distal to the cut degenerates (wallerian degeneration). Or thograde transport occurs along
microtubules that run along the length of the axon and requires two molecular motors, d ynein and kinesin (Figure 4–4). Orthograde
transport moves from the cell body toward the axon terminals. It has both fast and slow components; fast axonal transport occurs at
about 400 mm/day, and slow axonal transport occurs at 0.5 to 10 mm/day. Retrograde transpo rt, which is in the opposite direction
(from the
FIGURE 4–4 Axonal transport along microtubules by dynein and kinesin. Fast and slow axonal orthograde transport occurs along
microtubules that run along the length of the axon from the cell body to the terminal. Re trograde transport occurs from the terminal to
the cell body. (From Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology. McGraw -Hill, 2008.)
nerve ending to the cell body), occurs along microtubules at about 200 mm/day. Synap tic vesicles recycle in the membrane, but some
used vesicles are carried back to the cell body and deposited in lysosomes. Some materi als taken up at the ending by endocytosis,
including nerve growth factor (NGF) and various viruses, are also transported back to the cell body . A potentially important exception
to these principles seems to occur in some dendrites. In them, single strands of mRNA tr ansported from the cell body make contact with
appropriate ribosomes, and protein synthesis appears to create local protein domains.
EXCITATION & CONDUCTION
Nerve cells have a low threshold for excitation. The stimulus may be electrical, chemical , or mechanical. Two types of physicochemical
disturbances are produced: local, nonpropagated potentials called, depending on their lo cation, synaptic, generator, or electrotonic
potentials; and propagated potentials, the action potentials (or nerve impulses). These are the only electrical responses of neurons and
other excitable tissues, and they are the main language of the nervous system. They are due to changes in the conduction of ions across
the cell membrane that are produced by alterations in ion channels. The electrical events in neurons are rapid, being measured in
milliseconds (ms); and the potential changes are small, being measured in millivolts (mV).
The impulse is normally transmitted (conducted) along the axon to its termination . Nerves are not “telephone wires” that transmit
impulses passively; conduction of nerve impulses, although rapid, is much slower than th at of electricity. Nerve tissue is in fact a
relatively poor passive conductor, and it would take a potential of many volts to produc e a signal of a fraction of a volt at the other end of
a meter-long axon in the absence of active processes in the nerve. Conduction is an act ive, self-propagating process, and the impulse
moves along the nerve at a constant amplitude and velocity. The process is often compa red to what happens when a match is applied to
one end of a trail of gunpowder; by igniting the powder particles immediately in front o f it, the flame moves steadily down the trail to its
end as it is extinguished in its progression.
Mammalian neurons are relatively small, but giant unmyelinated nerve cells exist in a num ber of invertebrate species. Such cells are
found, for example, in crabs (Carcinus), cuttlefish (Sepia), and squid (Loligo). The fu ndamental properties of neurons were first
determined in these species and then found to be similar in mammals. The neck region o f the muscular mantle of the squid contains
single axons up to 1 mm in diameter. The fundamental properties of these long axons a re similar to those of mammalian axons.
RESTING MEMBRANE POTENTIAL
When two electrodes are connected through a suitable amplifier and placed on the surf ace of a single axon, no potential difference is
observed. However, if one electrode is inserted into the interior of the cell, a constant potential difference is observed, with the inside
negative relative to the outside of the cell at rest. A membrane potential results from sepa ration of positive and negative charges across
the cell membrane (Figure 4–5). In neurons, the resting membrane potential is usually ab out –70 mV, which is close to the equilibrium
potential for K
+
(Figure 4–6).
In order for a potential difference to be present across a membrane lipid bilayer, two c onditions must be met. First, there must be an
unequal distribution of ions of one or more species across the membrane (ie, a concent ration gradient). Two, the membrane must be
permeable to one or more of these ion species. The permeability is provided by the exis tence of channels or pores in the bilayer; these
channels are usually permeable to a single species of ions. The resting membrane poten tial represents an equilibrium situation at which
the driving force for the membrane-permeant ions down their concentration gradients across the membrane is equal and opposite to the
driving force for these ions down their electrical gradients.
+
+
+
+
– + –
Equal
+,–
+
+
+ +
+
– –
+ – +– +
– –
+
+
+
+
+
+
+
+
+
Extracellular side
Cytoplasmic side +
– –
+
+
+
––
Equal
+
+
+ –
+
+,– – – – + +
+
+
– –
– +–
FIGURE 4–5 This membrane potential results from separation of positive and negati ve charges across the cell membrane. The
excess of positive charges (red circles) outside the cell and negative charges (blue circles ) inside the cell at rest represents a small
fraction of the total number of ions present. (From Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.).
(a)
+30
4
0
3 5
2 7 –70 1 6
Na
+
K
+
(b)
Gated Na
+
channel
Gated K
+
channel K
+
600
P
Na
300 P
K
50
1 0 1 234 Time (ms)
FIGURE 4–6 The changes in (a) membrane potential (mV) and (b) relative membrane permeability (P) to Na+ and K+ during an action
potential. (From Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology. McGraw-Hill, 2008.)
In neurons, the concentration of K
+
is much higher inside than outside the cell, while the reverse is the case for Na
+
. This concentration
difference is established by the Na
+
-K
+
ATPase. The outward K
+
concentration gradient results in passive movement of K
+
out of the
cell when K
+
-selective channels are open. Similarly, the inward Na
+
concentration gradient results in passive movement of Na
+
into the
cell when Na
+
-selective channels are open. Because there are more open K
+
channels than Na
+
channels at rest, the membrane
permeability to K
+
is greater. Consequently, the intracellular and extracellular K
+
concentrations are the prime determinants of the
resting membrane potential, which is therefore close to the equilibrium potential for K
+
. Steady ion leaks cannot continue forever
without eventually dissipating the ion gradients. This is prevented by the Na
+
-K
+
ATPase, which actively moves Na
+
and K
+
against
their electrochemical gradient.
IONIC FLUXES DURING
THE ACTION POTENTIAL
The cell membranes of nerves, like those of other cells, contain many different types of ion channels. Some of these are voltage-gated
and others are ligand-gated. It is the behavior of these channels, and particularly Na
+
and K
+
channels, which explains the electrical
events in nerves.
The changes in membrane conductance of Na
+
and K
+
that occur during the action potentials are shown in Figure 4–6. The conductance
of an ion is the reciprocal of its electrical resistance in the membrane and is a measure of the membrane permeability to that ion. In
response to a depolarizing stimulus, some of the voltage-gated Na
+
channels become active, and when the threshold potential is
reached, the voltage-gated Na
+
channels overwhelm the K
+
and other channels and an action potential results (a positive feedback loop).
The membrane potential moves toward the equilibrium potential for Na
+
(+60 mV) but does not reach it during the action potential,
primarily because the increase in Na
+
conductance is short-lived. The Na
+
channels rapidly enter a closed state called the inactivated
state and remain in this state for a few milliseconds before returning to the resting state , when they again can be activated. In addition,
the direction of the electrical gradient for Na
+
is reversed during the overshoot because the membrane po tential is reversed, and this
limits Na
+
influx. A third factor producing repolarization is the opening of voltage-gated K
+
channels. This opening is slower and more
prolonged than the opening of the Na
+
channels, and consequently, much of the increase in K
+
conductance comes after the increase in
Na
+
conductance. The net movement of positive charge out of the cell due to K
+
efflux at this time helps complete the process of
repolarization. The slow return of the K
+
channels to the closed state also explains the after-hyperpolarization, fol lowed by a return to
the resting membrane potential. Thus, voltage-gated K
+
channels bring the action potential to an end and cause closure of their gates
through a negative feedback process. Figure 4–7 shows the sequential feedba ck control in voltagegated K
+
and Na
+
channels during the
action potential.
Decreasing the external Na
+
concentration reduces the size of the action potential but has little effect on the resting m embrane potential.
The lack of much effect on the resting membrane potential would be predicted, since th e permeability of the membrane to Na
+
at rest is
relatively low. Conversely, increasing the external K
+
concentration decreases the resting membrane potential.
Although Na
+
enters the nerve cell and K
+
leaves it during the action potential, the number of ions involved is minute relative to the total
numbers present. The fact that the nerve gains Na
+
and loses K
+
during activity has been demonstrated experimentally, but significant
differences in ion concentrations can be measured only after prolonged, repeated stimu lation.
Other ions, notably Ca
2+
, can affect the membrane potential through both channel movement and membrane int eractions. A decrease in
extracellular Ca
2+
concentration increases the excitability of nerve and muscle cells by decreasing the amo unt of depolarization
necessary to initiate the changes in the Na
+
and K
+
conductance that produce the action potential. Conversely, an increase in
extracellular Ca
2+
concentration can stabilize the membrane by decreasing excitability.
DISTRIBUTION OF ION CHANNELS IN MYELINATED NEURONS
The spatial distribution of ion channels along the axon plays a key role in the initiation an d regulation of the action potential. Voltage-
gated Na
+
channels are highly concentrated in the nodes of Ranvier and the initial segment in mye linated neurons. The initial segment
and, in sensory neurons, the first node of Ranvier are the sites where impulses are nor mally generated, and the other nodes of Ranvier
are the sites to which the impulses jump during saltatory conduction. The number of Na
+
channels per square micrometer of membrane
in myelinated mammalian neurons has been estimated to be 50–75 in the cell body, 350– 500 in the initial segment, less than 25 on the
surface of the myelin, 2000–12,000 at the nodes of Ranvier, and 20–75 at the axon te rminals. Along the axons of unmyelinated neurons,
the number is about 110. In many myelinated neurons, the Na
+
channels are flanked by K
+
channels that are involved in repolarization.
“ALL-OR-NONE” LAW
It is possible to determine the minimal intensity of stimulating current (threshold intensity) that, acting fo r a given duration, will just
produce an action potential. The threshold intensity varies with the duration; with weak s timuli it is long, and with strong stimuli it is
short. The relation between the strength and the duration of a threshold stimulus is called the strength–duration curve. Slowly rising
currents fail to fire the nerve because the nerve adapts to the applied stimulus, a process called adaptation.
(a) Start
Depolarizing stimulus
Opening of voltage-gated Na
+
channels Stop
Inactivation
of Na
+
channels
+
Depolarization of membrane potential
Positive feedback
Increased P
Na
Increased flow of Na
+
into
the cell
(b) Start
Depolarization of membrane by Na
+
influx Opening of voltage-gated K
+
channels
Repolarization of membrane potential
Negative feedback
Increased P
K
Increased flow of K
+
out of the cell
FIGURE 4–7 Feedback control in voltage-gated ion channels in the membrane . (a) Na
+
channels exert positive feedback. (b) K
+
channels exert negative feedback. (From Widmaier EP, Raff H, Strang KT: Vander’s Huma n Physiology. McGraw-Hill, 2008.)
Once threshold intensity is reached, a full-fledged action potential is produced. Further increases in the intensity of a stimulus produce
no increment or other change in the action potential as long as the other experimental co nditions remain constant. The action potential
fails to occur if the stimulus is subthreshold in magnitude, and it occurs with constant am plitude and form regardless of the strength of
the stimulus if the stimulus is at or above threshold intensity. The action potential is therefo re “all or none” in character and is said to
obey the all-or-none law.
change that rises sharply and decays exponentially with time. The magnitude of this resp onse drops off rapidly as the distance between
the stimulating and recording electrodes is increased. Conversely, an anodal current pro duces a hyperpolarizing potential change of
similar duration. These potential changes are called electrotonic potentials. A s the strength of the current is increased, the response is
greater due to the increasing addition of a local response of the membrane (Figure 4–8). Finally, at 7–15 mV of depolarization (potential
of –55 mV), the firing level is reached and an action potential occurs.
ELECTROTONIC POTENTIALS, LOCAL RESPONSE, & FIRING LEVEL
Although subthreshold stimuli do not produce an action potential, they do have an effec t on the membrane potential. This can be
demonstrated by placing recording electrodes within a few millimeters of a stimulating el ectrode and applying subthreshold stimuli of
fixed duration. Application of such currents leads to a localized depolarizing potential
CHANGES IN EXCITABILITY DURING ELECTROTONIC POTENTIAL S & THE ACTION POTENTIAL
During the action potential, as well as during electrotonic potentials and the local respons e, the threshold of the neuron to stimulation
changes. Hyperpolarizing responses elevate the threshold, and depolarizing potentials lo wer it as they move
Propagated
action potential Spike
potential Firing level −55
Local response Resting membrane potential
70 −
0.5 1.0 1.5 ms
After-depolarization
After-hyperpolarization
Local
response Period of latent addition
Supernormal period −85
FIGURE 4–8 Electrotonic potentials and local response. The ch anges in the membrane potential of a neuron following application of
stimuli of 0.2, 0.4, 0.6, 0.8, and 1.0 times threshold intensity are shown super imposed on the same time scale. The responses below the
horizontal line are those recorded near the anode, and the responses above the line are those recorded near the cathode. The stimulus of
threshold intensity was repeated twice. Once it caused a propagated action potential (top line), an d once it did not.
Refractory period
Subnormal period
Time
FIGURE 4–9 Relative changes in excitability of a nerve cell membra ne during the passage of an impulse. Note that excitability is
the reciprocal of threshold. (Modified from Morgan CT: Physiological Psychology. McGraw-Hill, 1943.)
the membrane potential closer to the firing level. During the local response, the threshold is lowered, but during the rising and much of
the falling phases of the spike potential, the neuron is refractory to stimulation. This re fractory period is divided into an absolute
refractory period, corresponding to the period from the time the firing level is reached until repolarization is about one-third complete,
and a relative refractory period, lasting from this point to the start of afterdepolarization. During the absolute re fractory period, no
stimulus, no matter how strong, will excite the nerve, but during the relative refractory p eriod, stronger than normal stimuli can cause
excitation. During after-depolarization, the threshold is again decreased, and during aft er-hyperpolarization, it is increased. These
changes in threshold are correlated with the phases of the action potential in Figure 4–9 .
SALTATORY CONDUCTION
Conduction in myelinated axons depends on a similar pattern of circular current flow. H owever, myelin is an effective insulator, and
current flow through it is negligible. Instead, depo
ECF ++ ++ –– ++ + – – – – + + – – – Axon
– – – – + + – – – ++ ++ –– ++ +
ELECTROGENESIS OF
THE ACTION POTENTIAL
The nerve cell membrane is polarized at rest, with positive charges lined up along the ou tside of the membrane and negative charges
along the inside. During the action potential, this polarity is abolished and for a brief per iod is actually reversed (Figure 4–10). Positive
charges from the membrane ahead of and behind the action potential flow into the area of negativity represented by the action potential
(“current sink”). By drawing off positive charges, this flow decreases the polarity of th e membrane ahead of the action potential. Such
electrotonic depolarization initiates a local response, and when the firing level is reached , a propagated response occurs that in turn
electrotonically depolarizes the membrane in front of it.
Active Inactive node node
ECF _ +
Myelin
+
_
Axon + _ _ +
Direction of propagation
FIGURE 4–10 Local current flow (movement of positive charges) around an im pulse in an axon. Top: Unmyelinated axon.
Bottom: Myelinated axon. Positive charges from the membrane ahead of and behind the actio n potential flow into the area of negativity
represented by the action potential (“current sink”). In myelinated axons, depolarization jumps from one node of Ranvier to the next
(salutatory conduction).
larization in myelinated axons jumps from one node of Ranvier to the next, with the cur rent sink at the active node serving to
electrotonically depolarize the node ahead of the action potential to the firing level (Figu re 4–10). This jumping of depolarization from
node to node is called saltatory conduction. It is a rapid process that allows myelinated axons t o conduct up to 50 times faster than the
fastest unmyelinated fibers.
+ + − + + + + + + + +
_ _ + _ _ _ _ _ _ _ _
+ + + + − + + + + + +
_ _ _ _ + _ _ _ _ _ _
ORTHODROMIC & ANTIDROMIC
CONDUCTION
An axon can conduct in either direction. When an action potential is initiated in the midd le of it, two impulses traveling in opposite
directions are set up by electrotonic depolarization on either side of the initial current sin k. In the natural situation, impulses pass in one
direction only, ie, from synaptic junctions or receptors along axons to their termination. Such conduction is called orthodromic.
Conduction in the opposite direction is called antidromic. Because synapses, unlike ax ons, permit conduction in one direction only, an
antidromic impulse will fail to pass the first synapse they encounter and die out at that po int.
BIPHASIC ACTION POTENTIALS
The descriptions of the resting membrane potential and action potential outlined above a re based on recording with two electrodes, one in
The descriptions of the resting membrane potential and action potential outlined above a re based on recording with two electrodes, one in
the extracellular space and the other inside it. If both recording electrodes are placed on the surface of the axon, there is no potential
difference between them at rest. When the nerve is stimulated and an impulse is conduc ted past the two electrodes, a characteristic
sequence of potential changes results. As the wave of depolarization reaches the electro de nearest the stimulator, this electrode becomes
negative relative to the other electrode (Figure 4–11). When the impulse passes to the p ortion of the nerve between the two electrodes,
the potential returns to zero, and then, as it passes the second electrode, the first electrod e becomes positive relative to the second. It is
conventional to connect the leads in such a way that when the first electrode becomes n egative relative to the second, an upward
deflection is recorded. Therefore, the record shows an upward deflection followed by an isoelectric interval and then a downward
deflection. This sequence is called a biphasic action potential (Figure 4–11).
PROPERTIES OF MIXED NERVES
Peripheral nerves in mammals are made up of many axons bound together in a fibrous envelope called the epineurium. Potential
changes recorded extracellularly from such nerves therefore represent an algebraic su mmation of the all-or-none action potentials of
many axons. The thresholds of the individual axons in the nerve and their distance from the stimulat
+ + + + + + + + − + +
_ _ _ _ − _ _ _ + _ _
+ + + + + + + + + + −
_ _ _ _ _ _ _ _ _ _ +
Nerve
mV Time
FIGURE 4–11 Biphasic action potential. Both recording electrodes are on th e outside of the nerve membrane. It is conventional to
connect the leads in such a way that when the first electrode becomes negative relative t o the second, an upward deflection is recorded.
Therefore, the record shows an upward deflection followed by an isoelectric interval a nd then a downward deflection.
ing electrodes vary. With subthreshold stimuli, none of the axons are stimulated and no response occurs. When the stimuli are of
threshold intensity, axons with low thresholds fire and a small potential change is observ ed. As the intensity of the stimulating current is
increased, the axons with higher thresholds are also discharged. The electrical response increases proportionately until the stimulus is
strong enough to excite all of the axons in the nerve. The stimulus that produces excitati on of all the axons is the maximal stimulus, and
application of greater, supramaximal stimuli produces no further increase in the size of the observed potential.
NERVE FIBER TYPES & FUNCTION
After a stimulus is applied to a nerve, there is a latent period before the start of the action potential. This inte rval corresponds to the time
it takes the impulse to travel along the axon from the site of stimulation to the recording e lectrodes. Its duration is proportionate to the
distance between the stimulating and recording electrodes and inversely proportionate to the speed of conduction. If the duration of the
latent period and the distance between the stimulating and recording electrodes are know n, axonal conduction velocity can be
calculated.
Erlanger and Gasser divided mammalian nerve fibers into A, B, and C groups, further subdividing the A group into α, β, γ, and δ fibers.
In Table 4–1, the various fiber types are listed
TABLE 4–1 Nerve fiber types in mammalian nerve.
a
Fiber Type Function Fiber
Diameter (
μ
m) Conduction Spike Absolute Refractory Velocity (m/s) Duration (ms) Period (ms)
A
α
Proprioception; somatic motor 12–20 70–120
β
Touch, pressure 5–12 30–70 0.4–0.5 0.4–1
γ
Motor to muscle spindles 3–6 15–30
δ
Pain, cold, touch 2–5 12–30
B Preganglionic autonomic <3 3–15 1.2 1.2
C
Dorsal root Pain, temperature, some mechano-reception 0.4–1.2 0.5–2 2 2
Sympathetic Postganglionic sympathetic 0.3–1.3 0.7–2.3 2 2
a
A and B fibers are myelinated; C fibers are unmyelinated.
with their diameters, electrical characteristics, and functions. By comparing the neurolog ic deficits produced by careful dorsal root
section and other nerve-cutting experiments with the histologic changes in the nerves, th e functions and histologic characteristics of each
of the families of axons responsible for the various peaks of the compound action pote ntial have been established. In general, the greater
the diameter of a given nerve fiber, the greater its speed of conduction. The large axon s are concerned primarily with proprioceptive
sensation, somatic motor function, conscious touch, and pressure, while the smaller axo ns subserve pain and temperature sensations and
autonomic function. The dorsal root C fibers conduct some impulses generated by touc h and other cutaneous receptors in addition to
impulses generated by pain and temperature receptors.
Further research has shown that not all the classically described lettered components are homogeneous, and a numerical system (Ia, Ib,
II, III, IV) has been used by some physiologists to classify sensory fibers. Unfortunate ly, this has led to confusion. A comparison of the
number system and the letter system is shown in Table 4–2.
In addition to variations in speed of conduction and fiber diameter, the various classes o f fibers in peripheral nerves differ in their
sensitivity to hypoxia and anesthetics (Table 4–3). This fact has clinical as well as physio logic significance. Local anesthetics depress
transmission in the group C fibers before they affect group A touch fibers. Conversely , pressure on a nerve can cause loss of conduction
in large-diameter motor, touch, and pressure fibers while pain sensation remains relative ly intact. Patterns of this type are sometimes
seen in individuals who sleep with their arms under their heads for long periods, causin g compression of the nerves in the arms. Because
of the association of deep sleep with alcoholic intoxication, the syndrome is most commo n on weekends and has acquired the interesting
name Saturday night or Sunday morning paralysis.
NEUROTROPHINS
TROPHIC SUPPORT OF NEURONS
A number of proteins necessary for survival and growth of neurons have been isolate d and studied. Some of these neurotrophins are
products of the muscles or other structures that the neurons innervate, but others are pr oduced by astrocytes. These proteins bind to
receptors at the endings of a neuron. They are internalized and then transported by ret rograde transport to the neuronal cell body, where
they foster the production of proteins associated with neuronal development, growth, a nd survival. Other neurotrophins are produced in
neurons and transported in an anterograde fashion to the nerve ending, where they ma intain the integrity of the postsynaptic neuron.
TABLE 4–2 Numerical classification sometimes used for sensory neurons.
Number Fiber Origin Type
Ia Muscle spindle, annulo-spiral ending A α
Ib Golgi tendon organ A α
II Muscle spindle, flower-spray ending; touch, A β pressure
III Pain and cold receptors; some touch receptors A
δ
IV Pain, temperature, and other receptors Dorsal root C
TABLE 4–3 Relative susceptibility of mammalian A, B, and C nerve fibers to condu ction block produced by various agents.
Most Least Susceptibility to: Susceptible Intermediate Susceptible
Hypoxia B A C
Pressure A B C
Local anesthetics C B A
RECEPTORS
Four established neurotrophins and their three high-affinity receptors are listed in Table 4–4. Each of these trk receptors dimerizes, and
this initiates autophosphorylation in the cytoplasmic tyrosine kinase domains of the recep tors. An additional low-affinity NGF receptor
that is a 75-kDa protein is called p75
NTR
. This receptor binds all four of the listed neurotrophins with equal affinity. There is som e
evidence that it can form a heterodimer with trk A monomer and that the dimer has incr eased affinity and specificity for NGF. However,
it now appears that p75
NTR
receptors can form homodimers that in the absence of trk receptors cause apoptosis, a n effect opposite to the
usual growth-promoting and nurturing effects of neurotrophins.
ACTIONS
The first neurotrophin to be characterized was NGF, a protein growth factor that is necessary for the growth and maintenance of
sympathetic neurons and some sensory neurons. It is present in a broad spectrum of an imal species, including humans, and is found in
many different tissues. In male mice, there is a particularly high concentration in the sub mandibular salivary glands, and the level is
reduced by castration to that seen in females. The factor is made up of two α, two β, an d two γ subunits. The β subunits, each of which
has a molecular mass of 13,200 Da, have all the nerve growth-promoting activity, the α subunits have trypsinlike activity, and the γ
subunits are serine proteases. The function of the proteases is unknown. The structure of the β unit of NGF resembles that of insulin.
TABLE 4–4 Neurotrophins.
Neurotrophin Receptor
Nerve growth factor (NGF) trk A
Brain-derived neurotrophic factor (BDNF) trk B
Neurotrophin 3 (NT-3) trk C, less on trk A and trk B
Neurotrophin 4/5 (NT-4/5) trk B
CLINICAL BOX 4–2 Axonal Regeneration
Peripheral nerve damage is often reversible. Although the axon will degenerate distal to the damage, connective elements of the so-
called distal stump often survive. Axonal sprouting occurs from the proximal st ump, growing toward the nerve ending. This results
from growth-promoting factors secreted by Schwann cells that attract axons toward the distal stump. Adhesio n molecules of the
immunoglobulin superfamily (eg, NgCAM/L1) promote axon growth along cell membr anes and extracellular matrices. Inhibitory
molecules in the perineurium assure that the regenerating axons grow in a correct trajec tory. Denervated distal stumps are able to
upregulate production of neurotrophins that promote growth. Once the regenerate d axon reaches its target, a new functional connection
(eg, neuromuscular junction) is formed. Regeneration allows for considerable, althoug h not full, recovery. For example, fine motor
control may be permanently impaired because some motor neurons are guided to an in appropriate motor fiber. Nonetheless, recovery of
peripheral nerves from damage far surpasses that of central nerve pathways. The prox imal stump of a damaged axon in the CNS will
form short sprouts, but distant stump recovery is rare, and the damaged axons are unlik ely to form new synapses. This is because CNS
neurons do not have the growth-promoting chemicals needed for regeneration. In fac t, CNS myelin is a potent inhibitor of axonal
growth. In addition, following CNS injury several events—astrocytic prolifera tion, activation of microglia, scar formation,
inflammation, and invasion of immune cells—provide an inappropriate environment for regeneration. T hus, treatment of brain and
spinal cord injuries frequently focuses on rehabilitation rather than reversing the nerve damage. New research is aiming to identify ways
to initiate and maintain axonal growth, to direct regenerating axons to reconnect with the ir target neurons, and to reconstitute original
neuronal circuitry.
NGF is picked up by neurons and is transported in retrograde fashion from the ending s of the neurons to their cell bodies. It is also
present in the brain and appears to be responsible for the growth and maintenance of c holinergic neurons in the basal forebrain and
striatum. Injection of antiserum against NGF in newborn animals leads to near total destr uction of the sympathetic ganglia; it thus
produces an immunosympathectomy. There is evidence that the maintenance of neurons by NGF is due to a reduction in apoptosis.
Brain-derived neurotrophic factor (BDNF), neurotrophin 3
(NT-3), NT-4/5, and NGF each maintain a different pattern of neurons, although there is some overlap. Disruption of NT-3
by gene knockout causes a marked loss of cutaneous mechanoreceptors, even in heter ozygotes. BDNF acts rapidly and can actually
depolarize neurons. BDNF-deficient mice lose peripheral sensory neurons and have se vere degenerative changes in their vestibular
ganglia and blunted long-term potentiation.
OTHER FACTORS AFFECTING
NEURONAL GROWTH
The regulation of neuronal growth is a complex process. Schwann cells and astrocytes produce ciliary neurotrophic factor (CNTF).
This factor promotes the survival of damaged and embryonic spinal cord neurons and may prove to be of value in treating human
diseases in which motor neurons degenerate. Glial cell line-derived neurotrophic facto r (GDNF) maintains midbrain dopaminergic
neurons in vitro. However, GDNF knockouts have dopaminergic neurons that appear normal, but they have no kidneys and fail to
develop an enteric nervous system. Another factor that enhances the growth of neuron s is leukemia inhibitory factor (LIF). In addition,
neurons as well as other cells respond to insulinlike growth factor I (IGF-I) an d the various forms of transforming growth factor
(TGF), fibroblast growth factor (FGF), and platelet-derived growth factor (PDGF).
Clinical Box 4–2 compares the ability to regenerate neurons after central and periphera l nerve injury.
CHAPTER SUMMARY
There are two main types of microglia and macroglia. Microglia are scavenger cells. M acroglia include oligodendrocytes, Schwann
cells, and astrocytes. The first two are involved in myelin formation; astrocytes produce substances that are tropic to neurons, and they
help maintain the appropriate concentration of ions and neurotransmitters.
Neurons are composed of a cell body (soma) which is the metabolic center of the neuron, dendrites that extend outward from the cell
body and arborize extensively, and a long fibrous axon that originates from a somewhat thick ened area of the cell body, the axon hillock.
The axons of many neurons acquire a sheath of myelin, a protein–lipid complex th at is wrapped around the axon. Myelin is an effective
insulator, and depolarization in myelinated axons jumps from one node of Ranvier to the nex t, with the current sink at the active node
serving to electrotonically depolarize to the firing level the node ahead of the action po tential.
Orthograde transport occurs along microtubules that run the length of the axon and req uires molecular motors, dynein, and kinesin.
Two types of physicochemical disturbances occur in neurons: local, nonprop agated potentials (synaptic, generator, or electrotonic
potentials) and propagated potentials (action potentials).
In response to a depolarizing stimulus, voltage-gated Na
+
channels become active, and when the threshold potential is reached, an
action potential results. The membrane potential moves toward the equilibrium potential f or Na
+
. The Na
+
channels rapidly enter a
closed state (inactivated state) before returning to the resting state. The direction of the elec trical gradient for Na
+
is reversed during the
overshoot because the membrane potential is reversed, and this limits Na
+
influx. Voltage-gated K
+
channels open and the net movement
of positive charge out of the cell helps complete the process of repolarization. The slow return of the K
+
channels to the closed state
explains afterhyperpolarization, followed by a return to the resting membrane potential.
Nerve fibers are divided into different categories based on axonal diameter, cond uction velocity, and function.
Neurotrophins are produced by astrocytes and transported by retrograde transport to the neuronal cell body, where they foster the
production of proteins associated with neuronal development, growth, and survival.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. The distance from between one stimulating electrode to
recording electrode is 4.5 cm. When the axon is stimulated, the latent period is 1.5 ms. W hat is the conduction velocity of the axon? A)
15 m/s
B) 30 m/s
C) 40 m/s
D) 67.5 m/s
E) This cannot be determined from the information given.
2. Which of the following has the slowest conduction velocity? A) Aα fibers
B) Aβ fibers
C) Aγ fibers
D) B fibers
E) C fibers
3. A man falls into a deep sleep with one arm under his head. This arm is paraly zed when he awakens, but it tingles, and pain sensation
in it is still intact. The reason for the loss of motor function without loss of pain sensatio n is that in the nerves to his arm, A) A fibers are
more susceptible to hypoxia than B fibers. B) A fibers are more sensitive to pressure th an C fibers. C) C fibers are more sensitive to
pressure than A fibers. D) motor nerves are more affected by sleep than sensory nerv es. E) sensory nerves are nearer the bone than
motor nerves and
hence are less affected by pressure.
4. Which part of a neuron has the highest concentration of Na
+
channels per square millimeter of cell membrane?
A) dendrites
B) cell body near dendrites
C) initial segment
D) axonal membrane under myelin
E) none of the above
5. Which of the following statements about nerve growth factor is not true?
A) It is made up of three polypeptide subunits.
B) It facilitates the process of apoptosis.
C) It is necessary for the growth and development of the sympathetic nervous system.
D) It is picked up by nerves from the organs they innervate. E) It is present in the brain .
CHAPTER RESOURCES
Aidley DJ: The Physiology of Excitable Cells, 4th ed. Cambridge University Press , 1998.
Boron WF, Boulpaep EL: Medical Physiology, Elsevier, 2005.
Bradbury EJ, McMahon SB: Spinal cord repair strategies: Why do they work? Nat Rev Neurosc i 2006;7:644.
Catterall WA: Structure and function of voltage-sensitive ion channels. Science 1988; 242:6 49.
Hille B: Ionic Channels of Excitable Membranes, 3rd ed. Sinauer Associates, 2001.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural Science,4th ed. McG raw-Hill, 2000.
Nicholls JG, Martin AR, Wallace BG: From Neuron to Brain: A Cellular and Molecular App roach to the Function of the Nervous
System, 4th ed. Sinauer Associates, 2001.
Thuret S, Moon LDF, Gage FH: Therapeutic interventions after spinal cord injur y. Nat Rev Neurosci 2006;7:628.
Volterra A, Meldolesi J: Astrocytes, from brain glue to communication elements: T he revolution continues. Nat Rev Neurosci
2005;6:626.
Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology. McGraw-Hill, 2008.
Excitable Tissue: Muscle
CH APTER
5
OBJEC TIV ES
After studying this chapter, you should be able to:
Differentiate the major classes of muscle in the body.
Describe the molecular and electrical makeup of muscle cell excitation– contraction cou pling.
Define thick and thick filaments and how they slide to create contraction.
Differentiate the role(s) for Ca
2+
in skeletal, cardiac, and smooth muscle contraction.
Appreciate muscle cell diversity.
INTRODUCTION
Muscle cells, like neurons, can be excited chemically, electrically, and mechanically to pr oduce an action potential that is transmitted
along their cell membranes. Unlike neurons, they respond to stimuli by activating a cont ractile mechanism. The contractile protein
myosin and the cytoskeletal protein actin are abundant in muscle, where they are the pri mary structural components that bring about
contraction.
Muscle is generally divided into three types: skeletal, cardiac, and smooth , although smooth muscle is not a homogeneous single
category. Skeletal muscle makes up the great mass of the somatic musculature. It has we ll-developed cross-striations, does not normally
contract in the absence of nervous stimulation, lacks anatomic and functional connection s between individual muscle fibers, and is
generally under voluntary control. Cardiac muscle also has cross-striations, but it is func tionally syncytial and, although it can be
modulated via the autonomic nervous system, it can contract rhythmically in the absence of external innervation owing to the presence in
the myocardium of pacemaker cells that discharge spontaneously (see Chapter 30). Sm ooth muscle lacks cross-striations and can be
further subdivided into two broad types: unitary (or visceral) smooth muscle and multiu nit smooth muscle. The type found in most
hollow viscera is functionally syncytial and contains pacemakers that discharge irregular ly. The multiunit type found in the eye and in
some other locations is not spontaneously active and resembles skeletal muscle in graded contractile ability.
SKELETAL MUSCLE MORPHOLOGY
ORGANIZATION
Skeletal muscle is made up of individual muscle fibers that are the “building blocks” of th e muscular system in the same sense that the
neurons are the building blocks of the nervous system. Most skeletal muscles begin and end in tendons, and the muscle fibers are
arranged in parallel between the tendinous ends, so that the force of contraction of the units is additive. Each muscle fiber is a single cell
that is multinucleated, long, cylindrical, and surrounded by a cell membrane, the sarcolemma (Figure 5–1). There are no syncytial
bridges between cells. The muscle fibers are made up of myofibrils, which are divisible into individual filaments. These myofilaments
contain several proteins that together make up the contractile machinery of the skeletal m uscle.
93
A
Terminal cistern Transverse tubules
Sarcoplasmic reticulum
Sarcolemma
(muscle fiber membrane)
Filaments
Mitochondrion
Myofibril
B Z disk Sarcomere Z disk
C
Thin filament (F-actin)
Tropomyosin Troponin
Actin
Thick filament (myosin)
FIGURE 5–1 Mammalian skeletal muscle. A single muscle fi ber surrounded by its sarcolemma has been cut away to show individual
myofibrils. The cut surface of the myofibrils shows the arrays of thick and thin filamen ts. The sarcoplasmic reticulum with its transverse
(T) tubules and terminal cisterns surrounds each myofibril. The T tubules invaginate from the sa rcolemma and contact the myofibrils
twice in every sarcomere. Mitochondria are found between the myofibrils and a basal la mina surrounds the sarcolemma. (Reproduced with
permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill , 2000.)
The contractile mechanism in skeletal muscle largely depends on the proteins myosin-II, actin, tro pomyosin, and troponin. Troponin is
made up of three subunits: troponin I, troponin T, and trop onin C. Other important proteins in muscle are involved in main taining the
proteins that participate in contraction in appropriate structural relation to one another an d to the extracellular matrix.
A band I band H band Z line M line
FIGURE 5–2 Electron micrograph of human gastrocnemius muscle. The various bands and lines are identified at the top (
×
13,500).
(Courtesy of Walker SM, Schrodt GR.)
STRIATIONS
Differences in the refractive indexes of the various parts of the
muscle fiber are responsible for the characteristic cross-stria
tions seen in skeletal muscle when viewed under the micro
scope. The parts of the cross-striations are frequently
identified by letters (Figure 5–2). The light I band is divided by
the dark Z line, and the dark A band has the lighter H band in
its center. A transverse M line is seen in the middle of the H
band, and this line plus the narrow light areas on either side of
it are sometimes called the pseudo-H zone. The area between
two adjacent Z lines is called a sarcomere. The orderly arrange
ment of actin, myosin, and related proteins that produces this
pattern is shown in Figure 5–3. The thick filaments, which are
about twice the diameter of the thin filaments, are made up of myosin; the thin filaments are made up of actin, tropomyosin, and
troponin. The thick filaments are lined up to form the A bands, whereas the array of th in filaments extends out of the A band and into the
less dense staining I bands. The lighter H bands in the center of the A bands are the reg ions where, when the muscle is relaxed, the thin
filaments do not overlap the thick filaments. The Z lines allow for anchoring of the thin filaments. If a transverse section through the A
band is examined under the electron microscope, each thick filament is seen to be surro unded by six thin filaments in a regular hexagonal
pattern.
The form of myosin found in muscle is myosin-II, with two globular heads and a long tail. The heads of the myosin
Sarcomere A band
Actin
Myosin
Actin
Z line Relaxed Z line Contracted
Z line Thick Thin AB
M line
Tropomyosin IC
T
Troponin IC T
Actin
Actin Myosin
CD FIGURE 5–3 A) Arrangement of thin (actin) and thick (myosin) filaments in skeletal muscle (compare to Figure 5–2). B) Sliding of
actin on myosin during contraction so that Z lines move closer together. C) Detail of re lation of myosin to actin in an individual
sarcomere, the functional unit of the muscle. D) Diagrammatic representation of the ar rangement of actin, tropomyosin, and troponin of
the thin filaments in relation to a myosin thick filament. The globular heads of myo sin interact with the thin filaments to create the
contraction. Note that myosin thick filaments reverse polarity at the M line in th e middle of the sarcomere, allowing for contraction. (C
and D are modified with permision from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science , 4th ed. McGraw-Hill, 2000.)
molecules form cross-bridges with actin. Myosin contains heavy chains and light chains , and its heads are made up of the light chains
and the amino terminal portions of the heavy chains. These heads contain an actin-bind ing site and a catalytic site that hydrolyzes ATP.
The myosin molecules are arranged symmetrically on either side of the center of the sar comere, and it is this arrangement that creates the
light areas in the pseudo-H zone. The M line is the site of the reversal of polarity of the myosin molecules in each of the thick filaments.
At these points, there are slender cross-connections that hold the thick filaments in prope r array. Each thick filament contains several
hundred myosin molecules.
The thin filaments are polymers made up of two chains of actin that form a long double helix. Tropomyosin molecules are long filaments
located in the groove between the two chains in the actin (Figure 5–3). Each thin filame nt contains 300 to 400 actin molecules and 40 to
60 tropomyosin molecules. Troponin molecules are small globular units located at interv als along the tropomyosin molecules. Each of
the three troponin subunits has a unique function: Troponin T binds the troponin compo nents to tropomyosin; troponin I inhibits the
interaction of myosin with actin; and troponin C contains the binding sites for the Ca
2+
that helps to initiate contraction.
Some additional structural proteins that are important in skeletal muscle function include actinin, titin, and desmin. Actinin binds actin
to the Z lines. Titin, the largest known protein (with a molecular mass near 3,000,000 D a), connects the Z lines to the M lines and
provides scaffolding for the sarcomere. It contains two kinds of folded domains that pr ovide muscle with its elasticity. At first when the
muscle is stretched there is relatively little resistance as the domains unfold, but with furth er stretch there is a rapid increase in resistance
that protects the structure of the sarcomere. Desmin adds structure to the Z lines in part b y binding the Z lines to the plasma membrane.
Although these proteins are important in muscle structure/ function, by no means do the y represent an exhaustive list.
SARCOTUBULAR SYSTEM
The muscle fibrils are surrounded by structures made up of membranes that appear in electron photomicrographs as vesicles and tubules.
These structures form the sarcotubular system, which is made up of a T system and a sarcoplasmic reticulum. The T system of
transverse tubules, which is continuous with the sarcolemma of the muscle fiber, forms a grid perforated by the individual muscle fibrils
(Figure 5–1). The space between the two layers of the T system is an extension of the e xtracellular space. The sarcoplasmic reticulum,
which forms an irregular curtain around each of the fibrils, has enlarged terminal ciste rns in close contact with the T system at the
junctions between the A and I bands. At these points of contact, the arrangement of the central T system with a cistern of the
sarcoplasmic reticulum on either side has led to the use of the term triads to describe the sy stem. The T system, which is continuous with
the sarcolemma, provides a path for the rapid transmission of the action potential from t he cell membrane to all the fibrils in the muscle.
The sarcoplasmic reticulum is an important store of Ca
2+
and also participates in muscle metabolism.
DYSTROPHIN–GLYCOPROTEIN COMPLEX
The large dystrophin protein (molecular mass 427,000 Da) forms a rod tha t connects the thin actin filaments to the transmembrane
protein β-dystroglycan in the sarcolemma by smaller proteins in the cytoplasm, syntrophins . β-dystroglycan is connected to merosin
(merosin refers to laminins that contain the α2 subunit in their trimeric makeup) in the ex tracellular matrix by α-dystroglycan (Figure 5–
4). The dystroglycans are in turn associated with a complex of four transmembrane gly coproteins: α-, β-, γ-, and δ-sarcoglycan. This
dystrophin–glycoprotein complex adds strength to the muscle by providing a sca ffolding for the fibrils and connecting them to the
extracellular environment. Disruption of the tightly choreographed structure can lead to several different pathologies, or muscular
dystrophies (see Clinical Box 5–1).
ELECTRICAL PHENOMENA
& IONIC FLUXES
ELECTRICAL CHARACTERISTICS
OF SKELETAL MUSCLE
The electrical events in skeletal muscle and the ionic fluxes that underlie them share distin ct similarities to those in nerve, with
quantitative differences in timing and magnitude. The resting membrane potential of ske letal muscle is about –90 mV. The action
potential lasts 2 to 4 ms and is conducted along the muscle fiber at about 5 m/s. The abso lute refractory period is 1 to 3 ms long, and the
after-polarizations, with their related changes in threshold to electrical stimulation, are rel atively prolonged. The initiation of impulses at
the myoneural junction is discussed in the next chapter.
ION DISTRIBUTION & FLUXES
The distribution of ions across the muscle fiber membrane is similar to that across the ne rve cell membrane. Approximate values for the
various ions and their equilibrium potentials are shown in Table 5–1. As in nerves, depo larization is largely a manifestation of Na
+
influx, and repolarization is largely a manifestation of K
+
efflux.
CONTRACTILE RESPONSES
It is important to distinguish between the electrical and mechanical events in skeletal musc le. Although one response
α
2
β1 γ1
Laminin 2
Functionally important
α
carbohydrate side chains Sarcoglycan
complex
Dystroglycans
δ
β
β
γ
α
Sarcospan Dystrophin
F-Actin
Syntrophins
FIGURE 5–4 The dystrophin–glycoprotein complex. Dystrophin co nnects F-actin to the two members of the dystroglycan (DG)
complex, α and β-dystroglycan, and these in turn connect to the merosin subunit of lamin in 211 in the extracellular matrix. The
sarcoglycan complex of four glycoproteins, α-, β-, γ-, and δ-sarcoglycan, sarcospan, and syn tropins are all associated with the
dystroglycan complex. There are muscle disorders associated with loss, abnormalities, or both o f the sarcoglycans and merosin.
(Reproduced with permission from Kandel ER, Scwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
does not normally occur without the other, their physiologic bases and characteristics ar e different. Muscle fiber membrane
depolarization normally starts at the motor end plate, the specialized structure under the m otor nerve ending. The action potential is
transmitted along the muscle fiber and initiates the contractile response.
THE MUSCLE TWITCH
A single action potential causes a brief contraction followed by relaxation. This respons e is called a muscle twitch. In Figure 5–5, the
action potential and the twitch are plotted on the same time scale. The twitch starts about 2 ms after the start of depolarization of the
membrane, before repolarization is complete. The duration of the twitch varies with the type of muscle being tested. “Fast” muscle
fibers, primarily those concerned with fine, rapid, precise movement, have twitch durat ions as short as 7.5 ms. “Slow” muscle fibers,
principally those involved in strong, gross, sustained movements, have twitch durations up to 100 ms.
MOLECULAR BASIS OF CONTRACTION
The process by which the contraction of muscle is brought about is a sliding of the thin filaments over the thick filaments. Note that this
shortening is not due to changes in the actual lengths of the thick and thin filaments, rath er, by their increased overlap within the muscle
cell. The width of the A bands is constant, whereas the Z lines move closer together wh en the muscle contracts and farther apart when it
relaxes (Figure 5–3).
The sliding during muscle contraction occurs when the myosin heads bind firmly to acti n, bend at the junction of the head with the neck,
and then detach. This “power stroke” depends on the simultaneous hydrolysis of ATP. Myosin-II molecules are dimers that have two
heads, but only one attaches to actin at any given time. The probable sequence of event s of the power stroke is outlined in Figure 5–6. In
resting muscle, troponin I is bound to actin and tropomyosin and covers the sites where myosin heads interact with actin. Also at rest, the
myosin head contains tightly bound ADP. Following an action potential cytosolic Ca
2+
is increased and free Ca
2+
binds to troponin C.
This binding results in a weakening of the troponin I interaction with actin and exposes the actin binding site for myosin to
text continues on p. 100
CLINICAL BOX 5–1
Disease of Muscle
Muscular Dystrophies
The term muscular dystrophy is applied to diseases that cause progressive weakness of skeletal muscle. About 50 such diseases have
been described, some of which include cardiac as well as skeletal muscle. They range f rom mild to severe and some are eventually fatal.
They have multiple causes, but mutations in the genes for the various components of the dystrophin–glycoprotein complex are a
prominent cause. The dystrophin gene is one of the largest in the body, and mutations c an occur at many different sites in it. Duchenne
muscular dystrophy is a serious form of dystrophy in which the dystrophin protein is absen t from muscle. It is X-linked and usually
fatal by the age of 30. In a milder form of the disease, Becker muscular dystrop hy, dystrophin is present but altered or reduced in
amount. Limb-girdle muscular dystrophies of various types are associated with mutation s of the genes coding for the sarcoglycans or
other components of the dystrophin–glycoprotein complex.
Metabolic Myopathies
Mutations in genes that code for enzymes involved in the metabolism of carbohydrates, fats, and proteins to CO
2
and H
2
O in muscle and
the production of ATP can cause metabolic myopathies (eg, McArdle syndrome) . Metabolic myopathies all
TABLE 5–1 Steady-state distribution of ions in the intracellular and extracellular compartm ents of mammalian skeletal muscle,
and the equilibrium potentials for these ions.
100
Concentration (mmol/L) 0
Ion
a
Intracellular Extracellular Fluid Fluid Equilibrium Potential (mV)
Na
+
12 145 +65 30
K
+
155 4 –95 0 H
+
13
×
10
–5
3.8
×
10
–5
–32
Cl
3.8 120 –90 0 5 10 15 20 25 ms
HCO3– 8 27 –32
A
155 0 …
Membrane potential = –90 mV
a
A
represents organic anions. The value for intracellular Cl
is calculated from the membrane potential, using the Nernst equation.
have in common exercise intolerance and the possibility of muscle breakdown due to ac cumulation of toxic metabolites.
Ion Channel Myopathies
In the various forms of clinical myotonia, muscle relaxation is prolonged after voluntary contraction. The molecular bases of myotonias
are due to dysfunction of channels that shape the action potential. Myotonia dystrophy is caused by an autosomal dominant mutation that
leads to overexpression of a K
+
channel (although the mutation is not at the K
+
channel). A variety of myotonias are associated with
mutations in Na
+
channels (eg, hyperkalemic periodic paralysis, paramyotonia congenita, or Na
+
channel congenita) or Cl
channels (eg,
dominant or recessive myotonia congenita).
Malignant hyperthermia is another disease related to dysfunctional muscle ion channels. Patients with malignant hyperthermia can
respond to general anesthetics such as halothane by eliciting rigidity in the muscles and a quick increase in body temperature. This
disease has been traced to a mutation in RyR, the Ca
2+
release channel in the sarcoplasmic reticulum. The mutation results in an
inefficient feedback mechanism to shut down Ca
2+
release after stimulation of the RyR, and thus, increased contractility and heat
generation.
FIGURE 5–5 The electrical and mechanical responses of a mammalian skel etal muscle fiber to a single maximal stimulus. The
electrical response (mV potential change) and the mechanical response (T, tension in ar bitrary units) are plotted on the same abscissa
(time). The mechanical response is relatively long-lived compared to the electrical resp onse that initiates contraction.
Troponin
Thin
filament
ADP Myosin
Actin
Tropomyosin Thick
filament
A
Ca
2+
Ca
2+
ADP
Exposed binding site
B
Longitudinal force
ADP
C
ATP
D
P i ADP
E FIGURE 5–6 Power stroke of myosin in skeletal muscle. A) At rest, myosin heads are bound to adenosine diphosphate and are said
to be in a “cocked” position in relation to the thin filament, which does not hav e Ca
2+
bound to the troponin–tropomyosin complex. B)
Ca
2+
bound to the troponin–tropomyosin complex induced a conformational change in the thin filame nt that allows for myosin heads to
cross-bridge with thin filament actin. C) Myosin heads rotate, move the attached actin and shorten t he muscle fiber, forming the power
stroke. D) At the end of the power stroke, ATP binds to a now exposed site, and causes a detac hment from the actin filament. E) ATP is
hydrolyzed into ADP and inorganic phosphate (P
i
) and this chemical energy is used to “re-cock” the myosin head. (Modified with permission
from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McG raw-Hill, 2000.)
allow for formation of myosin/actin cross-bridges. Upon formation of the cross-bridge , ADP is released, causing a conformational
change in the myosin head that moves the thin filament relative to the thick filament, com prising the crossbridge “power stroke.” ATP
quickly binds to the free site on the myosin, which leads to a detachment of the myosin h ead from the thin filament. ATP is hydrolyzed
and inorganic phosphate (P
i
) released, causing a “re-cocking” of the myosin head and completing the cycle. As lon g as Ca
2+
remains
elevated and sufficient ATP is available, this cycle repeats. Many myosin heads cycle at or near the same time, and they cycle
repeatedly, producing gross muscle contraction. Each power stroke shortens the sarcom ere about 10 nm. Each thick filament has about
500 myosin heads, and each head cycles about five times per second during a rapid co ntraction.
The process by which depolarization of the muscle fiber initiates contraction is called ex citation–contraction coupling. The action
potential is transmitted to all the fibrils in the fiber via the T system (Figure 5–7). It trigge rs the release of Ca
2+
from the terminal
cisterns, the lateral sacs of the sarcoplasmic reticulum next to the T system. Depolarization of the T tubule membrane activates the
sarcoplasmic reticulum via dihydropyridine receptors (DHPR), named for the drug dih ydropyridine, which blocks them (Figure 5–8).
DHPR are voltage-gated Ca
2+
channels in the T tubule membrane. In cardiac muscle, influx of Ca
2+
via these channels triggers the
release of Ca
2+
stored in the sarcoplasmic reticulum (calciuminduced calcium release) by activating the ryanodine receptor (RyR). The
RyR is named after the plant alkaloid ryanodine that was used in its discovery. It is a liga nd-gated Ca
2+
channel with Ca
2+
as its natural
ligand. In skeletal muscle, Ca
2+
entry from the extracellular fluid (ECF) by this route is not required for Ca
2+
release. Instead, the DHPR
that serves as the voltage sensor unlocks release of Ca
2+
from the nearby sarcoplasmic reticulum via physical interaction with the RyR.
The released Ca
2+
is quickly amplified through calciuminduced calcium release. Ca
2+
is reduced in the muscle cell by the sarcoplasmic
or endoplasmic reticulum Ca
2+
ATPase (SERCA) pump. The SERCA pump uses energy from ATP hydrolysis to rem ove Ca
2+
from the
cytosol back into the terminal cisterns, where it is stored until released by the next action potential. Once the Ca
2+
concentration outside
the reticulum has been lowered sufficiently, chemical interaction between myosin and ac tin ceases and the muscle relaxes. Note that ATP
provides the energy for both contraction (at the myosin head) and relaxation (via SERC A). If transport of Ca
2+
into the reticulum is
inhibited, relaxation does not occur even though there are no more action potentials; the resulting sustained contraction is called a
contracture.
TYPES OF CONTRACTION
Muscular contraction involves shortening of the contractile elements, but because muscl es have elastic and viscous elements in series
with the contractile mechanism, it is possible
Steps in contraction
a
Discharge of motor neuron
Release of transmitter (acetylcholine) at motor end-plate
Binding of acetylcholine to nicotinic acetylcholine receptors
Increased Na
+
and K
+
conductance in end-plate membrane
Generation of end-plate potential
Generation of action potential in muscle fibers
Inward spread of depolarization along T tubules
Release of Ca
2+
from terminal cisterns of sarcoplasmic reticulum and diffusion to thick and thin filaments
Binding of Ca
2+
to troponin C, uncovering myosin-binding sites on actin
Formation of cross-linkages between actin and myosin and sliding of thin on thick filam ents, producing movement
Steps in relaxation
Ca
2+
pumped back into sarcoplasmic reticulum
Release of Ca
2+
from troponin
Cessation of interaction between
actin and myosin
a
Steps 1–6 in contraction are discussed in Chapter 4. FIGURE 5–7 Flow of info rmation that leads to muscle contraction.
Dihydropyridine receptor A Extracellular space
+ + + +
TT
NH2
COOH Cytoplasm COOH
Pivot Recorder
Lumen of SR
B
To stimulator
Ryanodine receptor
FIGURE 5–8 Relation of the T tubule (TT) to the sarcoplasmic reticulum in C a
2+
transport. In skeletal muscle, the voltage-gated
dihydropyridine receptor in the T tubule triggers Ca
2+
release from the sarcoplasmic reticulum (SR) via the ryanodine receptor (RyR).
Upon sensing a voltage change, there is a physical interaction between the sarcolemmal-bound DHPR and the SR-bound RyR. This
interaction gates the RyR and allows for Ca
2+
release from the SR.
Force
transducer
Recorder
for contraction to occur without an appreciable decrease in the length of the whole mus cle (Figure 5–9). Such a contraction is called
isometric (“same measure” or length). Contraction against a constant load with a decrea se in muscle length is isotonic (“same tension”).
Note that because work is the product of force times distance, isotonic contractions do w ork, whereas isometric contractions do not. In
other situations, muscle can do negative work while lengthening against a constant weig ht.
SUMMATION OF CONTRACTIONS
The electrical response of a muscle fiber to repeated stimulation is like that of nerve. The fiber is electrically refractory only during the
rising phase and part of the falling phase of the spike potential. At this time, the contracti on initiated by the first stimulus is just
beginning. However, because the contractile mechanism does not have a refractory pe riod, repeated stimulation before relaxation has
occurred produces additional activation of the contractile elements and a response that is added to the contraction already present. This
phenomenon is
FIGURE 5–9 A) Muscle preparation arranged for recording isotonic contracti ons. B) Preparation arranged for recording isometric
contractions. In A, the muscle is fastened to a writing lever that swings on a pivot. In B, it is attached to an electronic transducer that
measures the force generated without permitting the muscle to shorten.
known as summation of contractions. The tension developed during summation is considerably greater th an that during the single
muscle twitch. With rapidly repeated stimulation, activation of the contractile mechanism o ccurs repeatedly before any relaxation has
occurred, and the individual responses fuse into one continuous contraction. Such a res ponse is called a tetanus (tetanic contraction). It
is a complete tetanus when no relaxation occurs between stimuli and an incomplete tetanus when periods of incomplete relaxation take
place between the summated stimuli. During a complete tetanus, the tension developed is about four times that developed by the
individual twitch contractions. The development of an incomplete and a complete tetanus in response to stimuli of increasing frequency
is shown in Figure 5–10.
FIGURE 5–10 Tetanus. Isometric tension of a single muscle fiber during continuou sly increasing and decreasing stimulation frequency.
Dots at the top are at intervals of 0.2 s. Note the development of incomplete and then co mplete tetanus as stimulation is increased, and
the return of incomplete tetanus, then full response, as stimulation frequency is de creased.
The stimulation frequency at which summation of contractions occurs is determined by the twitch duration of the particular muscle being
studied. For example, if the twitch duration is 10 ms, frequencies less than 1/10 ms (100 /s) cause discrete responses interrupted by
complete relaxation, and frequencies greater than 100/s cause summation.
RELATION BETWEEN MUSCLE LENGTH & TENSION & VELOCITY O F CONTRACTION
Both the tension that a muscle develops when stimulated to contract isometrically (the total tension ) and the passive tension exerted by
the unstimulated muscle vary with the length of the muscle fiber. This relationship can be studied in a whole skeletal muscle preparation
such as that shown in Figure 5–9. The length of the muscle can be varied by changing the distance between its two attachments. At each
length, the passive tension is measured, the muscle is then stimulated electrically, and the t otal tension is measured. The difference
between the two values at any length is the amount of tension actually generated by the contractile process, the active tension. The
records obtained by plotting passive tension and total tension against muscle length are s hown in Figure 5–11. Similar curves are
obtained when single muscle fibers are studied. The length of the muscle at which the ac tive tension is maximal is usually called its
resting length. The term comes originally from experiments demonstrating that th e length of many of the muscles in the body at rest is
the length at which they develop maximal tension.
The observed length–tension relation in skeletal muscle is explained by the sliding filame nt mechanism of muscle contraction. When the
muscle fiber contracts isometrically, the tension developed is proportional to the number of crossbridges between the actin and the
myosin molecules. When muscle is stretched, the overlap between actin and myosin is re duced and the number of cross-linkages is
therefore reduced. Conversely, when the muscle is appreciably shorter than resting len gth, the distance the thin filaments can move is
reduced.
The velocity of muscle contraction varies inversely with the load on the muscle. At a giv en load, the velocity is maximal at the resting
length and declines if the muscle is shorter or longer than this length.
FIBER TYPES
Although skeletal muscle fibers resemble one another in a general way, skeletal muscle is a heterogeneous tissue made up of fibers that
vary in myosin ATPase activity, contractile speed, and other properties. Muscles are fre quently classified into two types, “slow” and
“fast.” These muscles can contain a mixture of three fiber types: type I (or SO for slow -oxidative); type IIA (FOG for fast-oxidative-
glycolytic); or type IIB (FG for fast glycolytic). Some of the properties associated with type I, type IIA, and type IIB fibers are
summarized in Table 5–2. Although this classification scheme is valid for muscles across many mammalian species, there are significant
variations of fibers within and between muscles. For example, type I fibers in a given m uscle can be larger than type IIA fibers from a
different muscle in the same animal. Many of the differences in the fibers that make up muscles stem from differences in the proteins
within them. Most of these are encoded by multigene families. Ten different isoforms of the myosin heavy chains (MHCs) have been
characterized. Each of the two types of light chains also have isoforms. It appears that t here is only one form of actin, but multiple
isoforms of tropomyosin and all three components of troponin.
30
Resting length
20
Total tension
Active tension 10
ENERGY SOURCES & METABOLISM
Muscle contraction requires energy, and muscle has been called “a machine for conver ting chemical energy into mechanical work.” The
immediate source of this energy is ATP, and this is formed by the metabolism of carboh ydrates and lipids.
Passive tension
0 01 23 4 5
Increase in muscle length (cm)
FIGURE 5–11 Length–tension relationship for the human triceps muscle. The passive t ension curve measures the tension exerted by
this skeletal muscle at each length when it is not stimulated. The total tension curve represe nts the tension developed when the muscle
contracts isometrically in response to a maximal stimulus. The active tension is the difference b etween the two.
PHOSPHORYLCREATINE
ATP is resynthesized from ADP by the addition of a phosphate group. Some of the ene rgy for this endothermic reaction is supplied by
the breakdown of glucose to CO
2
and H
2
O, but there also exists in muscle another energy-rich phosphate compound that can su pply this
energy for short periods. This compound is phosphorylcreatine, which is hydrolyzed to cr eatine and phosphate groups with the release
of considerable energy (Figure 5–12). At rest, some ATP in the mitochondria transfers its phosphate to creatine, so that a
phosphorylcreatine
TABLE 5–2 Classification of fiber types in skeletal muscles.
Type 1 Type IIA Type IIB
Other names Fast, Oxidative, Glycolytic (FOG) Slow, Oxidative (SO) Fast, Glycolytic (FG )
Color Red Red White
Myosin ATPase Activity Fast Slow Fast
Ca
2+
-pumping capacity of sarcoplasmic reticulum High Moderate High
Diameter Large Small Large
Glycolytic capacity High Moderate High
Oxidative capacity Moderate High Low
Associated Motor Unit Type Fast Resistant to Fatigue (FR) Slow (S) Fast Fatigable (FF)
Membrane potential = –90 mV
Oxidative capacity Moderate High Low
store is built up. During exercise, the phosphorylcreatine is hydrolyzed at the junction b etween the myosin heads and actin, forming ATP
from ADP and thus permitting contraction to continue.
CARBOHYDRATE & LIPID BREAKDOWN
At rest and during light exercise, muscles utilize lipids in the form of free fatty acids as th eir energy source. As the intensity of exercise
increases, lipids alone cannot supply energy fast enough and so use of carbohydrate b ecomes the predominant component in the muscle
fuel mixture. Thus, during exercise, much of the energy for phosphorylcreatine and A TP resynthesis comes from the breakdown of
glucose to CO
2
and H
2
O. Glucose in the bloodstream enters cells, where it is degraded through a series of che mical reactions to
pyruvate. Another source of intracellular glucose, and consequently of pyruvate, is gly cogen, the carbohydrate polymer that is especially
abundant in liver and skeletal muscle. When adequate O
2
is present, pyruvate enters the citric acid cycle and is metabolized—through
this cycle and the so-called respiratory enzyme pathway—to CO
2
and H
2
O. This process is called aerobic glycolysis. The metabolism of
glucose or glycogen to CO
2
and H
2
O forms large quantities of ATP from ADP. If O
2
supplies are insufficient, the pyruvate formed from
glucose does not enter the tricarboxylic acid cycle but is reduced to lactate. This process of anaerobic glycolysis is associated with the
net production of much smaller quantities of energy-rich phosphate bonds, but it does n ot require the presence of O
2
. A brief overview of
the various reactions involved in supplying energy to skeletal muscle is shown in Figure 5–13.
ATP + H
2
O ADP + H
3
PO
4
+ 7.3 kcal H
2
N Rest HN PO
3
Phosphorylcreatine + ADP Creatine + ATP H
2
N
+ —
C + ATP H
2
N
+—
C +
ADP Glucose + 2 ATP (or glycogen + 1 ATP) CH
3
NCH
2
COO
Exercise CH
3
NCH
2
COO
Creatine Phosphorylcreatine
Anaerobic
2 Lactic acid + 4 ATP Glucose + 2 ATP (or glycogen + 1 ATP)
HN C
O Oxygen 6 CO
2
+ 6 H
2
O + 40 ATP
HN
C
CH
3
N CH
2
Creatinine
FIGURE 5–12 Creatine, phosphorylcreatine, and creatinine cycling in muscle. During periods of high activity, cycling of
phosphorylcreatine allows for quick release of ATP to sustain muscle activity.
FIGURE 5–13
by hydrolysis of 1 mol of ATP and reactions responsible for resynthesis of ATP . The amount of ATP formed per mole of free fatty acid
(FFA) oxidized is large but varies with the size of the FFA. For example, complete oxid ation of 1 mol of palmitic acid generates 140 mol
of ATP. FFA Oxygen CO
2
+ H
2
O + ATP
ATP turnover in muscle cells. Energy released
THE OXYGEN DEBT MECHANISM
During exercise, the muscle blood vessels dilate and blood flow is increased so that the a vailable O
2
supply is increased. Up to a point,
the increase in O
2
consumption is proportional to the energy expended, and all the energy needs are met by aerobic processes. However,
when muscular exertion is very great, aerobic resynthesis of energy stores cannot keep pace with their utilization. Under these
conditions, phosphorylcreatine is still used to resynthesize ATP. In addition, some ATP s ynthesis is accomplished by using the energy
released by the anaerobic breakdown of glucose to lactate. Use of the anaerobic pathw ay is self-limiting because in spite of rapid
diffusion of lactate into the bloodstream, enough accumulates in the muscles to eventuall y exceed the capacity of the tissue buffers and
produce an enzyme-inhibiting decline in pH. However, for short periods, the presence of an anaerobic pathway for glucose breakdown
permits muscular exertion of a far greater magnitude than would be possible without it. For example, in a 100-m dash that takes 10 s,
85% of the energy consumed is derived anaerobically; in a 2-mi race that takes 10 min , 20% of the energy is derived anaerobically; and
in a longdistance race that takes 60 min, only 5% of the energy comes from anaerobic metabolism.
After a period of exertion is over, extra O
2
is consumed to remove the excess lactate, replenish the ATP and phosphorylcreatine sto res,
and replace the small amounts of O
2
that were released by myoglobin. The amount of extra O
2
consumed is proportional to the extent to
which the energy demands during exertion exceeded the capacity for the aerobic synth esis of energy stores, ie, the extent to which an
oxygen debt was incurred. The O
2
debt is measured experimentally by determining O
2
consumption after exercise until a constant, basal
consumption is reached and subtracting the basal consumption from the total. The amou nt of this debt may be six times the basal O
2
consumption, which indicates that the subject is capable of six times the exertion that wou ld have been possible without it.
RIGOR
When muscle fibers are completely depleted of ATP and phosphorylcreatine, they dev elop a state of rigidity called rigor. When this
occurs after death, the condition is called rigor mortis. In rigor, almost all of the myosin heads attach to actin but in an abnormal, fixed,
and resistant way.
storage in phosphate bonds is a small factor. Consequently, heat production is considera ble. The heat produced in muscle can be
measured accurately with suitable thermocouples.
Resting heat, the heat given off at rest, is the external manifestation of basal metabo lic processes. The heat produced in excess of resting
heat during contraction is called the initial heat. This is made up of activation heat, the heat that muscle produces whenever it is
contracting, and shortening heat, which is proportionate in amount to the distance the musc le shortens. Shortening heat is apparently
due to some change in the structure of the muscle during shortening.
Following contraction, heat production in excess of resting heat continues for as long as 30 min. This recovery heat is the heat liberated
by the metabolic processes that restore the muscle to its precontraction state. The recover y heat of muscle is approximately equal to the
initial heat; that is, the heat produced during recovery is equal to the heat produced duri ng contraction.
If a muscle that has contracted isotonically is restored to its previous length, extra heat in addition to recovery heat is produced
(relaxation heat). External work must be done on the muscle to return it to its previous length, and relaxation heat is mainly a
manifestation of this work.
PROPERTIES OF SKELETAL
MUSCLES IN THE
INTACT ORGANISM
EFFECTS OF DENERVATION
In the intact animal or human, healthy skeletal muscle does not contract except in respon se to stimulation of its motor nerve supply.
Destruction of this nerve supply causes muscle atrophy. It also leads to abnormal excita bility of the muscle and increases its sensitivity to
circulating acetylcholine (denervation hypersensitivity; see Chapter 6). Fine, irregular co ntractions of individual fibers (fibrillations)
appear. This is the classic picture of a lower motor neuron lesion. If the mo tor nerve regenerates, the fibrillations disappear. Usually,
the contractions are not visible grossly, and they should not be confused with fasciculations, which are jerky, visible contractions of
groups of muscle fibers that occur as a result of pathologic discharge of spinal motor n eurons.
HEAT PRODUCTION IN MUSCLE
Thermodynamically, the energy supplied to a muscle must equal its energy output. The energy output appears in work done by the
muscle, in energy-rich phosphate bonds formed for later use, and in heat. The overall mechanical efficiency of skeletal muscle (work
done/total energy expenditure) ranges up to 50% while lifting a weight during isotonic contraction and is essentially 0% during isometric
contraction. Energy
THE MOTOR UNIT
Because the axons of the spinal motor neurons supplying skeletal muscle each branch t o innervate several muscle fibers, the smallest
possible amount of muscle that can contract in response to the excitation of a single moto r neuron is not one muscle fiber but all the fibers
supplied by the neuron. Each single motor neuron and the muscle fibers it innervates co nstitute a motor unit. The number of muscle
fibers in a motor unit varies. In muscles such as those of the hand and those concerned with motion of the eye (ie, muscles concerned
with fine, graded, precise movement), each motor unit innervates very few (on the ord er of three to six) muscle fibers. On the other hand,
values of 600 muscle fibers per motor unit can occur in human leg muscles. The group of muscle fibers that contribute to a motor unit
can be intermixed within a muscle. That is, although they contract as a unit, they are not necessarily “neighboring” fibers within the
muscle.
Each spinal motor neuron innervates only one kind of muscle fiber, so that all the musc le fibers in a motor unit are of the same type. On
the basis of the type of muscle fiber they innervate, and thus on the basis of the duration of their twitch contraction, motor units are
divided into S (slow), FR (fast, resistant to fatigue), and FF (fast, fatigable) units. Interes tingly, there is also a gradation of innervation of
these fibers, with S fibers tending to have a low innervation ratio (ie, small units) and FF fibers tending to have a high innervation ratio
(ie, large units). The recruitment of motor units during muscle contraction is not random , rather it follows a general scheme, the size
principle. In general, a specific muscle action is developed first by the recruitment of S muscle units that contract relatively slowly to
produce controlled contraction. Next, FR muscle units are recruited, resulting in more p owerful response over a shorter period of time.
Lastly, FF muscle units are recruited for the most demanding tasks. For example, in mus cles of the leg, the small, slow units are first
recruited for standing. As walking motion is initiated, their recruitment of FR units increa ses. As this motion turns to running or jumping,
the FF units are recruited. Of course, there is overlap in recruitment, but, in general, this principle holds true.
The differences between types of muscle units are not inherent but are determined by, among other things, their activity. When the nerve
to a slow muscle is cut and the nerve to a fast muscle is spliced to the cut end, the fast ne rve grows and innervates the previously slow
muscle. However, the muscle becomes fast and corresponding changes take place in its muscle protein isoforms and myosin ATPase
activity. This change is due to changes in the pattern of activity of the muscle; in stimulatio n experiments, changes in the expression of
MHC genes and consequently of MHC isoforms can be produced by changes in the p attern of electrical activity used to stimulate the
muscle. More commonly, muscle fibers can be altered by a change in activity initiated th rough exercise (or lack thereof). Increased
activity can lead to muscle cell hypertrophy, which allows for increase in contractile stre ngth. Type IIA and IIB fibers are most
susceptible to these changes. Alternatively, inactivity can lead to muscle cell atrophy and a loss of contractile strength. Type I fibers—
that is, the ones used most often—are most susceptible to these changes.
ELECTROMYOGRAPHY
Activation of motor units can be studied by electromyography, the process of recordin g the electrical activity of muscle on an
Biceps
500 μV
Triceps
0.5 s
FIGURE 5–14 Electromyographic tracings from human biceps and triceps muscles during alternate flexion and extension of the
elbow. Note the alternate activation and rest patterns as one muscle is used for flexio n and the other for extension. Electrical activity of
stimulated muscle can be recorded extracellularly, yielding typical excitation responses after stim ulation.
(Courtesy of Garoutte BC.)
oscilloscope. This may be done in unanaesthetized humans by using small metal disks on the skin overlying the muscle as the pick-up
electrodes or by using hypodermic needle electrodes. The record obtained with such el ectrodes is the electromyogram (EMG). With
needle electrodes, it is usually possible to pick up the activity of single muscle fibers. The measured EMG depicts the potential
difference between the two electrodes, which is altered by the activation of muscles in b etween the electrodes. A typical EMG is shown
in Figure 5–14.
It has been shown by electromyography that little if any spontaneous activity occurs in t he skeletal muscles of normal individuals at rest.
With minimal voluntary activity a few motor units discharge, and with increasing volunta ry effort, more and more are brought into play
to monitor the recruitment of motor units. Gradation of muscle res ponse is therefore in part a function of the number of motor units
activated. In addition, the frequency of discharge in the individual nerve fibers plays a role, the tension developed during a tetanic
contraction being greater than that during individual twitches. The length of the muscle i s also a factor. Finally, the motor units fire
asynchronously, that is, out of phase with one another. This asynchronous firing cause s the individual muscle fiber responses to merge
into a smooth contraction of the whole muscle. In summary, EMGs can be used to quic kly (and roughly) monitor abnormal electrical
activity associated with muscle responses.
THE STRENGTH OF SKELETAL MUSCLES
Human skeletal muscle can exert 3 to 4 kg of tension per square centimeter of cross-se ctional area. This figure is about the same as that
obtained in a variety of experimental animals and seems to be constant for mammalian sp ecies. Because many of the muscles in humans
have a relatively large cross-sectional area, the tension they can develop is quite large. T he gastrocnemius, for example, not only
supports the weight of the whole body during climbing but resists a force several times this great when the foot hits the ground during
running or jumping. An even more striking example is the gluteus maximus, which can exert a tension of 1200 kg. The total tension that
could be developed if all muscles in the body of an adult man pulled together is approx imately 22,000 kg (nearly 25 tons).
BODY MECHANICS
Body movements are generally organized in such a way that they take maximal advanta ge of the physiologic principles outlined above.
For example, the attachments of the muscles in the body are such that many of them are normally at or near their resting length when
they start to contract. In muscles that extend over more than one joint, movement at one joint may compensate for movement at another
in such a way that relatively little shortening of the muscle occurs during contraction. Ne arly isometric contractions of this type permit
development of maximal tension per contraction. The hamstring muscles extend from th e pelvis over the hip joint and the knee joint to
the tibia and fibula. Hamstring contraction produces flexion of the leg on the thigh. If th e thigh is flexed on the pelvis at the same time,
the lengthening of the hamstrings across the hip joint tends to compensate for the shorte ning across the knee joint. In the course of
various activities, the body moves in a way that takes advantage of this. Such factors as momentum and balance are integrated into body
movement in ways that make possible maximal motion with minimal muscular exertion. O ne net effect is that the stress put on tendons
and bones is rarely over 50% of their failure strength, protecting them from damage.
In walking, each limb passes rhythmically through a support or stance phase when the foot is on the ground and a swing phase when the
foot is off the ground. The support phases of the two legs overlap, so that two periods of double support occur during each cycle. There
is a brief burst of activity in the leg flexors at the start of each step, and then the leg is sw ung forward with little more active muscular
contraction. Therefore, the muscles are active for only a fraction of each step, and wal king for long periods causes relatively little
fatigue.
A young adult walking at a comfortable pace moves at a velocity of about 80 m/min an d generates a power output of 150 to 175 W per
step. A group of young adults asked to walk at their most comfortable rate selected a ve locity close to 80 m/min, and it was found that
they had selected the velocity at which their energy output was minimal. Walking more r apidly or more slowly took more energy.
CARDIAC MUSCLE MORPHOLOGY
The striations in cardiac muscle are similar to those in skeletal muscle, and Z lines are pre sent. Large numbers of elongated mitochondria
are in close contact with the muscle fibrils. The muscle fibers branch and interdigitate, bu t each is a complete unit surrounded by a cell
membrane. Where the end of one muscle fiber abuts on another, the membranes of bo th fibers parallel each other through an extensive
series of folds. These areas, which always occur at Z lines, are called intercalated d isks (Figure 5–15). They provide a strong union
between fibers, maintaining cell-to-cell cohesion, so that the pull of one contractile cell ca n be transmitted along its axis to the next.
Along the sides of the muscle fibers next to the disks, the cell membranes of adjacent fib ers fuse for considerable distances, forming gap
junctions. These junctions provide low-resistance bridges for the spread of excitation fr om one fiber to another. They permit cardiac
muscle to function as if it were a syncytium, even though no protoplasmic bridges are p resent between cells. The T system in cardiac
muscle is located at the Z lines rather than at the A–I junction, where it is located in mamm alian skeletal muscle.
ELECTRICAL PROPERTIES
RESTING MEMBRANE
& ACTION POTENTIALS
The resting membrane potential of individual mammalian cardiac muscle cells is about –8 0 mV. Stimulation produces a propagated
action potential that is responsible for initiating contraction. Although action potentials var y among the cardiomyocytes in different
regions of the heart (discussed in later chapters), the action potential of a typical ventricu lar cardiomyocyte can be used as an example
(Figure 5–16). Depolarization proceeds rapidly and an overshoot of the zero potential is present, as in skeletal muscle and nerve, but this
is followed by a plateau before the membrane potential returns to the baseline. In mamm alian hearts, depolarization lasts about 2 ms, but
the plateau phase and repolarization last 200 ms or more. Repolarization is therefore no t complete until the contraction is half over.
As in other excitable tissues, changes in the external K
+
concentration affect the resting membrane potential of cardiac muscle, whereas
changes in the external Na
+
concentration affect the magnitude of the action potential. The initial rapid depolarization and the overshoot
(phase 0) are due to opening of voltage-gated Na
+
channels similar to that occurring in nerve and skeletal muscle (Figure 5–17). The
initial rapid repolarization (phase 1) is due to closure of Na
+
channels and opening of one type of K
+
channel. The subsequent prolonged
plateau (phase 2) is due to a slower but prolonged opening of voltage-gated Ca
2+
channels. Final repolarization (phase 3) to the resting
membrane potential (phase 4) is due to closure of the Ca
2+
channels and a slow, delayed increase of K
+
efflux through various types of
K
+
channels. Cardiac myocytes contain at least two types of Ca
2+
channels (T- and Ltypes), but the Ca
2+
current is due mostly to
opening of the slower L-type Ca
2+
channels.
A
Intercalated disk
Nucleus 10 μm FIBER
2μm
Capillary N
Fibrils Sarcolemma
Sarcoplasmic reticulum
T system
Terminal cistern N
FIBRIL
B
Intercalated disk
SARCOMERE Mitochondria
Electron photomicrograph of cardiac muscle. Note the similarity of the A-I regions seen in the skeletal FIGURE 5–15 Cardiac muscle.
A)
muscle EM of Figure 3-2. The fuzzy thick lines are intercalated disks and function simil arly to the Z-lines but occur at cell membranes (
×
12,000). (Reproduced with permission from Bloom W, Fawcett DW: A Textbook of Histology, 10th ed. Saunde rs, 1975.) B) Artist interpretation of cardiac
muscle as seen under the light microscope (top) and the electron microscope (bo ttom). Again, note the similarity to skeletal muscle
structure. N, nucleus.
(Reproduced
with permission from Braunwald E, Ross J, Sonnenblick EH: Mechanisms of contraction of the normal and failing heart. N Engl
J Med 1967;277:794. Courtesy of Little, Brown.)
MECHANICAL PROPERTIES
CONTRACTILE RESPONSE
The contractile response of cardiac muscle begins just after the start of depolarization an d lasts about 1.5 times as long as the action
potential (Figure 5–16). The role of Ca
2+
in excitation– contraction coupling is similar to its role in skeletal muscle (see above).
However, it is the influx of extracellular Ca
2+
through the voltage-sensitive DHPR in the T system that triggers calcium-induced calciu m
release through the RyR at the sarcoplasmic reticulum. Because there is a net influx of C a
2+
during activation, there is also a more
prominent role for plasma membrane Ca
2+
ATPases and the Na
+
/Ca
2+
exchanger in recovery of intracellular Ca
2+
concentrations.
Specific effects
R
T
+20
1
Q
Action potential
S recorded with
surface electrode 2 0
0
Action potential recorded intracellularly
3
0.5 g 4 −90
INa
ARP
RRP
Mechanical response
0 100 200 300 ms
FIGURE 5–16 Comparison of action potentials and contractile response of a mammalian cardiac muscle fiber in a typical
ventricular cell. In the top-most trace, the most commonly viewed surface action potential recor ding can be seen and it is broken down
into four regions: Q, R, S, and T. In the middle trace, the intracellular recording of the action potential shows the quick depolarization
and extended recovery. In the bottom trace, the mechanical response is matched to th e extracellular and intracellular electrical activities.
Note that in the absolute refractory period (ARP), the cardiac myocyte cannot be excited , whereas in the relative refractory period (RRP)
minimal excitation can occur.
ICa
IK
0 200 Time (ms)
FIGURE 5–17 Dissection of the cardiac action potential. Top: The ac tion potential of a cardiac muscle fiber can be broken down into
several phases: 0, depolarization; 1, initial rapid repolarization; 2, plateau phase; 3, late ra pid repolarization; 4, baseline. Bottom:
Diagrammatic summary of Na
+
, Ca
2+
, and cumulative K
+
currents during the action potential. As is convention, inward currents are
downward, and outward currents are upward.
of drugs that indirectly alter Ca
2+
concentrations are discussed in Clinical Box 5–2.
During phases 0 to 2 and about half of phase 3 (until the membrane potential reaches a pproximately –50 mV during repolarization),
cardiac muscle cannot be excited again; that is, it is in its absolute refractory period. It remains re latively refractory until phase 4.
Therefore, tetanus of the type seen in skeletal muscle cannot occur. Of course, tetanizat ion of cardiac muscle for any length of time
would have lethal consequences, and in this sense, the fact that cardiac muscle cannot b e tetanized is a safety feature.
ISOFORMS
Cardiac muscle is generally slow and has relatively low ATPase activity. Its fibers are de pendent on oxidative metabolism and hence on
a continuous supply of O
2
. The human heart contains both the α and the β isoforms of the myosin heavy chain (α MHC and β MHC). β
MHC has lower myosin ATPase activity than α MHC. Both are present in the atria, with the α isoform predominating, whereas the β
isoform predominates in the ventricle. The spatial differences in expression contribute to the well-coordinated contraction of the heart.
CLINICAL BOX 5–2 Glycolysidic Drugs & Cardiac Contractions
Oubain and other digitalis glycosides are commonly used to treat failing hearts. These dr ugs have the effect of increasing the strength of
cardiac contractions. Although there is discussion as to full mechanisms, a working hyp othesis is based on the ability of these drugs to
inhibit the Na, K ATPase in cell membranes of the cardiomyocytes. The block of the Na , K ATPase in cardiomyocytes would result in
an increased intracellular Na
+
concentration. Such an increase would result in a decreased Na
+
influx and hence Ca
2+
efflux via the Na
+
-
Ca
2+
exchange antiport during the Ca
2+
recovery period. The resulting intracellular Ca
2+
concentration increase in turn increases the
strength of contraction of the cardiac muscle. With this mechanism in mind, these drugs can also be quite toxic. Overinhibition of the Na,
K ATPase would result in a depolarized cell that could slow conduction, or even sponta neously activate. Alternatively, overly increased
Ca
2+
concentration could also have ill effects on cardiomyocyte physiology.
CORRELATION BETWEEN MUSCLE FIBER LENGTH & TENSION
The relation between initial fiber length and total tension in cardiac muscle is similar to tha t in skeletal muscle; there is a resting length at
which the tension developed on stimulation is maximal. In the body, the initial length of th e fibers is determined by the degree of
diastolic filling of the heart, and the pressure developed in the ventricle is proportionate to the volume of the ventricle at the end of the
filling phase (Starling’s law of the heart). The developed tension (Figure 5–18) increases as the diasto lic volume increases until it
reaches a maximum, then tends to decrease. However, unlike skeletal muscle, the decre ase in developed tension at high degrees of
stretch is not due to a decrease in the number of cross-bridges between actin and myos in, because even severely dilated hearts are not
stretched to this degree. The decrease is due instead to beginning disruption of the myoc ardial fibers.
The force of contraction of cardiac muscle can be also increased by catecholamines, an d this increase occurs without a change in muscle
length. This positive ionotropic effect of catecholamines is mediated via innervated β
1
-adrenergic receptors, cyclic AMP, and their
effects on Ca
2+
homeostasis. The heart also contains noninnervated β
2
-adrenergic receptors, which also act via cyclic AMP, but their
ionotropic effect is smaller and is maximal in the atria. Cyclic AMP activates protein kina se A, and this leads to phosphorylation of the
voltage-dependent Ca
2+
channels, causing them to spend more time in the open state. Cyclic AMP also increases the active transport of
Ca
2+
to the sarcoplasmic reticulum, thus accelerating relaxation and consequently shortening systole. This is important when the cardiac
rate is increased because it permits adequate diastolic filling (see Chapter 31).
METABOLISM
Mammalian hearts have an abundant blood supply, numerous mitochondria, and a high content of myoglobin, a muscle pigment that can
function as an O
2
storage mechanism. Normally, less than 1% of the total energy liberated is provided by anaerobic metabolism. During
hypoxia, this figure may increase to nearly 10%; but under totally anaerobic conditions , the energy liberated is inadequate to sustain
ventricular contractions. Under basal conditions, 35% of the caloric needs of the huma n heart are provided by carbohydrate, 5% by
ketones and amino acids, and 60% by fat. However, the proportions of substrates utiliz ed vary greatly with the nutritional state. After
ingestion of large amounts of glucose, more lactate and pyruvate are used; during prolo nged starvation, more fat is used. Circulating free
fatty acids normally account for almost 50% of the lipid utilized. In untreated diabetics, t he carbohydrate utilization of cardiac muscle is
reduced and that of fat is increased.
SMOOTH MUSCLE MORPHOLOGY
Smooth muscle is distinguished anatomically from skeletal and cardiac muscle because it lacks visible cross-striations. Actin and
myosin-II are present, and they slide on each other to produce contraction. However, they are not arranged in regular arrays, as in
skeletal and cardiac muscle, and so the striations are absent. Instead of Z lines, there are dense bodies in the cytoplasm and attached to
the cell membrane, and these are bound by α-actinin to actin filaments. Smooth muscle a lso contains tropomyosin, but troponin appears
to be absent. The isoforms of actin and myosin differ from those in skeletal muscle. A s arcoplasmic reticulum is present, but it is less
extensive than those observed in skeletal or cardiac muscle. In general, smooth muscles contain few mitochondria and depend, to a large
extent, on glycolysis for their metabolic needs.
270
240
Systolic
intraventricular pressure
210
180
150
Developed tension 120
90
60
30
Diastolic
intraventricular pressure
0 10 20 30 40 50 60 70 Diastolic volume (mL)
FIGURE 5–18 Length–tension relationship for cardiac muscle. Comparison of the systolic intraventricular pressure (top trace) and
diastolic intraventricular pressure (bottom trace) display the developed tension in the car diomyocyte. Values shown are for canine heart.
TYPES
There is considerable variation in the structure and function of smooth muscle in differe nt parts of the body. In general, smooth muscle
can be divided into unitary (or visceral) smooth muscle and multiunit smooth muscle . Unitary smooth muscle occurs in large sheets,
has many low-resistance gap junctional connections between individual muscle cells, and functions in a syncytial fashion. Unitary
smooth muscle is found primarily in the walls of hollow viscera. The musculature of the intestine, the uterus, and the ureters are
examples. Multiunit smooth muscle is made up of individual units with few (or no) gap j unctional bridges. It is found in structures such
as the iris of the eye, in which fine, graded contractions occur. It is not under voluntary control, but it has many functional similarities to
skeletal muscle. Each multiunit smooth muscle cell has en passant endings of nerve fiber s, but in unitary smooth muscle there are en
passant junctions on fewer cells, with excitation spreading to other cells by gap junctions . In addition, these cells respond to hormones
and other circulating substances. Blood vessels have both unitary and multiunit smooth m uscle in their walls.
ELECTRICAL & MECHANICAL ACTIVITY
Unitary smooth muscle is characterized by the instability of its membrane potential and b y the fact that it shows continuous, irregular
contractions that are independent of its nerve supply. This maintained state of partial con traction is called tonus, or tone. The membrane
potential has no true “resting” value, being relatively low when the tissue is active and hi gher when it is inhibited, but in periods of
relative quiescence values for resting potential are on the order of –20 to –65 mV. Smo oth muscle cells can display divergent electrical
activity (eg, Figure 5–19). There are slow sine wave-like fluctuations a few millivolts in magnitude and spikes that sometimes overshoot
the zero potential line and sometimes do not. In many tissues, the spikes have a duration of about 50 ms, whereas in some tissues the
action potentials have a prolonged plateau during repolarization, like the action potentials in cardiac muscle. As in the other muscle
types, there are significant contributions of K
+
, Na
+
, and Ca
2+
channels and Na, K ATPase to this electrical activity. However,
discussion of contributions to individual smooth muscle types is beyond the scope of this text.
Because of the continuous activity, it is difficult to study the relation between the electrica l and mechanical events in unitary smooth
muscle, but in some relatively inactive preparations, a single spike can be generated. In such preparations the excitation–contraction
coupling in unitary smooth muscle can occur with as much as a 500-ms delay. Thus, it i s a very slow process compared with that in
skeletal and cardiac muscle, in which the time from initial depolarization to initiation of co ntraction is less than 10 ms. Unlike unitary
smooth muscle, multiunit smooth muscle is nonsyncytial and contractions do not spread w idely through it. Because of this, the
contractions of multiunit smooth muscle are more discrete, fine, and localized than those of unitary smooth muscle.
50 mV
4 s
FIGURE 5–19 Electrical activity of individual smooth muscle cells in the gu inea pig taenia coli. Left: Pacemaker-like activity with
spikes firing at each peak. Right: Sinusoidal fluctuation of membra ne potential with firing on the rising phase of each wave. In other
fibers, spikes can occur on the falling phase of sinusoidal fluctuations and there can be mixtures o f sinusoidal and pacemaker potentials
in the same fiber.
MOLECULAR BASIS OF CONTRACTION
As in skeletal and cardiac muscle, Ca
2+
plays a prominent role in the initiation of contraction of smooth muscle. However, the s ource of
Ca
2+
increase can be much different in unitary smooth muscle. Depending on the activating stimulus, Ca
2+
increase can be due to influx
through voltage- or ligand-gated plasma membrane channels, efflux from intracellular stores through the RyR, efflux from intracellular
stores through the inositol trisphosphate receptor (IP
3
R) Ca
2+
channel, or via a combination of these channels. In addition, the lack of
troponin in smooth muscle prevents Ca
2+
activation via troponin binding. Rather, myosin in smooth muscle must be phosphorylat ed for
activation of the myosin ATPase. Phosphorylation and dephosphorylation of myosin als o occur in skeletal muscle, but phosphorylation is
not necessary for activation of the ATPase. In smooth muscle, Ca
2+
binds to calmodulin, and the resulting complex activates
calmodulin-dependent myosin light chain kinase. This enzyme catalyzes the ph osphorylation of the myosin light chain on serine at
position 19. The phosphorylation increases the ATPase activity.
Myosin is dephosphorylated by myosin light chain phosphatase in the cell. However, dephosphorylation of myosin light chain kinase
does not necessarily lead to relaxation of the smooth muscle. Various mechanisms are in volved. One appears to be a latch bridge
mechanism by which myosin cross-bridges remain attached to actin for some time after the cytoplasmic Ca
2+
concentration falls. This
produces sustained contraction with little expenditure of energy, which is especially imp ortant in vascular smooth muscle. Relaxation of
the muscle presumably occurs when the Ca
2+
-calmodulin complex finally dissociates or when some other mechanism comes into play .
The events leading to contraction and relaxation of unitary smooth muscle are summariz ed in Figure 5–20. The events in multiunit
smooth muscle are generally similar.
Unitary smooth muscle is unique in that, unlike other types of muscle, it contracts when stretched in the absence of any extrinsic
innervation. Stretch is followed by a decline in membrane potential, an increase in the fr equency of spikes, and a general increase in tone.
If epinephrine or norepinephrine is added to a preparation of intestinal smooth muscle arranged for recording of intracellular potentials in
vitro, the membrane potential usually becomes larger, the spikes decrease in frequency, and the muscle relaxes (Figure 5–21).
Norepinephrine is the chemical mediator released at noradrenergic nerve endings, and stimulation of the noradrenergic nerves to the
preparation produces inhibitory potentials. Acetylcholine has an effect opposite to that o f norepinephrine on the membrane potential and
contractile activity of intestinal smooth muscle. If acetylcholine is added to the fluid bathin g a smooth muscle preparation in vitro, the
membrane potential decreases and the spikes become more frequent. The muscle becom es more active, with an increase in tonic tension
and the number of rhythmic contractions. The effect is mediated by phospholipase
CLINICAL BOX 5–3
Binding of acetylcholine to muscarinic receptors
Increased influx of Ca
2+
into the cell
Activation of calmodulin-dependent myosin light chain kinase
Phosphorylation of myosin
Increased myosin ATPase activity and binding of myosin to actin
Contraction
Dephosphorylation of myosin by myosin light chain phosphatase
Relaxation, or sustained contraction due to the latch bridge and other mechanisms
FIGURE 5–20 Sequence of events in contraction and relaxation of smooth muscl e. Flow chart illustrates many of the molecular
changes that occur from the initiation of contraction to its relaxation. Note the distinct dif ferences from skeletal and cardiac muscle
excitation.
Common Drugs That Act on Smooth Muscle
Overexcitation of smooth muscle in the airways, such as that observed during an asthm a attack, can lead to bronchoconstriction. Inhalers
that deliver drugs to the conducting airway are commonly used to offset this smooth mu scle bronchoconstriction, as well as other
symptoms in the asthmatic airways. The rapid effects of drugs in inhalers are related to smooth muscle relaxation. Rapid response inhaler
drugs (eg, ventolin, albuterol, sambuterol) frequently target β-adrenergic receptors in t he airway smooth muscle to elicit a relaxation.
Although these β-adrenergic receptor agonists targeting the smooth muscle do not treat all symptoms associated with bronchial
constriction (eg, inflammation and increased mucus), they are quick and frequently allo w for sufficient opening of the conducting airway
to restore airflow, and thus allow for other treatments to reduce airway obstruction.
Smooth muscle is also a target for drugs developed to increase blood flow. As discussed in the text, NO is a natural signaling molecule
that relaxes smooth muscle by raising cGMP. This signaling pathway is naturally down-r egulated by the action of phosphodiesterase
(PDE), which transforms cGMP into a nonsignaling form, GMP. The drugs sild enafil, tadalafil, and vardenafil are all specific inhibitors
of PDE V, an isoform found mainly in the smooth muscle in the corpus cavernosum of the penis (see Chapter 25). Thus, oral
administration of these drugs can block the action of PDE V, increasing blood flow in a very limited region in the body and offsetting
erectile dysfunction.
C, which produces IP
3
and allows for Ca
2+
release through IP
3
receptors. In the intact animal, stimulation of cholinergic nerves causes
release of acetylcholine, excitatory potentials, and increased intestinal contractions.
0 Acetylcholine, parasympathetic stimulation, cold, stretch
Like unitary smooth muscle, multiunit smooth muscle is very sensitive to circulating chem ical substances and is normally activated by
chemical mediators (acetylcholine and norepinephrine) released at the endings of its mo tor nerves. Norepinephrine in particular tends to
persist in the muscle and to cause repeated firing of the muscle after a single stimulus rat her than a single action potential. Therefore, the
contractile response produced is usually an irregular tetanus rather than a single twitch. When a single twitch response is obtained, it
resembles the twitch contraction of skeletal muscle except that its duration is 10 times as lo ng.
−50
Membrane potential
Epinephrine, sympathetic stimulation
FIGURE 5–21 Effects of various agents on the membrane potential of intestinal smoot h muscle. Drugs and hormones can alter
firing of smooth muscle action potentials by raising (top trace) or lowering (bottom trace) resting membrane potential.
RELAXATION
In addition to cellular mechanisms that increase contraction of smooth muscle, there are cellular mechanisms that lead to its relaxation
(Clinical Box 5–3). This is especially important in smooth muscle that surrounds the bloo d vessels to increase blood flow. It was long
known that endothelial cells that line the inside of blood cells could release a substance th at relaxed smooth muscle (endothelial derived
relaxation factor,
EDRF). EDRF was later identified as the gaseous second messenger molecule, nitric oxide (NO). NO produced in endothelial cells is
free to diffuse into the smooth muscle for its effects. Once in muscle, NO directly activa tes a soluble guanylate cyclase to produce
another second messenger molecule, cyclic guanosine monophosphate (cGMP). This molecule can activate cGMP-sp ecific protein
kinases that can affect ion channels, Ca
2+
homeostasis, or phosphatases, or all of those mentioned, that lead to smooth muscle rela xation
(see Chapters 7 and 33).
The consequences of plasticity can be demonstrated in humans. For example, the tensio n exerted by the smooth muscle walls of the
bladder can be measured at different degrees of distention as fluid is infused into the bl adder via a catheter. Initially, tension increases
relatively little as volume is increased because of the plasticity of the bladder wall. Howev er, a point is eventually reached at which the
bladder contracts forcefully (see Chapter 38).
FUNCTION OF THE NERVE
SUPPLY TO SMOOTH MUSCLE
The effects of acetylcholine and norepinephrine on unitary smooth muscle serve to emp hasize two of its important properties: (1) its
spontaneous activity in the absence of nervous stimulation, and (2) its sensitivity to chem ical agents released from nerves locally or
brought to it in the circulation. In mammals, unitary muscle usually has a dual nerve sup ply from the two divisions of the autonomic
nervous system. The function of the nerve supply is not to initiate activity in the muscle b ut rather to modify it. Stimulation of one
division of the autonomic nervous system usually increases smooth muscle activity, wher eas stimulation of the other decreases it.
However, in some organs, noradrenergic stimulation increases and cholinergic stimulati on decreases smooth muscle activity; in others,
the reverse is true.
FORCE GENERATION &
PLASTICITY OF SMOOTH MUSCLE
Smooth muscle displays a unique economy when compared to skeletal muscle. Despite a pproximately 20% of the myosin content and a
100-fold difference in ATP use when compared with skeletal muscle, they can generat e similar force per crosssectional area. One of the
tradeoffs of obtaining force under these conditions is the noticeably slower contractions when compared to skeletal muscle. There are
several known reasons for these noticeable changes, including unique isoforms of myo sin and contractile-related proteins expressed in
smooth muscle and their distinct regulation (discussed above). The unique architecture o f the smooth cell and its coordinated units also
likely contribute to these changes.
Another special characteristic of smooth muscle is the variability of the tension it exerts a t any given length. If a unitary smooth muscle
is stretched, it first exerts increased tension. However, if the muscle is held at the greater length after stretching, the tension gradually
decreases. Sometimes the tension falls to or below the level exerted before the muscle w as stretched. It is consequently impossible to
correlate length and developed tension accurately, and no resting length can be assigne d. In some ways, therefore, smooth muscle
behaves more like a viscous mass than a rigidly structured tissue, and it is this property th at is referred to as the plasticity of smooth
muscle.
CHAPTER SUMMARY
There are three main types of muscle cells: skeletal, cardiac, and smooth.
Skeletal muscle is a true syncytium under voluntary control. Skeletal muscles receiv e electrical stimuli from neurons to elicit
contraction: “excitation–contraction coupling.” Action potentials in muscle cells are devel oped largely through coordination of Na
+
, K
+
,
and Ca
2+
channels. Contraction in skeletal muscle cells is coordinated through Ca
2+
regulation of the actomyosin system that gives the
muscle its classic striated pattern under the microscope.
There are several different types of skeletal muscle fibers (I, IIA, IIB) t hat have distinct properties in terms of protein makeup and force
generation. Skeletal muscle fibers are arranged into motor units of like fibers within a m uscle. Skeletal motor units are recruited in a
specific pattern as the need for more force is increased.
Cardiac muscle is a collection of individual cells (cardiomyocytes) that are linked as a syncytium by gap junctional communication.
Cardiac muscle cells also undergo excitation– contraction coupling. Pacemaker cells in th e heart can initiate propagated action potentials.
Cardiac muscle cells also have a striated, actomyosin system that underlies contraction.
Smooth muscle cells are largely under control of the autonomic nervous system.
There are two broad categories of smooth muscle cells: unitary and multiunit. Unitary smooth muscle contraction is synchronized by
gap junctional communication to coordinate contraction among many cells. Multiunit smo oth muscle contraction is coordinated by motor
units, functionally similar to skeletal muscle.
Smooth muscle cells contract through an actomyosin system, but do not have w ell-organized striations. Unlike skeletal and cardiac
muscle, Ca
2+
regulation of contraction is primarily through phosphorylation–dephosphorylation reactions .
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed . 1. The action potential of skeletal muscle
A) has a prolonged plateau phase.
B) spreads inward to all parts of the muscle via the T tubules.
C) causes the immediate uptake of Ca
2+
into the lateral sacs of the sarcoplasmic reticulum.
D) is longer than the action potential of cardiac muscle.
E) is not essential for contraction.
2. The functions of tropomyosin in skeletal muscle include A) sliding on actin to produc e shortening.
B) releasing Ca
2+
after initiation of contraction.
C) binding to myosin during contraction.
D) acting as a “relaxing protein” at rest by covering up the sites where myosin bin ds to actin.
E) generating ATP, which it passes to the contractile mechanism.
3. The cross-bridges of the sarcomere in skeletal muscle are made
up of
A) actin.
B) myosin.
C) troponin.
D) tropomyosin.
E) myelin.
4. The contractile response in skeletal muscle
A) starts after the action potential is over.
B) does not last as long as the action potential.
C) produces more tension when the muscle contracts isometrically than when the muscle contracts isotonically.
D) produces more work when the muscle contracts isometrically than when the muscle contracts isotonically.
E) decreases in magnitude with repeated stimulation. 5. Gap junctions
A) are absent in cardiac muscle.
B) are present but of little functional importance in cardiac muscle.
C) are present and provide the pathway for rapid spread of excitation from one cardia c muscle fiber to another.
D) are absent in smooth muscle.
E) connect the sarcotubular system to individual skeletal muscle cells.
CHAPTER RESOURCES
Alberts B, et al: Molecular Biology of the Cell, 5th ed. Garland Science, 2007.
Fung YC: Biomechanics, 2nd ed. Springer, 1993.
Hille B: Ionic Channels of Excitable Membranes, 3rd ed. Sinaver Associates, 2001.
Horowitz A: Mechanisms of smooth muscle contraction. Physiol Rev 1996;76: 967.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural Science, 4th ed. Mc Graw-Hill, 2000.
Sperelakis N (editor): Cell Physiology Sourcebook, 3rd ed. Academic Press, 2001 .
This page intentionally left blank
Synaptic & Junctional Transmission
CH APTER
6
OBJEC TIV ES
After studying this chapter, you should be able to:
Describe the main morphologic features of synapses.
Distinguish between chemical and electrical transmission at synapses.
Define convergence and divergence in neural networks, and
discuss their implications.
Describe fast and slow excitatory and inhibitory postsynaptic potentials, outline the ionic fluxes that underlie them, and explain how
the potentials interact to generate action potentials.
Define and give examples of direct inhibition, indirect inhibition, presynaptic inhibition, and postsynaptic inhibition.
Describe the neuromuscular junction, and explain how action potentials in the m otor neuron at the junction lead to contraction of the
skeletal muscle.
Define and explain denervation hypersensitivity.
INTRODUCTION
The all-or-none type of conduction seen in axons and skeletal muscle has been discusse d in Chapters 4 and 5. Impulses are transmitted
from one nerve cell to another cell at synapses (Figure 6–1). These are the junctions where the axon or some other portion of one cell
(the presynaptic cell) terminates on the dendrites, soma, or axon of another neuron (Figure 6–2) or, in some cases, a muscle or gland cell
(the postsynaptic cell). Cell-to-cell communication occurs across either a chemical or electrical synapse. At chemical synapses, a
synaptic cleft separates the terminal of the presynaptic cell from the postsynaptic cell. An im pulse in the presynaptic axon causes
secretion of a chemical that diffuses across the synaptic cleft and binds to receptors on t he surface of the postsynaptic cell. This triggers
events that open or close channels in the membrane of the postsynaptic cell. In electrical synapses, the membranes of the presynaptic and
postsynaptic neurons come close together, and gap junctions form between the cells (se e Chapter 2). Like the intercellular junctions in
other tissues, these junctions form low-resistance bridges through which ions can pass w ith relative ease. There are also a few conjoint
synapses in which transmission is both electrical and chemical. Regardless of the type of synapse, transmission is not a simple jumping
of an action potential from the presynaptic to the postsynaptic cell. The effects of discha rge at individual synaptic endings can be
excitatory or inhibitory, and when the postsynaptic cell is a neuron, the summation of al l the excitatory and inhibitory effects determines
whether an action potential is generated. Thus, synaptic transmission is a complex proce ss that permits the grading and adjustment of
neural activity necessary for normal function. Because most synaptic transmission is che mical, consideration in this chapter is limited to
chemical transmission unless otherwise specified.
Transmission from nerve to muscle resembles chemical synaptic transmission from one neuron to another. The neuromuscular
junction, the specialized area where a motor nerve terminates on a skeletal muscle fib er, is the site of a stereotyped transmission
process. The contacts between autonomic neurons and smooth and cardiac muscle are less specialized, and transmission in these
locations is a more diffuse process. These forms of transmission are also considered in this chapter.
115
1
3 4
5
2
6
6
1
FIGURE 6–1 Synapses on a typical motor neuron. The neuron has dendrites (1), an axon (2), and a prominent nucleus (3). Note that
rough endoplasmic reticulum extends into the dendrites but not into the axon. Many diff erent axons converge on the neuron, and their
terminal boutons form axodendritic (4) and axosomatic (5) synapses. (6) Myelin sheath. (Reproduced with permission from Krstic RV:
Ultrastructure of the Mammalian Cell. Springer, 1979.)
SYNAPTIC TRANSMISSION:
FUNCTIONAL ANATOMY
TYPES OF SYNAPSES
The anatomic structure of synapses varies considerably in the different parts of the mam malian nervous system. The ends of the
presynaptic fibers are generally enlarged to form terminal boutons (synaptic kn obs) (Figure 6–2). In the cerebral and cerebellar cortex,
endings are commonly located on dendrites and frequently on dendritic spines, which are small knobs projecting from dendrites (Figure
6–3). In some instances, the terminal branches of the axon of the presynaptic neuron f orm a basket or net around the soma of the
postsynaptic cell (basket cells of the cerebellum and autonomic ganglia). In other locatio ns, they intertwine with the dendrites of the
postsynaptic cell (climbing fibers of the cerebellum) or end on the dendrites directly (ap ical dendrites of cortical pyramidal cells). Some
end on axons of postsynaptic neurons (axoaxonal endings). On average, each neuron divides to form over 2000 synaptic endings, and
because the human central nervous system (CNS) has 10
11
neurons, it follows that there are about 2 × 10
14
synapses. Ob
S
M
P
D
FIGURE 6–2 Electron photomicrograph of synaptic knob (S) ending on the s haft of a dendrite (D) in the central nervous system.
P, postsynaptic density; M, mitochondrion. (
×
56,000). (Courtesy of DM McDonald.)
viously, therefore, communication between neurons is extremely complex. It should be noted as well that synapses are dynamic
structures, increasing and decreasing in complexity and number with use and experienc e.
It has been calculated that in the cerebral cortex, 98% of the synapses are on dendrites and only 2% are on cell bodies. In the spinal cord,
the proportion of endings on dendrites is less; there are about 8000 endings on the den drites of a typical spinal neuron and about 2000 on
the cell body, making the soma appear encrusted with endings.
PRESYNAPTIC & POSTSYNAPTIC STRUCTURE & FUNCTION
Each presynaptic terminal of a chemical synapse is separated from the postsynaptic struc ture by a synaptic cleft that is 20 to 40 nm wide.
Across the synaptic cleft are many neurotransmitter receptors in the postsynaptic membr ane, and usually a postsynaptic thickening called
the postsynaptic density (Figures 6–2 and 6–3). The postsynaptic density is an o rdered complex of specific receptors, binding proteins,
and enzymes induced by postsynaptic effects.
Inside the presynaptic terminal are many mitochondria, as well as many membrane-encl osed vesicles, which contain neurotransmitters.
There are three kinds of synaptic vesicles: small, clear synaptic vesicles that c ontain acetylcholine, glycine, GABA, or glutamate; small
vesicles with a dense core that contain catecholamines; and large vesicles with a dense co re that contain
Presynaptic cell
Microtubules
Mitochondria
Clear vesicles
Postsynaptic density
Active zone
Dendritic spine
Axodendritic Postsynaptic cell
Dendrite
Axodendritic
Soma
Axosomatic
Axon
Axo-axonal
Axodendritic, axoaxonal, and axosomatic
FIGURE 6–3
synapses. Many presynaptic neurons terminate on dendritic spines, as sho wn at the top, but some also end directly on the shafts of
dendrites. Note the presence of clear and granulated synaptic vesicles in endings and clustering of clear vesicles at active zones.
neuropeptides. The vesicles and the proteins contained in their walls are synthesized in th e neuronal cell body and transported along the
axon to the endings by fast axoplasmic transport. The neuropeptides in the large dense- core vesicles must also be produced by the
protein-synthesizing machinery in the cell body. However, the small clear vesicles and t he small dense-core vesicles recycle in the nerve
ending. These vesicles fuse with the cell membrane and release transmitters through exo cytosis and are then recovered by endocytosis to
be refilled locally. In some instances, they enter endosomes and are budded off the end osome and refilled, starting the cycle over again.
The steps involved are shown in Figure 6–4. More commonly, however, the synaptic v esicle discharges its contents through a small hole
in the cell membrane, then the opening reseals rapidly and the main vesicle stays inside th e cell (kiss-and-run discharge). In this way, the
full endocytotic process is short-circuited.
The large dense-core vesicles are located throughout the presynaptic terminals that cont ain them and release their neuropeptide contents
by exocytosis from all parts of the terminal. On the other hand, the small vesicles are loc ated near the synaptic cleft and fuse to the
membrane, discharging their contents very rapidly into the cleft at areas of membrane th ickening called active zones (Figure 6–3). The
active zones contain many proteins and rows of calcium channels.
The Ca
2+
that triggers exocytosis of transmitters enters the presynaptic neurons, and transmitter re lease starts within 200 μs. Therefore, it
is not surprising that the voltage-gated Ca
2+
channels are very close to the release sites at the active zones. In addition, for the transm itter
to be effective on the postsynaptic neuron requires proximity of release to the postsyna ptic receptors. This orderly organization of the
synapse depends in part on neurexins, proteins bound to the membrane of the presyn aptic neuron that bind neurexin receptors in the
membrane of the postsynaptic neuron. In many vertebrates, neurexins are produced b y a single gene that codes for the α isoform.
However, in mice and humans they are encoded by three genes, and both α and β isof orms are produced. Each of the genes has two
regulatory regions and extensive alternative splicing of their mRNAs. In this way, over 1000 different neurexins are produced. This raises
the possibility that the neurexins not only hold synapses together, but also provide a mec hanism for the production of synaptic
specificity.
As noted in Chapter 2, vesicle budding, fusion, and discharge of contents with subsequ ent retrieval of vesicle membrane are fundamental
processes occurring in most, if not all, cells. Thus, neurotransmitter secretion at synapse s and the accompanying membrane retrieval are
specialized forms of the general processes of exocytosis and endocytosis. The details of the processes by which synaptic vesicles fuse
with the cell membrane are still being worked out. They involve the v-snare protein synaptobrevin in the v esicle membrane locking
with the t-snare protein syntaxin in the cell membrane; a multiprotein complex regulate d by small GTPases such as rab3 is also
involved in the process (Figure 6–5). The synapse begins in the presynaptic and not in the postsynaptic cell. The one-way gate at the
synapses is necessary for orderly neural function.
Clinical Box 6–1 describes the how neurotoxins can disrupt transmitter release in either the CNS or at the neuromuscular junction.
ELECTRICAL EVENTS IN
POSTSYNAPTIC NEURONS
EXCITATORY & INHIBITORY
POSTSYNAPTIC POTENTIALS
Penetration of an α-motor neuron is a good example of the techniques used to study po stsynaptic electrical activity. It is
NT Early endosome
NT uptake Budding Endosome fusion H+
Translocation Translocation
Docking
Plasma
membrane
ATP
Priming Fusion/
Endocytosis
exocytosis
4 Ca2+ Ca2+ ?
Synaptic cleft
Ca2+
FIGURE 6–4 Small synaptic vesicle cycle in presynaptic nerve terminals. Vesicles bud off the early endosome and then fill with
neurotransmitter (NT; top left). They then move to the plasma membrane, dock, and be come primed. Upon arrival of an action potential
at the ending, Ca
2+
influx triggers fusion and exocytosis of the granule contents to the synaptic cleft. The v esicle wall is then coated with
clathrin and taken up by endocytosis. In the cytoplasm, it fuses with the early e ndosome, and the cycle is ready to repeat. (Reproduced with
permission from Sdhof TC: The synaptic vesicle cycle: A cascade of proteinprotein interactions. Nature 19 95;375:645. Copyright by Macmillan Magazines.)
achieved by advancing a microelectrode through the ventral portion of the spinal cord. Puncture of a cell membrane is signaled by the
appearance of a steady 70-mV potential difference between the microelectrode and an electrode outside the cell. The cell can be
identified as a spinal motor neuron by stimulating the appropriate ventral root and obser ving the electrical activity of the cell. Such
stimulation initiates an antidromic impulse (see Chapter 4) that is conducted to the soma
Neuron:
Synaptic vesicle Plasma membrane
NSF
α/γ SNAPs Synaptobrevin
Syntaxin
rab3
munc18/
SNAP
rbSec1
25
GTP
FIGURE 6–5 Main proteins that interact to produce synaptic vesicle docking and fusi on in nerve endings. (Reproduced with permission
from Ferro-Novick S, John R: Vesicle fusion from yeast to man. Nature 1994;370:191. Copyright by Macmillan Magazines.)
and stops at this point. Therefore, the presence of an action potential in the cell after anti dromic stimulation indicates that the cell that has
been penetrated is an α-motor neuron. Stimulation of a dorsal root afferent (sensory ne uron) can be used to study both excitatory and
inhibitory events in α-motor neurons (Figure 6–6).
When an impulse reaches the presynaptic terminals, an interval of at least 0.5 ms, the synaptic delay, occurs before a response is
obtained in the postsynaptic neuron. It is due to the time it takes for the synaptic mediator to be released and to act on the membrane of
the postsynaptic cell. Because of it, conduction along a chain of neurons is slower if ma ny synapses are in the chain than if there are
only a few. Because the minimum time for transmission across one synapse is 0.5 ms, it is also possible to determine whether a given
reflex pathway is monosynaptic or polysynaptic (contains more than one synapse) by m easuring the delay in transmission from the
dorsal to the ventral root across the spinal cord.
A single stimulus applied to the sensory nerves characteristically does not lead to the for mation of a propagated action potential in the
postsynaptic neuron. Instead, the stimulation produces either a transient partial depolariz ation or a transient hyperpolarization. The initial
depolarizing response produced by a single stimulus to the proper input begins about 0 .5 ms after the afferent impulse enters the spinal
cord. It reaches its peak 11.5 ms later and then declines exponentially. During this poten tial, the excitability of the neuron to other stimuli
is increased, and consequently the potential is called an excitatory postsynaptic pote ntial (EPSP) (Figure 6–6).
CLINICAL BOX 6–1 Botulinum and Tetanus Toxins
Several deadly toxins which block neurotransmitter release are zinc endopeptidases that cleave and hence inactivate proteins in the
fusion–exocytosis complex. Tetanus toxin and botulinum toxins B, D, F, and G act on synaptobrevin, and botulinum toxin C acts on
syntaxin. Botulinum toxins A and B act on SNAP-25. Clinically, tetanus toxin causes spa stic paralysis by blocking presynaptic
transmitter release in the CNS, and botulism causes flaccid paralysis by blocking the rele ase of acetylcholine at the neuromuscular
junction. On the positive side, however, local injection of small doses of botulinum toxin (botox) has proved effective in the treatment of
a wide variety of conditions characterized by muscle hyperactivity. Examples include in jection into the lower esophageal sphincter to
relieve achalasia and injection into facial muscles to remove wrinkles.
A Stretch reflex circuit for knee jerk
Quadriceps (extensor) Muscle spindle Sensory neuron
Spinal cord
Hamstring (flexor)
Extensor motor
neuron
Flexor motor neuron Inhibitory interneuron
B Experimental setup for recording from cells in the circuit
Current passing
Recording
Extracellular stimulating electrodes Sensory neuron
Extensor
motor neuron Recording
EPSP
Ia afferent fibers from muscle
spindles of
quadriceps
Motor neuron
Sensory neuron
EPSP
Recording Current passing Inhibitory interneurons Recording
Flexor
motor
neuron
Ia afferent fibers from muscle
spindles of
quadriceps
EPSP
IPSP
IPSP Motor neuron Interneuron
FIGURE 6–6 Excitatory and inhibitory synaptic connections mediating the str etch reflex provide an example of typical circuits
within the CNS. A) The stretch receptor sensory neuron of the quadriceps musc le makes an excitatory connection with the extensor
motor neuron of the same muscle and an inhibitory interneuron projecting to fl exor motor neurons supplying the antagonistic hamstring
muscle. B) Experimental setup to study excitation and inhibition of the extensor motor neuron. T op panel shows two approaches to elicit
an excitatory (depolarizing) postsynaptic potential or EPSP in the extensor motor neuron–electri cal stimulation of the whole Ia afferent
nerve using extracellular electrodes and intracellular current passing through an electro de inserted into the cell body of a sensory neuron.
Bottom panel shows that current passing through an inhibitory interneuron elicits an inh ibitory (hyperpolarizing) postsynaptic potential
or IPSP in the flexor motor neuron. (From Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
The EPSP is produced by depolarization of the postsynaptic cell membrane immediately under the presynaptic ending. The excitatory
transmitter opens Na
+
or Ca
2+
ion channels in the postsynaptic membrane, producing an inward current. The area of current flow thus
created is so small that it does not drain off enough positive charge to depolarize the wh ole membrane. Instead, an EPSP is inscribed.
The EPSP due to activity in one synaptic knob is small, but the depolarizations produced by each of the active knobs summate.
EPSPs are produced by stimulation of some inputs, but stimulation of other inputs produ ces hyperpolarizing responses. Like the EPSPs,
they peak 11.5 ms after the stimulus and decrease exponentially. During this potential, th e excitability of the neuron to other stimuli is
decreased; consequently, it is called an inhibitory postsynaptic potential
(IPSP) (Figure 6–6).
An IPSP can be produced by a localized increase in Cl transport. When an inhibitory sy naptic knob becomes active,
the released transmitter triggers the opening of Cl
channels in the area of the postsynaptic cell membrane under the knob. Cl
moves
down its concentration gradient. The net effect is the transfer of negative charge into th e cell, so that the membrane potential increases.
The decreased excitability of the nerve cell during the IPSP is due to movement of the m embrane potential away from the firing level.
Consequently, more excitatory (depolarizing) activity is necessary to reach the firing lev el. The fact that an IPSP is mediated by Cl
can
be demonstrated by repeating the stimulus while varying the resting membrane potential of the postsynaptic cell. When the membrane
potential is at E
Cl
, the potential disappears (Figure 6–7), and at more negative membrane potentials, it bec omes positive (reversal
potential).
Because IPSPs are net hyperpolarizations, they can be produced by alterations in other ion channels in the neuron. For example, they can
be produced by opening of K
+
channels, with movement of K
+
out of the postsynaptic cell, or by closure of Na
+
or Ca
2+
channels.
–40 mV
5 mV
–60 mV RMP
–70 mV E
Cl
–90 mV E
K
–100 mV 5 ms
FIGURE 6–7 IPSP is due to increased Cl influx during stimulation. This can be demonstrated by repeating the stimulus while varying
the resting membrane potential (RMP) of the postsynaptic cell. When the membrane pot ential is at E
Cl
, the potential disappears, and at
more negative membrane potentials, it becomes positive (reversal potential).
A Temporal summation Recording
B Spatial summation
Recording
Synaptic current A A Axon Axon
A AAB
2 × 10–10 A
Synaptic potential
TEMPORAL & SPATIAL SUMMATION
Summation may be temporal or spatial (Figure 6–8). Temporal summation occurs if repeated affe rent stimuli cause new EPSPs before
previous EPSPs have decayed. A longer time constant for the EPSP allows for a greater opportunity for summation. When activity is
present in more than one synaptic knob at the same time, spatial summation occurs and activity in one synaptic knob summates with
activity in another to approach the firing level. The EPSP is therefore not an allor-none response but is proportionate in size to the
strength of the afferent stimulus.
Spatial summation of IPSPs also occurs, as shown by the increasing size of the response , as the strength of an inhibitory afferent volley
is increased. Temporal summation of IPSPs also occurs.
B
Long time Long length constant constant (100 ms) V
m
(1 mm) V
m
2 mV
Short time Short length
constant constant
(20 ms) V
m
(0.33 mm) V
m
2 mV
25 ms FIGURE 6–8 Central neurons integrate a variety of synaptic inputs through te mporal and spatial summation. A) The time
constant of the postsynaptic neuron affects the amplitude of the depolarization caused b y consecutive EPSPs produced by a single
presynaptic neuron. B) The length constant of a postsynaptic cell affects the amplitude of two EPSPs produced by two presynaptic
neurons, A and B. (From Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science , 4th ed. McGraw-Hill, 2000.)
SLOW POSTSYNAPTIC POTENTIALS
In addition to the EPSPs and IPSPs described previously, slow EPSPs and IPSPs have be en described in autonomic ganglia, cardiac and
smooth muscle, and cortical neurons. These postsynaptic potentials have a latency of 10 0 to 500 ms and last several seconds. The slow
EPSPs are generally due to decreases in K
+
conductance, and the slow IPSPs are due to increases in K
+
conductance.
GENERATION OF THE ACTION
POTENTIAL IN THE
POSTSYNAPTIC NEURON
The constant interplay of excitatory and inhibitory activity on the postsynaptic neuron p roduces a fluctuating membrane potential that is
the algebraic sum of the hyperpolarizing and depolarizing activity. The soma of the neu ron thus acts as a sort of integrator. When the 10
to 15 mV of depolarization sufficient to reach the firing level is attained, a propagated s pike results. However, the discharge of the
neuron is slightly more complicated than this. In motor neurons, the portion of the cell w ith the lowest threshold for the production of a
fullfledged action potential is the initial segment, the portion of the axon at and just beyond th e axon hillock. This unmyelinated
segment is depolarized or hyperpolarized electrotonically by the current sinks and sour ces under the excitatory and inhibitory synaptic
knobs. It is the first part of the neuron to fire, and its discharge is propagated in two dir ections: down the axon and back into the soma.
Retrograde firing of the soma in this fashion probably has value in wiping the slate clea n for subsequent renewal of the interplay of
excitatory and inhibitory activity on the cell.
drites. There, each can become associated with a single ribosome in a dendritic spine an d produce proteins, which alters the effects of
input from individual synapses on the spine. Changes in dendritic spines have been imp licated in motivation, learning, and long-term
memory.
ELECTRICAL TRANSMISSION
At synaptic junctions where transmission is electrical, the impulse reaching the presynapt ic terminal generates an EPSP in the
postsynaptic cell that, because of the low-resistance bridge between the two, has a much shorter latency than the EPSP at a synapse
where transmission is chemical. In conjoint synapses, both a short-latency response and a longer-latency, chemically mediated
postsynaptic response take place.
INHIBITION & FACILITATION AT SYNAPSES
DIRECT & INDIRECT INHIBITION
Inhibition in the CNS can be postsynaptic or presynaptic. Postsynaptic inhibition during the course of an IPSP is called direct
inhibition because it is not a consequence of previous discharges of the postsyna ptic neuron. There are various forms of indirect
inhibition, which is inhibition due to the effects of previous postsynaptic neuron discharge. For example, the postsynaptic cell can be
refractory to excitation because it has just fired and is in its refractory period. During af terhyperpolarization it is also less excitable. In
spinal neurons, especially after repeated firing, this after-hyperpolarization may be larg e and prolonged.
FUNCTION OF THE DENDRITES
For many years, the standard view has been that dendrites are simply the sites of curren t sources or sinks that electrotonically change the
membrane potential at the initial segment; that is, they are merely extensions of the soma t hat expand the area available for integration.
When the dendritic tree of a neuron is extensive and has multiple presynaptic knobs en ding on it, there is room for a great interplay of
inhibitory and excitatory activity.
It is now well established that dendrites contribute to neural function in more complex w ays. Action potentials can be recorded in
dendrites. In many instances, these are initiated in the initial segment and conducted in a retrograde fashion, but propagated action
potentials are initiated in some dendrites. Further research has demonstrated the malleabil ity of dendritic spines. Dendritic spines appear,
change, and even disappear over a time scale of minutes and hours, not days and mon ths. Also, although protein synthesis occurs mainly
in the soma with its nucleus, strands of mRNA migrate into the den
POSTSYNAPTIC INHIBITION IN THE SPINAL CORD
Various pathways in the nervous system are known to mediate postsynaptic inhibition, a nd one illustrative example is presented here.
Afferent fibers from the muscle spindles (stretch receptors) in skeletal muscle project di rectly to the spinal motor neurons of the motor
units supplying the same muscle (Figure 6–6). Impulses in this afferent fiber cause EPSP s and, with summation, propagated responses in
the postsynaptic motor neurons. At the same time, IPSPs are produced in motor neuron s supplying the antagonistic muscles which have
an inhibitory interneuron interposed between the afferent fiber and the motor neuron. Therefore, activity in the afferent fibers from the
muscle spindles excites the motor neurons supplying the muscle from which the impulse s come, and inhibits those supplying its
antagonists (reciprocal innervation).
PRESYNAPTIC INHIBITION &
FACILITATION
Another type of inhibition occurring in the CNS is presynaptic inhibition, a process med iated by neurons whose terminals are on
excitatory endings, forming axoaxonal synapses (Figure 6–3). The neurons responsible for postsynaptic and presynaptic inhibition are
compared in Figure 6–9. Three mechanisms of presynaptic inhibition have been describ ed. First, activation of the presynaptic receptors
increases Cl
conductance, and this has been shown to decrease the size of the action potentials reach ing the excitatory ending (Figure 6–
10). This in turn reduces Ca
2+
entry and consequently the amount of excitatory transmitter released. Voltage-gated K
+
channels are also
opened, and the resulting K
+
efflux also decreases the Ca
2+
influx. Finally, there is evidence for direct inhibition of transmitter release
independent of Ca
2+
influx into the excitatory ending.
The first transmitter shown to produce presynaptic inhibition was GABA. Acting via GA BA
A
receptors, GABA increases Cl
conductance. GABA
B
receptors are also present in the spinal cord and appear to mediate presynaptic inhibitio n via a G protein that
produces an increase in K
+
conductance. Baclofen, a GABA
B
agonist, is effective in the treatment of the spasticity of spinal cord injury
and multiple sclerosis, particularly when administered intrathecally via an implanted pump . Other transmitters also mediate presynaptic
inhibition by G protein-mediated effects on Ca
2+
channels and K
+
channels.
Conversely, presynaptic facilitation is produced when the action potential is prolonged (Figure 6–10) and the Ca
2+
channels are open
for a longer period. The molecular events responsible for the production of presynapt ic facilitation mediated by serotonin in the sea snail
Aplysia have been worked out in detail. Serotonin released at an axoaxonal ending incr eases intraneuronal cAMP levels, and the
resulting phosphorylation
Presynaptic inhibition Postsynaptic inhibition
Motor neuron
FIGURE 6–9 Arrangement of neurons producing presynaptic and postsynap tic inhibition. The neuron producing presynaptic
inhibition is shown ending on an excitatory synaptic knob. Many of these neurons ac tually end higher up along the axon of the excitatory
cell.
Presynaptic action
potential EPSP in postsynaptic neuron
Ca
2+
current in presynaptic neuron
Presynaptic inhibition
Presynaptic facilitation
FIGURE 6–10 Effects of presynaptic inhibition and facilitation on the action potential a nd the Ca
2+
current in the presynaptic
neuron and the EPSP in the postsynaptic neuron. In each case, the solid lin es are the controls and the dashed lines the records
obtained during inhibition or facilitation. (Modified from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill,
2000.)
of one group of K
+
channels closes the channels, slowing repolarization and prolonging the action potential .
ORGANIZATION OF
INHIBITORY SYSTEMS
Presynaptic and postsynaptic inhibition are usually produced by stimulation of certain sy stems converging on a given postsynaptic
neuron (afferent inhibition). Neurons may also inhibit themselves in a negative feedbac k fashion (negative feedback inhibition). For
instance, each spinal motor neuron regularly gives off a recurrent collateral that synaps es with an inhibitory interneuron, which
terminates on the cell body of the spinal neuron and other spinal motor neurons (Figure 6–11). This particular inhibitory neuron is
sometimes called a Renshaw cell after its discoverer. Impulses generated in the motor ne uron activate the inhibitory interneuron to
secrete inhibitory mediators, and this slows or stops the discharge of the motor neuron. Similar inhibition via recurrent collaterals is seen
in the cerebral cortex and limbic system. Presynaptic inhibition due to descending pathw ays that terminate on afferent pathways in the
dorsal horn may be involved in the gating of pain transmission.
Another type of inhibition is seen in the cerebellum. In this part of the brain, stimulation of basket cells produces IPSPs in the Purkinje
cells. However, the basket cells and the Purkinje
A Motor neuron X
Motor neuron
B
Y
Inhibitory interneuron C
Z
Axon FIGURE 6–12 Simple nerve net. Neurons A, B, and C have excitatory endin gs on neurons X, Y, and Z.
FIGURE 6–11 Negative feedback inhibition of a spinal motor neuron v ia an inhibitory interneuron (Renshaw cell).
cells are excited by the same parallel-fiber excitatory input. This arrangement, which ha s been called feed-forward inhibition,
presumably limits the duration of the excitation produced by any given afferent volley.
SUMMATION & OCCLUSION
As noted above, the axons of most neurons discharge onto many other neurons. Conv ersely, any given neuron receives input from many
other neurons (convergence).
In the hypothetical nerve net shown in Figure 6–12, neurons A and B converge on X , and neuron B diverges on X and Y. A stimulus
applied to A or to B will set up an EPSP in X. If A and B are stimulated at the same time and action potentials are produced, two areas of
depolarization will be produced in X and their actions will sum. The resultant EPSP in X will be twice as large as that produced by
stimulation of A or B alone, and the membrane potential may well reach the firing level of X. The effect of the depolarization caused by
the impulse in A adds to that due to activity in B, and vice versa; spatial summation has ta ken place. In this case, Y has not fired, but its
excitability has been increased, and it is easier for activity in neuron C to fire Y during t he EPSP. Y is therefore said to be in the
subliminal fringe of X. More generally stated, neurons are in the subliminal fringe if they are not discharged by an afferent volley (not
in the discharge zone) but do have their excitability increased. The neurons that ha ve few active knobs ending on them are in the
subliminal fringe, and those with many are in the discharge zone. Inhibitory impulses sh ow similar temporal and spatial facilitation and
subliminal fringe effects.
If action potentials are produced repeatedly in neuron B, X and Y will discharge as a r esult of temporal summation of the EPSPs that are
produced. If C is stimulated repeatedly, Y and Z will discharge. If B and C are fired re peatedly at the same time, X, Y, and Z will
discharge. Thus, the response to stimulation of B and C together is not as great as the su m of responses to stimulation of B and C
separately, because B and C both end on neuron Y. This decrease in expected respons e, due to presynaptic fibers sharing postsynaptic
neurons, is called occlusion.
NEUROMUSCULAR
TRANSMISSION:
NEUROMUSCULAR JUNCTION
ANATOMY
As the axon supplying a skeletal muscle fiber approaches its termination, it loses its myeli n sheath and divides into a number of terminal
As the axon supplying a skeletal muscle fiber approaches its termination, it loses its myeli n sheath and divides into a number of terminal
boutons, or endfeet (Figure 6–13). The endfeet contain many small, clear vesicles that c ontain acetylcholine, the transmitter at these
junctions. The endings fit into junctional folds, which are depressions in the mo tor end plate, the thickened portion of the muscle
membrane at the junction. The space between the nerve and the thickened muscle memb rane is comparable to the synaptic cleft at
synapses. The whole structure is known as the neuromuscular, or myoneural, junctio n. Only one nerve fiber ends on each end plate,
with no convergence of multiple inputs.
SEQUENCE OF EVENTS
DURING TRANSMISSION
The events occurring during transmission of impulses from the motor nerve to the musc le are somewhat similar to those occurring at
neuron-to-neuron synapses (Figure 6–14). The impulse arriving in the end of the moto r neuron increases the permeability of its endings
to Ca
2+
. Ca
2+
enters the endings and triggers a marked increase in exocytosis of the acetylcholine-co ntaining vesicles. The acetylcholine
diffuses to the
(a)
(b) Motor nerve fiber
Myelin
Axon terminal
Schwann cell
Synaptic vesicles (containing ACh) Active zone
Sarcolemma
Synaptic cleft
Junctional Nucleus of muscle fiber
Region of
folds
sarcolemma
with ACh receptors
FIGURE 6–13 The neuromuscular junction. (a) Scanning ele ctronmicrograph showing branching of motor axons with terminals
embedded in grooves in the muscle fiber’s surface. (b) Structure of a neuro muscular junction.
(From Widmaier EP, Raff H, Strang KT: Vanders
Human Physiology.
McGraw-Hill, 2008.)
muscle-type nicotinic acetylcholine receptors, which are concentrated at the tops of the j unctional folds of the membrane of the motor
end plate. Binding of acetylcholine to these receptors increases the Na
+
and K
+
conductance of the membrane, and the resultant influx of
Na
+
produces a depolarizing potential, the end plate potential. The current sink created by this local potential depolarizes the adjacent
muscle membrane to its firing level. Acetylcholine is then removed from the synaptic cle ft by acetylcholinesterase, which is present in
high concentration at the neuromuscular junction. Action potentials are generated on eith er side of the end plate and are conducted away
from the end plate in both directions along the muscle fiber. The muscle action potential, in turn, initiates muscle contraction, as
described in Chapter 5.
END PLATE POTENTIAL
An average human end plate contains about 15 to 40 million acetylcholine receptors. Ea ch nerve impulse releases about 60
acetylcholine vesicles, and each vesicle contains about 10,000
molecules of the neurotransmitter. This amount is enough to activate about 10 times the n umber of acetylcholine receptors needed to
produce a full end plate potential. Therefore, a propagated response in the muscle is re gularly produced, and this large response obscures
the end plate potential. However, the end plate potential can be seen if the tenfold safety factor is overcome and the potential is reduced
to a size that is insufficient to fire the adjacent muscle membrane. This can be accomplish ed by administration of small doses of curare, a
drug that competes with acetylcholine for binding to muscle-type nicotinic acetylcholine receptors. The response is then recorded only at
the end plate region and decreases exponentially away from it. Under these conditions, end plate potentials can be shown to undergo
temporal summation.
QUANTAL RELEASE OF TRANSMITTER
Small quanta (packets) of acetylcholine are released randomly from the nerve cell mem brane at rest. Each produces a minute
depolarizing spike called a miniature end plate potential, which is about 0.5 mV in amplitude. The size of the quanta of acetylcholine
released in this way varies directly with the Ca
2+
concentration and inversely with the Mg
2+
concentration at the end plate. When a nerve
impulse reaches the ending, the number of quanta released increases by several orders of magnitude, and the result is the large end plate
potential that exceeds the firing level of the muscle fiber.
Quantal release of acetylcholine similar to that seen at the myoneural junction has been o bserved at other cholinergic synapses, and
quantal release of other transmitters probably occurs at noradrenergic, glutaminergic, a nd other synaptic junctions.
Two diseases of the neuromuscular junction, myasthenia gravis and Lambert-Eaton syn drome, are described in Clinical Box 6–2 and
Clinical Box 6–3, respectively.
1
Motor neuron action potential
2
Ca
2+
enters voltage-gated channels
2
Ca
2+
enters voltage-gated channels
++ +
–– –
+
– –
+
+
Acetylcholine
vesicle
7 Propagated action potential in muscle plasma membrane
Voltage-gated Na
+
channels
3
Acetylcholine
+ +
+ + +
release
4
Na
+
entry
–––
+
+
+
+ +
+
+
+
+
– –
– – –
8 Acetylcholine degradation Acetylcholine receptor
6
Muscle fiber action potential Acetylcholinesterase initiation
Motor end plate
5
Local current between
depolarized end plate and adjacent muscle plasma membrane
FIGURE 6–14 Events at the neuromuscular junction that lead to an action po tential in the muscle fiber plasma membrane.
Although potassium exits the muscle cell when Ach receptors are open, sodium entry an d depolarization dominate. (From Widmaier EP, Raff
H, Strang KT: Vanders Human Physiology. McGraw-Hill, 2008.)
NERVE ENDINGS IN SMOOTH & CARDIAC MUSCLE
ANATOMY
The postganglionic neurons in the various smooth muscles that have been studied in deta il branch extensively and come in close contact
with the muscle cells (Figure 6–15). Some of these nerve fibers contain clear vesicles an d are cholinergic, whereas others contain the
characteristic dense-core vesicles that contain norepinephrine. There are no recognizab le end plates or other postsynaptic specializations.
The nerve fibers run along the membranes of the muscle cells and sometimes groove th eir surfaces. The multiple branches of the
noradrenergic and, presumably, the cholinergic neurons are beaded with enlargements (varicosities) and contain synaptic vesicles
(Figure 6–15). In noradrenergic neurons, the varicosities are about 5 μm apart, with u p to 20,000 varicosities per neuron. Transmitter is
apparently liberated at each varicosity, that is, at many locations along each axon. This a rrangement permits one neuron to innervate
many effector cells. The type of contact in which a neuron forms a synapse on the sur face of another neuron or a smooth muscle cell and
then passes on to make similar contacts with other cells is called a synapse en pass ant.
In the heart, cholinergic and noradrenergic nerve fibers end on the sinoatrial node, the atrioventricular node, and the bundle of His.
Noradrenergic fibers also innervate the ventricular muscle. The exact nature of the end ings on nodal tissue is not known. In the ventricle,
the contacts between the noradrenergic fibers and the cardiac muscle fibers resemble th ose found in smooth muscle.
JUNCTIONAL POTENTIALS
In smooth muscles in which noradrenergic discharge is excitatory, stimulation of the no radrenergic nerves produces discrete partial
depolarizations that look like small end plate potentials and are called excitatory ju nction potentials (EJPs). These potentials summate
with repeated stimuli. Similar EJPs are seen in tissues excited by cholinergic discharges. I n tissues inhibited by noradrenergic stimuli,
hyperpolarizing inhibitory junction potentials (IJPs) are produced by stimulation of th e noradrenergic nerves. Junctional potentials
spread electrotonically.
CLINICAL BOX 6–2 CLINICAL BOX 6–3 Myasthenia Gravis
Myasthenia gravis is a serious and sometimes fatal disease in which skeletal musc les are weak and tire easily. It occurs in 25 to 125 of
every 1 million people worldwide and can occur at any age but seems to have a bimod al distribution, with peak occurrences in
individuals in their 20s (mainly women) and 60s (mainly men). It is caused by the form ation of circulating antibodies to the muscle type
of nicotinic acetylcholine receptors. These antibodies destroy some of the recep tors and bind others to neighboring receptors, triggering
their removal by endocytosis. Normally, the number of quanta released from the motor nerve terminal declines with successive repetitive
stimuli. In myasthenia gravis, neuromuscular transmission fails at these low levels of qua ntal release. This leads to the major clinical
feature of the disease– muscle fatigue with sustained or repeated activity. There are two major forms of the disease. In one form, the
extraocular muscles are primarily affected. In the second form, there is a generalized w eakness of skeletal muscles. Weakness improves
after a period of rest or after administration of acetylcholinesterase inhibitors. Cholinesterase inhibitors prevent metabolism of
acetylcholine and can thus compensate for the normal decline in released neurotransmitt ers during repeated stimulation. In severe cases,
all muscles, including the diaphragm, can become weak and respiratory failure and dea th can ensue. The major structural abnormality in
myasthenia gravis is the appearance of sparse, shallow, and abnormally wide or absent synaptic clefts in the motor end plate. Studies
show that the postsynaptic membrane has a reduced response to acetylcholine and a 70 –90% decrease in the number of receptors per end
plate in affected muscles. Patients with mysathenia gravis have a greater than normal ten dency to also have rheumatoid arthritis, systemic
lupus erythematosus, and polymyositis. About 30% of mysathenia gravis patients have a maternal relative with an autoimmune disorder.
These associations suggest that individuals with myasthenia gravis share a genetic predisp osition to autoimmune disease. The thymus
may play a role in the pathogenesis of the disease by supplying helper T cells sensitized against thymic proteins that cross-react with
acetylcholine receptors. In most patients, the thymus is hyperplastic, and 10–15% have thymomas. Thymectomy is indicated if a
thymoma is suspected. Even in those without thymoma, thymectomy induces remission in 35% and improves symptoms in another 45%
of patients.
DENERVATION HYPERSENSITIVITY
When the motor nerve to skeletal muscle is cut and allowed to degenerate, the muscle gr adually becomes extremely sensitive to
acetylcholine. This denervation hypersensitivity or super
Lambert–Eaton Syndrome
Another condition that resembles myasthenia gravis is the relatively rare condition called Lambert–Eaton Syndrome (LEMS). In this
condition, muscle weakness is caused by an autoimmune attack against one of the Ca
2+
channels in the nerve endings at the
neuromuscular junction. This decreases the normal Ca
2+
influx that causes acetylcholine release. Proximal muscles of the lower
extremities are primarily affected, producing a waddling gait and difficulty raising the a rms. Repetitive stimulation of the motor nerve
facilitates accumulation of Ca
2+
in the nerve terminal and increases acetylcholine release, leading to an increase in musc le strength. This
is in contrast to myasthenia gravis in which symptoms are exasperated by repetitive stimu lation. About 40% of patients with LEMS also
have cancer, especially small cell cancer of the lung. One theory is that antibodies that h ave been produced to attack the cancer cells may
also attack Ca
2+
channels, leading to LEMS. LEMS has also been associated with lymphosarcoma, malig nant thymoma, and cancer of
the breast, stomach, colon, prostate, bladder, kidney, or gall bladder. Clinical signs usua lly precede the diagnosis of cancer. A syndrome
similar to LEMS can occur after the use of aminoglycoside antibiotics, which also impair Ca
2+
channel function.
sensitivity is also seen in smooth muscle. Smooth muscle, unlike skeletal muscle, does not atrophy when denervated, but it becomes
hyperresponsive to the chemical mediator that normally activates it. A good example of denervation hypersensitivity is the response of
the denervated iris. If the postganglionic sympathetic nerves to one iris are cut in an exp erimental animal and, after several weeks,
norepinephrine (the transmitter released by sympathetic postganglionic neurons) is injec ted intravenously, the denervated pupil dilates
widely. A much smaller, less prolonged response is observed on the intact side.
The reactions triggered by section of an axon are summarized in Figure 6–16. Hyperse nsitivity of the postsynaptic structure to the
transmitter previously secreted by the axon endings is a general phenomenon, largely d ue to the synthesis or activation of more
receptors. There is in addition orthograde degeneration (wallerian degeneration) and retrogr ade degeneration of the axon stump to the
nearest collateral (sustaining collateral). A series of changes occur in the cell b ody that include a decrease in Nissl substance
(chromatolysis). The nerve then starts to regrow, with multiple small branches projecting along the path the axon previously followed
(regenerative sprouting). Axons sometimes grow back to their original targets, especiall y in locations like the neuromuscular junction.
However, nerve regeneration is generally limited because axons often become entangle d in the area of tissue damage at the site where
they were disrupted. This
Autonomic
nerve fiber
Varicosity Sheet of
cells
Mitochondrion
Synaptic vesicles
Varicosities
FIGURE 6–15 Endings of postganglionic autonomic neurons on smooth muscl e. Neurotransmitter, released from varicosities along
the branched axon, diffuses to receptors on smooth muscle cell plasma memb ranes. (From Widmaier EP, Raff H, Strang KT: Vanders Human
Physiology. McGraw-Hill, 2008.)
difficulty has been reduced by administration of neurotro
phins. For example, sensory neurons torn when dorsal nerve
roots are avulsed from the spinal cord regrow and form func
tional connections in the spinal cord if experimental animals
are treated with NGF, neurotrophin 3, or GDNF.
Hypersensitivity is limited to the structures immediately
innervated by the destroyed neurons and fails to develop in neu
rons and muscle farther downstream. Suprasegmental spinal cord lesions do not lead to hypersensitivity of the paralyzed skeletal muscles
to acetylcholine, and destruction of the preganglionic autonomic nerves to visceral struc tures does not cause hypersensitivity of the
denervated viscera. This fact has practical implications in the treatment of diseases due to spasm of the blood vessels in the extremities.
For example, if the upper extremity is sympathectomized by removing the upper part of the ganglionic chain and the stellate ganglion,
Axon branch Receptor
(sustaining collateral)
the hypersensitive smooth muscle in the vessel walls is stimulated by circulating norepinep hrine, and episodic vasospasm continues to
occur. However, if preganglionic sympathectomy of the arm is performed by cutting th e ganglion chain below the third ganglion (to
interrupt ascending preganglionic
Retrograde
degeneration
Receptor
hypersensitive
fibers) and the white rami of the first three thoracic nerves, no hypersensitivity results.
Site of in jury Denervation hypersensitivity has multiple causes. As noted in Chapter 2, a deficiency of a given chemical messenger genX
erally produces an upregulation of its receptors. Another factor is lack of reuptake of secreted neurotransmitters. Retrograde
reaction:
Regenerative
sprouting
chromatolysis
Orthograde
(wallerian)
degeneration
CHAPTER SUMMARY
FIGURE 6–16 Summary of changes occurring in a neuron
and the structure it innervates when its axon is crushed or cut at
the point marked X. Hypersensitivity of the postsynaptic structure to
the transmitter previously secreted by the axon occurs largely due to
the synthesis or activation of more receptors. There is both orthograde
(wallerian) degeneration from the point of damage to the terminal and
retrograde degeneration of the axon stump to the nearest collateral
(sustaining collateral). Changes also occur in the cell body, including
chromatolysis. The nerve starts to regrow, with multiple small branch
es projecting along the path the axon previously followed (regenera
tive sprouting).
Presynaptic terminals are separated from the postsynaptic structure by a synaptic cleft. The postsynaptic membrane contains many
neurotransmitter receptors and usually a postsynaptic thickening called the postsynaptic density.
At chemical synapses, an impulse in the presynaptic axon causes secretion of a chemical that diffuses across the synaptic cleft and
binds to postsynaptic receptors, triggering events that open or close channels in the me mbrane of the postsynaptic cell. At electrical
synapses, the membranes of the presynaptic and postsynaptic neurons come close toget her, and gap junctions form low-resistance
bridges through which ions pass with relative ease from one neuron to the next.
A neuron receives input from many other neurons (convergence), and a neuron branches to innervate many other neurons (divergence).
An EPSP is produced by depolarization of the postsynaptic cell after a latency of 0 .5 ms; the excitatory transmitter opens Na
+
or Ca
2+
ion channels in the postsynaptic membrane, producing an inward current. An IPS P is produced by a hyperpolarization of the postsynaptic
cell; it can be produced by a localized increase in Cl
transport. Slow EPSPs and IPSPs occur after a latency of 100 to 500 ms in
autonomic ganglia, cardiac and smooth muscle, and cortical neurons. The slow EPSPs are d ue to decreases in K
+
conductance, and the
slow IPSPs are due to increases in K
+
conductance.
Postsynaptic inhibition during the course of an IPSP is called direct inhibition. Indir ect inhibition is due to the effects of previous
postsynaptic neuron discharge; for example, the postsynaptic cell cannot be activated during its r efractory period. Presynaptic inhibition
is a process mediated by neurons whose terminals are on excitatory endings, formi ng axoaxonal synapses; in response to activation of
the presynaptic terminal. Activation of the presynaptic receptors can increase Cl
conductance, decreasing the size of the action
potentials reaching the excitatory ending, and reducing Ca
2+
entry and the amount of excitatory transmitter released.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. Fast inhibitor y postsynaptic potentials (IPSPs)
A) are a consequence of decreased Cl
conductance. B) occur in skeletal muscle.
C) can be produced by an increase in Na
+
conductance. D) can be produced by an increase in Ca
2+
conductance. E) interact with other
fast and slow potentials to move the
membrane potential of the postsynaptic neuron toward or away from the firing level.
2. Fast excitatory postsynaptic potentials (EPSPs)
2. Fast excitatory postsynaptic potentials (EPSPs)
A) are a consequence of decreased Cl
conductance. B) occur in skeletal muscle.
C) can be produced by an increase in Na
+
conductance. D) can be produced by a decrease in Ca
2+
conductance. E) all of the above
3. Initiation of an action potential in skeletal muscle by stimulating its motor ner ve
A) requires spatial facilitation.
B) requires temporal facilitation.
C) is inhibited by a high concentration of Ca
2+
at the neuromuscular junction.
D) requires the release of norepinephrine.
E) requires the release of acetylcholine.
4. A 35-year-old woman sees her physician to report muscle weakness in the extraocu lar eye muscles and muscles of the extremities. She
states that she feels fine when she gets up in the morning, but the weakness begin s soon after she becomes active. The weakness is
improved by rest. Sensation appears normal. The physician treats her with an anticholinesterase inhibitor, and she notes immediate return
of muscle strength. Her physician diagnoses her with
A) Lambert–Eaton syndrome.
B) myasthenia gravis.
C) multiple sclerosis.
D) Parkinson disease.
E) muscular dystrophy.
CHAPTER RESOURCES
Boron WF, Boulpaep EL: Medical Physiology, Elsevier, 2005. Hille B: Ionic Channels of Excitable Membranes, 3rd ed. Sinauer
Associates, 2001.
Jessell TM, Kandel ER: Synaptic transmission: A bidirectional and a
self-modifiable form of cellcell communication. Cell
1993;72(Suppl):1.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural
Science, 4th ed. McGraw-Hill, 2000.
McPhee SJ, Ganong WF: Pathophysiology of Disease. An Introduction
to Clinical Medicine, 5th ed. McGraw-Hill, 2006.
Squire LR, et al (editors): Fundamental Neuroscience, 3rd ed.,
Academic Press, 2008.
Unwin N: Neurotransmitter action: Opening of ligand-gated ion
channels. Cell 1993; 72(Suppl):31.
Van der Kloot W, Molg J: Quantal acetylcholine release at the
vertebrate neuromuscular junction. Physiol Rev 1994;74:899.
Neurotransmitters & Neuromodulators
OBJEC TIV ES
After studying this chapter, you should be able to:
List neurotransmitters and the principal sites in the nervous system at which they
are released.
Describe the receptors for catecholamines, acetylcholine, 5-HT, amino acids, and opio ids.
Summarize the steps involved in the biosynthesis, release, action, and removal from the synap tic cleft of the various synaptic
transmitters.
Define opioid peptide, list the principal opioid peptides in the body, and name the
CH APTER
7
precursor molecules from which they originate.
INTRODUCTION
The fact that transmission at most synapses is chemical is of great physiologic and pharm acologic importance. Nerve endings have been
called biological transducers that convert electrical energy into chemical energy. In broa d terms, this conversion process involves the
synthesis of the neurotransmitters, their storage in synaptic vesicles, and their release by the nerve impu lses into the synaptic cleft. The
secreted transmitters then act on appropriate receptors on the membrane of the postsyna ptic cell and are rapidly removed from the
synaptic cleft by diffusion, metabolism, and, in many instances, reuptake into the presyn aptic neuron. Some chemicals released by
neurons have little or no direct effects on their own but can modify the effects of neuro transmitters. These chemicals are called
neuromodulators. All these processes, plus the postreceptor events in the postsynaptic neu ron, are regulated by many physiologic
factors and at least in theory can be altered by drugs. Therefore, pharmacologists (in th eory) should be able to develop drugs that regulate
not only somatic and visceral motor activity but also emotions, behavior, and all the othe r complex functions of the brain.
CHEMICAL TRANSMISSION
OF SYNAPTIC ACTIVITY
CHEMISTRY OF TRANSMITTERS
One suspects that a substance is a neurotransmitter if it is unevenly distributed in the nerv ous system and its distribution parallels that of
its receptors and synthesizing and catabolizing enzymes. Additional evidence includes d emonstration that it is released from appropriate
brain regions in vitro and that it produces effects on single target neurons when applied to their membranes by means of a micropipette
(microiontophoresis). Many transmitters and enzymes involved in their synthesis and ca tabolism have been localized in nerve endings by
immunohistochemistry, a technique in which antibodies to a given substance are labeled an d applied to brain and other tissues. The
antibodies bind to the substance, and the location of the substance is then determined by locating the label with the light microscope or
electron microscope. In situ hybridization histochemistry, which permits localization of the mRNAs for pa rticular synthesizing
enzymes or receptors, has also been a valuable tool.
Identified neurotransmitters and neuromodulators can be divided into two major catego ries: small-molecule transmitters and large-
molecule transmitters. Small-molecule transmitters
129 include monoamines (eg, acetylcholine, serotonin, histamine), catecholamines (dopamine, norepin ephrine, and epinephrine), and
amino acids (eg, glutamate, GABA, glycine). Large-molecule transmitter s include a large number of peptides called neuropeptides
including substance P, enkephalin, vasopressin, and a host of others. In general, neuro peptides are colocalized with one of the small-
molecule neurotransmitters (Table 7–1).
There are also other substances thought to be released into the synaptic cleft to act as eith er a transmitter or modulator of synaptic
transmission. These include purine derivatives like adenosine and adenosine triphosphat e (ATP) and a gaseous molecule, nitric oxide
(NO).
Figure 7–1 shows the biosynthesis of some common smallmolecule transmitters released by neurons in the central or peripheral nervous
system. Figure 7–2 shows the location of major groups of neurons that contain norepin ephrine, epinephrine, dopamine, and
acetylcholine. These are some of the major neuromodulatory systems.
RECEPTORS
Cloning and other molecular biology techniques have permitted spectacular advances in knowledge about the structure and function of
receptors for neurotransmitters and other chemical messengers. The individual receptor s, along with their
TABLE 7–1 Examples of colocalization of small-molecule transmitters with n europeptides.
Small-Molecule Transmitter
Neuropeptide
Monoamines
Acetylcholine Enkephalin, calcitonin-gene-related peptide,
galanin, gonadotropin-releasing hormone, neurotensin, somatostatin, substance P, vasoactive intestinal polypeptide
Serotonin
Catecholamines
Cholecystokinin, enkephalin, neuropeptide Y, substance P, vasoactive intestinal polypep tide
Dopamine Cholecystokinin, enkephalin, neurotensin
Norepinephrine Enkephalin, neuropeptide Y, neurotensin, somatostatin, vasopressin
Epinephrine
Amino Acids
Enkephalin, neuropeptide Y, neurotensin, substance P
Glutamate Substance P
Glycine Neurotensin
GABA Cholecystokinin, enkephalin, somatostatin, substance P, thyrotropin-releasing horm one
ligands, are discussed in the following parts of this chapter. However, five themes have emerged that should be mentioned in this
introductory discussion.
First, in every instance studied in detail to date, it has become clear that each ligand has m any subtypes of receptors. Thus, for example,
norepinephrine acts on α
1
and α
2
receptors, and three of each subtype have been cloned. In addition, there are β
1
, β
2
, and β
3
receptors.
Obviously, this multiplies the possible effects of a given ligand and makes its effects in a given cell more selective.
Second, there are receptors on the presynaptic as well as the postsynaptic elements for m any secreted transmitters. These presyna ptic
receptors, or autoreceptors, often inhibit further secretion of the ligand, providing feedback control. For example, norepinephrine acts
on α
2
presynaptic receptors to inhibit norepinephrine secretion. However, autoreceptors can also facilitate the release of
neurotransmitters.
Third, although there are many ligands and many subtypes of receptors for each ligand , the receptors tend to group in large families as
far as structure and function are concerned. Many receptors act via trimeric G proteins and protein kinases to produce their effects.
Others are ion channels. The receptors for a group of selected, established neurotransm itters and neuromodulators are listed in Table 7–2,
along with their principal second messengers and, where established, their net effect on ion channels. It should be noted that this table is
an oversimplification. For example, activation of α
2
-adrenergic receptors decreases intracellular cAMP concentrations, but there is
evidence that the G protein activated by α
2
-adrenergic presynaptic receptors also acts directly on Ca
2+
channels to inhibit norepinephrine
release by decreasing Ca
2+
increases.
Fourth, receptors are concentrated in clusters in postsynaptic structures close to the endi ngs of neurons that secrete the neurotransmitters
specific for them. This is generally due to the presence of specific binding proteins for them. In the case of nicotinic acetylcholine
receptors at the neuromuscular junction, the protein is rapsyn, and in the case of excitatory glutamatergic receptors, a family of PB2-
binding proteins is involved. GABA
A
receptors are associated with the protein gephyrin, which also binds glyci ne receptors, and
GABA
C
receptors are bound to the cytoskeleton in the retina by the protein MAP-1B. At least in the case of GABA
A
receptors, the
binding protein gephyrin is located in clumps in the postsynaptic membrane. With activit y, the free receptors move rapidly to the
gephyrin and bind to it, creating membrane clusters. Gephyrin binding slows and restric ts their further movement. Presumably, during
neural inactivity, the receptors are unbound and move again.
Fifth, prolonged exposure to their ligands causes most receptors to become unresponsiv e, that is, to undergo desensitization. This can be
of two types: homologous desensitization, with loss of responsiveness only to th e particular ligand and maintained responsiveness of the
cell to other ligands; and heterologous desensitization, in which the cell becomes unresponsive to other ligands as well. Desensitization
in β- adrenergic receptors has been studied in considerable detail. One form involves p hosphorylation of the carboxyl terminal
FIGURE 7–1 Biosynthesis of some common small molecule transmitters. (Reprodu ced with permission from Boron WF, Boulpaep EL: Medical
Physiology. Elsevier, 2005.)
FIGURE 7–2 Four diffusely connected systems of central neurons using modu latory transmitters. A) Norepinephrine-containing
neurons. B) Serotonin-containing neurons. C) D opamine-containing neurons. D) Acetylcholine-containing n eurons. (Reproduced with
permission from Boron WF, Boulpaep EL: Medical Physiology. Elsevier, 2005.)
region of the receptor by a specific β-adrenergic receptor
kinase (β-ARK) or binding β-arrestins. Four β-arrestins have
been described in mammals. Two are expressed in rods and
cones of the retina and inhibit visual responses. The other two,
β-arrestin 1 and β-arrestin 2, are more ubiquitous. They
desensitize β-adrenegic receptors, but they also inhibit other
heterotrimeric G protein-coupled receptors. In addition, they
foster endocytosis of ligands, adding to desensitization.
REUPTAKE
Neurotransmitters are transported from the synaptic cleft back into the cytoplasm of the neurons that secreted them, a process referred to
as reuptake (Figure 7–3). The high-affinity reuptake systems employ two families of transporter pr oteins. One family has 12
transmembrane domains and cotransports the transmitter with Na
+
and Cl
. Members of this family include transporters for
norepinephrine, dopamine, serotonin, GABA, and glycine, as well as transporters for p roline, taurine, and the acetylcholine precursor
choline. In addition, there
TABLE 7–2 Mechanism of action of selected small-molecule transmitters. Transmitte r
Monoamines Acetylcholine
Serotonin
Receptor Second Messenger Net Channel Effects
Nicotinic M
1,
M
3,
M
5
M
2,
M
4
5HT
1A
5HT
1B
5HT
1D
5HT
2A
5HT
2C
5HT
3
5HT
4
↑Na
+
, K
+
↑ IP
3
, DAG ↑Ca
2+
↓Cyclic AMP ↑K
+
↓Cyclic AMP ↑K
+
↓Cyclic AMP
↓Cyclic AMP ↓K
+
IP
3
, DAG ↓K
+
↑IP
3
, DAG
↑Na
+
↑Cyclic AMP Catecholamines Dopamine
D
1
, D
5
D
2
D
3
, D
4
Norepinephrine α
1
α
2
α
2
β
1
β
2
β
3
↑Cyclic AMP
↓Cyclic AMP ↑K
+
, ↓Ca
2+
Cyclic AMP↓
↑IP
3
, DAG ↓K
+ +
, ↓Ca
2+
↓Cyclic AMP ↑K ↑Cyclic AMP
↑Cyclic AMP
↑Cyclic AMP
Amino Acids
Glutamate Metabotropic
a
GABA
Glycine Ionotropic
AMPA, Kainate NMDA
GABA
A
GABA
B
Glycine
↑Na
+
, K
+
↑Na
+
, K
+
,Ca
2+
↑Cl
↑IP
3
, DAG ↑K
+
,↓Ca
2+
↑Cl
a
Eleven subtypes identified; all decrease cAMP or increase IP
3
and DAG, except one, which increases cAMP.
may be an epinephrine transporter. The other family is made up of at least three transpo rters that mediate glutamate uptake by neurons
and two that transport glutamate into astrocytes. These glutamate transporters are couple d to the cotransport of Na
+
and the
countertransport of K
+
, and they are not dependent on Cl
transport. There is a debate about their structure, and they may have 6, 8, or
10 transmembrane domains. One of them transports glutamate into glia rather than neur ons (see Chapter 4).
There are in addition two vesicular monoamine transporters, VMAT1 and VMAT2, tha t transport neurotransmitters from the cytoplasm
to synaptic vesicles. They are coded by different genes but have extensive homology. B oth have a broad specificity, moving dopamine,
norepinephrine, epinephrine, serotonin, and histamine from the cytoplasm into secretor y granules. Both are inhibited by reserpine, which
accounts for the marked monoamine depletion produced by this drug. Like the neurotr ansmitter membrane transporter family, they have
12 transmembrane domains, but they have little homology to the other transporters. The re is also a vesicular GABA transporter (VGAT)
that moves GABA and glycine into vesicles and a vesicular acetylcholine transporter.
Reuptake is a major factor in terminating the action of transmitters, and when it is inhibite d, the effects of transmitter release are
increased and prolonged. This has clinical consequences. For example, several effectiv e antidepressant drugs are inhibitors of the
reuptake of amine transmitters, and cocaine is believed to inhibit dopamine reuptake. Glu tamate uptake into neurons and glia is
important because glutamate is an excitotoxin that can kill cells by overstimulating them (s ee Clinical Box 7–1). There is evidence that
during ischemia
Neuron Presynaptic
Glial cellterminal 4
1 Amino acid precursor Transmitter
synthesis NTT
K+
Na
+
2 5
VMAT 3 H
+
R
Cl
R
4
R
Second
+ Other
messengers
receptors
Postsynaptic terminal
Neuron
FIGURE 7–3 Fate of monoamines secreted at synaptic junctions. In each monoamine-secreting neuron, the monoamine is synthesized
in the cytoplasm and the secretory granules (1) and its concentration in secretory granules is maintained (2) by the two vesicular
monoamine transporters (VMAT). The monoamine is secreted by exocytosis of the granules (3), and it acts (4) on receptors (Y-shaped
structures labeled R). Many of these receptors are postsynaptic, but some are presynaptic and some are located on glia. In addition, there
is extensive reuptake into the cytoplasm of the presynaptic terminal (5) via the mo noamine neurotransmitter transporter (NTT) for the
monoamine that is synthesized in the neuron. (Reproduced with permission from Hoffman BJ, et al: Distribution of monoamine neurotransmitter transporters
in the rat brain. Front Neuroendocrinol 1998;19:187.)
and anoxia, loss of neurons is increased because glutamate reuptake is inhibited.
SMALL-MOLECULE TRANSMITTERS
Synaptic physiology is a rapidly expanding, complex field that cannot be covered in de tail in this book. However, it is appropriate to
Synaptic physiology is a rapidly expanding, complex field that cannot be covered in de tail in this book. However, it is appropriate to
summarize information about the principal neurotransmitters and their receptors.
MONOAMINES
Acetylcholine
Acetylcholine, which is the acetyl ester of choline, is largely enclosed in small, clear syna ptic vesicles in high concentration in the
terminal boutons of neurons that release acetylcholine (cholinergic neurons). Synthesis of ac etylcholine involves the reaction of choline
with acetate (Figure 7–1). Acetylcholine is the transmitter at the neuromuscular junction, in autonomic ganglia, and in postganglionic
parasympathetic nerve-target organ junctions and some postganglionic sympathetic nerv e-target junctions. It is also found within the
brain, including the basal forebrain complex and pontomesencephalic cholinergic comp lex (Figure 7–2). These systems may be involved
in regulation of sleep-wake states, learning, and memory.
Cholinergic neurons actively take up choline via a transporter (Figure 7–4). Choline is also synthesized in neurons. The acetate is
activated by the combination of acetate groups with reduced coenzyme A. The reaction between active acetate (acetyl-coenzyme A,
acetyl-CoA) and choline is catalyzed by the enzyme choline acetyltransferase. This enzyme is foun d in high concentration in the
cytoplasm of cholinergic
CLINICAL BOX 7–1
Cholinergic neuron
Excitotoxins
Glutamate is usually cleared from the brain’s extracellular fluid by Na
+
-dependent uptake systems in neurons and glia, keeping only
micromolar levels of the chemical in the extracellular fluid despite millimolar levels inside neurons. However, excessive levels of
glutamate occur in response to ischemia, anoxia, hypoglycemia, or trauma. Glutamate an d some of its synthetic congeners are unique in
that when they act on neuronal cell bodies, they can produce so much Ca
2+
influx that neurons die. This is the reason why
microinjection of these excitotoxins is used in research to produce discrete lesions that destro y neuronal cell bodies without affecting
neighboring axons. Evidence is accumulating that excitotoxins play a significant role in t he damage done to the brain by a stroke. When
a cerebral artery is occluded, the cells in the severely ischemic area die. Surrounding pa rtially ischemic cells may survive but lose their
ability to maintain the transmembrane Na
+
gradient. The elevated levels of intracellular Na
+
prevent the ability of astrocytes to remove
glutamate from the brain’s extracellular fluid. Therefore, glutamate accumulates to the p oint that excitotoxic damage and cell death
occurs in the penumbra, the region around the completely infarcted area.
nerve endings. Acetylcholine is then taken up into synaptic vesicles by a vesicular transp orter, VAChT.
Cholinesterases
Acetylcholine must be rapidly removed from the synapse if repolarization is to occur. T he removal occurs by way of hydrolysis of
acetylcholine to choline and acetate, a reaction catalyzed by the enzyme acetylcholinesterase. This enzyme is also called true or specific
cholinesterase. Its greatest affinity is for acetylcholine, but it also hydrolyzes other ch oline esters. There are a variety of esterases in the
body. One found in plasma is capable of hydrolyzing acetylcholine but has different pr operties from acetylcholinesterase. It is therefore
called pseudocholinesterase or nonspecific cholinesterase. The plasma moiety is partly under endocr ine control and is affected by
variations in liver function. On the other hand, the specific cholinesterase molecules are clustered in the postsynaptic membrane of
cholinergic synapses. Hydrolysis of acetylcholine by this enzyme is rapid enough to exp lain the observed changes in Na
+
conductance
and electrical activity during synaptic transmission.
Acetylcholine Receptors
Historically, acetylcholine receptors have been divided into two main types on the basis of their pharmacologic properties. Muscarine,
the alkaloid responsible for the toxicity of
Acetyl-CoA +
Choline
ACh
Choline ACh
ASE
Postsynaptic tissue
FIGURE 7–4 Biochemical events at cholinergic endings. ACh, acetylcholine; ASE, acetylcholinesterase; X, recept or.
toadstools, has little effect on the receptors in autonomic ganglia but mimics the stimulator y action of acetylcholine on smooth muscle
and glands. These actions of acetylcholine are therefore called muscarinic actions, and the receptors involved are muscarinic
cholinergic receptors. They are blocked by the drug atropine. In sympathetic ganglia, small am ounts of acetylcholine stimulate
postganglionic neurons and large amounts block transmission of impulses from pregang lionic to postganglionic neurons. These actions
are unaffected by atropine but mimicked by nicotine. Consequently, these actions of ac etylcholine are nicotinic actions and the receptors
are nicotinic cholinergic receptors. Nicotinic receptors are subdivided into th ose found in muscle at neuromuscular junctions and those
found in autonomic ganglia and the central nervous system. Both muscarinic and nicotin ic acetylcholine receptors are found in large
numbers in the brain.
The nicotinic acetylcholine receptors are members of a superfamily of ligand-gated ion channels that also includes the GABA
A
and
glycine receptors and some of the glutamate receptors. They are made up of multiple su bunits coded by different genes. Each nicotinic
cholinergic receptor is made up of five subunits that form a central channel which, whe n the receptor is activated, permits the passage of
Na
+
and other cations. The 5 subunits come from a menu of 16 known subunits, α
1
–α
9
, β
2
–β
5
, γ, δ, and ε, coded by 16 different genes.
Some of the receptors are homomeric—for example, those that contain five α
7
subunits—but most are heteromeric. The muscle type
nicotinic receptor found in the fetus is made up of two α
1
subunits, a β
1
subunit, a γ subunit, and a δ subunit (Figure 7–5). In adult
mammals, the γ subunit is replaced by a δ subunit, which decreases the channel open tim e but increases its
No ACh bound: Two ACh molecules bound: Channel closed Channel open
ACh
Na+
K
+
FIGURE 7–5 Three-dimensional model of the nicotinic acetylcholine-gated ion channel. The receptor–channel complex consists
of five subunits, all of which contribute to forming the pore. When two molecules of acetylcho line bind to portions of the
α
-subunits
exposed to the membrane surface, the receptor–channel changes conformation. This opens the pore in the portion of the channel
emnbedded in the lipid bilayer, and both K
+
and Na
+
flow through the open channel down their electrochemical gradient. (From Kandel ER ,
Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
conductance. The nicotinic cholinergic receptors in autonomic ganglia are heteromers th at usually contain α
3
subunits in combination
with others, and the nicotinic receptors in the brain are made up of many other subunits . Many of the nicotinic cholinergic receptors in
the brain are located presynaptically on glutamate-secreting axon terminals, and they fac ilitate the release of this transmitter. However,
others are postsynaptic. Some are located on structures other than neurons, and some s eem to be free in the interstitial fluid, that is, they
are perisynaptic in location.
Each α subunit has a binding site for acetylcholine, and when an acetylcholine molecul e binds to each of them, they induce a
confirmational change in the protein so that the channel opens. This increases the condu ctance of Na
+
and other cations, and the resulting
influx of Na
+
produces a depolarizing potential. A prominent feature of neuronal nicotinic cholinerg ic receptors is their high
permeability to Ca
2+
.
Muscarinic cholinergic receptors are very different from nicotinic cholinergic receptor s. Five types, encoded by five separate genes, have
been cloned. The exact status of M
5
is uncertain, but the remaining four receptors are coupled via G proteins to adenylyl cy clase, K
+
channels, and/or phospholipase C (Table 7–2). The nomenclature of these receptors ha s not been standardized, but the receptor
designated M
1
in Table 7–2 is abundant in the brain. The M
2
receptor is found in the heart. The M
4
receptor is found in pancreatic acinar
and islet tissue, where it mediates increased secretion of pancreatic enzymes and insulin. The M
3
and M
4
receptors are associated with
smooth muscle.
Serotonin
Serotonin (5-hydroxytryptamine; 5-HT) is present in highest concentration in blood pla telets and in the gastrointestinal tract, where it is
found in the enterochromaffin cells and the myenteric plexus. It is also found within the brain stem in the midline raphé nuclei which
project to portions of the hypothalamus, the limbic system, the neocortex, the cerebellum , and the spinal cord (Figure 7–2).
Serotonin is formed in the body by hydroxylation and decarboxylation of the essential amino acid tryptophan (Figures 7–1 and 7–6).
After release from serotonergic neurons, much of the released serotonin is recaptured by an active reuptake mechanism and inactivated
by monoamine oxidase (MAO) to form 5-hydroxyindoleacetic acid (5-HIAA). This s ubstance is the principal urinary metabolite of
serotonin, and urinary output of 5-HIAA is used as an index of the rate of serotonin m etabolism in the body.
Tryptophan hydroxylase in the human CNS is slightly different from the tryptophan hy droxylase in peripheral tissues, and is coded by a
different gene. This is presumably why knockout of the TPH1 gene, which codes for tryptophan hydroxylase in peripheral tissues, has
much less effect on brain serotonin production than on peripheral serotonin production .
As described in Clinical Box 7–2, there is evidence for a relationship between behavior and brain serotonin content.
Serotonergic Receptors
The number of cloned and characterized serotonin receptors has increased rapidly. Th ere are at least seven types of 5-HT
Serotonergic neuron
CLINICAL BOX 7–2
L
-Tryptophan
5-HTP
MAO 5-HIAA
5-HT
Reuptake 5-HT
Postsynaptic tissue
FIGURE 7–6 Biochemical events at serotonergic synapses. Biochemical events at ser otonergic synapses. Biochemical events at
serotonergic synapses. HIAA, 5-hydroxyindoleacetic acid; X, serotonin receptor. F or clarity, the presynaptic receptors have been
omitted.
receptors (from 5-HT
1
through 5-HT
7
receptors). Within the
5-HT
1
group are the 5-HT
1A
, 5-HT
1B
, 5-HT
1D
, 5-HT
1E
, and
5-HT
1F
subtypes. Within the 5-HT
2
group there are 5-HT
2A
,
5-HT
2B
, and 5-HT
2C
subtypes. There are two 5-HT
5
subtypes: 5-HT
5A
and 5-HT
5B
. Most of these are G protein-coupled receptors and
affect adenylyl cyclase or phospholipase C (Table 7–2). However, the 5-HT
3
receptors, like nicotinic cholinergic receptors, are ligand-
gated ion channels. Some of the serotonin receptors are presynaptic, and others are po stsynaptic.
5-HT
2A
receptors mediate platelet aggregation and smooth muscle contraction. Mice in which th e gene for 5-HT
2C
receptors has been
knocked out are obese as a result of increased food intake despite normal responses to leptin, and they are prone to fatal seizures. 5-HT
3
receptors are present in the gastrointestinal tract and the area postrema and are related to vomiting. 5-HT
4
receptors are also present in
the gastrointestinal tract, where they facilitate secretion and peristalsis, and in the brain. 5 -HT
6
and 5-HT
7
receptors in the brain are
distributed throughout the limbic system, and the 5-HT
6
receptors have a high affinity for antidepressant drugs.
Histamine
Histaminergic neurons have their cell bodies in the tuberomammillary nucleus of the pos terior hypothalamus, and their axons project to
all parts of the brain, including the cerebral cortex and the spinal cord. Histamine is also found in cells in the gastric mucosa and in
heparin-containing cells called mast cells that are plentiful in the anterior and posterior lobes of the pituitary gland as well as at body
surfaces.
Role of Serotonin in Mood & Behavior
The hallucinogenic agent lysergic acid diethylamide (LSD) is a serotonin agonist that p roduces its effects by activating 5-HT
2
receptors
in the brain. The transient hallucinations and other mental aberrations produced by this d rug were discovered when the chemist who
synthesized it inhaled some by accident. Its discovery called attention to the correlation b etween behavior and variations in brain
serotonin content. Psilocin (and its phosphorylated form, psilocybin), a substance fo und in certain mushrooms, and N,N-
dimethyltryptamine (DMT) are also hallucinogenic and, like serotonin, are deriv atives of tryptamine. 2,5-Dimethoxy-4-
methylamphetamine (DOM) and mescaline and its congeners, the other true hal lucinogens, are phenylethylamines rather than
indolamines. However, all these hallucinogens appear to exert their effects by binding to 5-HT
2
receptors. 3,4-
Methylenedioxymethamphetamine, a drug known as MDMA or ecstasy, is a popular dru g of abuse. It produces euphoria, but this is
followed by difficulty in concentrating, depression, and, in monkeys, insomnia. The dr ug causes release of serotonin followed by
serotonin depletion; the euphoria may be due to the release and the later symptoms to the depletion.
Drugs that increase extracellular norepinephrine levels in the brain elevate mood, and d rugs that decrease extracellular norepinephrine
levels cause depression. However, individuals with congenital dopamine β-hydroxylase (DBH) deficiency are normal as far as mood is
concerned. Drugs that inhibit norepinephrine reuptake were of considerable value in th e treatment of depression, but these drugs also
inhibit serotonin reuptake. It is also known that the primary serotonin metabolite 5-HIAA is l ow in CSF of depressed individuals. Drugs
such as fluoxetine (Prozac), which inhibit serotonin reuptake without affecting norepinephrine reuptake, are effective as
antidepressants. Thus, the focus in treating clinical depression has shifted from norepine phrine to serotonin.
Histamine is formed by decarboxylation of the amino acid histidine (Figure 7–1). Histam ine is converted to methylhistamine or,
alternatively, to imidazoleacetic acid. The latter reaction is quantitatively less important in h umans. It requires the enzyme diamine
oxidase (histaminase) rather than MAO, even though MAO catalyzes the oxidation of m ethylhistamine to methylimidazoleacetic acid.
The three known types of histamine receptors—H
1
, H
2
, and H
3
—are all found in both peripheral tissues and the brain. Most, if not all,
of the H
3
receptors are presynaptic, and they mediate inhibition of the release of histamine and o ther transmitters via a G protein. H
1
receptors activate phospholipase C, and H
2
receptors increase the intracellular cAMP concentration. The function of this diffuse
histaminergic system is unknown, but evidence links brain histamine to arousal, sexual b ehavior, blood pressure, drinking, pain
thresholds, and regulation of the secretion of several anterior pituitary hormones.
CATECHOLAMINES
Norepinephrine & Epinephrine
The chemical transmitter present at most sympathetic postganglionic endings is norepinep hrine. It is stored in the synaptic knobs of the
neurons that secrete it in characteristic small vesicles that have a dense core (granulated v esicles; see above). Norepinephrine and its
methyl derivative, epinephrine, are secreted by the adrenal medulla, but epinephrine is n ot a mediator at postganglionic sympathetic
endings. As discussed in Chapter 6, each sympathetic postganglionic neuron has multipl e varicosities along its course, and each of these
varicosities appears to be a site at which norepinephrine is secreted.
There are also norepinephrine-secreting and epinephrinesecreting neurons in the brain . Norepinephrine-secreting neurons are properly
called noradrenergic neurons, although the term adrenergic neurons is also applied. However, it seems appropriate to reserve the
latter term for epinephrine-secreting neurons. The cell bodies of the norepinephrine-co ntaining neurons are located in the locus ceruleus
and other medullary and pontine nuclei (Figure 7–2). From the locus ceruleus, the axon s of the noradrenergic neurons form the locus
ceruleus system. They descend into the spinal cord, enter the cerebellum, and asc end to innervate the paraventricular, supraoptic, and
periventricular nuclei of the hypothalamus, the thalamus, the basal telencephalon, and th e entire neocortex.
Biosynthesis & Release of Catecholamines
The principal catecholamines found in the body—norepinephrine, epinephrin e, and dopamine—are formed by hydroxylation and
decarboxylation of the amino acid tyrosine (Figure 7–1). Some of the tyrosine is forme d from phenylalanine, but most is of dietary
origin. Phenylalanine hydroxylase is found primarily in the liver (see Clinical Bo x 7–3). Tyrosine is transported into catecholamine-
secreting neurons and adrenal medullary cells by a concentrating mechanism. It is conv erted to dopa and then to dopamine in the
cytoplasm of the cells by tyrosine hydroxylase and dopa decarboxylase. The decarboxylase, which is also called aromatic L-amino
acid decarboxylase, is very similar but probably not identical to 5-hydroxytryptophan d ecarboxylase. The dopamine then enters the
granulated vesicles, within which it is converted to norepinephrine by dopamine β-hydroxylase (DBH). L-Dopa is the isomer involved,
but the norepinephrine that is formed is in the D configuration. The rate-limiting step in synthesis is the conversion of tyrosine to dopa.
Tyrosine hydroxylase, which catalyzes this step, is subject to feedback inhibition by dop amine and norepinephrine, thus providing
internal control of the synthetic process. The cofactor for tyrosine hydroxylase is
CLINICAL BOX 7–3 Phenylketonuria
Phenylketonuria is a disorder characterized by severe mental deficiency and the accumulation in the blood, tissues, and urine of large
amounts of phenylalanine and its keto acid derivatives. It is usually due to de creased function resulting from mutation of the gene for
phenylalanine hydroxylase. This gene is located on the long arm of chromoso me 12. Catecholamines are still formed from tyrosine,
and the cognitive impairment is largely due to accumulation of phenylalanine and its der ivatives in the blood. Therefore, it can be treated
with considerable success by markedly reducing the amount of phenylalanine in the die t. The condition can also be caused by
tetrahydrobiopterin (BH4) deficiency. Because BH4 is a cofactor for tyrosine hydr oxylase and tryptophan hydroxylase, as well as
phenylalanine hydroxylase, cases due to tetrahydrobiopterin deficiency have catechola mine and serotonin deficiencies in addition to
hyperphenylalaninemia. These cause hypotonia, inactivity, and developmental problems . They are treated with tetrahydrobiopterin,
levodopa, and 5-hydroxytryptophan in addition to a low-phenylalanine diet. BH4 is als o essential for the synthesis of nitric oxide (NO)
by nitric oxide synthase. Severe BH4 deficiency can lead to impairment of NO formatio n, and the CNS may be subjected to increased
oxidative stress.
tetrahydrobiopterin, which is converted to dihydrobiopterin when tyrosine is con verted to dopa.
Some neurons and adrenal medullary cells also contain the cytoplasmic enzyme phenylethanolamine-Nmethyltransferase (PNMT),
which catalyzes the conversion of norepinephrine to epinephrine. In these cells, norepi nephrine apparently leaves the vesicles, is
converted to epinephrine, and then enters other storage vesicles.
In granulated vesicles, norepinephrine and epinephrine are bound to ATP and associat ed with a protein called chromogranin A. In some
but not all noradrenergic neurons, the large granulated vesicles also contain neuropepti de Y. Chromogranin A is a 49-kDa acid protein
that is also found in many other neuroendocrine cells and neurons. Six related chro mogranins have been identified. They have been
claimed to have multiple intracellular and extracellular functions. Their level in the plasma is elevated in patients with a variety of tumors
and in essential hypertension, in which they probably reflect increased sympathetic activ ity. However, their specific functions remain
unsettled.
The catecholamines are transported into the granulated vesicles by two vesicular transpo rters, and these transporters are inhibited by the
drug reserpine.
Catecholamines are released from autonomic neurons and adrenal medullary cells by ex ocytosis. Because they are present in the
granulated vesicles, ATP, chromogranin A, and the dopamine β hydroxylase that is not mem brane-bound are released with
norepinephrine and epinephrine. The half-life of circulating dopamine β-hydroxylase is much longer than that of the catecholamines, and
circulating levels of this substance are affected by genetic and other factors in addition t o the rate of sympathetic activity.
Noradrenergic
neuron
Dopa Tyrosine
Dopamine
Catabolism of Catecholamines
Norepinephrine, like other amine and amino acid transmitters, is removed from the syna ptic cleft by binding to postsynaptic receptors,
binding to presynaptic receptors (Figure 7–3), reuptake into the presynaptic neurons, o r catabolism. Reuptake is a major mechanism in
the case of norepinephrine, and the hypersensitivity of sympathetically denervated struc tures is explained in part on this basis. After the
noradrenergic neurons are cut, their endings degenerate with loss of reuptake in them. Consequently, more norepinephrine from other
sources is available to stimulate the receptors on the autonomic effectors.
Epinephrine and norepinephrine are metabolized to biologically inactive products by ox idation and methylation. The former reaction is
catalyzed by MAO and the latter by catechol-Omethyltransferase (COMT). MAO is located on the o uter surface of the mitochondria.
It has two isoforms, MAO-A and MAO-B, which differ in substrate specificity and sen sitivity to drugs. Both are found in neurons. MAO
is widely distributed, being particularly plentiful in the nerve endings at which catecholam ines are secreted. COMT is also widely
distributed, particularly in the liver, kidneys, and smooth muscles. In the brain, it is prese nt in glial cells, and small amounts are found in
postsynaptic neurons, but none is found in presynaptic noradrenergic neurons. Conse quently, catecholamine metabolism has two
different patterns.
Extracellular epinephrine and norepinephrine are for the most part O-methylated, and m easurement of the concentrations of the O-
methylated derivatives normetanephrine and metanephrine in the urine is a good index of the rate of secretion of norepinephrine and
epinephrine. The O-methylated derivatives that are not excreted are largely oxidized, an d 3-methoxy-4-hydroxymandelic acid
(vanillylmandelic acid, VMA) is the most plentiful catecholamine metabolite in the urine. Small amounts of the O-methylated
derivatives are also conjugated to sulfate and glucuronide.
In the noradrenergic nerve terminals, on the other hand, some of the norepinephrine is constantly being converted by intracellular MAO
(Figure 7–7) to the physiologically inactive deaminated derivatives, 3,4-dihydroxymand elic acid (DOMA) and its corresponding glycol
(DHPG). These are subsequently converted to their corresponding O-methyl derivativ es, VMA and 3-methoxy-4-hydroxyphenylglycol
(MHPG).
α & β Receptors
Epinephrine and norepinephrine both act on α and β receptors, with norepinephrine ha ving a greater affinity for α-adrenergic
MAO
Deaminated
NE
derivatives
Reuptake NE
COMT
Normetanephrine
Postsynaptic tissue
FIGURE 7–7 Biochemical events at noradrenergic endings. NE, norepinephrine; COMT, c atecholOmethyltransferase; MAO,
monoamine oxidase; X, receptor. For clarity, the presynaptic receptors have b een omitted. Note that MAO is intracellular, so that
norepinephrine is being constantly deaminated in noradrenergic endings. COMT acts primarily on secreted norepinephrine.
receptors and epinephrine for β-adrenergic receptors. As noted previously, the α a nd β receptors are typical G protein-coupled receptors,
and each has multiple forms. They are closely related to the cloned receptors for dopam ine and serotonin and to muscarinic acetylcholine
receptors.
Clonidine lowers blood pressure when administered centrally. It is an α
2
agonist and was initially thought to act on presynaptic α
2
receptors, reducing central norepinephrine discharge. However, its structure resembles that of imidazoline, and it binds to imidazoline
receptors with higher affinity than to α
2
adrenergic receptors. A subsequent search led to the discovery that imidazoline recepto rs occur
in the nucleus tractus solitarius and the ventrolateral medulla. Administration of imidazolin es lowers blood pressure and has a depressive
effect. However, the full significance of these observations remains to be explored.
Dopamine
In certain parts of the brain, catecholamine synthesis stops at dopamine (Figure 7–1) wh ich can then be secreted into the synaptic cleft.
Active reuptake of dopamine occurs via a Na
+
- and Cl
-dependent dopamine transporter. Dopamine is metabolized to inactive
compounds by MAO and COMT in a tabolized to inactive compounds by MAO and C OMT in a Dihydroxyphenylacetic acid (DOPAC)
and homovanillic acid (HVA) are conjugated, primarily to sulfate.
Dopaminergic neurons are located in several brain regions including the nigrostriatal system, which p rojects from the substantia nigra
to the striatum and is involved in motor control, and the mesocortical system, which a rises primarily in the ventral tegmental area
(Figure 7–2). The mesocortical system projects to the nucleus accumbens and limbic sub cortical areas, and it is involved in reward
behavior and addiction. Studies by PET scanning in normal humans show that a steady loss of dopamine receptors occurs in the basal
ganglia with age. The loss is greater in men than in women.
Dopamine Receptors
Five different dopamine receptors have been cloned, and several of these exist in multip le forms. This provides for variety in the type of
responses produced by dopamine. Most, but perhaps not all, of the responses to these r eceptors are mediated by heterotrimeric G
proteins. One of the two forms of D
2
receptors can form a heterodimer with the somatostatin SST5 receptor, further increasi ng the
dopamine response menu. Overstimulation of D
2
receptors is thought to be related to schizophrenia (see Clinical Box 7–4). D
3
receptors
are highly localized, especially to the nucleus accumbens (Figure 7–2). D
4
receptors have a greater affinity than the other dopamine
receptors for the “atypical” antipsychotic drug clozapine, which is effective in schizophre nia but produces fewer extrapyramidal side
effects than the other major tranquilizers do.
EXCITATORY & INHIBITORY
AMINO ACIDS
Glutamate
The amino acid glutamate is the main excitatory transmitter in the brain and spinal cord, and it has been c alculated that it is the
transmitter responsible for 75% of the excitatory transmission in the brain. Glutamate is formed by reductive amination of the Krebs
cycle intermediate α-ketoglutarate in the cytoplasm. The reaction is reversible, but in glut aminergic neurons, glutamate is concentrated in
synaptic vesicles by the vesicle-bound transporter BPN1. The cytoplasmic store of glutamine is enriched by three transporters that
import glutamate from the interstitial fluid, and two additional transporters carry glutamat e into astrocytes, where it is converted to
glutamine and passed on to glutaminergic neurons. The interaction of astrocytes and glu taminergic neurons is shown in Figure 7–8.
Released glutamate is taken up by astrocytes and converted to glutamine, which passes b ack to the neurons and is converted back to
glutamate, which is released as the synaptic transmitter. Uptake into neurons and astrocy tes is the main mechanism for removal of
glutamate from synapses.
Glutamate Receptors
Glutamate receptors are of two types: metabotropic receptors and ionotropic receptors. The metabo tropic receptors are G protein-
coupled receptors that increase intracellular IP
3
and
CLINICAL BOX 7-4 Schizophrenia
Schizophrenia is an illness that involves deficits of multiple brain systems that alter a n individual’s inner thoughts as well as their
interactions with others. Individuals with schizophrenia suffer from hallucinations, delus ions, and racing thoughts (positive symptoms);
and they experience apathy, difficulty dealing with novel situations, and little spontaneity or motivation (negative symptoms).
Worldwide, about 1–2% of the population lives with schizophrenia. A combination of genetic, biological, cultural, and psychological
factors contributes to the illness. A large amount of evidence indicates that a defect in the mesocortical system is responsible for the
development of at least some of the symptoms of schizophrenia. Attention was initially fo cused on overstimulation of limbic D
2
dopamine receptors. Amphetamine, which causes release of dopamine as well as norepine phrine in the brain, causes a
schizophrenialike psychosis; brain levels of D
2
receptors are said to be elevated in schizophrenics; and there is a clear positive
correlation between the antischizophrenic activity of many drugs and their ability to bloc k D
2
receptors. However, several recently
developed drugs are effective antipsychotic agents but bind D
2
receptors to a limited degree. Instead, they bind to D
4
receptors, and there
is active ongoing research into the possibility that these receptors are abnormal in individ uals with schizophrenia.
Lactate Capillary
Glucose
34 ATP
Gln
Gln Lactate
Glu ATP
Glu Glu ATP
3Na
+ 3Na
+
2K+
Glutaminergic synapse
Astrocyte
FIGURE 7–8 The glutamate–glutamine cycle through glutaminergic neurons and as trocytes. Glutamate released into the synaptic
cleft is taken up by a Na
+
-dependent glutamate transporter, and in the astrocyte it is converted to glutamine. The glutamin e enters the
neuron and is converted to glutamate. Glucose is transported out of capillaries and enters astrocytes and neurons. In astrocytes, it is
metabolized to lactate, producing two ATPs. One of these powers the conversion of glut amate to glutamine, and the other is used by
Na
+
– K
+
ATPase to transport three Na
+
out of the cell in exchange for two K
+
. In neurons, the glucose is metabolized further through
the citric acid cycle, producing 34 ATPs.
DAG levels or decrease intracellular cAMP levels. Eleven subtypes have been identified (Table 7–2). They are both presynaptic and
postsynaptic, and they are widely distributed in the brain. They appear to be involved in the production of synaptic plasticity, particularly
in the hippocampus and the cerebellum. Knockout of the gene for one of these recepto rs, one of the forms of mGluR1, causes severe
motor incoordination and deficits in spatial learning.
The ionotropic receptors are ligand-gated ion channels that resemble nicotinic cholinerg ic receptors and GABA and glycine receptors.
There are three general types, each named for the congeners of glutamate to which the y respond in maximum fashion. These are the
kainate receptors (kainate is an acid isolated from seaweed), AMPA receptors (for αα hydroxy-5-methylisoxazole-4-propionate), and
NMDA receptors (for Nmethyl-D-aspartate). Four AMPA, five kainate, and s ix NMDA subunits have been identified, and each is coded
by a different gene. The receptors were initially thought to be pentamers, but some may be tetramers, and their exact stoichiometry is
unsettled.
The kainate receptors are simple ion channels that, when open, permit Na
+
influx and K
+
efflux. There are two populations of AMPA
receptors: one is a simple Na
+
channel and one also passes Ca
2+
. The balance between the two in a given synapse can be shifted by
activity.
The NMDA receptor is also a cation channel, but it permits passage of relatively large a mounts of Ca
2+
, and it is unique in several ways
(Figure 7–9). First, glycine facilitates its function by binding to it, and glycine appears to be essential for its normal response to
glutamate. Second, when glutamate binds to it, it opens, but at normal membrane potentia ls, its channel is blocked by a Mg
2+
ion. This
block is removed only when the neuron containing the receptor is partially depolarized by activation of AMPA or other channels that
produce rapid depolarization via other synaptic circuits. Third, phencyclidine and ketam ine, which produce amnesia and a feeling of
dissociation from the environment, bind to another site inside the channel. Most target ne urons for glutamate have both AMPA and
NMDA receptors. Kainate receptors are located presynaptically on GABA-secreting n erve endings and postsynaptically at various
localized sites in the brain. Kainate and AMPA receptors are found in glia as well as neu rons, but it appears that NMDA receptors occur
only in neurons.
The concentration of NMDA receptors in the hippocampus is high, and blockade of th ese receptors prevents long-term potentiation, a
long-lasting facilitation of transmission in neural pathways following a brief period of h igh-frequency stimulation. Thus, these receptors
may well be involved in memory and learning.
GABA
GABA is the major inhibitory mediator in the brain, including being responsible for pre synaptic inhibition. GABA, which exists as β-
aminobutyrate in the body fluids, is formed
L-Glutamate Ca
2+
Na
+
K + Glycine
Mg
2+
Extracellular MK-801
Intracellular
Channel blocker
Open ion channel
Closed ion channel
FIGURE 7–9 Diagrammatic representation of the NMDA receptor. When glyc ine and glutamate bind to the receptor, the closed ion
channel (left) opens, but at the resting membrane potential, the channel is bloc ked by Mg
2+
(right). This block is removed if partial
depolarization is produced by other inputs to the neuron containing the receptor, and C a
2+
and Na
+
enter the neuron. Blockade can also
be produced by the drug dizocilpine maleate (MK-801).
by decarboxylation of glutamate (Figure 7–1). The enzyme that catalyzes this reaction is glutamate decarboxylase (GAD), which is
present in nerve endings in many parts of the brain. GABA is metabolized primarily by transamination to succinic semialdehyde and
thence to succinate in the citric acid cycle. GABA transaminase (GABA-T) is the enzyme that catalyzes the transamination. Pyridoxal
phosphate, a derivative of the B complex vitamin pyridoxine, is a cofactor for GAD an d GABA-T. In addition, there is an active
reuptake of GABA via the GABA transporter. A vesicular GABA transporter (VGAT ) transports GABA and glycine into secretory
vesicles.
GABA Receptors
Three subtypes of GABA receptors have been identified: GABA
A
, GABA
B
, and GABA
C
. The GABA
A
and GABA
B
receptors are
widely distributed in the CNS, whereas in adult vertebrates the GABA
C
receptors are found almost exclusively in the retina. The
GABA
A
and GABA
C
receptors are ion channels made up of five subunits surrounding a pore, like the nicot inic acetylcholine receptors
and many of the glutamate receptors. In this case, the ion is Cl
(Figure 7–10). The GABA
B
receptors are metabotropic and are coupled
to heterotrimeric G proteins that increase conductance in K
+
channels, inhibit adenylyl cyclase, and inhibit Ca
2+
influx. Increases in Cl
influx and K
+
efflux and decreases in Ca
2+
influx all hyperpolarize neurons, producing an IPSP. The G protein mediation of GAB A
B
receptor effects is unique in that a G protein heterodimer, rather than a single protein, is involved.
The GABA
C
receptors are relatively simple in that they are pentamers of three ρ subunits in various combinations. On
GABA
A
GABA
B
NH
2
Extracellular
K+ Ca
2
+
β
α α
β
γ γ
AC
Cl−
COOH
Intracellular
FIGURE 7–10 Diagram of GABA
A
and GABA
B
receptors, showing their principal actions. The G protein that mediates the effects
of GABA
B
receptors is a heterodimer. (Reproduced with permission from Bowery NG, Brown DA: T he cloning of GABA
B
receptors. Nature 1997;386:223.
Copyright © 1997 by Macmillan Magazines.)
the other hand, the GABA
A
receptors are pentamers made up of various combinations of six α subunits, four β, fo ur γ, one δ, and one ε.
This endows them with considerably different properties from one location to another.
An observation of considerable interest is that there is a chronic low-level stimulation of GABA
A
receptors in the CNS that is aided by
GABA in the interstitial fluid. This background stimulation cuts down on the “noise” cau sed by incidental discharge of the billions of
neural units and greatly improves the signal-to-noise ratio in the brain. It may be that this GABA discharge declines with advancing age,
resulting in a loss of specificity of responses of visual neurons. Support for this hypoth esis comes from studies in which microinjection of
GABA in older monkeys resulted in restoration of the specificity of visual neurons.
The increase in Cl
conductance produced by GABA
A
receptors is potentiated by benzodiazepines, drugs that have marked anti-anxiety
activity and are also effective muscle relaxants, anticonvulsants, and sedatives. Benzodia zepines bind to the α subunits. Diazepam and
other benzodiazepines are used throughout the world. At least in part, barbiturates and alcohol also act by facilitating Cl
conductance
through the Cl
channel. Metabolites of the steroid hormones progesterone and deoxycorticosterone b ind to GABA
A
receptors and
increase Cl
conductance. It has been known for many years that progesterone and deoxycorticos terone are sleep-inducing and
anesthetic in large doses, and these effects are due to their action on GABA
A
receptors.
A second class of benzodiazepine receptors is found in steroid-secreting endocrine gla nds and other peripheral tissues, and hence these
receptors are called peripheral benzodiazepine receptors. They may be involved in s teroid biosynthesis, possibly performing a
function like that of the StAR protein in moving steroids into the mitochondria. Another possibility is a role in the regulation of cell
proliferation. Peripheral-type benzodiazepine receptors are also present in astrocytes in the brain, and they are found in brain tumors.
Glycine
Glycine has both excitatory and inhibitory effects in the CNS. When it binds to NMDA receptors, it makes them more sensitive. It
appears to spill over from synaptic junctions into the interstitial fluid, and in the spinal cor d, for example, this glycine may facilitate pain
transmission by NMDA receptors in the dorsal horn. However, glycine is also respons ible in part for direct inhibition, primarily in the
brain stem and spinal cord. Like GABA, it acts by increasing Cl
conductance. Its action is antagonized by strychnine. The clinical
picture of convulsions and muscular hyperactivity produced by strychnine emphasizes the importance of postsynaptic inhibition in
normal neural function. The glycine receptor responsible for inhibition is a Cl
channel. It is a pentamer made up of two subunits: the
ligand-binding α subunit and the structural β subunit. Recently, solid evidence has been presented that three kinds of neurons are
responsible for direct inhibition in the spinal cord: neurons that secrete glycine, neurons that secrete GABA, and neurons that secrete
both. Presumably, neurons that secrete only glycine have the glycine transporter GLYT 2, those that secrete only GABA have GAD, and
those that secrete glycine and GABA have both. This third type of neuron is of special interest because the neurons seem to have glycine
and GABA in the same vesicles.
Anesthesia
Although general anesthetics have been used for millennia, little has been understood ab out their mechanisms of action. However, it now
Although general anesthetics have been used for millennia, little has been understood ab out their mechanisms of action. However, it now
appears that alcohols, barbiturates, and many volatile inhaled anesthetics as well act on io n channel receptors and specifically on
GABA
A
and glycine receptors to increase Cl
conductance. Regional variation in anesthetic actions in the CNS seems to parallel the
variation in subtypes of GABA
A
receptors. Other inhaled anesthetics do not act by increasing GABA receptor activity, b ut appear to act
by inhibiting NMDA and AMPA receptors instead.
In contrast to general anesthetics, local anesthetics produce anesthesia by blocking cond uction in peripheral nerves via reversibly binding
to and inactivating Na
+
channels. Na
+
influx through these channels normally causes depolarization of nerve cell membranes and
propagation of impulses toward the nerve terminal. When depolarization and propagati on are interrupted, the individual loses sensation
in the area supplied by the nerve.
pled receptors. Activation of the substance P receptor causes activation of phospholipas e C and increased formation of IP
3
and DAG.
Substance P is found in high concentration in the endings of primary afferent neurons in the spinal cord, and it is probably the mediator
at the first synapse in the pathways for pain transmission in the dorsal horn. It is also fou nd in high concentrations in the nigrostriatal
system, where its concentration is proportional to that of dopamine, and in the hypothala mus, where it may play a role in neuroendocrine
regulation. Upon injection into the skin, it causes redness and swelling, and it is probably the mediator released by nerve fibers that is
responsible for the axon reflex. In the intestine, it is involved in peristalsis. It has recently been reported that a centrally active NK-1
receptor antagonist has antidepressant activity in humans. This antidepressant effect take s time to develop, like the effect of the
antidepressants that affect brain monoamine metabolism, but the NK-1 inhibitor does no t alter brain monoamines in experimental
animals. The functions of the other tachykinins are unsettled.
LARGE-MOLECULE TRANSMITTERS: NEUROPEPTIDES
Substance P & Other Tachykinins
Substance P is a polypeptide containing 11 amino acid residues that is found in the intesti ne, various peripheral nerves, and many parts
of the CNS. It is one of a family of six mammalian polypeptides called tachykinins that d iffer at the amino terminal end but have in
common the carboxyl terminal sequence of Phe-X-Gly-LeuMet-NH
2
, where X is Val, His, Lys, or Phe. The members of the family are
listed in Table 7–3. There are many related tachykinins in other vertebrates and in inver tebrates.
The mammalian tachykinins are encoded by two genes. The neurokinin B gene encodes only o ne known polypeptide, neurokinin B.
The substance P/neurokinin A gene encodes the remaining five polypeptides. Three are formed by alternative processing of the
primary RNA and two by posttranslational processing.
There are three neurokinin receptors. Two of these, the substance P and the neuropep tide K receptors, are G protein-cou
Opioid Peptides
The brain and the gastrointestinal tract contain receptors that bind morphine. The search for endogenous ligands for these receptors led to
the discovery of two closely related pentapeptides (enkephalins; Table 7–4) that bind to th ese opioid receptors.
TABLE 7–4 Opioid peptides and their precursors.
Precursor Opioid
Peptides Structures
Proenkephalin Tyr-Gly-Gly-Phe-Met Met
enkephalin
Leu
enkephalin Tyr-Gly-Gly-Phe-Leu
Octapeptide Tyr-Gly-Gly-Phe-Met-Arg-GlyLeu
Heptapeptide Tyr-Gly-Gly-Phe-Met-Arg-Phe
TABLE 7–3 Mammalian tachykinins.
Proopiomelanocortin β-Endorphin Tyr-Gly-Glu-Phe-Met-Thr-Ser
Lys-Ser-Gln-Thr-Pro-Leu-ValThr-Leu-Phe-Lys-Asn-Ala-Ile-ValLys-Asn-Ala-His-L ys-Lys-Gly-Gln
Gene Polypeptide Products
SP/NKA Substance P Tyr-Gly-Gly-Phe-Leu-Arg-Arg-lle
Receptors
Substance P (NK-1) Prodynorphin Dynorphin 1–8
Neurokinin A
Neuropeptide K Neuropeptide K (NK-2)
Dynorphin 1–17
Tyr-Gly-Gly-Phe-Leu-Arg-Arglle-Arg-Pro-Lys-Leu-Lys-TrpAsp-Asn-Gln
Neuropeptide α α-Neoendorphin Tyr-Gly-Gly-Phe-Leu-Arg-LysTyr-Pro-Lys
Neurokinin A (3–10)
NKB Neurokinin B Neurokinin B (NK-3)
β
-Neoendorphin Tyr-Gly-Gly-Phe-Leu-Arg-LysTyr-Pro
One contains methionine (met-enkephalin), and one contains leucine (leu-enkephalin). These and oth er peptides that bind to opioid
receptors are called opioid peptides. The enkephalins are found in nerve endings i n the gastrointestinal tract and many different parts of
the brain, and they appear to function as synaptic transmitters. They are found in the su bstantia gelatinosa and have analgesic activity
when injected into the brain stem. They also decrease intestinal motility.
Like other small peptides, the opioid peptides are synthesized as part of larger precurso r molecules. More than 20 active opioid peptides
have been identified. Unlike other peptides, however, the opioid peptides have a numb er of different precursors. Each has a prepro form
and a pro form from which the signal peptide has been cleaved. The three precursors that have been characterized, and the opioid
peptides they produce, are shown in Table 7–4. Proenkephalin was first identified in the adrenal medulla , but it is also the precursor for
met-enkephalin and leu-enkephalin in the brain. Each proenkephalin molecule contains four met-enkephalins, one leuenkephalin, one
octapeptide, and one heptapeptide. Proopiomelanocortin, a large precur sor molecule found in the anterior and intermediate lobes of the
pituitary gland and the brain, contains β-endorphin, a polypeptide of 31 amino acid res idues that has metenkephalin at its amino terminal.
There are separate enkephalin-secreting and β endorphin-secreting systems of neurons in the brain. β-Endorphin is also secreted into the
bloodstream by the pituitary gland. A third precursor molecule is prodynorphin, a protein that contains three leuenkephalin residues
associated with dynorphin and neoendorphin. Dynorphin 1-17 is found in the duoden um and dynorphin 1-8 in the posterior pituitary and
hypothalamus. Alpha- and β-neoendorphins are also found in the hypothalamus. The r easons for the existence of multiple opioid peptide
precursors and for the presence of the peptides in the circulation as well as in the brain and the gastrointestinal tract are presently
unknown.
Enkephalins are metabolized primarily by two peptidases: enkephalinase A, which splits the Gly-Phe bond, and enkephalinase B, which
splits the Gly-Gly bond. Aminopeptidase, which splits the Tyr-Gly bond, also contribute s to their metabolism.
Opioid receptors have been studied in detail, and three are now established: μ, κ, and δ . They differ in physiologic effects (Table 7–5),
distribution in the brain and elsewhere, and affinity for various opioid peptides. All thre e are G proteincoupled receptors, and all inhibit
adenylyl cyclase. In mice in which the μ receptors have been knocked out, morphine f ails to produce analgesia, withdrawal symptoms,
and self-administration of nicotine. Selective knockout of the other system fails to produ ce this blockade. Activation of μ receptors
increases K
+
conductance, hyperpolarizing central neurons and primary afferents. Activation of κ r eceptors and δ receptors closes Ca
2+
channels.
The affinities of individual ligands for the three types of receptors are summarized in Fi gure 7–11. Endorphins bind only to μ receptors,
the main receptors that mediate analgesia. Other opioid peptides bind to multiple opioid r eceptors.
TABLE 7–5 Physiologic effects produced by stimulation of opiate receptors.
Receptor Effect
μ Analgesia
Site of action of morphine
Respiratory depression
Constipation
Euphoria
Sedation
Increased secretion of growth hormone and prolactin
Meiosis
κ Analgesia
Diuresis
Sedation
Meiosis
Dysphoria
δ Analgesia
Other Polypeptides
Numerous other polypeptides are found in the brain. For example, somatostatin is foun d in various parts of the brain, where it apparently
functions as a neurotransmitter with effects on sensory input, locomotor activity, and co gnitive function. In the hypothalamus, this
growth hormone-inhibiting hormone is secreted into the portal hypophysial vessels; in th e endocrine pancreas, it inhibits insulin
secretion and the secretion of other pancreatic hormones; and in the gastrointestinal trac t, it is an important inhibitory gastrointestinal
regulator. A family of five different somatostatin receptors have been
Endomorphins
Dynorphins β-Endorphin Enkephalins
κ δ
FIGURE 7–11 Opioid receptors. The ligands for the κ, μ, and δ receptors are sh own with the width of the arrows proportionate to the
affinity of the receptor for each ligand. (Reproduced with permission from Julius DJ: Another spark for the masses ? Nature 1997;386:442. Copyright © 1997 by
Macmillan Magazines.)
identified (SSTR1 through SSTR5). All are G protein-coupled receptors. They inhibit a denylyl cyclase and exert various other effects on
intracellular messenger systems. It appears that SSTR2 mediates cognitive effects and inh ibition of growth hormone secretion, whereas
SSTR5 mediates the inhibition of insulin secretion.
Vasopressin and oxytocin are not only secreted as hormones but also are present in ne urons that project to the brain stem and spinal cord.
The brain contains bradykinin, angiotensin II, and endothelin. The gastrointestinal horm ones VIP, CCK-4, and CCK-8 are also found in
the brain. There are two kinds of CCK receptors in the brain, CCK-A and CCK-B. CC K-8 acts at both binding sites, whereas CCK-4 acts
at the CCK-B sites. Gastrin, neurotensin, galanin, and gastrin-releasing peptide are also found in the gastrointestinal tract and brain.
Neurotensin and VIP receptors have been cloned and shown to be G protein-coupled receptors. The hypothalamus contains both gastrin
17 and gastrin 34. VIP produces vasodilation and is found in vasomotor nerve fibers. The functions of these peptides in the nervous
system are unknown.
Calcitonin gene-related peptide (CGRP) is a polypeptide that exists in two forms in ra ts and humans: CGRPα and CGRPβ. In humans,
these two forms differ by only three amino acid residues, yet they are encoded by diff erent genes. In rats, and presumably in humans,
CGRPβ is present in the gastrointestinal tract, whereas CGRPβ is found in primary affer ent neurons, neurons that project which taste
impulses to the thalamus, and neurons in the medial forebrain bundle. It is also present a long with substance P in the branches of
primary afferent neurons that end near blood vessels. CGRP-like immunoreactivity is p resent in the circulation, and injection of CGRP
causes vasodilation. CGRPα and the calcium-lowering hormone calcitonin are both prod ucts of the calcitonin gene. In the thyroid gland,
splicing produces the mRNA that codes for calcitonin, whereas in the brain, alternative splicing produces the mRNA that codes for
CGRPα. CGRP has little effect on Ca
2+
metabolism, and calcitonin is only a weak vasodilator.
Neuropeptide Y is a polypeptide containing 36 amino acid residues that acts on at le ast two of the four known G proteincoupled
receptors: Y
1
, Y
2
, Y
4
, and Y
5
. Neuropeptide Y is found throughout the brain and the autonomic nervous system. W hen injected into the
hypothalamus, this polypeptide increases food intake, and inhibitors of neuropeptide Y synthesis decrease food intake. Neuropeptide Y-
containing neurons have their cell bodies in the arcuate nuclei and project to the parave ntricular nuclei.
OTHER CHEMICAL TRANSMITTERS Purine & Pyrimidine Transmitters
After extended debate, it now seems clear that ATP, uridine, adenosine, and adenosine metabolites are neurotransmitters or
neuromodulators. Adenosine is a neuromodulator that acts as a general CNS depressan t and has additional widespread effects throughout
the body. It acts on four receptors: A
1
, A
2A
, A
2B
, and A
3
. All are G protein-coupled receptors and increase (A
2A
and A
2B
) or decrease
(A
1
and A
3
) cAMP concentrations. The stimulatory effects of coffee and tea are due to blockade o f adenosine receptors by caffeine and
theophylline. Currently, there is considerable interest in the potential use of A
1
antagonists to decrease excessive glutamate release and
thus to minimize the effects of strokes.
ATP is also becoming established as a transmitter, and it has widespread receptor-mediat ed effects in the body. It appears that soluble
nucleotidases are released with ATP, and these accelerate its removal after it has produc ed its effects. ATP has now been shown to
mediate rapid synaptic responses in the autonomic nervous system and a fast response i n the habenula. ATP binds to P2X receptors
which are ligand-gated ion channel receptors; seven subtypes (P2X
1
–P2X
7
) have been identified. P2X receptors have widespread
distributions throughout the body; for example, P2X
1
and P2X
2
receptors are present in the dorsal horn, indicating a role for ATP in
sensory transmission. ATP also binds to P2Y receptors which are G proteincoupled rec eptors. There are eight subtypes of P2Y receptors:
P2Y
1
, P2Y
2
, P2Y
4
, P2Y
6
, P2Y
11
, P2Y
12
, P2Y
13
, and P2Y
14
.
Cannabinoids
Two receptors with a high affinity for Δ
9
-tetrahydrocannabinol (THC), the psychoactive ingredient in marijuana, have been clon ed. The
CB
1
receptor triggers a G protein-mediated decrease in intracellular cAMP levels and is com mon in central pain pathways as well as in
parts of the cerebellum, hippocampus, and cerebral cortex. The endogenous ligand for the receptor is anandamide, a derivative of
arachidonic acid. This compound mimics the euphoria, calmness, dream states, drowsine ss, and analgesia produced by marijuana. There
are also CB
1
receptors in peripheral tissues, and blockade of these receptors reduces the vasodilator effect of anandamide. However, it
appears that the vasodilator effect is indirect. A CB
2
receptor has also been cloned, and its endogenous ligand may be
palmitoylethanolamide (PEA). However, the physiologic role of this compound is unsettled.
Gases
Nitric oxide (NO), a compound released by the endothelium of blood vessels as endoth elium-derived relaxing factor (EDRF), is also
produced in the brain. It is synthesized from arginine, a reaction catalyzed in the brain b y one of the three forms of NO synthase. It
activates guanylyl cyclase and, unlike other transmitters, it is a gas, which crosses cell me mbranes with ease and binds directly to
guanylyl cyclase. It may be the signal by which postsynaptic neurons communicate with presynaptic endings in long-term potentiation
and long-term depression. NO synthase requires NADPH, and it is now known that NA DPH-diaphorase (NDP), for which a
histochemical stain has been available for many years, is NO synthase.
Other Substances
Prostaglandins are derivatives of arachidonic acid found in the nervous system. They are present in nerve-ending fractions of brain
homogenates and are released from neural tissue in vitro. A putative prostaglandin tran sporter with 12 membrane-spanning domains has
been described. However, prostaglandins appear to exert their effects by modulating r eactions mediated by cAMP rather than by
functioning as synaptic transmitters.
Many steroids are neuroactive steroids; that is, they affect brain fun ction, although they are not neurotransmitters in the usual sense.
Circulating steroids enter the brain with ease, and neurons have numerous sex steroid a nd glucocorticoid receptors. In addition to acting
in the established fashion by binding to DNA (genomic effects), some steroids seem to a ct rapidly by a direct effect on cell membranes
(nongenomic effects). The effects of steroids on GABA receptors have been discusse d previously. Evidence has now accumulated that
the brain can produce some hormonally active steroids from simpler steroid precursors , and the term neurosteroids has been coined to
refer to these products. Progesterone facilitates the formation of myelin, but the exact ro le of most steroids in the regulation of brain
function remains to be determined.
CHAPTER SUMMARY
Neurotransmitters and neuromodulators are divided into two major categories: small-mo lecule transmitters (monoamines,
catecholamines, and amino acids) and large-molecule transmitters (neuropeptides). Usu ally neuropeptides are colocalized with one of the
small-molecule neurotransmitters.
Monoamines include acetylcholine, serotonin, and histamine. Catecholami nes include norepinephrine, epinephrine, and dopamine.
Amino acids include glutamate, GABA, and glycine.
Acetylcholine is found at the neuromuscular junction, in autonomic ganglia, and in postganglionic parasympathetic nervetarget organ
junctions and some postganglionic sympathetic nerve-target junctions. It is also found in the basal forebrain complex and
pontomesencephalic cholinergic complex. There are two major types of cholinergic receptors: muscarinic (G protein-coupled receptors)
and nicotinic (ligand-gated ion channel receptors).
Serotonin (5-HT) is found within the brain stem in the midline raphé nuclei w hich project to portions of the hypothalamus, the limbic
system, the neocortex, the cerebellum, and the spinal cord. There are at least seven types of 5-HT receptors, and many of these contain
subtypes. Most are G protein-coupled receptors.
Norepinephrine-containing neurons are in the locus ceruleus and other medullary and p ontine nuclei. Some neurons also contain
PNMT, which catalyzes the conversion of norepinephrine to epinephrine. Epinephrine and norepinephrine act on α and β receptors, with
norepinephrine having a greater affinity for α-adrenergic receptors and epinephrin e for β-adrenergic receptors. They are G protein-
coupled receptors, and each has multiple forms.
The amino acid glutamate is the main excitatory transmitter in the CNS. There are two ma jor types of gluatamate receptors:
metabotropic (G protein-coupled receptors) and ionotropic (ligand-gated ion channels recep tors, including kainite, AMPA, and NMDA).
GABA is the major inhibitory mediator in the brain. Three subtypes of GABA rec eptors have been identified: GABA
A
and GABA
C
(ligand-gated ion channel) and GABA
B
(G proteincoupled). The GABA
A
and GABA
B
receptors are widely distributed in the CNS.
There are three types of G protein-coupled opioid receptors (μ, κ, and δ) that dif fer in physiological effects, distribution in the brain
and elsewhere, and affinity for various opioid peptides.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. Which of the following is a ligand-gated ion channel? A) VIP
receptor
B) norepinephrine receptor
C) GABA
A
receptor
D) GABA
B
receptor
E) metabotropic glutamate receptor
2. Which of the following synaptic transmitters is not a peptide, polypeptide, or p rotein?
A) substance P
B) met-enkephalin
C) β-endorphin
D) serotonin
E) dynorphin
3. Activation of which of the following receptors would be expected to decrease anxie ty?
A) nicotinic cholinergic receptors
B) glutamate receptors
C) GABA
A
receptors
D) glucocorticoid receptors
E) α
1
-adrenergic receptors
4. Which of the following receptors is coupled to a heterotrimeric G protein?
A) glycine receptor
B) GABA
B
receptor
C) nicotinic acetylcholine receptor at myoneural junction D) 5-HT
3
receptor
E) ANP receptor
5. Which of the following would not be expected to enhance noradrenergic tra nsmission?
A) A drug that increases the entry of arginine into neurons. B) A drug that enhances ty rosine hydroxylase activity. C) A drug that
enhances dopamine β-hydroxylase activity. D) A drug that inhibits monoamine oxidase .
E) A drug that inhibits norepinephrine reuptake.
CHAPTER RESOURCES
Boron WF, Boulpaep EL: Medical Physiology. Elsevier, 2005. Cooper JR, Bloom FE, Roth RH: The Biochemical Basis of
Neuropharmacology, 8th ed. Oxford University Press, 2002. Fink KB, Göthert M: 5-HT recep tor regulation of neurotransmitter
release. Pharmacol Rev 2007;59:360.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural
Science, 4th ed. McGraw-Hill, 2000.
Monaghan DT, Bridges RJ, Cotman CW: The excitatory amino acid
receptors: Their classes, pharmacology, and distinct properties in
the function of the central nervous system. Ann Rev Pharmacol
Toxicol 1989;29:365.
Nadeau SE, et al: Medical Neuroscience, Sauders, 2004.
Olsen RW: The molecular mechanism of action of general
anesthetics: Structural aspects of interactions with GABA
A
receptors. Toxicol Lett 1998;100:193.
Owens DF, Kriegstein AR: Is there more to GABA than synaptic
inhibition? Nat Rev Neurosci 2002;3:715.
Squire LR, et al (editors): Fundamental Neuroscience, 3rd ed.
Academic Press, 2008.
This page intentionally left blank
Properties of Sensory Receptors
CH APTER
8
OBJEC TIV ES
After studying this chapter, you should be able to:
Describe the classification of sensory receptors.
Name the types of sensory receptors found in the skin, and discuss their relation to touc h, cold, warmth, and pain.
Define generator potential.
Explain the essential elements of sensory coding.
INTRODUCTION
Information about the internal and external environment activates the CNS via a variety of sensory receptors. These receptors are
transducers that convert various forms of energy in the environment into action potentia ls in neurons. The characteristics of these
receptors, the way they generate impulses in afferent neurons, and the general principl es or “laws” that apply to sensation are considered
in this chapter. Emphasis is placed on receptors mediating the sensation of touch, and lat er chapters focus on other sensory processes.
We learn in elementary school that there are “five senses,” but this dictum takes into acc ount only some of the senses that reach our
consciousness. In addition, some sensory receptors relay information that does not reac h consciousness. For example, the muscle
spindles provide information about muscle length, and other receptors provide informa tion about arterial blood pressure, the temperature
of the blood in the head, and the pH of the cerebrospinal fluid. The list of senses in Tab le 8–1 is somewhat simplified. The rods and
cones, for example, respond maximally to light of different wavelengths, and three diff erent types of cones are present, one for each of
the three primary colors. There are five different modalities of taste: sweet, salt, sour, bi tter, and umami. Sounds of different pitches are
heard primarily because different groups of hair cells in the cochlea are activated maxim ally by sound waves of different frequencies.
Whether these various responses to light, taste, and sound should be considered separa te senses is a semantic question that in the present
context is largely academic.
SENSE RECEPTORS
& SENSE ORGANS
It is worth noting that the term receptor is used in physiology to refer not only to sensory recept ors but also, in a very different sense, to
proteins that bind neurotransmitters, hormones, and other substances with great affinity and specificity as a first step in initiating specific
physiologic responses.
CLASSIFICATION OF
SENSORY RECEPTORS
Numerous attempts have been made to classify sensory receptors, but none has been en tirely successful. One classification divides them
into (1) teleceptors (“distance receivers”), which are concerned with events at a distanc e; (2) exteroceptors, which are concerned with the
external environment near at hand; (3) interoceptors, which are concerned with the inte rnal
149
TABLE 8–1 Principle sensory modalities.
Sensory System Modality Stimulus Energy Receptor Class Receptor Cell Types
Somatosensory Touch Tap, flutter 5–40 Hz Cutaneous mechanoreceptor Meissner cor puscles
Somatosensory Touch Motion Cutaneous mechanoreceptor Hair follicle receptors
Somatosensory Touch Deep pressure, vibration 60–300 Hz
Cutaneous mechanoreceptor Pacinian corpuscles
Somatosensory Touch Touch, pressure Cutaneous mechanoreceptor Merkel cells
Somatosensory Touch Sustained pressure Cutaneous mechanoreceptor Ruffini corpusc les
Somatosensory Proprioception Stretch Mechanoreceptor Muscle spindles
Somatosensory Proprioception Tension Mechanoreceptor Golgi tendon organ
Somatosensory Temperature Thermal Thermoreceptor Cold and warm receptors
Somatosensory Pain
Chemical, thermal, and mechanical
Chemoreceptor, thermoreceptor, and mechanoreceptor Polymodal receptors or chemic al, thermal, and mechanical nociceptors
Somatosensory Itch Chemical Chemoreceptor Chemical nociceptor
Visual Vision Light Photoreceptor Rods, cones
Auditory Hearing Sound Mechanoreceptor Hair cells (cochlea)
Vestibular Balance Angular acceleration Mechanoreceptor Hair cells (semicircular cana ls)
Vestibular Balance Linear acceleration, gravity Mechanoreceptor Hair cells (otolith org ans)
Olfactory Smell Chemical Chemoreceptor Olfactory sensory neuron
Gustatory Taste Chemical Chemoreceptor Taste buds
environment; and (4) proprioceptors, which provide information about the position of the body in space at any given instant. However,
the conscious component of proprioception (“body image”) is actually synthesized from information coming not only from receptors in
and around joints but also from cutaneous touch and pressure receptors.
Other special terms are frequently used to identify sensory receptors. The cutaneous re ceptors for touch and pressure are
mechanoreceptors. Potentially harmful stimuli such as pain, extreme heat, and extreme cold are said to be mediated by nociceptors.
The term chemoreceptor is used to refer to receptors stimulated by a change in the chemical composition of the environment in which
they are located. These include receptors for taste and smell as well as visceral receptors such as those sensitive to changes in the plasma
level of O
2
, pH, and osmolality. Photoreceptors are those in the rods and con es in the retina that respond to light.
SENSE ORGANS
Sensory receptors can be specialized dendritic endings of afferent nerve fibers, and th ey are often associated with nonneural cells that
surround it, forming a sense organ. Touch and pressure are sensed by four types o f mechanoreceptors (Figure 8–1). Meissner
corpuscles are dendrites encapsulated in connective tissue and respond to changes in tex ture and slow vibrations. Merkel cells are
expanded dendritic endings, and they respond to sustained pressure and touch. Ruffin i corpuscles are enlarged dendritic endings with
elongated capsules, and they respond to sustained pressure. Pacinian corpuscles consist of unmyelinated dendritic endings of a sensory
nerve fiber, 2 μm in diameter, encapsulated by concentric lamellae of connective tissue that give the organ the appearance of a cocktail
onion. Theses receptors respond to deep pressure and fast vibration.
The Na
+
channel BNC1 is closely associated with touch receptors. This channel is one of the degenerins, so called because when they
are hyperexpressed, they cause the neurons they are in to degenerate. However, it is n ot known if BNC1 is part of the receptor complex
or the neural fiber at the point of initiation of the spike potential. The receptor may be o pened mechanically by pressure on the skin.
Some sensory receptors are not specialized organs but rather are free nerve endings. P ain and temperature sensations arise from
unmyelinated dendrites of sensory neurons located around hair follicles throughout the glaborous and hairy skin as well as deep tissue.
A Modality Touch
Receptors
Meissner’s Merkel Pacinian corpuscle cells corpuscle Ruffini endings
B Location
Receptive field
C Intensity and time course
Neural
spike train
Stimulus
FIGURE 8–1 Sensory systems encode four elementary attributes of stimuli: m odality, location (receptive field), intensity, and
duration (timing). A) The human hand has four types of mechanoreceptors; their combined act ivation produces the sensation of contact
with an object. Selective activation of Merkel cells and Ruffini endings causes sensation of steady pressure; selective activation of
Meissner’s and Pacinian corpuscles causes tingling and vibratory sensation. B) Location of a stim ulus is encoded by spatial distribution
of the population of receptors activated. A receptor fires only when the skin close to its sensory terminals is touched. These receptive
fields of mechanoreceptors (shown as red areas on fingertips) differ in size and respo nse to touch. Merkel cells and Meissner’s corpuscles
provide the most precise localization as they have the smallest receptive fields and are most sens itive to pressure applied by a small
probe. C) Stimulus intensity is signaled by firing rates of individual receptors; duration of stimu lus is signaled by time course of firing.
The spike trains indicate action potentials elicited by pressure from a small probe at the cen ter of each receptive field. Meissner’s and
Pacinian corpuscles adapt rapidly, the others adapt slowly. (From Kandel ER, Schwartz JH, Jessell TM [ed itors]: Principles of Neural Science, 4th ed.
McGraw-Hill, 2000.)
GENERATION OF IMPULSES IN CUTANEOUS RECEPTORS
PACINIAN CORPUSCLES
The way receptors generate action potentials in the sensory nerves that innervate them v aries with the complexity of the sense organ. In
the skin, the Pacinian corpuscle has been studied in some detail. As noted above, the Pac inian corpuscles are touch receptors. Because of
their relatively large size and accessibility, they can be isolated, studied with microelectrod es, and subjected to microdissection. The
myelin sheath of the sensory nerve begins inside the corpuscle (Figure 8–2). The first n ode of Ranvier is also located inside, whereas the
second is usually near the point at which the nerve fiber leaves the corpuscle.
GENERATOR POTENTIALS
Recording electrodes can be placed on the sensory nerve as it leaves a Pacinian corpus cle and graded pressure applied to the corpuscle.
When a small amount of pressure is applied, a nonpropagated depolarizing potential res embling an EPSP is recorded. This is called the
generator potential or receptor potential (Figure 8–2). As the pressure is increased, the magn itude of the receptor potential increases.
When the magnitude of the generator potential is about 10 mV, an action potential is gen erated in the sensory nerve. As the pressure is
further increased, the generator potential becomes even larger and the sensory nerve f ires repetitively.
SOURCE OF THE GENERATOR POTENTIAL
By microdissection techniques, it has been shown that removal of the connective tissue l amellas from the unmyelinated nerve
2
1
e e
dcb
dcb
a a
3 4
dcb abcd a
FIGURE 8–2 Demonstration that the generator potential in a Pacinian corpus cle originates in the unmyelinated nerve terminal.
(1) The electrical responses to a pressure of 1
×
(record a), 2
×
(b), 3
×
(c), and 4
×
(d) were recorded. The strongest stimulus produced an
action potential in the sensory nerve (e). (2) Similar responses persisted after remo val of the connective tissue capsule, except that the
responses were more prolonged because of partial loss of adaptation. (3) The generator r esponses persisted but the action potential was
absent when the first node of Ranvier was blocked by pressure or with narcotics (arrow). (4) All responses disappeared when the sensory
nerve was cut and allowed to degenerate before the experiment.
ending in a Pacinian corpuscle does not abolish the generator potential. When the first n ode of Ranvier is blocked by pressure or
narcotics, the generator potential is unaffected but conducted impulses are abolished (Fi gure 8–2). When the sensory nerve is sectioned
and the nonmyelinated terminal is allowed to degenerate, no generator potential is forme d. These and other experiments have established
that the generator potential is produced in the unmyelinated nerve terminal. The recepto r therefore converts mechanical energy into an
electrical response, the magnitude of which is proportionate to the intensity of the stimulu s. The generator potential in turn depolarizes
the sensory nerve at the first node of Ranvier. Once the firing level is reached, an actio n potential is produced and the membrane
repolarizes. If the generator potential is great enough, the neuron fires again as soon as it repolarizes, and it continues to fire as long as
the generator potential is large enough to bring the membrane potential of the node to th e firing level. Thus, the node converts the graded
response of the receptor into action potentials, the frequency of which is proportionate to the magnitude of the applied stimuli.
SENSORY CODING
Converting a receptor stimulus to a recognizable sensation is termed sensory coding. All sen sory systems code for four elementary
attributes of a stimulus: modality, location, intensity, and duration. Modality is the type of energy tr ansmitted by the stimulus. Location
is the site on the body or space where the stimulus originated. Intensity is signaled by the response amp litude or frequency of action
potential generation. Duration refers to the time from start to end of a response in the receptor. The se attributes of sensory coding are
shown for the modality of touch in Figure 8–1.
MODALITY
Humans have four basic classes of receptors based on their sensitivity to one predomin ant form of energy: mechanical, thermal,
electromagnetic, or chemical. The particular form of energy to which a receptor is mos t sensitive is called its adequate stimulus. The
adequate stimulus for the rods and cones in the eye, for example, is light (an example o f electromagnetic energy). Receptors do respond
to forms of energy other than their adequate stimuli, but the threshold for these nonspec ific responses is much higher. Pressure on the
eyeball will stimulate the rods and cones, for example, but the threshold of these recepto rs to pressure is much higher than the threshold
of the pressure receptors in the skin.
LOCATION
The term sensory unit is applied to a single sensory axon and all its peri pheral branches. These branches vary in number but may be
numerous, especially in the cutaneous senses. The receptive field of a sensory unit is the spatial distribution from which a stimulus
produces a response in that unit (Figure 8–1). Representation of the senses in the skin is punctate. If the skin is carefully mapped,
millimeter by millimeter, with a fine hair, a sensation of touch is evoked from spots over lying these touch receptors. None is evoked
from the intervening areas. Similarly, temperature sensations and pain are produced by stimulation of
CLINICAL BOX 8–1 Two-Point Discrimination
The size of the receptive fields for light touch can be measured by the two-point threshold test. In this procedure, the two points on a
pair of calipers are simultaneously positioned on the skin and one determines the minimu m distance between the two caliper points that
can be perceived as separate points of stimulation. This is called the two-point discriminatio n threshold. If the distance is very small,
each caliper point is touching the receptive field of only one sensory neuron. If the dist ance between stimulation points is less than this
threshold, only one point of stimulation can be felt. Thus, the two-point discrimination th reshold is a measure of tactile acuity. The
magnitude of two-point discrimination thresholds varies from place to place on the body and is smallest where touch receptors are most
abundant. Stimulus points on the back, for instance, must be separated by at least 65 mm before they can be distinguished as separate,
whereas on the fingertips two stimuli are recognized if they are separated by as little as 2 mm. Blind individuals benefit from the tactile
acuity of fingertips to facilitate the ability to read Braille; the dots forming Braille symbols are separated by 2.5 mm. Two-point
discrimination is used to test the integrity of the dorsal column (medial lemnisc us) system, the central pathway for touch and
proprioception.
the skin only over the spots where the receptors for these modalities are located. In the cornea and adjacent sclera of the eye, the surface
area supplied by a single sensory unit is 50–200 mm
2
. Generally, the areas supplied by one unit overlap and interdigitate with the areas
supplied by others.
One of the most important mechanisms that enable localization of a stimulus site is lateral inhibition. Information from sensory neurons
whose receptors are at the peripheral edge of the stimulus is inhibited compared to infor mation from the sensory neurons at the center of
the stimulus. Thus, lateral inhibition enhances the contrast between the center and periph ery of a stimulated area and increases the ability
of the brain to localize a sensory input. Lateral inhibition underlies two-point discr imination (see Clinical Box 8–1).
INTENSITY
The intensity of sensation is determined by the amplitude of the stimulus applied to the re ceptor. This is illustrated in Figure 8–3. As a
greater pressure is applied to the skin, the receptor potential in the mechanoreceptor inc reases (not shown), and the frequency of the
action potentials in a single axon transmitting information to the CNS is also increased. In addition to increasing the firing rate in a single
axon, the greater intensity of stimulation also will recruit more receptors into the receptiv e field.
It has long been taught that the magnitude of the sensation felt is proportional to the log o f the intensity of the stimulus (Weber–Fechner
law). It now appears, however, that a power function more accurately describes this relation. In other words,
A
R = KS
where R is the sensation felt, S is the intensity of the stimulus, and, for any specific senso ry modality, K and A are constants. The
frequency of the action potentials generated in a sensory nerve fiber is also related to th e intensity of the initiating stimulus by a power
function. An example of this relation is shown is shown in Figure 8–4, in which the calc ulated exponent is 0.52. However, the relation
between direct stimulation of a sensory nerve and the sensation felt is linear. Consequen tly, it appears that for any given sensory
modality, the relation between sensation and stimulus intensity is determined primarily by the properties of the peripheral receptors.
DURATION
When a maintained stimulus of constant strength is applied to a receptor, the frequency of the action potentials in its sensory nerve
declines over time. This phenomenon is known as adaptation or desensitization. The degre e to which adaptation occurs varies from one
sense to another. Receptors can be classified into rapidly adapting (phasic) receptors and slowly adapti ng (tonic) receptors. This is
illustrated for different types of touch receptors in Figure 8–1. Meissner and Pacinian c orpuscles are examples of rapidly adapting
receptors, and Merkel cells and Ruffini endings are examples of slowly adapting recep tors. Other examples of slowly adapting receptors
are muscle spindles and nociceptors. Different types of sensory adaptation appear to ha ve some value to the individual. Light touch
would be distracting if it were persistent; and, conversely, slow adaptation of spindle inp ut is needed to maintain posture. Similarly, input
from nociceptors provides a warning that would lose its value if it adapted and disappea red.
SENSORY INFORMATION
The speed of conduction and other characteristics of sensory nerve fibers vary, but ac tion potentials are similar in all nerves. The action
potentials in the nerve from a touch receptor, for example, are essentially identical to tho se in the nerve from a warmth receptor. This
raises the question of why stimulation of a touch receptor causes a sensation of touch an d not of warmth. It also raises the question of
how it is possible to tell whether the touch is light or heavy.
LAW OF SPECIFIC NERVE ENERGIES
The sensation evoked by impulses generated in a receptor depends in part on the specif ic part of the brain they ultimately activate. The
specific sensory pathways are discrete from sense
Afferent neuron
Skin
Glass probe
180
120
60
Time
FIGURE 8–3 Relationship between stimulus and impulse frequency in an affere nt fiber. Action potentials in an afferent fiber from a
mechanoreceptor of a single sensory unit increase in frequency as branches of the aff erent neuron are stimulated by pressure of
increasing magnitude. (From Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology. McGraw-Hil l, 2008.)
100
R = 9.4 S
0.52
90
2.0 1.9
80 1.8
70
1.7
1.6
60
1.5
1.4
50 1.3
40 1.2
1.1
30
1.0
20
0.9
10
.2 .3 .4 .5 .6 .7 .8 .9 1.0 1.11.2 1.31.4 1.51.6 1.71.81.9 2.0 2.1 Log S
10 20 30 40 50 60 70 80 90 100 S (% maximum stimulus)
FIGURE 8–4 Relation between magnitude of touch stimulus (S) and frequency of action potentials in sensory nerve fibers (R).
Dots are individual values from cats and are plotted on linear coordinates (left) and log–log c oordinates (right). The equation shows the
calculated power function relationship between R and S. (Reproduced, with per mission, from Werner G, Mountcastle VB: Neural activity in
mechanoreceptive cutaneous afferents. Stimulus–response relations, Weber functions, and information tr ansmission. J Neurophysiol 1965;28:359.)
CLINICAL BOX 8–2 CLINICAL BOX 8–3 Vibratory Sensibility
Vibratory sensibility
Vibratory sensibility Hz) tuning fork to the skin on the fingertip, tip of the toe, or bony prominences o f the toes. The normal response is
a “buzzing” sensation. The sensation is most marked over bones. The term pallesthesia is also us ed to describe this ability to feel
mechanical vibrations. The receptors involved are the receptors for touch, especially Pacinian corpuscles, but a time factor is also
necessary. A pattern of rhythmic pressure stimuli is interpreted as vibration. The impulse s responsible for the vibrating sensation are
carried in the dorsal columns. Degeneration of this part of the spinal cord occurs in poorly controlled diabetes, pernicious anemia,
vitamin B
12
deficiencies, or early tabes dorsalis. Elevation of the threshold for vibratory stimuli is an early symptom of this
degeneration. Vibratory sensation and proprioception are closely related; when one is d iminished, so is the other.
Stereognosis
Stereognosis is the perception of the form and nature of an object without looking at it. Normal per sons can readily identify objects such
as keys and coins of various denominations. This ability depends on relatively intact touc h and pressure sensation and is compromised
when the dorsal columns are damaged. The inability to identify an object by touch is cal led tactile agnosia. It also has a large cortical
component; impaired stereognosis is an early sign of damage to the cerebral cortex and sometimes occurs in the absence of any
detectable defect in touch and pressure sensation when there is a lesion in the parietal lob e posterior to the postcentral gyrus.
Stereoagnosia can also be expressed by the failure to identify an object by sight (visual agnosia) , the inability to identify sounds or
words (auditory agnosia) or color (color agnosia), or the inability to identify the loca tion or position of an extremity (position
agnosia).
organ to cortex. Therefore, when the nerve pathways from a particular sense organ a re stimulated, the sensation evoked is that for which
the receptor is specialized no matter how or where along the pathway the activity is initia ted. This principle, first enunciated by Müller in
1835, has been given the rather cumbersome name of the law of specific nerve energie s. For example, if the sensory nerve from a
Pacinian corpuscle in the hand is stimulated by pressure at the elbow or by irritation from a tumor in the brachial plexus, the sensation
evoked is touch. Similarly, if a fine enough electrode could be inserted into the appropr iate fibers of the dorsal columns of the spinal
cord, the thalamus, or the postcentral gyrus of the cerebral cortex, the sensation produc ed by stimulation would be touch. The general
principle of specific nerve energies remains one of the cornerstones of sensory physio logy.
LAW OF PROJECTION
No matter where a particular sensory pathway is stimulated along its course to the cortex , the conscious sensation produced is referred to
the location of the receptor. This principle is called the law of projection. Cortical stimulation e xperiments during neurosurgical
procedures on conscious patients illustrate this phenomenon. For example, when the co rtical receiving area for impulses from the left
hand is stimulated, the patient reports sensation in the left hand, not in the head.
RECRUITMENT OF SENSORY UNITS
As the strength of a stimulus is increased, it tends to spread over a large area and gener ally not only activates the sense organs
immediately in contact with it but also “recruits” those in the surrounding area. Furtherm ore, weak stimuli activate the receptors with the
lowest thresholds, and stronger stimuli also activate those with higher thresholds. Some o f the receptors activated are part of the same
sensory unit, and impulse frequency in the unit therefore increases. Because of overlap and interdigitation of one unit with another,
however, receptors of other units are also stimulated, and consequently more units fire. In this way, more afferent pathways are
activated, which is interpreted in the brain as an increase in intensity of the sensation.
NEUROLOGICAL EXAM
The sensory component of a neurological exam includes an assessment of various sens ory modalities including touch, proprioception,
vibratory sense, and pain. Clinical Box 8–2 describes the test for vibratory sensibility. C ortical sensory function can be tested by placing
familiar objects in a patient’s hands and asking him or her to identify it with the eyes clos ed (see Clinical Box 8–3).
CHAPTER SUMMARY
Sensory receptors are commonly classified as mechanoreceptors, nociceptors, che moreceptors, or photoreceptors.
Touch and pressure are sensed by four types of mechanoreceptors: Meissner’ s corpuscles (respond to changes in texture and slow
vibrations), Merkel’s cells (respond to sustained pressure and touch), Ruffini corpuscles (respond to sustained pressure), and Pacinian
corpuscles (respond to deep pressure and fast vibrations).
Nociceptors and thermoreceptors are free nerve endings on unmyelinated or ligh tly myelinated fibers in hairy and glaborous skin and
deep tissues.
The generator or receptor potential is the nonpropagated depolarizing potential rec orded in a sensory organ after an adequate stimulus
is applied. As the stimulus is increased, the magnitude of the receptor potential increases. Whe n it reaches a critical threshold, an action
potential is generated in the sensory nerve.
Converting a receptor stimulus to a recognizable sensation is termed sensory codin g. All sensory systems code for four elementary
attributes of a stimulus: modality, location, intensity, and duration.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. Pacinian corp uscles are
A) a type of thermoreceptor.
B) usually innervated by Aδ nerve fibers.
C) rapidly adapting touch receptors.
D) slowly adapting touch receptors.
E) nociceptors.
2. Adaptation to a sensory stimulus produces
A) a diminished sensation when other types of sensory stimuli are withdrawn.
B) a more intense sensation when a given stimulus is applied repeatedly.
C) a sensation localized to the hand when the nerves of the brachial plexus are stimulate d.
D) a diminished sensation when a given stimulus is applied repeatedly over time.
E) a decreased firing rate in the sensory nerve from the receptor when one’s attention is directed to another matter. 3. Sensory systems
code for the following attributes of a stimulus:
A) modality, location, intensity, and duration
B) threshold, receptive field, adaptation, and discrimination
C) touch, taste, hearing, and smell
D) threshold, laterality, sensation, and duration
E) sensitization, discrimination, energy, and projection 4. In which of the following is th e frequency of stimulation not linearly related to
the strength of the sensation felt?
A) sensory area of the cerebral cortex
B) specific projection nuclei of the thalamus
C) lateral spinothalamic tract
D) dorsal horn
E) cutaneous receptors
5. Which of the following receptors and sense organs are incorrectly
paired?
A) rods and cones : eye
B) receptors sensitive to sodium : taste buds
C) hair cells : olfactory epithelium
D) receptors sensitive to stretch : carotid sinus
E) glomus cells : carotid body
6. Which best describes the law of specific nerve energies?
A) No matter where a particular sensory pathway is stimulated along its course to the c ortex, the conscious sensation produced is
referred to the location of the receptor.
B) A nerve can only be stimulated by electrical energy.
C) Receptors can respond to forms of energy other than their adequate stimuli, but the th reshold for these nonspecific responses is much
higher.
D) For any given sensory modality, the specific relationship between sensation and stimulu s intensity is determined by the properties of
the peripheral receptors.
E) The sensation evoked by impulses generated in a receptor depends in part on the specific part of the brain they ultimately activate.
7. Which of the following does not contain cation channels that are
activated by mechanical distortion, producing depolarization?
A) olfactory receptors
B) Pacinian corpuscles
C) hair cells in cochlea
D) hair cells in semicircular canals
E) hair cells in utricle
CHAPTER RESOURCES
Barlow HB, Mollon JD (editors): The Senses. Cambridge University Press, 1982 .
Bell J, Bolanowski S, Holmes MH: The structure and function of Pacinian corp uscles: A review. Prog Neurobiol 1994;42:79.
Haines DE (editor): Fundamental Neuroscience for Basic and Clinical Applications, 3rd ed. Elsevier, 2006.
Iggo A (editor): Handbook of Sensory Physiology. Vol 2, Somatosensory System . Springer-Verlag, 1973.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural Science, 4th ed. Mc Graw-Hill, 2000.
Mountcastle VB: Perceptual Neuroscience. Harvard University Press, 1999.
Squire LR, et al (editors): Fundamental Neuroscience, 3rd ed. Academic Press, 20 08.
Reflexes
CH APTER
9
OBJEC TIV ES
After studying this chapter, you should be able to:
Describe the components of a reflex arc.
Describe the muscle spindles and their role in the stretch reflex.
Describe the Golgi tendon organs and analyze their function as part of a feedback system that maintains muscle force.
Define reciprocal innervation, inverse stretch reflex, clonus, and lengthening reaction.
INTRODUCTION
The basic unit of integrated reflex activity is the reflex arc. This arc consists of a sense organ, an afferent neuron, one or more synapses
within a central integrating station, an efferent neuron, and an effector. In mammals, th e connection between afferent and efferent
somatic neurons is generally in the brain or spinal cord. The afferent neurons enter via the dorsal roots or cranial nerves and have their
cell bodies in the dorsal root ganglia or in the homologous ganglia on the cranial nerves . The efferent fibers leave via the ventral roots or
corresponding motor cranial nerves. The principle that in the spinal cord the dorsal roo ts are sensory and the ventral roots are motor is
known as the Bell–Magendie law.
Activity in the reflex arc starts in a sensory receptor with a receptor potential whose mag nitude is proportional to the strength of the
stimulus (Figure 9–1). This generates all-ornone action potentials in the afferent nerve, the number of action potentials being proportional
to the size of the generator potential. In the central nervous system (CNS), the response s are again graded in terms of excitatory
postsynaptic potentials (EPSPs) and inhibitory postsynaptic potentials (IPSPs) at the synap tic junctions. All-or-none responses are
generated in the efferent nerve. When these reach the effector, they again set up a gra ded response. When the effector is smooth muscle,
responses summate to produce action potentials in the smooth muscle, but when the effe ctor is skeletal muscle, the graded response is
always adequate to produce action potentials that bring about muscle contraction. The co nnection between the afferent and efferent
neurons is usually in the CNS, and activity in the reflex arc is modified by the multiple in puts converging on the efferent neurons or at
any synaptic station within the reflex loop.
The simplest reflex arc is one with a single synapse between the afferent and efferent n eurons. Such arcs are monosynaptic, and ref lexes
occurring in them are called monosynaptic reflexes. Reflex arcs in which one or more interneuron is in terposed between the afferent
and efferent neurons are called polysynaptic reflexes. There can be anywhere from two to hundre ds of synapses in a polysynaptic reflex
arc.
157
Sense organ Afferent Synapse neuron
Efferent Neuromuscular Muscle neuron junction
Generator potential Action EPSPs potentials (and IPSPs) Action Endplate Action potentia ls potentials potentials
FIGURE 9–1 The reflex arc. Note that at the receptor and in the CNS a nonpr opagated graded response occurs that is proportionate to
the magnitude of the stimulus. The response at the neuromuscular junction is a lso graded, though under normal conditions it is always
large enough to produce a response in skeletal muscle. On the other hand, in the portion s of the arc specialized for transmission (afferent
and efferent axons, muscle membrane), the responses are all-or-none action potentials.
MONOSYNAPTIC REFLEXES: THE STRETCH REFLEX
When a skeletal muscle with an intact nerve supply is stretched, it contracts. This respons e is called the stretch reflex. The stimulus that
initiates the reflex is stretch of the muscle, and the response is contraction of the muscle b eing stretched. The sense organ is a small
encapsulated spindlelike or fusiform shaped structure called the muscle spindle, located within the fleshy part of the muscle. The
impulses originating from the spindle are transmitted to the CNS by fast sensory fibers th at pass directly to the motor neurons which
supply the same muscle. The neurotransmitter at the central synapse is glutamate. The str etch reflex is the best known and studied
monosynaptic reflex and is typified by the knee jerk reflex (see Clinical Box 9–1).
STRUCTURE OF MUSCLE SPINDLES
Each muscle spindle has three essential elements: (1) a group of specialized intrafusal mu scle fibers with contractile polar ends and a
noncontractile center, (2) large diameter myelinated afferent nerves (types Ia and II) o riginating in the central portion of the intrafusal
fibers, and (3) small diameter myelinated efferent nerves supplying the polar contractile regions of the intrafusal fibers (Figure 9–2A). It
is important to understand the relationship of these elements to each other and to the mus cle itself to appreciate the role of this sense
organ in signaling changes in the length of the muscle in which it is located. Changes in muscle length are associated with changes in
joint angle; thus muscle spindles provide information on position (ie, proprioception).
The intrafusal fibers are positioned in parallel to the extrafusal fib ers (the regular contractile units of the muscle) with the ends of the
spindle capsule attached to the tendons at either end of the muscle. Intrafusal fibers do n ot contribute to the overall contractile force of
the muscle, but rather serve a pure sensory function. There are two types of intrafusal fibers in mammalian muscle spindles. The first type
contains many nuclei in a dilated central area and is called a nuclear bag fiber (Figure 9–2B). There are two subtypes of nuclear bag
fibers, dynamic and static. Typically, there are two or three nuclear bag fibers per spindle. The s econd intrafusal fiber type, the nuclear
chain fiber, is thinner and shorter and lacks a definite bag. Each spindle has about five of these fibers.
There are two kinds of sensory endings in each spindle, a single primary (group Ia) ending a nd up to eight secondary (group II)
endings. The Ia afferent fiber wraps around the center of the dynamic and static nuclear bag fib ers and nuclear chain fibers. Group II
sensory fibers are located adjacent to the centers of the static nuclear bag and nuclear c hain fibers; these fibers do not innervate the
dynamic nuclear bag fibers. Ia afferents are very sensitive to the velocity of the change in muscle length during a stretch (dynamic
response); thus they provide information about the speed of movements and allow for qu ick corrective movements. The steady-state
(tonic) activity of group Ia and II afferents provide information on steady-state length of the muscle (static response). The top trace in
Figure 9–2C shows the dynamic and static components of activity in a Ia afferent durin g muscle stretch. Note that they discharge most
rapidly while the muscle is being stretched (shaded area of graphs) and less rapidly dur ing sustained stretch.
The spindles have a motor nerve supply of their own. These nerves are 3–6 μm in diam eter, constitute about 30% of the fibers in the
ventral roots, and are called γ-motor neurons. There are two types of γ-motor neurons: dynamic, which supply the dynamic nuclear bag
fibers and static, which supply the static nuclear bag fibers and the nuclear chain fib ers. Activation of dynamic γ-motor neurons
increases the dynamic sensitivity of the group Ia endings. Activation of the static γ- moto r neurons increases the tonic level of activity in
both group Ia and II endings, decreases the dynamic sensitivity of group Ia afferents, and can prevent silencing of Ia afferents during
muscle stretch (Figure 9–2C).
CLINICAL BOX 9–1 Knee Jerk Reflex
Tapping the patellar tendon elicits the knee jerk, a stretch reflex of the quadriceps femoris mus cle, because the tap on the tendon
stretches the muscle. A similar contraction is observed if the quadriceps is stretched man ually. Stretch reflexes can also be elicited from
most of the large muscles of the body. Tapping on the tendon of the triceps brachii, for example, causes an extensor response at the
elbow as a result of reflex contraction of the triceps; tapping on the Achilles tendon cau ses an ankle jerk due to reflex contraction of the
gastrocnemius; and tapping on the side of the face causes a stretch reflex in the masseter . The knee jerk reflex is an example of a deep
tendon reflex (DTR) in a neurological exam and is graded on the following s cale: 0 (absent), 1+ (hypoactive), 2+ (brisk, normal), 3+
(hyperactive without clonus), 4+ (hyperactive with mild clonus), and 5+ (hyperactive w ith sustained clonus). Absence of the knee jerk
can signify an abnormality anywhere within the reflex arc, including the muscle spindle , the Ia afferent nerve fibers, or the motor
neurons to the quadriceps muscle. The most common cause is a peripheral neuropathy from such things as diabetes, alcoholism, and
toxins. A hyperactive reflex can signify an interruption of corticospinal and other desce nding pathways that influence the reflex arc.
A Muscle spindle B Intrafusal fibers of the muscle spindle
Static nuclear bag fiber
C Response of Ia sensory fiber to selective activation of motor neurons
200
Intrafusal muscle
fibers
Dynamic nuclear bag fiber
Nuclear
chain fiber Dynamic response Steady-state response 0 Stretch alone
200 Capsule
Sensory endings II
Ia
Afferent axons
0
Stimulate static gamma fiber Static 200
Efferent axons
Dynamic
Gamma motor
endings 0
Stimulate dynamic gamma fiber 6
0 0.2 s
FIGURE 9–2 Mammalian muscle spindle. A) Diagrammatic repre sentation of the main components of mammalian muscle spindle
including intrafusal muscle fibers, afferent sensory fiber endings, and efferent motor f ibers (γ-motor neurons). B) Three types of
intrafusal muscle fibers: dynamic nuclear bag, static nuclear bag, and nuclear chain fibers. A single Ia afferent fiber innervates all three
types of fibers to form a primary sensory ending. A group II sensory fiber inne rvates nuclear chain and static bag fibers to form a
secondary sensory ending. Dynamic γ-motor neurons innervate dynamic bag fibers; s tatic γ-motor neurons innervate combinations of
chain and static bag fibers. C) Comparison of discharge pattern of Ia afferent activ ity during stretch alone and during stimulation of static
or dynamic γ-motor neurons. Without γ-stimulation, Ia fibers show a small dynamic re sponse to muscle stretch and a modest increase in
steady-state firing. When static γ-motor neurons are activated, the steadystate response i ncreases and the dynamic response decreases.
When dynamic γ-motor neurons are activated, the dynamic response is markedly increased b ut the steady-state response gradually
returns to its original level. (From Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of N eural Science, 4th ed. McGraw-Hill, 2000.)
CENTRAL CONNECTIONS
OF AFFERENT FIBERS
Ia fibers end directly on motor neurons supplying the extrafusal fibers of the same mus cle (Figure 9–3). The time between the application
of the stimulus and the response is called the reaction time. In humans, the reaction time f or a stretch reflex such as the knee jerk is 19–
24 ms. Weak stimulation of the sensory nerve from the muscle, known to stimulate only Ia fibers, causes a contractile response with a
similar latency. Because the conduction velocities of the afferent and efferent fiber type s are known and the distance from the muscle to
the spinal cord can be measured, it is possible to calculate how much of the reaction time was taken up by conduction to and from the
spinal cord. When this value is subtracted from the reaction time, the remainder, called t he central delay, is the time taken for the reflex
activity to traverse the spinal cord. In humans, the central delay for the knee jerk is 0.6– 0.9 ms, and figures of similar magnitude have
been found in experimental animals. Because the minimal synaptic delay is 0.5 ms, only one synapse could have been traversed.
Muscle spindles also make connections that cause muscle contraction via polysynaptic pa thways, and the afferents involved are probably
those from the secondary endings. However, group II fibers also make monosynaptic connections to the motor neurons and make a small
contribution to the stretch reflex.
Dorsal root
Interneuron releasing inhibitory mediator
Motor neuron Ib fiber
from Ia fiber
Golgi fromtendon muscle
Ventral rootorgan
spindle
Motor endplate on extrafusal fiber
FIGURE 9–3 Diagram illustrating the pathways responsible for the stretch reflex and the inverse stretch reflex. Stretch stimulates
the muscle spindle, which activates Ia fibers that excite the motor neuron. Stretch also stimulates the Golgi tendon organ, which activates
Ib fibers that excite an interneuron that releases the inhibitory mediator glycine. With strong stretch, the resulting hyperpolarization of
the motor neuron is so great that it stops discharging.
FUNCTION OF MUSCLE SPINDLES
When the muscle spindle is stretched, its sensory endings are distorted and receptor pote ntials are generated. These in turn set up action
potentials in the sensory fibers at a frequency proportional to the degree of stretching. B ecause the spindle is in parallel with the
extrafusal fibers, when the muscle is passively stretched, the spindles are also stretched, referred to as “loading the spindle.” This
initiates reflex contraction of the extrafusal fibers in the muscle. On the other hand, the s pindle afferents characteristically stop firing
when the muscle is made to contract by electrical stimulation of the α-motor neurons to th e extrafusal fibers because the muscle shortens
while the spindle is unloaded (Figure 9–4).
Thus, the spindle and its reflex connections constitute a feedback device that operates to maintain muscle length; if the muscle is
stretched, spindle discharge increases and reflex shortening is produced, whereas if the muscle is shortened without a change in γ-motor
neuron discharge, spindle afferent activity decreases and the muscle relaxes. Dynamic a nd static responses of muscle spindle afferents
influence physiological tremor (see Clinical Box 9–2).
EFFECTS OF γ-MOTOR
NEURON DISCHARGE
Stimulation of γ-motor neurons produces a very different picture from that produced by stimulation of the extrafusal fibers. Such
stimulation does not lead directly to detectable contraction of the muscles because the intr afusal fibers are not strong enough or plentiful
enough to cause shortening. However, stimulation does cause the contractile ends of the intrafusal fibers to shorten and therefore
stretches the nuclear bag portion of the spindles, deforming the endings and initiating im pulses in the Ia fibers (Figure 9–4). This in turn
can lead to reflex contraction of the muscle. Thus, muscle can be made to contract via s timulation of the α-motor neurons that innervate
the extrafusal fibers or the γ-motor neurons that initiate contraction indirectly via the stre tch reflex.
If the whole muscle is stretched during stimulation of the γ- motor neurons, the rate of discharge in the Ia fibers is further increased
(Figure 9–4). Increased γ-motor neuron activity thus increases spindle sensitivity during stretch.
In response to descending excitatory input to spinal motor circuits, both α- and γ-motor neurons are activated. Because of this “α–γ
coactivation,” intrafusal and extrafusal fibers shorten together, and spindle afferent acti vity can occur throughout the period of muscle
contraction. In this way, the spindle remains capable of responding to stretch and reflex ly adjusting α-motor neuron discharge.
Spindle
Tendon
Extrafusal fiber Sensory nerve
Impulses in sensory nerve Muscle at rest
Muscle stretched
CLINICAL BOX 9–2 Physiological Tremor
The response of the Ia sensory fiber endings to the dynamic (phasic) as well as the stati c events in the muscle is important because the
prompt, marked phasic response helps to dampen oscillations caused by conduction del ays in the feedback loop regulating muscle
length. Normally a small oscillation occurs in this feedback loop. This physiologic tremor has a low am plitude (barely visible to the
naked eye) and a frequency of approximately 10 Hz. Physiological tremor is a normal phenomenon which affects everyone while
maintaining posture or during movements. However, the tremor would be worse if it w ere not for the sensitivity of the spindle to
velocity of stretch. It can become exaggerated in some situations such as when we are a nxious or tired or because of drug toxicity.
Numerous factors contribute to the genesis of physiological tremor. It is likely dependen t on not only central (inferior olive) sources but
also from peripheral factors including motor unit firing rates, reflexes, and mechanical resonance.
Muscle contracted
Increased γ efferent discharge
Increased γ efferent
discharge—muscle stretched
FIGURE 9–4 Effect of various conditions on muscle spindle discharge. When the whole muscle is stretched, the muscle spindle is
also stretched and its sensory endings are activated at a frequency proportional to the d egree of stretching (“loading the spindle”).
Spindle afferents stop firing when the muscle contracts (“unloading the spindle”). Stimulation of
γ
-motor neurons cause the contractile
ends of the intrafusal fibers to shorten. This stretches the nuclear bag region, initiating imp ulses in sensory fibers. If the whole muscle is
stretched during stimulation of the
γ
-motor neurons, the rate of discharge in sensory fibers is further increased.
CONTROL OF γ-MOTOR
NEURON DISCHARGE
The γ-motor neurons are regulated to a large degree by descending tracts from a nu mber of areas in the brain. Via these pathways, the
sensitivity of the muscle spindles and hence the threshold of the stretch reflexes in vario us parts of the body can be adjusted and shifted
to meet the needs of postural control.
Other factors also influence γ-motor neuron discharge. Anxiety causes an increased disch arge, a fact that probably explains the
hyperactive tendon reflexes sometimes seen in anxious patients. In addition, unexpected movement is associated with a greater efferent
discharge. Stimulation of the skin, especially by noxious agents, increases γ-motor neur on discharge to ipsilateral flexor muscle spindles
while decreasing that to extensors and produces the opposite pattern in the opposite limb . It is well known that trying to pull the hands
apart when the flexed fingers are hooked together facilitates the knee jerk reflex (Jend rassik’s maneuver), and this may also be due to
increased γ-motor neuron discharge initiated by afferent impulses from the hands.
RECIPROCAL INNERVATION
When a stretch reflex occurs, the muscles that antagonize the action of the muscle involv ed (antagonists) relax. This phenomenon is said
to be due to reciprocal innervation. Impulses in the Ia fibers from the muscle spindles of the p rotagonist muscle cause postsynaptic
inhibition of the motor neurons to the antagonists. The pathway mediating this effect is b isynaptic. A collateral from each Ia fiber passes
in the spinal cord to an inhibitory interneuron that synapses on a motor neuron supplyin g the
Nerve fiber Tendon bundles
Organ of Golgi, showing ramification of nerve fibrils Muscular fibers
FIGURE 9–5 Golgi tendon organ. (Reproduced, with permission, from Goss CM [editor]: Gray’s An atomy of the Human Body, 29th ed. Lea & Febiger,
1973.)
antagonist muscles. This example of postsynaptic inhibition is discussed in Chapter 6, and the pathway is illustrated in Figure 6–6.
INVERSE STRETCH REFLEX
Up to a point, the harder a muscle is stretched, the stronger is the reflex contraction. Ho wever, when the tension becomes great enough,
contraction suddenly ceases and the muscle relaxes. This relaxation in response to stron g stretch is called the inverse stretch reflex or
autogenic inhibition.
The receptor for the inverse stretch reflex is in the Golgi tendon organ (Figure 9–5). This organ consists of a netlike collection of
knobby nerve endings among the fascicles of a tendon. There are 3–25 muscle fibers per tendon organ. The fibers from the Golgi tendon
organs make up the Ib group of myelinated, rapidly conducting sensory nerve fibers. Stimulation of these Ib fibers leads to the
production of IPSPs on the motor neurons that supply the muscle from which the fibers arise. The Ib fibers end in the spinal cord on
inhibitory interneurons that in turn terminate directly on the motor neurons (Figure 9–3) . They also make excitatory connections with
motor neurons supplying antagonists to the muscle.
Because the Golgi tendon organs, unlike the spindles, are in series with the muscle fiber s, they are stimulated by both passive stretch and
active contraction of the muscle. The threshold of the Golgi tendon organs is low. The d egree of stimulation by passive stretch is not
great because the more elastic muscle fibers take up much of the stretch, and this is why it takes a strong stretch to produce relaxation.
However, discharge is regularly produced by contraction of the muscle, and the Golgi tendon organ thus functions as a transducer in a
feedback circuit that regulates muscle force in a fashion analogous to the spindle feedba ck circuit that regulates muscle length.
The importance of the primary endings in the spindles and the Golgi tendon organs in r egulating the velocity of the muscle contraction,
muscle length, and muscle force is illustrated by the fact that that section of the afferent n erves to an arm causes the limb to hang loosely
in a semiparalyzed state. The organization of the system is shown in Figure 9–6. The int eraction of
Interneuronal
control
signal
Inter
neurons Force feedback Tendon organs
α Control − signal +
α Internal disturbances
Efferent signal +
Muscle External forces
Muscular + Muscle force
Load
length −
Length and velocity
Length and velocity
-Dynamic
feedback
γ
control
signal
γd
γ -Static
control
signal
γs
Spindles
FIGURE 9–6 Block diagram of peripheral motor control system. The dashe d line indicates the nonneural feedback from muscle that
limits length and velocity via the inherent mechanical properties of muscle. γ
d
, dynamic γ-motor neurons; γ
s
, static γ-motor neurons.
(Reproduced, with permission, from Houk J in: Medical Physiology, 13th ed. Mount-Castle VB [e ditor]. Mosby, 1974.)
CLINICAL BOX 9–3 Clonus
A characteristic of states in which increased γ-motor neuron discharge is present is clonus. This neurologic sign is the occurrence of
regular, repetitive, rhythmic contractions of a muscle subjected to sudden, maintained str etch. Only sustained clonus with five or more
beats is considered abnormal. Ankle clonus is a typical example. This is initiated by brisk , maintained dorsiflexion of the foot, and the
response is rhythmic plantar flexion at the ankle. The stretch reflex–inverse st retch reflex sequence may contribute to this response.
However, it can occur on the basis of synchronized motor neuron discharge without G olgi tendon organ discharge. The spindles of the
tested muscle are hyperactive, and the burst of impulses from them discharges all the mo tor neurons supplying the muscle at once. The
consequent muscle contraction stops spindle discharge. However, the stretch has been m aintained, and as soon as the muscle relaxes it is
again stretched and the spindles stimulated. Clonus may also occur after disruption of de scending cortical input to a spinal glycinergic
inhibitory interneuron called the Renshaw cell. This cell receives excitatory input from α-motor neuron s via an axon collateral (and in
turn it inhibits the same). In addition, cortical fibers activating ankle flexors contact Rens haw cells (as well as type Ia inhibitory
interneurons) that inhibit the antagonistic ankle extensors. This circuitry prevents reflex stimulation of the extensors when flexors are
active. Therefore, when the descending cortical fibers are damaged (upper motor neuron lesion), the inhibition of antagonists is absent.
The result is repetitive, sequential contraction of ankle flexors and extensors (clonus). C lonus may be seen in patients with amyotrophic
lateral sclerosis, stroke, multiple sclerosis, spinal cord damage, and hepatic encephalopat hy.
ation is clearly seen. Passive flexion of the elbow, for example, meets immediate resistan ce as a result of the stretch reflex in the triceps
muscle. Further stretch activates the inverse stretch reflex. The resistance to flexion sudd enly collapses, and the arm flexes. Continued
passive flexion stretches the muscle again, and the sequence may be repeated. This sequ ence of resistance followed by give when a limb
is moved passively is known as the clasp-knife effect because of its resemblanc e to the closing of a pocket knife. It is also known as the
lengthening reaction because it is the response of a spastic muscle to lengthening.
POLYSYNAPTIC REFLEXES:
THE WITHDRAWAL REFLEX
Polysynaptic reflex paths branch in a complex fashion (Figure 9–7). The number of sy napses in each of their branches varies. Because of
the synaptic delay at each synapse, activity in the branches with fewer synapses reaches the motor neurons first, followed by activity in
the longer pathways. This causes prolonged bombardment of the motor neurons from a single stimulus and consequently prolonged
responses. Furthermore, some of the branch pathways turn back on themselves, permi tting activity to reverberate until it becomes unable
to cause a propagated transsynaptic response and dies out. Such reverberating circuits are common in the brain and spinal cord.
WITHDRAWAL REFLEX
The withdrawal reflex is a typical polysynaptic reflex that occurs in response to a usuall y painful stimulation of the skin or subcutaneous
tissues and muscle. The response is flexor muscle contraction and inhibition of extensor muscles, so that the body part stimulated is
flexed and withdrawn from the stimulus. When a strong stimulus is applied to a limb, the response includes not only flexion and
withdrawal of that limb but also
spindle discharge, tendon organ discharge, and reciprocal innervation determines the r ate of discharge of α-motor neurons (see Clinical
Box 9–3).
Sensory
B Cneuron
A
MUSCLE TONE
The resistance of a muscle to stretch is often referred to as its tone or tonus. If the motor nerve to a muscle is cut, the m uscle offers very
little resistance and is said to be flaccid. A hypertonic (spastic) muscle is one in wh ich the resistance to stretch is high because of
hyperactive stretch reflexes. Somewhere between the states of flaccidity and spasticity is the ill-defined area of normal tone. The
muscles are generally hypotonic when the rate of γ-motor neuron discharge is low and hypertonic when it is high.
When the muscles are hypertonic, the sequence of moderate stretch → muscle contract ion, strong stretch → muscle relax
Motor neuron
FIGURE 9–7 Diagram of polysynaptic connections between afferent and efferent neurons in the spinal cord. The dorsal root fiber
activates pathway A with three interneurons, pathway B with four interneurons, and pathway C with four interneurons. Note that one of
the interneurons in pathway C connects to a neuron that doubles back to othe r interneurons, forming reverberating circuits.
extension of the opposite limb. This crossed extensor response i s properly part of the withdrawal reflex. Strong stimuli in experimental
animals generate activity in the interneuron pool that spreads to all four extremities. This is difficult to demonstrate in normal animals but
is easily demonstrated in an animal in which the modulating effects of impulses from the brain have been abolished by prior section of
the spinal cord (spinal animal). For example, when the hind limb of a spinal cat is pinched, the stimulate d limb is withdrawn, the
opposite hind limb extended, the ipsilateral forelimb extended, and the contralateral fore limb flexed. This spread of excitatory impulses
up and down the spinal cord to more and more motor neurons is called irradiati on of the stimulus, and the increase in the number of
active motor units is called recruitment of motor units.
IMPORTANCE OF THE
WITHDRAWAL REFLEX
Flexor responses can be produced by innocuous stimulation of the skin or by stretch of the muscle, but strong flexor responses with
withdrawal are initiated only by stimuli that are noxious or at least potentially harmful to t he animal. These stimuli are therefore called
nociceptive stimuli. Sherrington pointed out the survival value of the withdrawal response. Flexi on of the stimulated limb gets it away
from the source of irritation, and extension of the other limb supports the body. The pa ttern assumed by all four extremities puts the
animal in position to run away from the offending stimulus. Withdrawal reflexes are prepotent; that is, they preempt the spinal
pathways from any other reflex activity taking place at the moment.
Many of the characteristics of polysynaptic reflexes can be demonstrated by studying th e withdrawal reflex. A weak noxious stimulus to
one foot evokes a minimal flexion response; stronger stimuli produce greater and great er flexion as the stimulus irradiates to more and
more of the motor neuron pool supplying the muscles of the limb. Stronger stimuli also cause a more prolonged response. A weak
stimulus causes one quick flexion movement; a strong stimulus causes prolonged flexion and sometimes a series of flexion movements.
This prolonged response is due to prolonged, repeated firing of the motor neurons. Th e repeated firing is called after-discharge and is
due to continued bombardment of motor neurons by impulses arriving by complicated and circuitous polysynaptic paths.
As the strength of a noxious stimulus is increased, the reaction time is shortened. Spatial and temporal facilitation occurs at synapses in
the polysynaptic pathway. Stronger stimuli produce more action potentials per second in the active branches and cause more branches to
become active; summation of the EPSPs to the firing level therefore occurs more rapidly .
FRACTIONATION & OCCLUSION
Another characteristic of the withdrawal response is the fact that supramaximal stimulatio n of any of the sensory nerves from a limb
never produces as strong a contraction of the flexor muscles as that elicited by direct ele ctrical stimulation of the muscles themselves.
This indicates that the afferent inputs fractionate the motor neuron p ool; that is, each input goes to only part of the motor neuron pool
for the flexors of that particular extremity. On the other hand, if all the sensory inputs a re dissected out and stimulated one after the
other, the sum of the tension developed by stimulation of each is greater than that produ ced by direct electrical stimulation of the muscle
or stimulation of all inputs at once. This indicates that the various afferent inputs share so me of the motor neurons and that occlusion
occurs when all inputs are stimulated at once.
GENERAL PROPERTIES
OF REFLEXES
It is apparent from the preceding description of the properties of monosynaptic and po lysynaptic reflexes that reflex activity is
stereotyped and specific in terms of both the stimulus and the response; a particular stimu lus elicits a particular response. The fact that
reflex responses are stereotyped does not exclude the possibility of their being modified by experience. Reflexes are adaptable and can
be modified to perform motor tasks and maintain balance. Descending inputs from high er brain regions play an important role in
modulating and adapting spinal reflexes.
ADEQUATE STIMULUS
The stimulus that triggers a reflex is generally very precise. This stimulus is called the adequate stimulus for the particular reflex. A
dramatic example is the scratch reflex in the dog. This spinal reflex is adequately stimula ted by multiple linear touch stimuli such as
those produced by an insect crawling across the skin. The response is vigorous scratch ing of the area stimulated. If the multiple touch
stimuli are widely separated or not in a line, the adequate stimulus is not produced and n o scratching occurs. Fleas crawl, but they also
jump from place to place. This jumping separates the touch stimuli so that an adequate sti mulus for the scratch reflex is not produced. It
is doubtful if the flea population would survive long without the ability to jump.
FINAL COMMON PATH
The motor neurons that supply the extrafusal fibers in skeletal muscles are the efferent s ide of many reflex arcs. All neural influences
affecting muscular contraction ultimately funnel through them to the muscles, and they a re therefore called the final common paths.
Numerous inputs converge on them. Indeed, the surface of the average motor neuron and its dendrites accommodates about 10,000
synaptic knobs. At least five inputs go from the same spinal segment to a typical spinal m otor neuron. In addition to these, there are
excitatory and inhibitory inputs, generally relayed via interneurons, from other levels o f the spinal cord and multiple long-descending
tracts from the brain. All of these pathways converge on and determine the activity in th e final common paths.
CENTRAL EXCITATORY
& INHIBITORY STATES
The spread up and down the spinal cord of subliminal fringe effects from excitatory sti mulation has already been mentioned. Direct and
presynaptic inhibitory effects can also be widespread. These effects are generally trans ient. However, the spinal cord also shows
prolonged changes in excitability, possibly because of activity in reverberating circuits o r prolonged effects of synaptic mediators. The
terms central excitatory state and central inhibitory state have been used to describe pr olonged states in which excitatory influences
overbalance inhibitory influences and vice versa. When the central excitatory state is ma rked, excitatory impulses irradiate not only to
many somatic areas of the spinal cord but also to autonomic areas. In chronically parap legic humans, for example, a mild noxious
stimulus may cause, in addition to prolonged withdrawal-extension patterns in all four lim bs, urination, defecation, sweating, and blood
pressure fluctuations (mass reflex).
CHAPTER SUMMARY
A reflex arc consists of a sense organ, an afferent neuron, one or more synapses w ithin a central integrating station, an efferent neuron,
and an effector response.
A muscle spindle is a group of specialized intrafusal muscle fibers with contractile p olar ends and a noncontractile center that is located
in parallel to the extrafusal muscle fibers and is innervated by types Ia and II afferent f ibers and γ-motor neurons. Muscle stretch
activates the muscle spindle to initiate reflex contraction of the extrafusal muscle fibers in the same muscle (stretch reflex).
A Golgi tendon organ is a netlike collection of knobby nerve endings among the fa scicles of a tendon that is located in series with
extrafusal muscle fibers and innervated by type Ib afferents. They are stimulated by both passive stretch and active contraction of the
muscle to relax the muscle (inverse stretch reflex) and function as a transducer to regulate mu scle force.
A collateral from an Ia afferent branches to terminate on an inhibitory interneuron that synapses on an antagonistic muscle (reciprocal
innervation) to relax that muscle when the agonist contracts. Clonus is the occurrence of regular, rhythmic contractions of a muscle
subjected to sudden, maintained stretch. A sequence of increased resistance followed by reduced resistance when a limb is moved
passively is known as the lengthening reaction.
MULTIPLE-CHOICE QUESTIONS
For all questions, select the single best answer unless otherwise directed. 1. The inverse stretch reflex
A) has a lower threshold than the stretch reflex.
B) is a monosynaptic reflex.
C) is a disynaptic reflex with a single interneuron inserted
between the afferent and efferent limbs.
D) is a polysynaptic reflex with many interneurons inserted between the afferent and effe rent limbs.
E) requires the discharge of central neurons that release acetylcholine.
2. When γ-motor neuron discharge increases at the same time as
α-motor neuron discharge to muscle,
A) prompt inhibition of discharge in spindle Ia afferents takes place.
B) the contraction of the muscle is prolonged.
C) the muscle will not contract.
D) the number of impulses in spindle Ia afferents is smaller than when α discharge alone i s increased.
E) the number of impulses in spindle Ia afferents is greater than when α discharge alone is increased.
3. Which of the following is not characteristic of a reflex?
A) Modification by impulses from various parts of the CNS
B) May involve simultaneous contraction of some muscles and relaxation of others
C) Chronically suppressed after spinal cord transection
D) Always involves transmission across at least one synapse
E) Frequently occurs without conscious perception 4. Withdrawal reflexes are not
A) initiated by nociceptive stimuli.
B) prepotent.
C) prolonged if the stimulus is strong.
D) an example of a flexor reflex.
E) accompanied by the same response on both sides of the body.
CHAPTER RESOURCES
Haines DE (editor): Fundamental Neuroscience for Basic and Clinical Applications, 3rd e d. Elsevier, 2006.
Hulliger M: The mammalian muscle spindle and its central control. Rev Physiol Bio chem Pharmacol 1984;101:1.
Hunt CC: Mammalian muscle spindle: Peripheral mechanisms. Physiol Rev 1990; 70: 643.
Jankowska E: Interneuronal relay in spinal pathways from proprioceptors. Prog Neuro biol 1992;38:335.
Kandel ER, Schwartz JH, Jessell TM (editors): Principles of Neural Science, 4th ed. Mc Graw-Hill, 2000.
Lundberg A: Multisensory control of spinal reflex pathways. Prog Brain Res 1979;50:11.
Matthews PBC: Mammalian Muscle Receptors and Their Central Actions, Williams & Wilkins, 1972.
This page intentionally left blank
SECTION III CENTRAL &
PERIPHERAL NEUROPHYSIOLOGY
CH APTER
Pain & Temperature 10
OBJEC TIV ES
After studying this chapter, you should be able to:
Name the types of peripheral nerve fibers and receptor types that mediate
warmth, cold, and nociception.
Explain the difference between pain and nociception.
Explain the differences between fast and slow pain and acute and chronic pain.
Explain hyperalgesia and allodynia.
Describe and explain referred pain.
INTRODUCTION
One of the most common reasons an individual seeks the advice of a physician is becau se he or she is in pain. Pain was called by
Sherrington, “the physical adjunct of an imperative protective reflex.” Painful stimuli gen erally initiate potent withdrawal and avoidance
responses. Pain differs from other sensations in that it sounds a warning that something is wrong, preempts other signals, and is
associated with an unpleasant affect. It turns out to be immensely complex because whe n pain is prolonged and tissue is damaged, central
nociceptor pathways are sensitized and reorganized.
NOCICEPTORS &
THERMORECEPTORS
Pain and temperature sensations arise from unmyelinated dendrites of sensory neurons located around hair follicles throughout the
glabrous and hairy skin as well as deep tissue. Impulses from nociceptors (pain) are transmitted via two fiber types. One system
comprises thinly myelinated Aδ fibers (2–5 μm in diameter) which conduct at rates of 1 2–30 m/s. The other is unmyelinated C fibers
(0.4–1.2 μm in diameter) which conduct at low rates of 0.5–2 m/s. Thermorecepto rs also span these two fiber types. Cold receptors are
on dendritic endings of Aδ fibers and C fibers, whereas warmth (heat) receptors are o n C fibers.
Mechanical nociceptors respond to strong pressure (eg, from a sharp object). Thermal n ociceptors are activated by skin temperatures
above 45 °C or by severe cold. Chemically sensitive nociceptors respond to various ag ents like bradykinin, histamine, high acidity, and
environmental irritants. Polymodal nociceptors respond to combinations of these stimuli.
167
Mapping experiments show that the skin has discrete coldsensitive and heat-sensitive spo ts. There are 4 to 10 times as many cold-
sensitive as heat-sensitive spots. The threshold for activation of warmth receptors is 30 °C, and th ey increase their firing rate up to 46
°C. Cold receptors are inactive at temperatures of 40 °C, but then steadily increase t heir firing rate as skin temperature falls to about 24
°C. As skin temperature further decreases, the firing rate of cold receptors decreases u ntil the temperature reaches 10 °C. Below that
temperature, they are inactive and the cold becomes an effective local anesthetic.
Because the sense organs are located subepithelially, it is the temperature of the subcutan eous tissues that determines the responses. Cool
metal objects feel colder than wooden objects of the same temperature because the meta l conducts heat away from the skin more rapidly,
cooling the subcutaneous tissues to a greater degree.
A major advance in this field has been the cloning of three thermoreceptors and nocice ptors. The receptor for moderate cold is the cold
and menthol-sensitive receptor 1 (CMR 1). Two types of vanilloid receptors respond to noxious heat (VR1 and VRL-1). Vanillins are
a group of compounds, including capsaicin, that cause pain. The VR1 receptors respon d not only to capsaicin but also to protons and to
potentially harmful temperatures above 43 °C. VRL-1, which responds to temperatur es above 50 °C but not to capsaicin, has been
isolated from C fibers. There may be many types of receptors on single peripheral C fi ber endings, so single fibers can respond to many
different noxious stimuli. However, the different properties of the VR1 and the VRL-1 receptors make it likely that there are many
different nociceptor C fibers systems as well.
CMR1, VR1, and VRL1 are members of the transient receptor potential (TRP) f amily of excitatory ion channels. VR1 has a PIP
2
binding site, and when the amount of PIP
2
bound is decreased, the sensitivity of the receptors is increased. Aside from the fact tha t
activation of the cool receptor causes an influx of Ca
2+
, little is known about the ionic basis of the initial depolarization they produce. In
the cutaneous receptors in general, depolarization could be due to inhibition of K
+
channels, activation of Na
+
channels, or inhibition of
the Na
+
–K
+
pump, but the distinction between these possibilities has not been made.
CLASSIFICATION OF PAIN
For scientific and clinical purposes, pain is defined by the International Association for t he Study of Pain (IASP) as, “an unpleasant
sensory and emotional experience associated with actual or potential tissue damage, or d escribed in terms of such damage.” This is to be
distinguished from the term nociception which the IASP defines as the unconscious activity indu ced by a harmful stimulus applied to
sense receptors.
Pain is sometimes classified as fast and slow pain. A painful stimulus causes a “bright,” sh arp, localized sensation (fast pain) followed
by a dull, intense, diffuse, and unpleasant feeling (slow pain). Evidence suggests that fast pain is due to activity in the
CLINICAL BOX 10–1 Itch & Tickle
Itching (pruritus) is not much of a problem for normal individuals, but severe itchi ng that is difficult to treat occurs in diseases such as
chronic renal failure, some forms of liver disease, atopic dermatitis, and HIV infection. Especially in areas where many naked endings of
unmyelinated nerve fibers occur, itch spots can be identified on the skin by careful map ping. In addition, itch-specific fibers have been
demonstrated in the ventrolateral spinothalamic tract. This and other evidence implicate th e existence of an itch-specific path. Relatively
mild stimulation, especially if produced by something that moves across the skin, produc es itch and tickle. Scratching relieves itching
because it activates large, fast-conducting afferents that gate transmission in the dorsal ho rn in a manner analogous to the inhibition of
pain by stimulation of similar afferents. It is interesting that a tickling sensation is usually r egarded as pleasurable, whereas itching is
annoying and pain is unpleasant. Itching can be produced not only by repeated local m echanical stimulation of the skin but also by a
variety of chemical agents. Histamine produces intense itching, and injuries cause its liberation in the skin. However, in most instances
of itching, endogenous histamine does not appear to be the responsible agent; doses of histamine that are too small to produce itching still
produce redness and swelling on injection into the skin, and severe itching frequently o ccurs without any visible change in the skin. The
kinins cause severe itching.
A δ pain fibers, whereas slow pain is due to activity in the C pain fibers. Itch and tickle are related to pain sensation (see Clinical Box
10–1).
Pain is frequently classified as physiologic or acute pain and pathologic or chronic pain, which includes inflammatory pain and
neuropathic pain. Acute pain typically has a sudden onset and recedes during the h ealing process. Acute pain can be considered as
“good pain” as it serves an important protective mechanism. The withdrawal reflex is an example of this protective role of pain.
Chronic pain can be considered “bad pain” because it persists long after recovery from an injury and is often refractory to common
analgesic agents, including nonsteroidal anti-inflammatory drugs (NSAIDs) and opiates . Chronic pain can result from nerve injury
(neuropathic pain) including diabetic neuropathy, toxin-induced nerve damage, and ischemia. Causalgia is a type of neuropathic pain
(see Clinical Box 10–2).
Pain is often accompanied by hyperalgesia and allodynia. Hyperalgesia is an exaggerated response to a noxious stimulus, whereas
allodynia is a sensation of pain in response to an innocuous stimulus. An example of the latter is the painful sensation from a warm
shower when the skin is damaged by sunburn.
CLINICAL BOX 10–2 Neuropathic Pain
Neuropathic pain may occur when nerve fibers are injured. Commonly, it is excruciating and a d ifficult condition to treat. It occurs in
various forms in humans. For example, in causalgia, spontaneous burning pain occurs long after seemingly trivial injuries. The pain is
often accompanied by hyperalgesia and allodynia. Reflex sympathetic dystrophy is often present as well. In this condition, the skin in
the affected area is thin and shiny, and there is increased hair growth. Research in anim als indicates that nerve injury leads to sprouting
and eventual overgrowth of noradrenergic sympathetic nerve fibers into the dorsal roo t ganglia of the sensory nerves from the injured
area. Sympathetic discharge then brings on pain. Thus, it appears that the periphery has been short-circuited and that the relevant altered
fibers are being stimulated by norepinephrine at the dorsal root ganglion level. Alpha-a drenergic blockade produces relief of causalgia-
type pain in humans, though for unknown reasons α
1
-adrenergic blockers are more effective than α
2
-adrenergic blocking agents.
Treatment of painful sensory neuropathy is a major challenge and current therapies are often inadequate.
Hyperalgesia and allodynia signify increased sensitivity of nociceptive afferent fibers. F igure 10–1 shows how chemicals released at the
site of injury can further activate nociceptors leading to inflammatory pain. Injured cells release chemicals such as K
+
that depolarize
nerve terminals, making nociceptors more responsive. Injured cells also release bradyk inin and Substance P, which can further sensitize
nociceptive terminals. Histamine is released from mast cells, serotonin (5-HT) from plate lets, and prostaglandins from cell membranes,
all contributing to the inflammatory process and they activate or sensitize the nociceptors . Some released substances act by releasing
another one (eg, bradykinin activates both Aδ and C fibers and increases synthesis and release of prostaglandins). Prostaglandin E
2
(a
cyclooxygenase metabolite of arachidonic acid) is released from damaged cells and pro duces hyperalgesia. This is why aspirin and other
NSAIDs (inhibitors of cyclooxygenase) alleviate pain.
DEEP PAIN
The main difference between superficial and deep sensibility is the different nature of th e pain evoked by noxious stimuli. This is
probably due to a relative deficiency of Aδ nerve fibers in deep structures, so there is l ittle rapid, bright pain. In addition, deep pain and
visceral pain are poorly localized, nauseating, and frequently associated with sweating a nd changes in blood pressure. Pain can be
elicited experimentally from the periosteum and ligaments by injecting hypertonic saline i nto them. The pain produced in this fashion
initiates reflex contraction of nearby skeletal muscles. This reflex contraction is similar to the muscle spasm associated with injuries to
bones, tendons, and joints. The steadily contracting muscles become ischemic, and ische mia stimulates the pain receptors in the muscles
(see Clinical Box 10–3). The pain in turn initiates more spasm, setting up a vicious cycle .
VISCERAL PAIN
In addition to being poorly localized, unpleasant, and associated with nausea and autono mic symptoms, visceral pain often radiates or is
referred to other areas.
Mast cell
CGRP Substance P
Histamine Bradykinin
Lesion
5-HT
Prostaglandin
K
+
Dorsal root
ganglion neuron
CGRP
Substance P Blood vessel Spinal cord
FIGURE 10–1 In response to tissue injury, chemical mediators can sensitize an d activate nociceptors. These factors contribute to
hyperalgesia and allodynia. Tissue injury releases bradykinin and prostaglandins that se nsitize or activate nociceptors, which in turn
releases substance P and calcitonin gene-related peptide (CGRP). Substance P acts on m ast cells to cause degranulation and release
histamine, which activates nociceptors. Substance P causes plasma extravasation and CGRP dilates blood vessels; the resulting edema
causes additional release of bradykinin. Serotonin (5-HT) is released from plate lets and activates nociceptors. (From Kandel ER, Schwartz JH,
Jessell TM [editors]: Principles of Neural Science. McGraw-Hill, 2000.)
CLINICAL BOX 10–3 Muscle Pain
If a muscle contracts rhythmically in the presence of an adequate blood supply, pain do es not usually result. However, if the blood
supply to a muscle is occluded, contraction soon causes pain. The pain persists after the contraction until blood flow is reestablished.
These observations are difficult to interpret except in terms of the release during contrac tion of a chemical agent (Lewis’s “P factor”)
that causes pain when its local concentration is high enough. When the blood supply is r estored, the material is washed out or
metabolized. The identity of the P factor is not settled, but it could be K
+
. Clinically, the substernal pain that develops when the
myocardium becomes ischemic during exertion (angina pectoris) is a classic exam ple of the accumulation of P factor in a muscle.
Angina is relieved by rest because this decreases the myocardial O
2
requirement and permits the blood supply to remove the factor.
Intermittent claudication, the pain produced in the leg muscles of persons with occ lusive vascular disease, is another example. It
characteristically comes on while the patient is walking and disappears on stopping. Visc eral pain, like deep somatic pain, initiates reflex
contraction of nearby skeletal muscle. This reflex spasm is usually in the abdominal wall and makes the abdominal wall rigid. It is most
marked when visceral inflammatory processes involve the peritoneum. However, it can occur without such involvement. The spasm
protects the underlying inflamed structures from inadvertent trauma. Indeed, this reflex spasm is sometimes called “guarding.”
The autonomic nervous system, like the somatic, has afferent components, central integr ating stations, and effector pathways. The
receptors for pain and the other sensory modalities present in the viscera are similar to th ose in skin, but there are marked differences in
their distribution. There are no proprioceptors in the viscera, and few temperature and touch receptors. Nociceptors are present, although
they are more sparsely distributed than in somatic structures.
Afferent fibers from visceral structures reach the CNS via sympathetic and parasympat hetic nerves. Their cell bodies are located in the
dorsal roots and the homologous cranial nerve ganglia. Specifically, there are visceral a fferents in the facial, glossopharyngeal, and vagus
nerves; in the thoracic and upper lumbar dorsal roots; and in the sacral roots (Figure 10 –2). There may also be visceral afferent fibers
from the eye in the trigeminal nerve.
As almost everyone knows from personal experience, visceral pain can be very severe . The receptors in the walls of the hollow viscera
are especially sensitive to distention of these organs. Such distention can be produced ex perimentally in the gastrointestinal tract by
inflation of a swallowed balloon attached to a tube. This produces pain that waxes and w anes (intestinal colic) as the intestine contracts
and relaxes on the balloon. Similar colic is produced in intestinal obstruction by the contr actions of the dilated intestine above the
obstruction. When a visceral organ is inflamed or hyperemic, relatively minor stimuli ca use severe pain. This is probably a form of
hyperalgesia.
REFERRED PAIN
Irritation of a visceral organ frequently produces pain that is felt not at that site but in so me somatic structure that may be a considerable
distance away. Such pain is said to be referred to the somatic structure. Obviously, kno wledge of referred pain and the common sites of
pain referral from each of the viscera is of great importance to the physician. Perhaps t he bestknown example is referral of cardiac pain
to the inner aspect of the left arm. Other examples include pain in the tip of the shoulder caused by irritation of the central portion of the
diaphragm and pain in the testicle due to distention of the ureter. Additional instances ab ound in the practices of medicine, surgery, and
dentistry. However, sites of reference are not stereotyped, and unusual reference sites occur with considerable frequency. Cardiac pain,
for instance, may be referred to the right arm, the abdominal region, or even the back and neck.
When pain is referred, it is usually to a structure that developed from the same embryon ic segment or dermatome as the structure in
which the pain originates. This principle is called the dermatomal rule. For example, the heart and the arm have the same segmental
origin, and the testicle has migrated with its nerve supply from the primitive urogenital ri dge from which the kidney and ureter have
developed.
The basis for referred pain may be convergence of somatic and visceral pain fibers on the same second-order neurons in the dorsal horn
that project to the thalamus and then to the somatosensory cortex (Figure 10–3). This is called the convergence–projection theory.
Somatic and visceral neurons converge in lamina I–VI of the ipsilateral dorsal horn, bu t neurons in lamina VII receive afferents from
both sides of the body—a requirement if convergence is to explain referral to the side opposite that of the source of pain. The somatic
nociceptive fibers normally do not activate the second-order neurons, but when the vis ceral stimulus is prolonged, facilitation of the
somatic fiber endings occurs. They now stimulate the secondorder neurons, and of cou rse the brain cannot determine whether the
stimulus came from the viscera or from the area of referral.
Glossopharyngeal nerve
Superior laryngeal nerve Vagus
THORACIC PAIN LINE Splanchnic nerves
(T7–9)
R splanchnic
nerves (T7–9)
Duodenum and jejunum (splanchnic nerves) Upper thoracic vagal rami
Brachial plexus
a
i (
i
(Intercostal nerves)
Lower
splanch
nic nerves (T10–L1)
Ureter
(T11–L1)
Peripheral
Central
diaphragm
diaphragm
(intercostal nerves)
(phrenic nerve)
Small intestine splanchnic nerves
e t a l peritoneum(T9–11) r i
Somatic nerves (T11–L1)
Fundus (T11–L1)
PELVIC lleum
PAIN LINE
Trigone
Prostate Cervix andUrethra upper vagina
(pelvic nerves, S2–4)
(S2–4)
Parasympathetic
Testicle
rami (S2–4)
(sacral nerves, S2–4) (genitofemoral nerves, L1–2) (spermatic plexus, T10)