You are on page 1of 801

Medical Physiology

A Systems Approach

Hershel Raff, PhD


Professor
Departments of Medicine and Physiology
Medical College of Wisconsin
Endocrine Research Laboratory
Aurora St. Luke’s Medical Center
Milwaukee, Wisconsin

Michael Levitzky, PhD


Professor of Physiology and Anesthesiology
Louisiana State University Health Sciences Center
New Orleans, Louisiana

Medical
New York Chicago San Francisco Lisbon London Madrid Mexico City
Milan New Delhi San Juan Seoul Singapore Sydney Toronto
Copyright © 2011 by The McGraw-Hill Companies, Inc. All rights reserved. Except as permitted under the United States Copyright Act of 1976, no part of this
publication may be reproduced or distributed in any form or by any means, or stored in a database or retrieval system, without the prior written permission of the
publisher.

ISBN: 978-0-07-176663-0

MHID: 0-07-176663-4

The material in this eBook also appears in the print version of this title: ISBN: 978-0-07-162173-1,
MHID: 0-07-162173-3.

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 owner, with no intention of infringement of the trademark. Where such designations appear in this
book, they have been printed with initial 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.

NOTICE
Medicine is an ever-changing science. As new research and clinical experience broaden our knowledge, changes in treatment and drug therapy are required. The
authors and the publisher of this work have checked with sources believed to be reliable in their efforts to provide information that is complete and generally
in accord with the standards accepted at the time of publication. However, in view of the possibility of human error or changes in medical sciences, neither the
authors nor the publisher nor any other party who has been involved in the preparation or publication of this work warrants that the information contained herein
is in every respect accurate or complete, and they disclaim all responsibility for any errors or omissions or for the results obtained from use of the information
contained in this work. Readers are encouraged to confi rm the information contained herein with other sources. For example and in particular, readers are advised
to check the product information sheet included in the package of each drug they plan to administer to be certain that the information contained in this work is
accurate and that changes have not been made in the recommended dose or in the contraindications for administration. This recommendation is of particular
importance in connection with new or infrequently used drugs.

TERMS OF USE

This is a copyrighted work and The McGraw-Hill Companies, Inc. (“McGrawHill”) and its licensors reserve all rights in and to the work. Use of this work is
subject to these terms. Except as permitted under the Copyright Act of 1976 and the right to store and retrieve one copy of the work, you may not decompile,
disassemble, reverse engineer, reproduce, modify, create derivative works based upon, transmit, distribute, disseminate, sell, publish or sublicense the work or
any part of it without 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 MAKE NO GUARANTEES OR WARRANTIES AS TO THE ACCURACY,
ADEQUACY OR COMPLETENESS OF OR RESULTS TO BE OBTAINED FROM USING THE WORK, INCLUDING ANY INFORMATION THAT CAN BE
ACCESSED THROUGH THE WORK VIA HYPERLINK OR OTHERWISE, AND EXPRESSLY DISCLAIM ANY WARRANTY, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO IMPLIED WARRANTIES OF MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. McGraw-Hill
and its licensors do not warrant or guarantee that the functions contained in the work will meet your requirements or that its operation will be uninterrupted or
error free. Neither McGraw-Hill nor its licensors shall be liable to you or anyone else for any inaccuracy, error or omission, regardless of cause, in the work or for
any damages resulting therefrom. McGraw-Hill has no responsibility for the content of any information accessed through the work. Under no circumstances shall
McGraw-Hill and/or its licensors be liable for any indirect, incidental, special, punitive, consequential or similar damages that result from the use of or inability
to use the 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 cause whatsoever
whether such claim or cause arises in contract, tort or otherwise.
To our students, mentors, colleagues, and families.
iv
KEY FEATURES v
About the Authors

Hershel Raff Michael Levitzky


Hershel Raff received his Michael Levitzky is Professor of
Ph.D. in Environmental Physiology and Anesthesiology
Physiology from the Johns at the Louisiana State University
Hopkins University in 1981 Health Sciences Center and
and did postdoctoral training is Director of Basic Science
in Endocrinology at the Curriculum at the LSU School
University of California at of Medicine in New Orleans.
San Francisco. He joined the He received a B.A. from the
faculty at the Medical College University of Pennsylvania in
of Wisconsin in 1983, and rose 1969 and a Ph.D. in Physiology
to the rank of Professor of Medicine (Endocrinology, from Albany Medical College in 1975. He joined
Metabolism, and Clinical Nutrition) and Physiology the faculty of the LSU School of Medicine in 1975,
in 1991. He is also Director of the Endocrine Research rising to the rank of Professor in 1985. He has also
Laboratory at Aurora St. Luke’s Medical Center. At the been Adjunct Professor of Physiology at Tulane
Medical College of Wisconsin, he teaches physiology University School of Medicine since 1991. Dr.
and pharmacology to medical and graduate students. Levitzky teaches physiology to medical students,
He was an inaugural inductee into the Society of residents, fellows, and graduate students. He has
Teaching Scholars, received the Beckman Basic received numerous teaching awards from student
Science Teaching Award and the Outstanding Teacher organizations at both LSU and Tulane. He received
Award, and has been one of the MCW’s Outstanding the inaugural LSUHSC Allen A. Copping Award for
Medical Student Teachers each year the award has Excellence in Teaching in the Basic Sciences in 1997
been given. Dr Raff was elected to Alpha Omega and the American Physiological Society’s Arthur C.
Alpha (AOA) Honor Medical Society as a faculty Guyton Teacher of the Year Award in 1998. He was
teacher in 2005. He is also an Adjunct Professor of elected to the Alpha Omega Alpha (AOA) Honor
Biomedical Sciences at Marquette University. He is Medical Society as a faculty teacher in 2006. Dr.
Associate Editor of Advances in Physiology Education. Levitzky has served the American Physiological
He was Secretary-Treasurer of the Endocrine Society Society as a member of the Education Committee
and is currently Chair of the Publications Committee and as a member of the Steering Committee of the
of the American Physiological Society. He was Teaching Section. He served as a member of the
elected a Fellow of the American Association for the National Board of Medical Examiners United States
Advancement of Science in 2005. Dr. Raff ’s basic Medical Licensing Examination (USMLE) Step 1
research focuses on the adaptation to low oxygen Physiology Test Material Development Committee
(hypoxia). His clinical interest focuses on pituitary and from 2007-2011. He is the author or co-author of
adrenal diseases, with a special focus on the diagnosis several other textbooks, one of which, Pulmonary
of Cushing’s syndrome. Dr. Raff is also a co-author of Physiology (Lange/McGraw-Hill), is currently in its
Vander’s Human Physiology (McGraw-Hill) currently 7th edition.
in its 12th Edition, and Physiology Secrets, currently in
its 2nd Edition.
vi
Contents
Contributors xi
Preface xiii

S E C T I O N I 10. Cardiac Muscle Structure and Function 93


INTRODUCTION 1 Kathleen H. McDonough
11. Smooth Muscle Structure and Function 99
1. General Physiological Concepts 1 Kathleen H. McDonough
Hershel Raff and Michael Levitzky

S E C T I O N II
S E C T I O N IV
CNS/NEURAL PHYSIOLOGY 105
CELL PHYSIOLOGY 9
12. Introduction to the Nervous System 105
2. Cells and Cellular Processes 9 Susan M. Barman
David Landowne
13. General Sensory Systems: Touch, Pain,
3. Cell Membranes and Transport and Temperature 115
Processes 15 Susan M. Barman
David Landowne
14. Spinal Reflexes 125
4. Channels and the Control of Membrane Susan M. Barman
Potential 33
David Landowne 15. Special Senses I: Vision 133
Susan M. Barman
5. Sensory Generator Potentials 43
David Landowne 16. Special Senses II: Hearing and
Equilibrium 147
6. Action Potentials 47 Susan M. Barman
David Landowne
17. Special Senses III: Smell and Taste 159
7. Synapses 59 Susan M. Barman
David Landowne
18. Control of Posture and Movement 167
Susan M. Barman
S E C T I O N III 19. Autonomic Nervous System 177
MUSCLE PHYSIOLOGY 79 Susan M. Barman
8. Overview of Muscle Function 79 20. Electrical Activity of the Brain, Sleep–Wake
Kathleen H. McDonough States, and Circadian Rhythms 185
Susan M. Barman
9. Skeletal Muscle Structure and Function 83
Kathleen H. McDonough 21. Learning, Memory, Language,
and Speech 191
Susan M. Barman

vii
viii CONTENTS

37. Acid–Base Regulation


S E C T I O N V and Causes of Hypoxia 375
CARDIOVASCULAR Michael Levitzky
PHYSIOLOGY 199 38. Control of Breathing 385
Michael Levitzky
22. Overview of the Cardiovascular
System 199
Lois Jane Heller and David E. Mohrman S E C T I O N VII
23. Cardiac Muscle Cells 211 RENAL PHYSIOLOGY 397
Lois Jane Heller and David E. Mohrman
39. Renal Functions, Basic Processes,
24. The Heart Pump 223 and Anatomy 397
Lois Jane Heller and David E. Mohrman Douglas C. Eaton and John P. Pooler
25. Cardiac Function Assessments 235 40. Renal Blood Flow and
Lois Jane Heller and David E. Mohrman Glomerular Filtration 409
26. Peripheral Vascular System 251 Douglas C. Eaton and John P. Pooler
David E. Mohrman and Lois Jane Heller 41. Clearance 417
27. Vascular Control 263 Douglas C. Eaton and John P. Pooler
David E. Mohrman and Lois Jane Heller 42. Tubular Transport Mechanisms 423
28. Venous Return and Cardiac Output 275 Douglas C. Eaton and John P. Pooler
David E. Mohrman and Lois Jane Heller 43. Renal Handling of Organic Substances 429
29. Arterial Pressure Regulation 285 Douglas C. Eaton and John P. Pooler
David E. Mohrman and Lois Jane Heller 44. Basic Renal Processes for Sodium,
30. Cardiovascular Responses to Chloride, and Water 437
Physiological Stress 295 Douglas C. Eaton and John P. Pooler
Lois Jane Heller and David E. Mohrman 45. Regulation of Sodium and
Water Excretion 449
S E C T I O N VI Douglas C. Eaton and John P. Pooler
PULMONARY PHYSIOLOGY 305 46. Regulation of Potassium Balance 463
Douglas C. Eaton and John P. Pooler
31. Function and Structure of the 47. Regulation of Acid–Base Balance 471
Respiratory System 305 Douglas C. Eaton and John P. Pooler
Michael Levitzky
48. Regulation of Calcium and
32. Mechanics of the Respiratory System 313 Phosphate Balance 485
Michael Levitzky
Douglas C. Eaton and John P. Pooler
33. Alveolar Ventilation 331
Michael Levitzky S E C T I O N VIII
34. Pulmonary Perfusion 341 GI PHYSIOLOGY 491
Michael Levitzky
35. Ventilation–Perfusion Relationships 49. Overview of the GI System—Functional
and Respiratory Gas Exchange 353 Anatomy and Regulation 491
Michael Levitzky Kim E. Barrett
36. Transport of Oxygen and 50. Gastric Secretion 507
Carbon Dioxide 363 Kim E. Barrett
Michael Levitzky
CONTENTS ix

51. Pancreatic and Salivary Secretion 517 62. Anterior Pituitary Gland 623
Kim E. Barrett Patricia E. Molina
52. Water and Electrolyte Absorption 63. Thyroid Gland 633
and Secretion 527 Patricia E. Molina
Kim E. Barrett
64. Parathyroid Gland and Calcium
53. Intestinal Mucosal Immunology and Phosphate Regulation 643
and Ecology 535 Patricia E. Molina
Kim E. Barrett
65. Adrenal Gland 655
54. Intestinal Motility 543 Patricia E. Molina
Kim E. Barrett
66. Endocrine Pancreas 671
55. Functional Anatomy of the Liver Patricia E. Molina
and Biliary System 559
67. Male Reproductive System 683
Kim E. Barrett
Patricia E. Molina
56. Bile Formation, Secretion, 68. Female Reproductive System 695
and Storage 565
Patricia E. Molina
Kim E. Barrett
69. Endocrine Integration of Energy
57. Handling of Bilirubin and Ammonia and Electrolyte Balance 715
by the Liver 575
Patricia E. Molina
Kim E. Barrett
58. Digestion and Absorption of
Carbohydrates, Proteins, S E C T I O N X
and Water-soluble Vitamins 583 INTEGRATIVE PHYSIOLOGY 729
Kim E. Barrett
59. Lipid Assimilation 593 70. Control of Body Temperature 729
Kim E. Barrett Hershel Raff and Michael Levitzky
71. Hypoxia and Hyperbaria 735
S E C T I O N IX Michael Levitzky and Hershel Raff
ENDOCRINE AND METABOLIC 72. Exercise 745
PHYSIOLOGY 601 Michael Levitzky and Kathleen H. McDonough
73. Aging 753
60. General Principles of Endocrine Hershel Raff
Physiology 601
Patricia E. Molina
61. The Hypothalamus and Posterior Answers to Study Questions 757
Pituitary Gland 613 Index 761
Patricia E. Molina
This page intentionally left blank
Contributors

Susan M. Barman, PhD Kathleen H. McDonough, PhD


Professor Professor
Department of Pharmacology & Toxicology and Department of Physiology
Neuroscience Program Associate Dean, School of Graduate Studies
Michigan State University Louisiana State University Health Sciences Center
East Lansing, Michigan New Orleans, Louisiana
Kim E. Barrett, PhD Patricia E. Molina, MD, PhD
Professor of Medicine and Dean of Graduate Studies Richard Ashman, PhD Professor and Head of Physiology
University of California, San Diego Department of Physiology
La Jolla, California Louisiana State University Health Sciences Center
New Orleans, Louisiana
Douglas C. Eaton, PhD
Distinguished Professor and Chair of Physiology and David E. Mohrman, PhD
Professor of Pediatrics Associate Professor, Emeritus
Department of Physiology and Department of Physiology and Pharmacology
Center for Cell & Molecular Signaling University of Minnesota Medical School
Emory University School of Medicine Duluth, Minnesota
Atlanta, Georgia
John P. Pooler, PhD
Lois Jane Heller, PhD Professor of Physiology Emeritus
Professor Emory University School of Medicine
Department of Physiology and Pharmacology Atlanta, Georgia
University of Minnesota Medical School
Hershel Raff, PhD
Duluth, Minnesota
Professor
David Landowne, PhD Departments of Medicine and Physiology
Professor Medical College of Wisconsin
Department of Physiology and Biophysics Endocrine Research Laboratory
University of Miami, Miller School of Medicine Aurora St. Luke’s Medical Center
Miami, Florida Milwaukee, Wisconsin
Michael Levitzky, PhD
Professor of Physiology and Anesthesiology
Louisiana State University Health Sciences Center
New Orleans, Louisiana

xi
This page intentionally left blank
Preface

Medical Physiology: A Systems Approach is intended to pro- Review of Medical Physiology. Finally, the Introduction, Muscle
vide first-year medical and graduate students and advanced Physiology, and Integrative Physiology sections are new.
undergraduate students with the basis of the major physiolog- Each chapter begins with a list of objectives and concludes
ical processes necessary for understanding both health and with a chapter summary. Most chapters also end with a clinical
disease. The curriculum of many medical schools is changing; correlation that reinforces the major physiological principles
most medical schools have undergone, or are in the midst of, just learned, and illustrates their importance to understanding
a transition from the block approach, with each discipline disease states. Each chapter ends with multiple-choice ques-
having its own course, to a vertically integrated structure. One tions designed to test the knowledge of some of the major con-
of the goals of an integrated curriculum is the presentation of cepts covered in the chapter.
much more clinical material during the first two years of med- The authors are indebted to our mentors who provided us
ical school as well as the reinforcement of basic concepts in the with a foundation for advances in physiological education in
two primarily clinical years. As a result, there is an increasing the 21st century. We also thank our students for providing a
focus on the essential concepts necessary to understand sounding board for the pedagogical approaches exploited in
pathophysiology. this book. The authors are thankful to Michael Weitz, Karen
Therefore, this book is considerably shorter than the full- Davis, and Brian Kearns at McGraw-Hill for their outstanding
length, standard physiology textbook. It focuses on major editorial help. Finally, we give special thanks to our families:
physiological concepts and clinical correlates, and leaves the Judy and Jonathan; and Elizabeth, Edward, and Sarah.
minute details to larger books. Most of this book evolved from
the Lange Physiology Series of monographs. The section on the Hershel Raff
central nervous system arose from the 23rd edition of Ganong’s Michael Levitzky

xiii
This page intentionally left blank
SECTION I INTRODUCTION

1
C H A P T E R

General Physiological
Concepts
Hershel Raff and Michael Levitzky

O B J E C T I V E S

■ Understand the general properties of a eukaryotic cell.


■ Explain the general organization of the internal organs of
the body.
■ Compare and contrast the composition of extracellular versus
intracellular fluid.
■ Describe the different types of membrane transport.
■ Understand the general concepts of pressure, flow, resistance,
and compliance.
■ Explain mass balance.
■ Define negative and positive feedback.

INTRODUCTION caused by disease or injury. Therefore, physiology is one of


the foundations of the health sciences.
Physiology is the science of the function of organisms. The
object of physiology is to explain how the organ systems, cells,
and even molecules interact to maintain normal function. THE CELL
The hallmark of physiology is the concept of homeostasis,
which is the maintenance of a normal internal environment The basic building block of the organs of the body is the
in the face of external and internal perturbations so that the cell. The details of cell physiology are covered in Section 2.
functions of the cells and organ systems of the body are main- Figure 1–1 shows the general structure of a nucleated
tained. This is accomplished primarily by feedback systems (eukaryotic) cell. It is surrounded by a cell membrane that is
such that when a system is disturbed, a variety of local composed of a lipid bilayer, membrane proteins, and car-
responses, systemic reflexes (automatic, rapid reactions bohydrates in association with lipids (glycolipids) or pro-
to stimuli), and long-term adjustments are activated to teins (glycoproteins). The cell membrane is the gatekeeper
return the system to its normal set point. By understanding for anything that enters or leaves the cell and is a barrier that
how things work under normal conditions, one can appreci- helps to maintain the internal composition of the cell. Some
ate when and why there is a malfunction. This is called membrane proteins and glycoproteins function as sensors,
pathophysiology—a lasting disturbance in normal function or receptors, which sense the external environment and

Ch01_001-008.indd 1 11/26/10 9:50:25 AM


2 SECTION I Introduction

Secretory granules

Golgi
apparatus

Centrioles

Rough
endoplasmic Smooth
reticulum endoplasmic
reticulum

Lysosomes
Nuclear envelope Lipid
droplets

Mitochondrion

Globular heads
Nucleolus

FIGURE 1–1 Diagram showing a hypothetical cell in the center seen with a light microscope. (Adapted with permission from Fawcett DW, et al.
The ultrastructure of endocrine glands, Recent Prog Horm Res. 1969;25:315–380.)

chemical signals, and then signal the interior of the cell, usu- response to stimuli. Some cells contain numerous lipid drop-
ally through chemical second messengers or changes in elec- lets, because fat is hydrophobic and does not dissolve readily
trical activity of the membrane. Other membrane proteins in the aqueous environment of the cytosol. Mitochondria have
function as transporters that regulate the entry or exit of two lipid bilayer membranes in apposition, and are the energy-
substances into or out of the cell. The lipid bilayer structure
and associated proteins of the cell membrane are shown in
Figure 1–2.
The inside of the cell is composed of cytosol, which is a liq- Extracellular fluid
uid consisting primarily of water in which proteins, metabo- Transmembrane
Carbohydrate proteins Phospholipids
lites, fuel, and inorganic ions (called electrolytes) are dissolved. portion of
Also dispersed in the cytosol are a variety of subcellular parti- glycoprotein
cles and organelles. Altogether, the combination of cytosol
and the intracellular structures is called the cytoplasm. The Channel
organelles include the endoplasmic reticulum, which is an
extensive network of membranes inside of which are proteins Integral
proteins
and other important chemicals. The endoplasmic reticulum is
important in many metabolic functions and the packaging of
secretory products. The ribosomes are involved in translation,
Peripheral
which is the synthesis of proteins from messenger protein
RNA (mRNA). These ribosomes are associated with endoplas-
mic reticulum in a combined structure called the rough endo- Polar regions
plasmic reticulum (RER). The Golgi apparatus is associated
with endoplasmic reticulum; the Golgi apparatus packages Nonpolar regions Intracellular fluid
material synthesized in the RER. Lysosomes are intracellu-
lar, membrane-surrounded structures that contain digestive FIGURE 1–2 Organization of the phospholipid bilayer and
enzymes located in granules that are involved in intracellular associated proteins in a biological cell membrane. (Reproduced with
metabolism. Secretory granules contain molecules that the permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed.
cell will release into the extracellular fluid by exocytosis, in McGraw-Hill, 2008.)
CHAPTER 1 General Physiological Concepts 3

generating organelles. The cytoplasmic organelles are held in plasm and nucleus that respond to signals that enter the cell.
position by filaments and microtubules, arising from the cen- Examples of such signals are steroid hormones such as estro-
trosomes, which are also important in the movement of chro- gen and testosterone that are lipophilic (“fat-loving”) and, as a
mosomes during cell division. Finally, the nucleus, also result, can readily diffuse through the cell membrane to exert
surrounded by a lipid bilayer membrane called the nuclear an intracellular action.
envelope, contains chromatin that is composed of DNA con-
taining the nucleic acid code for cellular differentiation, func-
tion, and replication. DNA contains the genes that encode
mRNAs that are produced from DNA by transcription. Also GENERAL STRUCTURE
contained within the nucleus is the nucleolus, which is the site OF THE BODY
of ribosome synthesis.
As you will learn in many chapters in this book, the cell Figure 1–3 is a diagrammatic representation of the human
membrane contains many different types of receptors that body. The organs (e.g., brain and heart) receive nutrients and
sense extracellular signals that are transduced into intracellu- eliminate waste products via the circulatory system. The heart
lar signals. In addition, there are receptors within the cyto- is illustrated as two parts—right and left—as a functional

Central nervous system

Afferent and efferent nerves

O2 CO2 Atmosphere

Right O2 CO2 Lung Left

Heart Heart
Venous Arterial
blood blood
Tissues
Nutrients

Waste products

Endocrine glands

Hormones

Liver GI tract Food &


Synthesis water
Nutrients intake

Metabolism Bile Waste

Kidney Reabsorption

FIGURE 1–3 General


organization of the major organs
Filtration Waste
of the body. Arrows show the
direction of blood flow and flux
Feces
Urine of gases, nutrients, hormones, and
waste products.
4 SECTION I Introduction

depiction even though it is actually one organ. The right side TABLE 1–1 Composition of extracellular and
of the heart receives partially deoxygenated blood returning intracellular fluids.
from the tissues and pumps blood to the lungs. In the lungs,
oxygen diffuses into the blood from the gas phase for use in Extracellular Intracellular
cellular respiration in the body, and carbon dioxide, a waste Concentration (mM) Concentration (mM)
product of cellular respiration, is eliminated by diffusion from Na+ 140 12
the blood into the gas phase. The left side of the heart receives +
K 5 150
oxygenated blood from the lung and pumps the blood into the
2+
arterial tree to perfuse the organs of the body. Nutrients, min- Ca 1 0.0001
erals, vitamins, and water are taken in by the ingestion of food Mg 2+
1.5 12
and liquids and absorption in the gastrointestinal (GI) tract. −
Cl 100 7
The liver, usually considered part of the GI system, processes

substances absorbed into the blood from the GI tract, and also HCO3 24 10
synthesizes new molecules such as glucose from precursors. Amino acids 2 8
Metabolic waste products are eliminated by the GI system in
Glucose 4.7 1
the feces and by the kidney in the urine. The two main integra-
tive controllers of the internal environment are the nervous Protein 0.2 4
and endocrine systems. The nervous system is composed of The intracellular concentrations are slightly different for different tissues. The Ca2+
the brain, spinal cord, sensory systems, and nerves. The endo- concentrations shown are the free, biologically active ions not bound to proteins.
Total Ca2+ (bound plus free) are considerably higher in extracellular (2.5 mM) and
crine system is composed of ductless glands and scattered intracellular (1.5 mM) fluids.
secretory cells distributed throughout the body that release Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human
hormones into the blood in response to metabolic, hormonal, Physiology, 11th ed. McGraw-Hill, 2008.

and neural signals. It is the function of the nervous and endo-


crine systems to coordinate the behavior and interactions of
the organ systems described throughout this book.
the cell membrane is governed by the concentration gradient
Water is the most abundant molecule in the body, constitut-
and the ease with which each molecule can go through the
ing about 50–60% of the total body weight. All cells and organs
cell membrane (permeability); energy expenditure is not
exist in an aqueous environment. The intracellular water is the
directly required for diffusion, which is why it is sometimes
main component of the cytosol. Water is also the main compo-
called passive diffusion. There are also protein transporters
nent of the extracellular fluid. The extracellular fluid includes
located in the cell membrane that mediate facilitated diffu-
the interstitial fluid, which bathes the cells of the body, the
sion of molecules that are too large or hydrophilic to perme-
blood plasma, which is the liquid component of the blood,
ate the membrane by simple diffusion. Facilitated diffusion
cerebrospinal fluid, which is found only in the central ner-
does not require energy and moves molecules down a concen-
vous system, synovial fluid, which is found in joints such as
tration gradient. By contrast, active transport is a process of
the knee, and lymph, which is a liquid formed from interstitial
moving molecules across a cell membrane against a concen-
fluid that flows back to the circulatory system via the lym-
tration gradient; it can be thought of as a pump that uses
phatic system. There are significant differences in the compo-
energy to do work.
sition of intracellular and extracellular fluids that are very
The movement of water molecules across the cell membrane
important in many aspects of cellular function (Table 1–1).
also occurs by diffusion from a higher to a lower water “con-
centration.” This is termed osmosis; water moves from a com-
partment with fewer osmotically active particles (higher water
GENERAL PHYSICAL concentration) to a compartment with more osmotically active
FACTORS AND CONCEPTS particles (lower water concentration). Examples of osmotically
active particles are ions such as sodium, potassium, and chlo-
It is not an accident that physiology and physics come from the ride, and organic molecules such as glucose and amino acids.
same Greek word physis (nature). It is important that students
of physiology understand the physical forces and factors that
govern body function. BUFFERING AND pH
One of the most tightly controlled variables in the body is the
MEMBRANE TRANSPORT hydrogen ion concentration of the intracellular and extracellu-
lar fluids. This is because most proteins have optimal function
There are several different mechanisms by which molecules within a very narrow range of pH. Remember that the pH is the
cross the cell membrane either coming into or going out of negative logarithm (base 10) of the hydrogen ion concentration
the cell. These are all described in detail in Section 2. The sim- in molar units—when pH is low, the fluid is acidic; when pH is
plest is diffusion in which the rate at which a molecule crosses high, the fluid is alkaline. The body has several mechanisms for
CHAPTER 1 General Physiological Concepts 5

maintaining a normal pH. These are extensively covered in change, increasing the resistance will decrease the flow. If the
Sections 6 and 7. The body can rid itself of acid by increasing flow through the tube does not change, increasing the resis-
the elimination of carbon dioxide from the lungs. This is tance will increase the pressure difference between the ends
because carbon dioxide and hydrogen ion are linked through of the tube. The pressure difference between the two ends of
chemical reactions to bicarbonate, one of the main buffers in the tube represents energy conversion to heat by the internal
the body. A buffer is an ionic compound that attenuates changes friction of the fluid with itself and with the wall of the vessel.
in pH by combining with or releasing hydrogen ions. The kid- You will notice that the relationship between pressure, flow,
neys can also remove hydrogen ion from the body via the com- and resistance for liquid flowing through a tube is analogous
plex processes involved in producing urine. Finally, changes in to Ohm’s law for electricity in which the voltage drop across a
intracellular and extracellular pH can be prevented by a variety circuit (analogous to a pressure drop in a tube with liquid
of buffers in addition to bicarbonate. flowing through it) is proportional to the product of current
(analogous to flow) and resistance.
Most of the vessels or chambers in the body will stretch pas-
HYDROSTATIC FORCES AND sively if the pressure difference across their walls increases.
PRESSURE, RESISTANCE, AND This results in an increased volume of the vessel. This ability to
COMPLIANCE stretch in response to an increased transmural (across the
wall) pressure difference is called compliance. A less specific
Pressure is defined as force per unit of area. The pressure at the term for compliance is distensibility.
bottom of a column of liquid increases with the height of the The inverse of compliance is elastance. Elastance can there-
column and is also dependent on the density of the liquid and fore be thought of as the resistance to stretch when the trans-
on gravity. The pressure at any point in a column of liquid is mural pressure difference increases or as the ability of a vessel
called the hydrostatic pressure, and is the pressure difference to return to its original volume after the increased transmural
between that point and the top of the column. Hydrostatic pres- pressure difference is removed. It is directly related to Hooke’s
sure differences have many important physiologic consequences, law of elasticity for mechanical springs.
particularly in blood vessels, as will be seen in Section 5.
The flow of a fluid (a liquid or a gas) is quantified as the
volume of the fluid moving through a vessel per unit of time. MASS BALANCE AND
The relationships among pressure, flow, and the resistance METABOLISM
offered by the vessels through which a fluid flows can be com-
plex, but are simplified as follows. The rate of flow of liquid In order to achieve the steady state that defines homeostasis,
through a tube is proportional to the difference in pressure any substance taken in by the body must be nearly equal to
between the two ends of the tube and inversely proportional to the amount of the substance leaving the body plus that
the resistance to flow through the tube. Resistance cannot be removed by metabolism (Figure 1–4). The influx of a sub-
determined directly, but is calculated from the pressure and stance is the sum of uptake in the lung, absorption in the
flow. If the resistance does not change, increasing the pressure GI tract, synthesis in the body (e.g., liver synthesis of glucose
difference through a tube will increase the flow. If the pressure from molecular precursors), and release from cells (e.g., fatty
difference from one end of the tube to the other does not acid release from adipose tissue). The efflux of a substance is

NET GAIN TO BODY DISTRIBUTION WITHIN NET LOSS FROM


BODY BODY

Food GI tract Storage depots Metabolism

Air Lungs POOL

Excretion from body


Reversible via lungs, GI tract,
Synthesis in body
incorporation kidneys, skin,
into other menstrual flow
molecules

FIGURE 1–4 Concept of mass balance. The central compartment is usually extracellular fluid (which includes blood plasma). It receives
substances from intake, synthesis, and release from cells. It loses substances by excretion, metabolism, and uptake into cells. In the steady state,
when a substance is said to be “in balance,” intake and excretion are nearly equal. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s
Human Physiology, 11th ed. McGraw-Hill, 2008.)
6 SECTION I Introduction

the sum of metabolism, uptake into cells, losses via the GI


tract, respiratory system, sweat, and urinary excretion. In the
CONTROL SYSTEMS
steady state, the difference between total influx and efflux The main focus of physiology is the understanding of the
should be very close to zero. From minute to minute, there are mechanisms by which cells, organs, and organ systems main-
obviously large differences between influx and efflux, but over tain homeostasis. This is accomplished primarily by negative
days to weeks when the substance is usually in balance, the feedback. The general concept is that the body tries to increase
difference should be close to zero. Examples of this are sodium a variable when it is below its optimum (termed the set
balance described in Section 7 and calcium and phosphate point), and decrease a variable when it is above its optimum.
balance described in Section 9. This is analogous to the thermostat that controls room
temperature by adjusting the heating and/or cooling of the
room. For example, if you open a window on a cold day, the
EXCITABILITY room temperature decreases from the set point of the thermo-
stat. This is called perturbation. The thermostat contains a
As you will learn in Sections 2–4, living cells have an electrical sensor that detects the difference between the room tempera-
charge difference across the cell membrane created primarily ture and the set point. The thermostat signals the furnace to
by differences in ion concentration and movement between generate heat, and the room temperature is returned toward
the outside and inside of the cell (see Table 1–1). As a result, normal. The difference between the low point in room tem-
the membranes have a resting electrical potential that can be perature and the final room temperature at steady state is called
changed by a variety of inputs. Dramatic changes in ion flux the correction. Because the window is left open in this exam-
across the cell membrane lead to large changes in electrical ple, room temperature does not quite return to the set point;
potential that can result in major cellular responses. For exam- the remaining difference between the final room temperature
ple, muscular contraction described in Section 3 results from and its thermostatic set point is called the error. The ability of
the depolarization of the muscle cell membrane that is trans- the control system to restore the system to its set point is called
duced into a chemical signal within the cell that leads to the gain, which is represented by the following equation:
generation of force and movement.
Correction
Gain = ____________
Remaining error
(1)

CELL–CELL INTERACTIONS A classic example of this is shown in Figure 1–5 that shows
the response of the cardiovascular system to rapid blood loss
As you will learn in Sections 2–4, 8, and 9, cells interact with (hemorrhage). In this example, the rapid loss of 1 L of blood
each other locally. One mechanism is by direct contact between leads to a decrease in mean blood pressure from the set point
cells via tight junctions and gap junctions. Another is the of 100 to 75 mm Hg. As you will learn in Chapter 29, there are
synapse, in which neurons can release chemicals called neu- sensors in the cardiovascular system called baroreceptors that
rotransmitters to alter the function of a neighboring cell. detect blood pressure. These sensors change their neural input
Finally, there are a variety of chemical signals by which cells to the brain to activate systemic reflexes to restore blood pres-
can communicate with neighboring cells by diffusion. An sure to normal. In this example, these reflexes restore blood
example of this is paracrine signaling by which humoral fac- pressure to 95 mm Hg. The correction, therefore, is 20 mm Hg
tors are released by one cell, diffuse through the interstitial and the remaining error is 5 mm Hg. Using equation (1), this
fluid, and bind to a receptor on a neighboring cell within the gives a gain of about 4. Although clinicians do not usually cal-
same tissue. culate gain when taking care of patients, it is a convenient way

Set Rapid blood loss Final blood pressure


point 100
95 100-95=5 Remaining error
Original
95-75=20 Correction due perturbation
75 to reflexes
Nadir
blood pressure (mm Hg)

Correction 20
Gain= = =4
Remaining error 5
Mean arterial

FIGURE 1–5 Moderate hemorrhage as


an example of the gain of a feedback control
system. The higher the gain of a system, the
better able it is to restore a controlled variable
Time (min)
to its set point in response to a perturbation.
CHAPTER 1 General Physiological Concepts 7

to think of the ability of reflexes to restore a perturbed system ■ There are significant concentration gradients between
to normal via negative feedback. The higher the gain, the intracellular and extracellular fluid for sodium, potassium,
higher the ratio of correction to remaining error and the better calcium, magnesium, chloride, and bicarbonate, as well as
the control system is at restoring the system to its set point. For organic compounds.
example, as you will learn in Chapter 70, the control of body ■ Molecules can enter the cell by passive diffusion, and through
temperature has a very high gain. transporters that do not (facilitated transport) and do directly
use cellular energy (active transport).
Included in many feedback systems is a change in behavior.
■ The rate of flow of a liquid through a tube is determined by the
For example, drinking extra water when blood volume is
pressure difference between the inflow and outflow, and the
decreased helps to restore plasma volume. Putting on warm
resistance to flow of the tube.
clothes and curling up helps to minimize heat loss in a cold
■ Most important substances in the body are in balance, with
environment. Finally, set points of control systems can change.
the influx and efflux being approximately the same over time.
Examples of this are resetting of the baroreceptor set point
■ Most systems are controlled by negative feedback with the
during chronic increases in blood pressure (hypertension) that
controlled variable being able to shut off its own release much
you will learn about in Chapter 29, and during the acclimatiza-
like a thermostat controls room temperature.
tion to the low ambient oxygen of high altitude (hypoxia) that
you will learn about in Chapter 71.
Although most control systems of the body are negative
feedback, there are a few examples of positive feedback, which
STUDY QUESTIONS
are feedback loops that amplify themselves. You will learn 1. Which of the following organelles is primarily response for
about several examples of this in Chapter 68. One is the stimu- generation of energy?
lation of the anterior pituitary hormone LH by estrogen just A) Golgi apparatus
before ovulation that causes a large increase in LH, which then B) mitochondria
stimulates more estrogen release, and so on. Another example C) lysosomes
is the birth of a baby during which stretch of the cervix stimu- D) ribosomes
lates the release of oxytocin from the posterior pituitary gland 2. Which of the following has an intracellular fluid concentration
that, in turn, stimulates stronger uterine contractions. This much higher than its extracellular fluid concentration?
causes additional cervical stretch, more oxytocin release, and A) sodium ion
greater uterine contractions. Positive feedback is also respon- B) chloride ion
C) glucose
sible for detrimental effects in the body. One example is heart
D) potassium ion
failure during which the pumping of the heart decreases due
3. Which of the following would result in an increase in flow of
to, for example, an infection of the heart muscle. The resultant
a liquid through a tube?
decrease in blood pressure leads to reflexes that stimulate the
A) increase in resistance
heart to pump harder in an effort to raise blood pressure. This
B) increase in pressure at the downstream end of the tube
additional stress on the heart actually makes it work less well, C) increase in pressure at the inflow end of the tube
and the heart failure feeds on itself. D) increase in length of the tube
Another important concept in homeostatic control is poten- 4. Which of the following has the highest feedback gain?
tiation. This is when one substance augments the response to
A) starting blood pressure = 100; low point in blood
another substance, even though the first substance does not pressure = 70; final blood pressure after feedback
exert a significant response on its own. An example of this that correction = 90
you will learn in Chapters 49 and 66 is the release of the GI B) starting body temperature = 37.2°C; high point in body
hormones from the intestine in response to a meal. These hor- temperature = 38.9°C; final body temperature after
mones can potentiate the pancreatic insulin response to feedback correction = 37.4°C
absorbed glucose. This is an example of feedforward potentia- C) starting blood glucose = 80 mg/dL; high point in blood
tion, because these GI hormones “announce” the impending glucose = 110 mg/dL; final blood glucose after feedback
increase in blood glucose before glucose absorption actually correction = 85 mg/dL
occurs in the small intestine. When glucose finally arrives via D) starting plasma osmolality = 280 mOsm/kg; low point in
the bloodstream at the pancreas, there is a potentiated insulin plasma osmolality = 270 mOsm/kg; final osmolality after
feedback correction = 278 mOsm/kg
response such that hyperglycemia is prevented.

CHAPTER SUMMARY
■ The cell is surrounded by a membrane that regulates the
intracellular composition and the flux of molecules in and
out of the cell.
■ Water is the most abundant molecule in the body, and its
concentration and balance is highly regulated.
This page intentionally left blank
SECTION II CELL PHYSIOLOGY

2
C H A P T E R

Cells and Cellular


Processes
David Landowne

O B J E C T I V E S

■ Recognize and describe the types of electrophysiological events.


■ Describe the types of membrane channels and their roles.
■ Describe physiological control systems.

INTRODUCTION logical events in terms of inputs, processes, and outputs


(Figure 2–1). Complex processes can be broken down into
Life is cellular, and cells are the fundamental units of life. simpler ones, with the outputs of one or more processes be-
Without cells, there would be no living beings. All the cells of coming the inputs to the next one.
In order to survey the processes discussed here, it is useful
a given individual are ultimately derived from a single fertil-
to consider a three-cell model of the body. Figure 2–2 shows
ized ovum. Most of the cells of multicellular organisms reside a sensory neuron or nerve cell, a motor neuron, and a skel-
within their tissues and organs. This chapter concentrates on etal muscle cell. These cells represent the hardware the body
the cellular processes and leaves the discussion of their higher uses to carry out the dynamic cell processes described in the
organization to chapters concerned with the various organ previous paragraph. The cells have specialized portions for
systems. Drugs, toxins, and diseases are introduced to illus- the different processes. Starting from the left, the sensory
cell has one end that is specialized for the transduction of a
trate the cellular processes.
stimulus into a cellular signal. The various senses have dif-
ferent specializations to accomplish this transduction.
COMMUNICATION Besides the classic five senses (touch, hearing, vision, taste,
and smell), there are sensors or proprioceptors inside the
Dynamic cell processes support sensory perception of the en- body that sense internal parameters—for example, body
vironment, communication, and the integration of informa- temperature, blood pressure, blood oxygen levels, or the
tion within and between cells, as well as their expression, or lengths of various muscles.
actions on the environment. These are the processes that If it is sufficiently large, the initial signal causes another
enable the cell to contribute to the functioning of tissues, or- signal to propagate over the axon (the long cylindrical portion
gans, and individuals. These processes make up one of the of the nerve cell) until it reaches the other end, where the sen-
phenomena of cells—excitability. The others, reproduction sory neuron makes a synaptic connection with dendrites of
and metabolism, are not covered in depth here. Perception, the motor neuron, located in the central nervous system
integration, and expression can be best considered as physio- (CNS). The message is transmitted from the presynaptic cell

Ch02_009-014.indd 9 11/26/10 9:40:02 AM


10 SECTION II Cell Physiology

branes. Every living cell has a surface membrane that separates


its intracellular and extracellular spaces. All cells, not just
Input Output
Process those of nerve and muscle, are electrically negative inside the
cell with respect to outside. This is called the membrane
potential. When the cells are “resting”—that is, not signal-
FIGURE 2–1 The input–process–output structural framework ing—their membrane potential is called the resting potential.
is a specification of causal relationships in a system. Complex Chapter 4 is about the origins of the resting potential.
systems can be considered as composed of simple units. (Modified with Even though the signals described above are changes in
permission from Landowne D: Cell Physiology. New York: Lange Medical potential, they are generally referred to as named poten-
Books/McGraw-Hill, 2006.) tials. On the left in Figure 2–2, there is the sensory genera-
tor potential, which has two properties to distinguish it
from the next signal, the action potential. The sensory
generator potential is local; it occurs only within a few mil-
to the postsynaptic cell, where it is integrated or evaluated limeters of the sensory ending. The action potential is prop-
along with messages from other neurons that synapse on the agated; it travels from the sensory ending to the presynaptic
same motor neuron. In the complete organism, this integra- terminal, perhaps more than a meter away. The sensory gen-
tion and comparison occurs in many cells and at different lev- erator potential is also graded; a larger-amplitude stimulus
els within the CNS, so the decision to move or not move can produces a larger-amplitude sensory generator potential. In
be made considering more than one input and also anything contrast, the action potential has a stereotyped amplitude
the organism has learned from the past. and duration; it is all-or-none. The information about the
If the motor neuron is sufficiently excited, it will send an- stimulus is encoded in the number of action potentials, or
other message along the axon that leads to a synapse on a the number per second. A larger-amplitude stimulus will
muscle cell. In healthy people, this neuromuscular synapse result in a higher frequency of action potentials, each with
leads to a signal that propagates over the length of the mus- the same stereotyped amplitude. Because the all-or-none
cle cell and activates contraction, which can act on the envi- character of neurons is similar to the true-or-false character
ronment. Other actions on the environment are effected by of logical propositions, cyberneticists (people who study
the secretions of various glands; these too may be controlled control and communication in the animal and the machine)
by synaptic connections. These muscles and glands may act have considered that neural events and the relations among
internally (e.g., to control heart rate or blood pressure) or them can be treated by means of propositional logic.
externally (for locomotion or communication with other Chapters 5 and 6 are about sensory generator potentials and
people). action potentials, respectively.
These signals are all electrical; they all represent changes in The presynaptic terminals contain a mechanism to re-
the electrical potential difference across the various cell mem- lease the contents of vesicles containing chemical transmit-

Hardware Sensory ending Axon Synapse Axon Muscle

Signals Sensory Action Synaptic Action Endplate


(potentials) generator
Local Propagated Local Propagated Local

Graded All-or-none Graded All-or-none Graded

Channels Mechano Voltage Chemo Voltage Chemo


sensitive sensitive sensitive sensitive sensitive

Cybernetics Input Transmission Process Transmission Output

FIGURE 2–2 The cellular processes of a hypothetical three-celled organism. Different types of channels underlie different physiological
processes that support the input–process–output functions of animals, including humans. (Modified with permission from Landowne D: Cell Physiology.
New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 2 Cells and Cellular Processes 11

ters that diffuse across the narrow synaptic cleft and react Voltage-sensitive channels underlie action potentials. They
with the postsynaptic cell to produce a postsynaptic poten- open in response to a change in membrane potential. When
tial. The postsynaptic potential is also local and graded. It is they are open, ions flow through them, and this changes the
only seen within a few millimeters of the site of the presynap- membrane potential as well. The generator potential or the
tic ending and its amplitude depends on how much transmit- synaptic potentials activate these channels, and then they open
ter is released. There are excitatory postsynaptic potentials the remaining adjacent voltage-sensitive channels. This ac-
(EPSPs) and inhibitory postsynaptic potentials (IPSPs), de- counts for the propagation and all-or-none, stereotyped qual-
pending on whether the postsynaptic potential makes the cell ity of the action potentials. Nerve and skeletal muscle action
more or less likely to initiate an action potential. If there is potentials are produced by the successive activation of volt-
sufficient excitation to overwhelm any inhibition that may be age-sensitive sodium channels, followed by voltage-sensitive
occurring, an action potential will be initiated in the postsyn- potassium channels. There are also voltage-sensitive calcium
aptic cell. There are many presynaptic cells ending on each channels in the presynaptic nerve endings. When the action
postsynaptic neuron as well as various different transmitters potential reaches the presynaptic terminal, these calcium
in different synapses. These transmitters, the release mecha- channels open and permit calcium to enter the cell. The calci-
nism, and the resulting postsynaptic potentials are discussed um binds to intracellular components and initiates the release
in Chapter 7. of synaptic transmitters.
The action potential in the motor neuron and the synapse Chemosensitive channels are responsible for the synaptic
with the muscle cell are very similar to the neuron to neuron potentials. The transmitters bind to these channels, causing
synapse discussed above. In the light microscope, the neuro- them to open. There are different channels for different trans-
muscular junction looks like a small plate; hence, the junction mitters and also different channels for EPSPs and IPSPs.
is often called an endplate and the postsynaptic potential an Chemosensitive channels also subserve the chemical senses of
endplate potential. The neuromuscular junction differs from smell and taste. There are also channels that open or close in
most other synapses because there is only one presynaptic cell, response to intracellular chemicals such as adenosine triphos-
its effect is always excitatory, and—in healthy people—the phate (ATP) or the cyclic nucleotides, cyclic adenosine mono-
endplate potential is always large enough to initiate an action phosphate (cAMP) or cyclic guanosine monophosphate
potential in the muscle cell. (cGMP). Vision is supported by a reaction series whereby light
The muscle action potential propagates along the length absorption leads to a decrease in cGMP, which produces a clo-
of the cell and into the interior by small transverse tubules, sure of cyclic nucleotide–gated (chemosensitive) channels.
whose membranes are continuous with the surface mem- When sodium ions stop flowing through these channels, the
brane. The action potential excitation is coupled to the mus- membrane potential changes.
cular contraction by processes described in Chapter 10. That From a cybernetic viewpoint, Figure 2–2 indicates that the
chapter also discusses the control of cardiac and smooth body has mechanisms to input information, to transmit it
muscle cells. within the body, to process the information, and to provide
The resting potential, the sensory generator potentials, the output. This type of analysis appears frequently in physiolo-
action potentials, and the synaptic potentials all occur by gy. Much of what you will learn can be broken into various
the opening and closing of channels in the cell membranes. steps where the output of one process becomes the input for
These channels are made of proteins that are embedded in and the next. For example, the sensory generator potentials are
span the membrane connecting the intracellular and extracel- an input to the action potential–generation process and the
lular spaces. Each has a small pore through the middle, which action potential is the input to the voltage-sensitive calcium
may be opened or closed and is large enough to allow specific channel, which permits calcium to enter the presynaptic ter-
ions to flow through and small enough to keep metabolites and minal. This calcium is the input for the transmitter release
proteins from flowing out of the cell. There are many channels, process, and so on.
and a good part of Chapter 3 is devoted to their description.
They are generally named either for the ion that passes through
them or for the agent that causes them to open. CONTROL
There are three classes of channels that act to produce the
changes in potential described in Figure 2–2. All these chan- Although most of this book focuses on isolating the different
nels will be discussed individually in Chapter 3 and then again processes so as to analyze them more easily, an understanding of
in the context of the various potentials in subsequent chapters. the value and true significance of each physiological quality must
Mechanosensitive channels subserve the sensations of refer to the whole organism. A recurring theme throughout all of
touch and hearing and the many proprioceptors that provide physiology is the maintenance of a stable internal environment
information on muscle length, muscle tension, joint position, through homeostasis. Many internal properties (e.g., body tem-
the orientation and angular acceleration of the head, and blood perature or blood glucose levels) are homeostatically controlled
pressure. These channels open when the membrane of the sen- within narrow limits by feedback control systems.
sory ending is stretched, sodium ions flow through the chan- Homeostasis is a property of many complex open systems.
nels, and the membrane potential changes. Feedback control is the central feature of organized activity.
12 SECTION II Cell Physiology

Desired value

Comparator

Effector

Controlled parameter

Sensor

A
From higher centers

Motoneuron

Muscle

Sensory neuron Muscle spindle

FIGURE 2–3 Homeostasis and feedback control. A and B) By having inputs that sense the output and feed information back to the
controller, machines and humans can gain control of their operating conditions. (Modified with permission from Landowne D: Cell Physiology. New York:
Lange Medical Books/McGraw-Hill, 2006.)

A homeostatic system (e.g., a cell, the body, an ecosystem) is an stretched, mechanosensitive channels in sensory organs open,
open system that maintains itself by controlling many dynamic changing membrane potentials in the sensory endings that in-
equilibria. The system maintains its internal balance by reacting duce action potentials to propagate to the spinal cord. Trans-
to changes in the environment with responses of opposite direc- mitter is released, which excites the nerve leading back to the
tion to those that created the disturbance. The balance is main- muscle, where the synaptic process is repeated and the muscle
tained by negative feedback. Perhaps the most familiar negative shortens to compensate for the initial stretch.
feedback control system is the thermostat that controls the tem- There are a few positive feedback systems that are physio-
perature of a room, which was described in Chapter 1. logically important. A positive feedback system is unstable;
The basic steps (Figure 2–3A) in negative feedback control the signal from the sensor increases the effect, which increas-
of any measurable parameter are the measurement by a sen- es the signal from the sensor in a “vicious cycle,” which is lim-
sor, communication of that measurement to a comparator, ited only by the availability of resources. The all-or-none
making the comparison, and communicating the compari- property of action potentials is due to a positive feedback
son to an effector that changes the parameter of interest. The loop. Some other examples of positive feedback were de-
feedback is called negative because the signal to the effector scribed in Chapter 1.
is in the opposite direction to any disturbance and reduces
the difference between the measured value and the desired
value. CHAPTER SUMMARY
The three cells in Figure 2–2, arranged as a negative feed- ■ Communication in excitable cells occurs via electrical signals
back loop (Figure 2–3B), represent the process used to control within the cells and via chemical signals at synapses between
the length of muscles both to maintain posture and to achieve the cells.
movement in response to signals from the brain. This feedback ■ There are two classes of electrical signals: those that are local
loop can be easily demonstrated by the stretch reflex—that is, and graded and those that are propagated and stereotyped, or
the knee-jerk reflex (see Chapter 14). When a muscle is all-or-none.
CHAPTER 2 Cells and Cellular Processes 13

■ The chemical transmitters are released presynaptically and 2. Negative feedback control systems do not
produce an electrical signal in the postsynaptic cell. A) improve the reliability of control.
■ Three classes of ion channels produce the electrical signals: B) require the sensing or measurement of the controlled
mechanosensitive, chemosensitive, and voltage-sensitive process.
channels. C) require communication between separate parts of the
■ Homeostasis by negative feedback control is an important system.
feature of living systems. D) regulate blood pressure and body temperature.
■ There are three basic elements of a negative feedback loop: a E) cause the all-or-none property of the action potential.
sensor, a comparator, an effector, and two communication links
connecting them.

STUDY QUESTIONS
1. Which of the following changes in electrical potential require
voltage-sensitive channels?
A) excitatory synaptic potentials
B) mechanical sensory generator potentials
C) propagated action potentials
D) light sensory generator potentials
E) inhibitory synaptic potentials
This page intentionally left blank
3
C H A P T E R

Cell Membranes and


Transport Processes
David Landowne

O B J E C T I V E S

■ Describe the molecular composition of biological membranes.


■ Describe the functional biophysical properties of biological membranes.
■ Describe classes of ion channels, their molecular structure, and their
biophysical properties.
■ Describe the molecular organization, properties, control, and functional roles
of cell–cell channels.
■ Describe the movement and transport of substances across biological
membranes by passive processes.
■ Describe the movement and transport of substances across biological
membranes by active processes.
■ Describe the physiological importance of two examples of active transport
and two examples of passive transport.
■ Define osmotic pressure.
■ Calculate the osmolarity of simple solutions.
■ Calculate the changes in osmolarity in body compartments caused by
drinking various simple solutions.
■ Describe physiological mechanisms to regulate osmolarity.

Every living cell has a surface membrane that defines its limits LIPIDS
and the connectivity of the intracellular and extracellular com-
partments. Cell membranes are about 10-nm thick and consist Most of the membrane lipids are glycerophospholipids, which
of a 3–4-nm-thick lipid bilayer with various embedded proteins have a glycerol backbone with two of its three –OH groups
that may protrude into either compartment (see Figure 1–2). esterified by fatty acids and the third esterified to a phosphate
Membranes also delimit intracellular organelles, including the group, which is in turn esterified to a small molecule that gives
nuclear envelope, Golgi apparatus, endoplasmic reticulum (ER), its name to the whole molecule (Figure 3–1). The most com-
mitochondria, and various vesicles (see Figure 1–1). The lipid mon glycerophospholipids are phosphatidylcholine (PC),
bilayer is impermeable to charged or polar substances. The pro- phosphatidylethanolamine (PE), and phosphatidylserine
teins handle the transport of specific ions or molecules across (PS). Membranes also contain phosphatidylinositol (PI),
the membranes and thus control the composition of different which plays an important role in signaling within the cyto-
solutions on either side. They support communication across plasm. Notice that PS and PI have a net negative charge.
the membranes and along the surface of the cell and provide Animal cell membranes may also contain sphingolipids, in-
mechanical coupling between cells. cluding the phosphosphingolipid, sphingomyelin, which has

15

Ch03_015-032.indd 15 11/26/10 9:40:20 AM


16 SECTION II Cell Physiology

R
O +
N(CH3)3
O P O–
HCH
O Phosphatidylcholine (PC)
HCH
O O

C=O C=O +
NH3
HCH Phosphatidylethanolamine (PE)
HCH

+
NH3

COO– Phosphatidylserine (PS)

HCH

OH
OH
OH Phosphatidylinositol (PI)
OH HO

FIGURE 3–1 Glycerophospholipids. Along with cholesterol, these form the bilayer that separates the inside of cells and supports the
embedded membrane proteins. (Modified with permission from Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)

two acyl chains and a phosphate-linked choline head linked to The ease of lateral diffusion, or membrane fluidity, is in-
a serine backbone, and glycosphingolipids, which have sugars creased by the presence of unsaturation or double bonds in the
in the head group. Membranes also contain cholesterol, which hydrocarbon tails. This forms a kink in the tail and therefore
has a steroid ring structure. looser packing. At the concentrations generally found in bio-
All of these lipids are amphipathic because they have logical membranes, cholesterol reduces the fluidity because of
hydrophilic, or “water-loving,” head groups and hydropho- its rigid ring structure. Glycosphingolipid head groups tend to
bic, or “water-fearing,” acyl tails. The –OH group of associate with each other and reduce fluidity. Lipid protein
cholesterol is hydrophilic and the rest is hydrophobic. A hy- interactions may also reduce fluidity. There are cholesterol–
drophobic effect arises from the lack of interactions of sphingolipid microdomains, or “lipid rafts,” involved in intra-
hydrocarbons with water and the strong attraction of water cellular trafficking of proteins and lipids.
for itself. Thus, when placed in an aqueous environment,
these lipids spontaneously assemble into closed bilayer
membrane vesicles. PROTEINS
The lipids are relatively free to diffuse laterally within the
plane of the membranes, but—with the exception of The intrinsic proteins of the membrane support the selec-
cholesterol—they are unlikely to flip-flop from one half of tive movement of ions and small molecules from one side of
the bilayer to the other owing to the hydrophilicity of the head the membrane to the other, sense a ligand on one side of the
groups. The bilayer is asymmetrical, with the choline- membrane and transmit a signal to the other side, and pro-
containing phospholipids, PC and sphingomyelin, in the out- vide mechanical linkage for other proteins on either side of
er half and the amino-containing phospholipids, PE and PS, the membrane. The proteins that move materials across the
in the inner half. In addition, the glycosphingolipids are in membrane can be functionally divided into channels,
the noncytoplasmic half and PI is facing the cytoplasm. The pumps, and transporters. Channels may be specific and
asymmetrical arrangement is produced as the membranes are may open and close, but, when open, they facilitate the
assembled in the ER. The phospholipids are synthesized and movement of materials only with their electrochemical
inserted on the cytoplasmic side of the membrane; then a gradients. Ion channels control the flow of electrical cur-
phospholipid translocator or “flippase” transfers PC to the rent through the membrane. Pumps move ions against their
noncytoplasmic side. Sphingomyelin and the glycosphingo- electrochemical gradient at the expense of consuming ATP.
lipids are produced in the Golgi apparatus on the noncyto- The pumps maintain the gradients that allow the channels
plasmic side. and transporters to do their jobs. Transporters can link the
CHAPTER 3 Cell Membranes and Transport Processes 17

movement of two (or more) substances and can move one of analysis of entire genomes suggests that about 20% of the pro-
them against its gradient at the expense of moving the other teins contain one or more TM segments and are thus mem-
one with its gradient. brane proteins.
A protein is the product of translating a gene; it is a folded, Only a few membrane proteins have been crystallized and
linked sequence of alpha amino acids chosen from a palette subjected to x-ray diffraction analysis. These crystals must
with 20 possible different side chains. The peptide link between include lipid or detergent molecules to satisfy the hydrophobic
amino acids –CO–NH– has a planar transconformation; the needs of the TM segments. Most of the solved structures are of
folding occurs according to the torsion angles between the bacterial proteins that have been genetically modified to en-
amino group and the alpha carbon (Φ) and between the alpha hance crystallization. A strong sequence homology between
carbon and the carboxyl group (ψ). The alpha helix axis and the crystallized molecule and part of the human protein is
the beta sheet are secondary structures, with particular torsion taken to indicate that they have similar structures.
angles, that are found in proteins. Channels, pumps, transporters, receptors, and cell adhesion
The conformation or tertiary structure of the entire pro- molecules come in many varieties to serve many roles. The
tein is the three-dimensional relationship of all its atoms. following five sections will describe a taxonomy and the anat-
Proteins have regions of various secondary structures con- omy of examples of each functional class. It may be useful to
nected by linkers with less easily characterized structure. return to this section while reading the later part of this chap-
Most of the proteins discussed in this book have more than ter and those parts of the rest of the book that describe the role
one conformation. For example, a channel may be open or of these molecules in physiological processes.
closed. The local secondary structures do not change very
much during these conformational changes; rather, change
occurs in the relationship between larger portions of the
molecule.
CHANNELS
There is also a supermolecular or quaternary level of organi- In the previous chapter, channels were distinguished by the
zation. Some channels are made of a single polypeptide chain, mechanism by which they open. There are mechanosensitive
while others are made of four to six chains. Many channels channels involved in sensory processes, voltage-sensitive
also have accessory proteins that modulate their function. In channels involved in action potential propagation, and
addition, the lipid matrix imposes structural restrictions on chemosensitive channels involved in synaptic transmission.
the embedded proteins. There are also channels that are usually open, such as chan-
In general, proteins are amphipathic and have regions that nels that maintain resting potential, water channels, and spe-
are more hydrophobic or hydrophilic, depending on the na- cialized cell–cell channels that connect the cytoplasm of one
ture of the side chains. The membrane proteins discussed here cell with the cytoplasm of another. This section describes
have one or more transmembrane (TM) alpha-helical seg- some channels that support various cell processes discussed
ments with hydrophobic side chains in contact with the hy- later in the book. It is not exhaustive; many channels and
drocarbon of the lipid. If more than one helix is involved, it is many classes of channels are not mentioned. This is a “golden
possible to have hydrophobic residues facing the lipid and age” for ion channels. Electrophysiology and molecular and
other groups facing each other in the more interior parts of the structural biology are revealing some amazing membrane
protein. The general pattern is for the protein to cross the proteins.
membrane several times, with intracellular and extracellular Many ion channels are selective and are named according to
loops between TM segments. There is also an N-terminal re- the ion that passes through them. The first channel to be crys-
gion before the first segment and a C-terminal region after the tallized is the resting potential potassium channel, also known
last; an example is shown in Figure 3–3. The N-terminal re- as the inward rectifier or Kir. The reason for this name is
gion can be on either side, but the C-terminal region is usually discussed in the next chapter, along with its function. Kir is a
cytoplasmic. Either or both terminal regions can be quite large tetramer with four identical subunits arranged with radial
compared to the TM regions. symmetry and a pore that permits ion flow at the axis (Figure
The TM folding occurs as the protein is synthesized in the 3–2A). Each monomer has two TM segments with an extracel-
ER. The noncytoplasmic portions of the protein may be glyco- lular P loop in between (Figure 3–2B; see also Figure 3–4,
sylated in the Golgi apparatus before being inserted in the sur- segments 5 and 6). The four P loops dip back into the mem-
face membrane. Subunit assembly may also occur in the ER or brane and together form the lining of a pore that goes about
Golgi apparatus. one third of the way through the membrane. This pore emp-
For most membrane proteins, only the primary sequence is ties into a larger intramembranous cavity that communicates
known. Secondary structure can be predicted by sequence with the cytoplasmic space. The eight helices form a wall for
analysis. The presence of putative hydrophobic helices of suf- the cavity and also surround the inserted P loops. The TM
ficient length is taken as a suggestion of a TM segment. A to- helices form a conical structure with the point toward the
pology or pattern of loops and TM segments can be predicted; cytoplasm.
such a prediction has been tested for many proteins by prepar- The selectivity of the pore for potassium ions depends on the
ing antibodies for the putative extracellular portions. Sequence specific amino acids forming the lining. VGYGD is the
18 SECTION II Cell Physiology

A B

C
FIGURE 3–2 The crystal structure of an inward rectifier K channel (Kir). A) Top view of a ribbon-structure representation
with stick-and-ball for the GYG sequences. B) Side view with two monomers removed; the GYG sequence is a space-filling representation.
C) Close-up view of two VGYGD sequences and an ion. (Modified with permission from Landowne D: Cell Physiology, New York: Lange Medical
Books/McGraw-Hill, 2006.)

K-channel signature sequence (Figure 3–2C); it has been nal helices. Ca2+ and cyclic nucleotides increase the open
found in K channels from more than 200 organisms. This por- probability of some other 2- and 6-TM channels by a similar
tion of the molecule is the selectivity filter because it accepts mechanism.
K+ ions and excludes other ions. The pore is lined with the There are eight subfamilies of 2-TM Kir channels in the hu-
carbonyl oxygen groups; these are in the same relation to each man genome. Several are important in cardiac electrophysiol-
other as the oxygen of the water molecules that coordinate ogy. Kir2 (or IK1) is the original inward rectifier discovered in
around K+ ions in solution because of its positive charge and cardiac muscle; it is responsible for maintaining the resting
the oxygen’s electronegativity. Two of the coordinating oxy- potential. Kir3 channels open via G protein–coupled receptors
gens from glycines just below the tyrosines can be seen in (GPCRs); in the heart, they are referred to as KACh. Kir6 chan-
Figure 3–2C. Ions with different charges or radii will coordi- nels open when the ADP/ATP ratio rises. In the heart, they are
nate water differently and thus will be less likely than K+ ions referred to as KATP.
to leave the water and enter the K channel.
It is thought that Figure 3–2 represents a closed Kir channel.
The structure of another prokaryotic 2-TM channel has been MECHANOSENSITIVE CHANNELS
solved; its inner helices are bent and splayed open, creating a
wide entryway. This second Kir channel responds to Ca2+ on Mechanosensitive channels are a diverse class of structur-
its intracellular side by increasing its open probability. The ally unrelated channels that subserve many different func-
Ca2+ binds to the regulator of K conductance (RCK) domain tions in different cells. Mechanosensation is important for
in the C-terminal part of the protein, not shown in Figure touch and hearing and also for sensing blood pressure and
3–2, inducing a conformational change that splays the inter- for proprioception, providing information about position,
CHAPTER 3 Cell Membranes and Transport Processes 19

G
Y
P G
+
+
S1 S2 S3 S4 S5 S6
+
+

C
N
FIGURE 3–3 The topology of one monomer of voltage-dependent K channels (KV). The six transmembrane helices (S1–S6)
are characteristic of all voltage-dependent ion channels. (Modified with permission from Landowne D: Cell Physiology, New York: Lange Medical Books/
McGraw-Hill, 2006.)

orientation, velocity, and acceleration of the body and its is a signature feature of voltage-sensitive channels. It is
parts. The channels are associated with accessory molecules thought to be the voltage sensor that moves toward the extra-
and cellular structures that enhance their particular func- cellular surface when the membrane potential changes and
tions. Somatic nonsensory cells also respond to mechanical causes the conformational changes that lead to channel open-
stress without informing the nervous system—for instance, ing. There are nine subfamilies of KV channels and several
to compensate for osmotic swelling or modulate secretion more 6-TM-channel subfamilies, including the Ca-activated
or contraction. K channels, the hyperpolarization-activated channels im-
Many mechanosensitive channels are relatively nonselective portant for pacemaker activity in the heart, and cyclic nucle-
cation channels. Some are very large and permit electrolytes otide–gated channels. The last two families are nonselective
and small metabolites, but not proteins, to cross the mem- cation channels.
brane. The two structures that have been solved are bacterial. Voltage-sensitive Na channels (NaV) are responsible for
One is a homopentamer, with each subunit containing two the upstroke of the action potential and support its propaga-
TM helices. The other is a heptamer, with each subunit con- tion. Voltage-sensitive Ca2+ channels (CaV) couple mem-
taining three TM helices. These are beautiful structures, but brane potential changes with an increase in intracellular Ca2+
they do not shed much light on the many other forms of concentration, which acts as a second messenger to control a
mechanosensitive channels. variety of intracellular processes. NaV and CaV channels have
a structure similar to the KV channels except that they are
single larger molecules incorporating four domains, each
VOLTAGE-SENSITIVE CHANNELS with slightly different 6-TM segments (Figure 3–4). The se-
lectivity filters have four different walls. The CaV channel has
Voltage-sensitive K channels (KV) are responsible for the four characteristic glutamates (EEEE) in its pore lining, one
return to the resting state, which ends an action potential. KV on each domain. The NaV channel has a DEKA pattern on
has a core structure similar to that of Kir and an additional the four walls of its pore. These side chains must be exposed
four TM helices on each subunit (see Figure 3–3). The fourth to the lumen of the pore. The charges they expose to the
TM segment (S4) is distinguished because it has between lumen and the size of the pore determine the selectivity of
four and eight positively charged side chains (Arg or Lys). S4 the channel.

P P P P
+ + +
+ + +
S1 S2 S3 S4 S5 S6 S1 S2 S3 S4 S5 S6 S1 S2 S3 S4 S5 S6 S1 S2 S3 S4 S5 S6
+ + +
+ + +

C
N

FIGURE 3–4 The topology of voltage-dependent Na channels (NaV). Four slightly different domains are linked together in one protein.
(Modified with permission from Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)
20 SECTION II Cell Physiology

CHEMOSENSITIVE CHANNELS a large extracellular N-terminal region. At the neuromuscular


junction, the nAChR has two alpha subunits, with Ach-binding
There are many different chemosensitive or ligand-gated sites at the interface between subunits and far from the lipid
channels. These control the flow of ions and thus generate elec- membrane. ACh binding induces a conformational change
trical signals in response to specific chemicals, such as acetyl- that opens the pore formed at the level of the lipid membrane
choline (ACh), glutamate, or ATP. They can be grouped into and lined by the second TM segment of each of the monomer’s
three different superfamilies according to the stoichiometry five subunits. The open channels are highly permeable to both
and membrane topology of their subunits. Many of these were Na+ and K+, slightly permeable to Ca2+, and not permeable to
first discovered pharmacologically by noticing that certain anions. They are not as selective as the Kir or voltage-sensitive
compounds, called agonists, produced membrane currents or channels. Functionally, the Na permeability is most important,
altered the electrical activity of cells and other compounds; as discussed in Chapter 7.
antagonists could block these effects. For some agonist-induced The CNS postsynaptic receptors for glycine (glyR), gamma-
currents, the ligand binds the same molecule that contains the aminobutyric acid (GABAAR), and serotonin (5HT3R) have
pore. These are the ligand-gated channels, which are sometimes similar pentameric architecture, although some are homomer-
called ionotropic ligand receptors to distinguish them from ic, as are some nAChRs. glyRs and GABAARs are selectively
metabotropic ligand receptors, where the ligand binds a GPCR permeable to anions and produce inhibitory postsynaptic po-
and triggers a biochemical cascade that may include opening of tentials (IPSPs.) 5HT3Rs are cation-selective, similar to nACh-
other channels, for example, KACh, described above. Rs, and produce excitatory postsynaptic potentials (EPSPs).
The ACh receptor channels (AChRs) are referred to as The most common CNS EPSP channels are glutamate re-
nicotinic AChRs, or nAChRs. The term nicotinic indicates ceptors (gluR), which have an architecture (Figure 3–6) remi-
these receptor bind nicotine, which also opens the channels. niscent of an inverted Kir molecule with extra TM segments.
nAChRs are distinguished from muscarinic AChRs, which gluRs are heteromeric tetramers with three TMs per subunit.
are not channels but rather GPCRs. nAChRs are found on the They have a large extracellular region with four glutamate-
postsynaptic membranes at skeletal neuromuscular junctions binding sites and a cytoplasmic-facing P loop. Several func-
and in the autonomic and central nervous systems. tionally different gluRs are discussed in more detail in
The best-studied nAChRs are heteromeric pentamers Chapter 7. They are all cation-selective; some allow Ca2+ entry
(Figure 3–5). The monomers have four TM segments each and and others do not.

N
N

Out
Out

In
In

α β

γ α
δ

FIGURE 3–5 The topology of one monomer of nicotinic FIGURE 3–6 The topology of one monomer of glutamate
acetylcholine receptor channels (nAChR), with a top view showing receptor channels (gluR), with a top view showing the arrangement
the arrangement of the five monomers. (Modified with permission from of the four monomers. (Modified with permission from Landowne D: Cell
Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.) Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 3 Cell Membranes and Transport Processes 21

The ATP-sensitive channels or P2X receptors (P2XRs) CELL–CELL CHANNELS


have two TMs per subunit and three subunits per channel. “P”
refers to the purine sensitivity; adenine is a purine. P2 distin- In most tissues, there are channels that connect the cytoplasm
guishes them from P1 receptors, which are sensitive to of one cell to the cytoplasm of its neighbor. The exceptions are
adenosine and act through adenylyl cyclase (AC). The P1 re- free-floating cells in the blood and skeletal muscle cells.
ceptors are often referred to as A receptors (A for adenosine); These channels are mostly between cells of the same type, but
they are GPCRs. Caffeine is an antagonist of some of the A there are some cells of different type with junctions between
receptors. P2 receptors prefer ADP or ATP to adenosine. them. These channels were originally detected electrically by
P2XRs are channels and P2YRs are GPCRs. Purinergic recep- showing that current could pass from one cell to another
tors are best known as regulators of blood flow in tissues; they through an electrical synapse. Later they were associated with
have also been implicated in several sensory processes. an anatomic structure called the gap junction, named for its
Two additional channel families have chemosensitive mem- appearance in electron micrographs. Actually this gap is
bers but also have important members without known ligands. spanned by matching arrays of proteins from each cell, with up
These are the epithelial sodium channel (ENaC) and the to thousands of cell–cell channels per gap junction.
inositol triphosphate (IP3) receptor (IP3R) family. Each cell–cell channel is made of two hemichannels, one
ENaCs are important in the reabsorption of sodium from from each cell (Figure 3–7). They are also called connexons. A
the nascent urine in the tubules of the nephron. They are hemichannel is a homomeric or heteromeric hexamer of pro-
thought to be heteromeric tetramers each with two TM seg- teins called connexins. There are more than 15 different con-
ments; they are not voltage-dependent. It is known that they nexins with molecular weights between 25 and 50 kDa. They
are regulated by control of their insertion and removal from all have four TM segments and two extracellular loops and
the membrane, and there may be an unknown ligand for this their N- and C-terminals are in the cytoplasm. Some but not
channel. There are structurally related channels in inverte- all connexins can form hybrid channels joining different he-
brates that have known ligands. michannels on the two cells.
IP3Rs and the related ryanodine receptors (RyR) are found
in the membrane of the ER. When open, they permit the re-
lease of Ca2+ from the ER. IP3 is a second messenger produced
by the action of phospholipase C (PLC) on the membrane
lipid phosphotidylinositol, which has been previously phos-
phorylated to be PIP2. RyRs also control the release of calcium, Out
primarily in muscle, from the sarcoplasmic reticulum. Ryano-
dine refers to a toxin that partially opens these channels. RyRs
are opened by direct interaction with a modified CaV channel
In
in skeletal muscle and by intracellular Ca2+ in cardiac muscle.
The functions of IP3Rs and RyRs are discussed in further detail
in Chapters 9 and 10.
RyRs are homotetramers with an enormous 20-nm-diameter
cytoplasmic N-terminal region. The total molecular weight
for the tetramer is above 2,000 kDa, about 10 times larger than
N C
NaV or KV channels. IP3R channels are also homotetramers
about half the size of RyRs. It has been predicted that IP3Rs
have 6 TM segments per monomer and RyRs have 4–12.
In
WATER CHANNELS
Some cells require more permeability to water than is provided
by the lipid bilayer. Red blood cells, which must quickly change Out
shape to pass through narrow capillaries, and many epithelial
cells, most notably those in the kidney, have specialized water
channels or aquaporins (AQPs), which permit the passage of
water but exclude ions. The AQPs are found as tetramers with In
four functional pores, one in each subunit. The subunits have
FIGURE 3–7 The topology of connexin, a monomer
six TM segments and two regions similar to the P loop of KV of cell–cell channels, top view showing the arrangement of
channels. One of the loops faces the extracellular surface and six monomers in a hemichannel and side view showing two
the other faces the cytoplasm, and they meet in the middle of cell membranes with aligned hemichannels. (Modified with
the membrane. The functions of AQPs and ENaCs are dis- permission from Landowne D: Cell Physiology, New York: Lange Medical Books/
cussed toward the end of this chapter. McGraw-Hill, 2006.)
22 SECTION II Cell Physiology

The pore is much larger than the ion channels described


above. It is about 1.2 nm and is permeable to anions, cations, Out
2Ko 3Nao
and small metabolites as well as second messengers such as
ATP, cAMP, or IP3 but not proteins. Experimentally, the pore E2
3Na 2K
is permeable to molecules with molecular weights below
1 kDa. Cell–cell channels allow cells in a tissue to work in a In
coordinated manner.
If a cell is damaged, it can close its cell–cell channels leading P P
to its neighbors and thus prevent the loss of small molecules
from the whole tissue. This gating is controlled by intracellular
Ca2+, H+, or transjunctional voltage. Different connexons have P
relatively different sensitivity to these three changes. Gating
can also be induced by octanol and anesthetics such as halo-
thane. Out
In some situations unpaired connexons, or the related
pannexons, can open and allow small molecules to move from Occluded
the cytoplasm to the extracellular space. Pannexons have been
suggested to have a role in inflammation and the response to 3Na 2K
In
ischemia by allowing the release of ATP to signal to cells near
a site of tissue insult.
P

ADP ATP
PUMPS
Ions move across cell membranes via channels, pumps, and
Out
transporters. These are three fundamentally distinct mecha-
nisms and the student should be careful not to confuse them.
Pumps create and maintain ionic gradients, moving ions against E1
the gradient at the expense of ATP. Channels use these gradi-
3Na 2Ki 3Nai 2K
ents to produce the various electrical signals. Transporters use In
one or more gradients; the with-gradient movement of an ion
(often Na+) is coupled to the against-gradient movement of an- ATP ATP
other substance. Because they consume ATP, pumps are often
referred to as ATPases. Five pumps will be described in detail: FIGURE 3–8 The Na/K pump cycle. Operating in the clockwise
the Na/K pump, the Ca pump, and three types of proton pump. direction the pump moves three Na+ out and then moves two K+
in at the expense of converting one ATP to ADP. (Modified with
The first three of these are called P-type pumps, because they
permission from Landowne D: Cell Physiology, New York: Lange Medical Books/
are autophosphorylated during the reaction cycle, or E1–E2
McGraw-Hill, 2006.)
pumps, because they have two major conformational states.

Na/K PUMP Na+ and ATP bind to the E1 form of the alpha subunit, which is
then phosphorylated and converts to the E2 form (Figure 3–8,
The Na/K pump, often referred to more simply as the Na lower left, proceeding clockwise). The E2 form releases the Na+
pump, moves three Na+ ions out of the cell and two K+ ions into the extracellular space and binds extracellular K+. The crys-
into the cell in a cycle that converts one ATP molecule to tal structure of the E2-2K+–Pi form has recently been solved.
ADP + Pi. At maximum speed, the pump completes about The overall structure of domains and helices is similar to the Ca
100 cycles per second (cps), which means the movement of pump described below but there are differences related to the
ions per molecule is much less than a NaV channel, which may specific functions of each pump. The cycle continues when the
allow 1,000 ions/ms to flow into the cell. The NaV channels are phosphate is hydrolyzed off the protein; the protein changes
open only briefly when the cell is active; the pump runs con- back to the E1 form, releases the K+ inside the cell, and then
tinuously to recover from the activity. Pump activity increases binds the next Na load. As the Na+ and K+ alternately move
when intracellular Na+ or extracellular K+ increase and the through the membrane, the pump passes through an occluded
pump acts homeostatically to restore the original levels. state where the ions are not accessible to either solution.
The Na pump is a heterodimer with an alpha subunit that has Digitalis and ouabain, a related cardiac glycoside, stop the
the Na+, K+, and ATP-binding sites and a beta subunit thought action of the pump by binding extracellularly to the E2 form
to be important for membrane insertion. The beta subunit has near the K+-binding location. Digitalis is used to treat a variety
1 TM segment; the alpha subunit probably has 10. Intracellular of cardiac conditions. It is a relatively dangerous drug and must
CHAPTER 3 Cell Membranes and Transport Processes 23

be used cautiously so as to block only some of the pump mole- TABLE 3–1 Localization of membrane pumps.
cules and leave others functional. The danger is complicated
because extracellular K+ antagonizes the binding of digitalis by Pump Cell Type Membrane Inhibitor
driving the pump toward the E1 form; the prudent clinician will Na/K All Surface Digitalis
monitor blood potassium levels during digitalis treatment.
The Na pump is electrogenic, because each cycle moves one Ca All Surface and ER Thapsigargin
net charge out of the cell. This current has only a small effect on H/K Gut, kidney Surface Omeprazol
the membrane potential compared to ion flow through channels, F-type H All Mitochondria Oligomycin
which is discussed in the next chapter. The net movement of Na+
out of the cell prevents NaCl from accumulating in the cell. If the V-type H All Surface and Bafilomycin
vesicles
pump is blocked with cardiac glycosides, the cell will swell be-
cause of the osmotic influx of water following the NaCl.

The pump has 8 different subunits and more than 20 poly-


Ca PUMP peptide chains. The F0 portion spans the membrane and car-
ries the H ions; the F1 extends into the mitochondrial matrix.
There are two important Ca pumps, one that pumps Ca2+ from Part of the complex rotates about an axis perpendicular to the
the cytoplasm into the extracellular space and another, the plane of the membrane, similar to a turbine, as the H ions flow
SERCA pump, that pumps Ca2+ from the cytoplasm into through. Another portion, the stator, stays fixed in position,
the lumen of the sarcoplasmic or ER. They are thought to have and the interaction between the rotator and the stator produc-
similar mechanisms; both are P-type E1–E2 pumps that move es a sequence of conformational states that favor the synthesis
two Ca2+ ions out of the cytoplasm and two or three H+ ions of ATP. In the presence of high ATP, low ADP, and no proton
into the cytoplasm for each ATP consumed. gradient, the process can be reversed to pump H+.
The SERCA pump structure has been solved in several differ-
ent states. It is a tall molecule, about 15-nm high and 8-nm thick,
mostly extending out of the membrane on the cytoplasmic side. V-TYPE H PUMPS
There are 10 TM segments. The cytoplasmic headpiece consists
of the actuator (A), nucleotide binding (N), and phosphorylation V-type H pumps are also protein complexes of up to 14 sub-
(P) domains. The three cytoplasmic domains are widely split in units with rotors and stators. They move protons into vacuoles
the E1 • 2Ca state but gather to form a compact headpiece in the and other intracellular organelles such as lysosomes, the Golgi
other states. This motion is transmitted to the membrane por- apparatus, and secretory vesicles. The H+ gradient produced
tion through helices 1–3, attached to the A domain, and 4 and 5, across synaptic vesicle membranes is used to drive the packag-
attached to the P domain, to allow the Ca2+ to be released on the ing of neurotransmitters (see Figures 7–3 to 7–5). These
noncytoplasmic side. The distance between the Ca2+-binding pumps are responsible for the H+ that is secreted by osteoclasts
sites and the phosphorylation site is greater than 5 nm. to dissolve bones and also for H+ secretion in the kidney and
epididymus (Table 3–1).

H/K PUMP
TRANSPORTERS
The H/K pump secretes acid into the stomach by pumping two
H+ ions out of the parietal cells of the gastric glands and two K+ Transporters move ions and other small molecules across the
ions into the cell while splitting one ATP molecule. Similar membrane and are not channels or pumps. Sometimes the
pumps also operate in epithelial cells in the intestine and kidney. word transporter is used in the general sense to include all
This is an E1–E2 P-type pump and has a beta subunit, similar to transport mechanisms and secondary transporter is used to
the Na/K pump. The H/K pump is inhibited by omeprazole distinguish this group. Transporters undergo a conformational
(Prilosec) and other similar drugs used in the treatment of fre- change as they transport; in this aspect they are similar to
quent heartburn. pumps and different from an open channel. Unlike a pump,
they do not consume ATP. Most transporters are thought to
have 12 TM segments in two groups of 6 with a larger cyto-
F-TYPE H PUMPS plasmic loop between them. Some have a 2-fold pseudosym-
metry and P loops facing both surfaces. There are three gen-
The most significant F-type H pump usually runs in reverse as eral categories of transporters: uniporters, symporters or
the F0–F1 ATP synthase found in mitochondria. This protein cotransporters, and antiporters or exchangers (Figure 3–9).
complex allows protons to flow with their electrochemical The glucose transporter (GLUT) is a uniporter that fa-
gradient and converts the flow of 10 protons to form three cilitates the diffusion of glucose with its concentration gra-
ATPs from ADP. The hydrogen gradients are produced by dient into many cells that are consuming glucose. It also
oxidative metabolism in mitochondria. facilitates movement of glucose from cells that are releasing
24 SECTION II Cell Physiology

The Cl/HCO3 exchanger, also known as the anion


exchanger (AE), is important for moving CO2 from the tissues
to the lungs. CO2, produced by metabolism in the cells, is con-
verted to bicarbonate by carbonic anhydrase in the red blood
cells, and the HCO3– moves into the plasma exchanging for
chloride via AE. The process is reversed as the blood passes
through the lungs and the CO2 moves into the air to be ex-
haled. This process will be covered in Chapter 37.
Uniporter Symporter Antiporter
FIGURE 3–9 Three types of transporters. (Modified with
permission from Landowne D: Cell Physiology, New York: Lange Medical Books/
ABC TRANSPORTERS
McGraw-Hill, 2006.)
This mixed group of 12 TM transport proteins contains a
characteristic ATP-binding cassette (ABC) amino acid se-
quence and, in the absence of more specific information, is
glucose by breaking down glycogen and from basal surfaces
assumed to consume ATP while transporting some material
of epithelial cells that line the intestines and kidney tubules
across the membrane. Two ABC transporters deserve men-
(see Figure 3–14).
tion here, the multidrug resistance (MDR) transporter,
The Na–glucose cotransporter (SGLT) is a symport that
which is a pump, and the cystic fibrosis transmembrane
carries glucose into intestinal and kidney epithelial cells across
regulator (CFTR), which is a channel.
their apical surfaces against the glucose concentration gradi-
MDR1 extrudes hydrophobic drugs across the cell mem-
ent. The energy required for this transport comes from the
brane. It is thought to act somewhat like the flippase and ex-
movement of one or two sodium ions with their electrochemi-
trudes the drugs without much specificity. A wide variety of
cal gradient for each transported glucose molecule.
cells in the GI tract, liver, and kidney express MDR proteins.
A Na/glutamate cotransporter recovers glutamate that is
These can frustrate the physician who is attempting to provide
used as a neurotransmitter at CNS synapses (see Figure
drugs to treat cancer among these cells.
7–4). It couples the downhill movement of three Na+ ions
CFTR is a protein that, when mutated, leads to cystic fibrosis.
and one K+ ion to the uphill transport of one glutamate. The
The wild-type protein is a chloride channel that requires phos-
structure of a bacterial glutamate transporter, which is
phorylation by protein kinase A (PKA) and additional ATP
thought to be similar to that of humans, has recently been
hydrolysis by the activated CFTR protein in order to open.
solved. It has eight TM segments and two P loops, one fac-
The Cl− moves with its electrochemical gradient. Cystic fibrosis
ing the cytoplasm and one facing the outside. It is thought
occurs because of the lack of Cl− transport in the pancreatic
that relatively small movements of the protein can transfer
duct (hence cystic). The decreased Cl− leads to decreased wa-
the glutamate from one P loop to the other and thus across
ter and the protein-rich secretion thickens and can block the
the membrane.
ducts that then become fibrotic. Before the development of
There is an H/glutamate antiporter that uses the H+ gradi-
oral replacement therapy for the missing pancreatic enzymes,
ent, established by a V-type pump, across the membrane en-
many CF patients died of complications of malnutrition. Now
closing synaptic vesicles to concentrate glutamate inside the
the major problem is the thickening of mucus in the lungs be-
vesicle (see Figure 7–4).
cause of insufficient fluid secretion.
There are many other Na-driven cotransporters to move
other small molecules into cells and H-driven transporters to
move some material into vesicles. Some of these transporters
are targets for pharmacologic intervention. For example,
MEMBRANE RECEPTORS
fluoxetine (Prozac) acts on a Na/serotonin cotransporter. The word receptor comes from pharmacologic studies, where
Others are discussed further in Chapter 7. Some anions are it designates the site of action or the molecule that a small
cotransported with sodium; for example, the Na/I symporter molecule of interest, perhaps a hormone or neurotransmitter,
concentrates iodine into thyroid follicle cells. acts on. Here it is used in a more restrictive sense to mean
The Na/Ca exchanger (NCX) is an important regulator of molecules that span the membrane, are acted on the external
intracellular Ca2+ concentration. Three sodium ions moving surface by the small molecule, and trigger some action inside
with their electrochemical gradient into the cell can move one the cell when the small molecule is present. There are also
calcium ion out, or vice versa; all of the exchangers can run intracellular receptors, for example, the steroid hormone
either way depending on the relative gradients. The effect of receptor. Steroid hormones and related drugs can cross the
digitalis on cardiac muscle is to raise intracellular Na by inhib- lipid bilayer and bind these intracellular proteins. Chemosen-
iting the Na/K pump. Elevated Nai+ means that there is less sitive channels are excluded as well, although some pharma-
inward gradient for Na+ and therefore less Ca2+ efflux via NCX. cologists like to call them ionotropic receptors. There are two
This increases Cai2+ and produces a stronger contraction (see major categories of these membrane receptors: the GPCRs and
also Chapter 23). the enzyme-linked or catalytic receptors.
CHAPTER 3 Cell Membranes and Transport Processes 25

Agonist AC Effector

GPCR
β/γ P

G protein α
ATP cAMP

PKA

FIGURE 3–10 The Gαs signaling pathway. Binding of agonist to the G protein–coupled receptor causes the dissociation of the α subunit,
which causes adenylyl cyclase to raise cAMP levels. This, in turn, causes protein kinase A to phosphorylate an effector protein (in this case a
channel). (Modified with permission from Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)

G PROTEIN–COUPLED RECEPTORS phospholipid PI to produce IP3 and diacylglycerol (DAG). As


described above, IP3 binds the IP3R channels, which increases
GPCRs have seven TM segments with an extracellular N termi- Cai, which in turn triggers various reactions. Several exam-
nus. They are coupled to a trimeric GTP-binding protein com- ples of GPCR-initiated cascades are described more fully in
plex. When a hormone or neurotransmitter interacts with a later chapters.
GPCR, it induces a conformation in the receptor that activates The toxins that underlie two infectious diseases, cholera
a heterotrimeric G protein on the inner membrane surface of and pertussis, ADP-ribosylate Gα subunits leading to consti-
the cell (Figure 3–10). In the inactive heterotrimeric state, GDP tutive activation. In cholera, activated Gα in intestinal epithe-
is bound to the Gα subunit. On activation, GDP is released, lial tissue stimulates AC, cAMP levels increase, and CFTR
GTP binds to Gα, and subsequently Gα–GTP dissociates from chloride channels open, leading to a watery diarrhea. People
Gβγ and from the receptor. Both Gα–GTP and Gβγ are then with cystic fibrosis can be resistant to cholera because they
free to activate other membrane proteins. Most Gα and Gγ are have fewer functional chloride channels. The cellular patho-
lipidated; they have a covalently attached lipid anchor into the genesis of pertussis is not clear.
membrane bilayer. The duration of the G-protein signal is de-
termined by the intrinsic GTP hydrolysis rate of the Gα subunit ENZYME-LINKED RECEPTORS
and the subsequent reassociation of Gα–GDP with Gβγ.
There are more than 2,000 predicted GPCRs in the human Most enzyme-linked receptors are receptor tyrosine kinases
genome, more than 5% of all the genes. More than 800 are (RTK) and act by phosphorylating tyrosine side chains on
olfactory receptors; others detect almost all neurotransmit- other proteins, which may in turn phosphorylate other
ters and many hormones. Light also is detected by GPCRs in proteins. Some enzyme-linked receptors are not kinases
the eye (Figure 5–2). Different cells have different palettes of themselves but are coupled to an associated protein that phos-
GPCRs coupled to different G proteins controlling different phorylates other proteins. Some enzyme-linked receptors are
sets of intracellular reactions. guanylyl cyclases, tyrosine phosphatases, or serine kinases.
There are only about 16 different Gα subunits and even Most growth and differentiation factors act by binding spe-
fewer Gβγ. Three classes of Gα subunits initiate most of the cific RTKs.
subsequent events described in this book. Gαs stimulates ad- The insulin receptor is an RTK that phosphorylates a fam-
enylyl cyclase (AC), Gαi inhibits AC and its associated βγ ily of substrates known as insulin-receptor substrates; these
directly activates KAch channels, and Gαq stimulates a phos- stimulate changes in glucose, protein, and lipid metabolism
pholipase (PLCβ). AC produces cAMP that can directly in- and also trigger the Ras signaling pathway, activating tran-
fluence some channels. cAMP also activates protein kinase A scription factors that promote growth. Interferon receptors
(PKA) by dissociating the regulatory subunits from the cata- and the CD4 and CD8 molecules on the surface of T lympho-
lytic subunits. The PKA phosphorylates many proteins, thus cytes are examples of receptors that are coupled to cytoplas-
altering the activity of the cells. PLCβ splits the membrane mic tyrosine kinases.
26 SECTION II Cell Physiology

CELL ADHESION MOLECULES gradient, and active transport, which creates or maintains
these gradients.
Most cells except red blood cells have integral membrane pro-
teins that attach to the extracellular matrix or with adhesion PASSIVE TRANSPORT
molecules on neighboring cells. These molecules hold the
tissue together and can allow the transmission of mechanical Simple Diffusion
forces from one cell to another. They can act as signals during Some materials can move with their concentration gradient by
development, so one cell can recognize another. Many also act simple diffusion though the lipid bilayer. Small, uncharged
as receptors, informing the inside of the cell that they have molecules such as O2, CO2, NH3, NO, H2O, steroids, and lipo-
bound something. Some are controlled from the inside, bind- philic drugs can enter or leave cells by simple diffusion. The
ing only when some signal has been received. net flux of these compounds through the membrane is pro-
The integrins are examples of cell-matrix adhesion mole- portional to difference in their concentrations on the two
cules. They have a single TM segment and link cells to sides, or as expressed in the following equation:
fibronectin or laminin in the extracellular matrix.
Cadherins are Ca-dependent cell–cell adhesion molecules; J1→2 = –P(C2 – C1) = –PΔC (1)
they are glycoproteins with a single TM segment and are Using the centimeter–gram–second (CGS) system of units,
thought to bind homophilically (to another copy of the same J1→2 is the number of moles that move through a square centi-
molecule) to cadherins on the other cell. Cadherins have been meter of membrane from side 1 into side 2 each second and C1
found at many neuron–neuron synapses. There is a large fam- and C2 are the numbers of moles of the material per cubic cen-
ily of cell adhesion molecules, of which the N-CAMs are the timeter of solution on the two sides. P, the proportionality
best understood. N-CAMs are found on a variety of cell types constant, is called the permeability of the membrane to this
and most nerve cells. Like cadherins, N-CAMs have a single material in centimeters per second. The equation is written
TM segment and bind homophilically, but they differ in that with the leading minus sign as an aid to remembering that the
they do not require Ca2+ for binding. Intercellular adhesion flux is moving with the concentration gradient. This relation-
molecules (ICAMs) are a related class expressed on the surface ship is shown graphically in Figure 3–11.
of capillary endothelial cells that have been activated by an in- Equation (1) is Fick’s first law. It can be used to describe the
fection in the surrounding tissue. They bind heterophilically flux by simple uncharged substances through any membrane.
(to a different molecule) to integrins on white blood cells and For example, it is useful to describe the movement of oxygen
help them move to the site of infection. Selectins are carbohy- from the air into the alveoli of the lungs and into the blood,
drate-binding proteins on the endothelial cell membrane that across the cells of the alveolar epithelium and the capillary en-
recognize sugars on the surface of the white blood cell and dothelium. A charged species will also be influenced by the
form the initial binding, which is strengthened by the ICAMs. electrical potential difference across the membrane in a man-
ner to be discussed in the next chapter. If there is no potential
difference across the membrane, Fick’s law is also applicable to
TRANSPORT ACROSS charged substances.
CELL MEMBRANES Permeability describes a property of a particular mem-
brane in relation to a particular substance. The membrane is
From a functional point of view, discussion of the transport of considered permeable, while the substances are said to be
materials across cell membranes can be divided into passive permeant or to permeate. The permeability will be propor-
transport, where the materials move with their concentration tional to the ability of the substance to partition into the

Flux Flux

Vmax

Concentration Km Concentration
FIGURE 3–11 The concentration dependence of simple diffusion (left) and facilitated diffusion (right). (Reproduced with permission from
Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 3 Cell Membranes and Transport Processes 27

membrane and to diffuse within the membrane. The perme- ACTIVE TRANSPORT
ability will be inversely proportional to the thickness of the
membrane. It is usually not easy or necessary to know these Pumps provide primary active transport, moving materials
three factors separately, but one should appreciate that thick- against their electrochemical gradients at the expense of ATP.
ening of the complex membrane between the alveolus and the The Na/K pump moves Na+ out of the cell and K+ into the cell;
pulmonary capillary blood will reduce the movement of oxy- both ions are moving against their respective gradients. The
gen into blood. SERCA pump moves Ca2+ against its gradient from the cyto-
It is sometimes convenient to think of Fick’s law as saying that plasm into the lumen of the ER.
the net influx of a substance is equal to the unidirectional in- Cotransporters and exchangers can provide secondary
flux (PC0) minus the unidirectional efflux (PCi). active transport, using a gradient produced by primary active
The permeability is a measure of the ease with which a sol- transport to move another material against its gradient. Many
ute crosses through a membrane. Plain lipid bilayers are rela- transporters couple the movement of Na+ or H+ with its elec-
tively permeable to small, uncharged molecules; the perme- trochemical gradient with the movement of another substance
ability to water is about 10−3 cm/s. Thus, water equilibrates against its gradient. The Na-GLUT and the H/glutamate anti-
across a cell membrane in a few seconds. Urea is moderately porter are two examples of secondary transport mechanisms.
permeable (P = 10−6 cm/s), and its equilibration time is a few The flux by pumps and transporters can be described by
minutes. Hydrophilic small organic molecules such as equations similar to equation (2), modified to include the affin-
glucose or uncharged amino acids are less permeable, with ity for each substance and for ATP. When more than one ion at
P = 10−7 and equilibration times of hours; ions are essentially a time is involved in the reactions at one pump or transporter
impermeable, with P = 10−12 cm/s and equilibration times of molecule, the equation must also be modified to reflect this co-
many years. operativity. Thus, the efflux of sodium through the Na/K pump
has a sigmoidal relationship to the internal Na+ concentration.
Facilitated Diffusion
Many substances, such as glucose or urea, easily enter cells in
spite of the fact that the lipid bilayer is relatively impermeable OSMOSIS
to them. The flux of these materials is described by Fick’s law
Life is intimately associated with the movement of water. Our
only for low concentrations. At higher concentrations, the flux
bodies are mostly water and are vitally dependent on its sup-
saturates at a maximum value (see Figure 3–11). This behavior
ply. Water is a small but abundant molecule. It is not much
can be described by the Michaelis–Menten equation, which is
larger than an oxygen atom, about 0.2 nm across—small
also used to describe enzyme kinetics. The unidirectional flux
enough to intercalate between other molecules, even in some
is given by the following equation:
crystals. A mole of water is 18 mL; thus, pure water is 55 mol/L.
J C
J = _____
max
(2) This concentration is several hundred-fold higher than the
C+K m Na+, K+, or Cl− concentrations in the body that are the next
where Jmax is the maximum flux and Km the affinity or the con- highest. More than 99% of the molecules in the body are H2O.
centration at which the flux is half its maximum value. This Because the molecules are small, they move easily; because
saturable property of the flux suggests that there are a fixed they are so abundant, their movements are important to our
number of sites at which the flux can take place. Also, as in the health. There are two or three distinct mechanisms for water
case of enzymes, it may be possible to demonstrate the compe- movement: bulk flow, molecular diffusion, and, perhaps, mo-
tition of different substances for the same site or noncompeti- lecular pumping. When you pull the plug in a bathtub or your
tive inhibition of the transport sites. The sites are selective for heart beats, there is bulk flow of fluid in response to an exter-
a particular substance or group of substances that they will nal mechanical force—a push or a pull. The driving force for
transport or that allow competition for transport. Selectivity, bulk flow is the mechanical pressure commonly produced by
affinity, and Jmax are three independent qualities of the sites; pushing or by gravity.
they will be found with different values in different systems. Molecular diffusion or osmosis is a passive process by which
Facilitated diffusion is now understood in terms of channels water diffuses from areas of high water concentration to those
or transporters. of low. There is a high water concentration where there is low
Most channels have low affinity or high Km values; they are solute concentration, and vice versa. Water can diffuse across
not saturated under normal physiological conditions. Three most cell membranes directly through the lipid bilayer or by
glucose uniporters, GLUT1, GLUT3, and GLUT4 (which is traveling through AQPs. Many cells produce AQPs, because
regulated by insulin), are found in many tissues and have simple diffusion does not permit adequate water flux. Some
a high affinity for glucose; they are saturated at all physiologi- kidney cells insert AQPs in response to antidiuretic hormone
cal concentrations. GLUT2, which is found in tissues carrying (ADH), so as to increase water flow from the forming urine
large glucose fluxes (such as intestine, kidney, and liver), has a back into the blood, thereby conserving water. This passive
low affinity for glucose, and the influx through GLUT2 trans- type of water movement is called osmotic flow, and the associ-
porters increases as the glucose concentration increases. ated driving force is the concentration gradient of the water.
28 SECTION II Cell Physiology

Water may also be transported across membranes at the ex- the terms used interchangeably. Also, the actual pressures are
pense of energy by the Na–glucose cotransporter (SGLT). The rarely discussed; rather, the osmoles are mentioned directly.
TM transport of two Na+ ions and one sugar molecule has Tonicity is a concept that is related to osmolarity but is a
been associated with the influx of 210 water molecules, inde- special case for cells. A solution is said to be isotonic if it
pendent of the osmotic gradient. The energy could come from causes neither shrinking nor swelling of cells. A 150-mM NaCl
allowing Na+ to move with its concentration gradient. This solution (9 g/L or 0.9%) is isotonic for mammalian cells and
molecular pumping would be a secondary active transport also isosmotic to the cell contents. A 300-mM urea solution is
mechanism and might account for almost half the daily uptake also isosmotic to the cell contents, but a cell placed in this so-
of water from the small intestine. lution will swell and eventually lyse or burst (Figure 3–12).
Osmotic pressure is the mechanical pressure needed to The urea solution is hypotonic; it has insufficient tonicity to
produce a flow of water equal and opposite to the osmotic flow keep the cell from swelling. It differs from the NaCl solution
produced by a water concentration gradient. In animal cells, because the urea can cross the cell membrane. The addition of
this pressure does not develop across the cell membrane be- a permeable material to a solution increases its osmolarity but
cause the cells will change their volume in response to osmotic not its tonicity. The addition of more impermeable solutes
flow. The concept of osmotic pressure is similar to (and his- makes a hypertonic solution; a 300-mM NaCl solution is hy-
torically preceded) the Nernst equilibrium potential, an elec- pertonic and will cause cells to shrink.
trical potential that produces a flow of ions equal and opposite If a moderately permeable solute is added to an isotonic solu-
to a flow produced by a concentration gradient. The Nernst tion (e.g., 300-mM urea + 150-mM NaCl), the cells will tran-
potential is discussed in Chapter 4. siently shrink and then return to their original volume
If two different solutions are in contact, the osmotic pres- (Figure 3–13). The rate at which they shrink is proportional to
sure, π, between them is: the water permeability of the membrane; the rate at which the
volume recovers is proportional to the urea permeability. If the
π = RTΔc (3)
original 150-mM NaCl solution is replaced, the opposite effects
where R is the molar gas constant (Avogadro’s number times will occur. The cells will swell as water rushes in and then return
Boltzmann’s constant), T the absolute temperature, and Δc the to their original volume as the urea (and water) leaves the cell.
concentration difference of all of the impermeable solutes. The In some cases, it is convenient to consider a reflection coef-
concentration difference refers to the summated molar con- ficient as a description of the permeability of solutes. Water
centration of all the particles created when the solute is dis- movement across capillary walls depends on the mechanical
solved in water. It is measured as the osmolarity, that is, the or hydrostatic pressure difference and on the difference in col-
sum of the moles of each component of the solution. A 2-mM loid osmotic pressure due to differences in protein concentra-
solution of MgCl2 contains 6 milliosmoles (mOsm) per liter tion in the plasma and the interstitial fluid. If the capillary wall
of solution, 2 for the Mg2+ and 2 for each Cl−. The osmolarity is completely impermeable to the proteins, it is said to have a
of this solution is 6 mOsm. A 3-mM NaCl solution and a reflection coefficient of 1.0. If the walls become leaky, the re-
6-mM urea solution have the same osmolarity because they flection coefficient decreases, proteins enter the interstitial
have the same number of particles per liter of solution. They space, and water follows.
are said to be isosmotic. Water movement in the whole body is concerned with two
The osmolality of a solution can be measured by the change compartments, intracellular and extracellular. The extracellu-
it produces in the freezing point or vapor pressure. Osmolality lar compartment has two subcompartments: the plasma fluid
refers to moles of solute per kilogram of solvent, whereas os- in the blood vessels and the interstitial fluid that bathes the
molarity refers to moles of solute per liter of solution. Because rest of the cells. The plasma and the interstitial fluid are sepa-
1 L of any body fluid contains very close to 1 kg of water, rated by the capillary walls, which are freely permeable to all
the distinction is moot in clinical situations, and you may hear the small molecules and ions but normally prevent the plasma

150 mM 300 mM 150 mM 300 mM


NaCl NaCl NaCl urea
Cell volume

Time
FIGURE 3–12 Cells shrink in hypertonic solutions and swell in hypotonic solutions. (Modified with permission from Landowne D: Cell Physiology,
New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 3 Cell Membranes and Transport Processes 29

150 mM 150 mM NaCl + 150 mM


NaCl 300 mM urea NaCl

Cell volume

Time
FIGURE 3–13 The addition of urea causes transient shrinking but does not change the steady-state tonicity. (Modified with permission
from Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)

proteins from entering the interstitial fluid. The proteins have 13, so the new osmolarity is 13/41 = 317 mosm. The extracel-
an overall net negative charge at blood pH. The equilibrium lular compartment will have 5.8 osm, so its new volume will be
that arises with impermeable proteins and freely permeable 5.8/0.317 = 18.3 L. The new intracellular volume will be
ions is called the Gibbs–Donnan equilibrium. This effect 7.2/0.317 = 22.7 L. Notice that water moved out of the cells to
produces small ion concentration gradients (<3%) and a small dilute the seawater.
potential (a few millivolts, lumen-negative) across the capil-
lary walls. For most clinical purposes, these can be ignored
and the ionic concentrations in the plasma, which are easily TRANSPORT ACROSS
measured, can be considered to represent the extracellular EPITHELIAL CELLS
fluid in general.
The plasma proteins are osmotically important; they tend to Many epithelial cell layers functionally separate two solutions
keep water in the blood vessels. The balance between the hy- with different compositions and act in a coordinated way to
drostatic pressure and this “colloid” osmotic pressure is selectively transport solutes and water across the layer. This is
described by the Starling hypothesis discussed in detail in achieved by having tight junctions between the sides of the
Chapter 26. It regulates the flow of water across capillary en- epithelial cells so that the sheet of cells is impermeable to sub-
dothelia. Edema represents the loss of this balance. stances that cannot pass through the cell membranes and by
Intracellular proteins promote the entry of water into cells incorporating selective pumps and channels appropriately on
that is counteracted by the extrusion of sodium ions by Na/K the two surfaces of the sheet. The two sides may be called by
pumps. Cell membranes are not freely permeable to ions and different names in different epithelia. The apical membrane
the intracellular compartment is not in Gibbs–Donnan equi- faces the lumen or outside of the body; it can be known as the
librium with the extracellular space. luminal or mucosal membrane or the brush border after the
Short-term changes in the volumes and osmolarity of body appearance of its microvilli. The basolateral membrane that
compartments can be calculated using the following four faces the inside of the body can be known as the serosal or
principles: in every compartment, the volume times the peritubular membrane.
osmolarity is the total number of osmoles. Water will move Figure 3–14 shows pathways for Na+ and glucose trans-
between the compartments to make the osmolarity equal in port across epithelial cell layers. Na/K pumps in the basolat-
all compartments. The total amount of water and the total eral membrane keep the intracellular Na+ low by moving it
number of osmoles is the sum of the amounts in the compart- into the extracellular fluid. Na+ can enter the cell by moving
ments. Any added impermeant osmotically active substances with its concentration gradient through ENaC channels on
will remain extracellular; added water will distribute among the apical membrane and leave via the pump on the other
the compartments according to the first three principles. side. Glucose may be brought into the cell through the api-
These calculations do not include the effects of the renal sys- cal membrane, against its concentration gradient by the
tem (Chapter 45) that will act to restore the original volumes SGLT, and then move with its concentration gradient
and osmolarities. through the glucose uniporter (GLUT) on the basolateral
For example, consider a 70-kg medical student with 16 L of surface.
extracellular fluid and 24 L of intracellular fluid for a total of When solutes are moved across epithelial membranes, water
40 L. If the osmolarity of these compartments is 300 mosm, may flow osmotically, “following” the solute. This effect is im-
the extracellular compartment contains 16 × 0.3 = 4.8 osm and portant for rehydration therapy to combat the water loss of
the intracellular compartment contains 7.2 osm, for a total of diarrhea. Adding glucose and salt to the drinking water will
12 osm. If this student should happen to swallow 1 L of seawa- stimulate SGLT to move Na+, glucose, and water into the cell.
ter containing 1 osm of salts, mostly NaCl, the total water in- The Na/K pump and GLUT transporter will then move the
creases to 41 L and the total number of osmoles increases to solutes into the body and the water will follow.
30 SECTION II Cell Physiology

Tight junction

ENaC

Na Na/K pump

Na
Lumenal K
apical
mucosal Basal
ATP
serosal
Na ADP
Glucose

SGLT

Glucose
GLUT

FIGURE 3–14 Sodium and glucose are transported through epithelial cell layers by a combination of pumps, channels, and
transporters. (Modified with permission from Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)

Water and water-soluble material may move across epithelia


by transcytosis or by receptor-mediated endocytosis. The Hyponatremia, defined by a blood sodium concentra-
substance is taken up into vesicles by endocytosis on one sur- tion less than 135 mmol/L, may lead to hypotonic enceph-
face and either released unchanged by exocytosis on the other alopathy with fatal cerebral edema. Of 488 runners in the
surface or broken down in endosomes and the products 2002 Boston Marathon providing a usable blood sample at
released by transporters. Transcytosis occurs across capillary the finish line, 13% were hyponatremic and 0.6% had crit-
endothelia; receptor-mediated endocytosis is important in the ical hyponatremia (120 mmol/L or less). The study con-
kidney and liver. cluded that hyponatremia occurs in a substantial fraction
of nonelite marathon runners and can be severe. It is usu-
ally caused by drinking excessive amounts of fluid that ex-
ceed the kidney’s capacity to excrete water during exercise.
Considerable weight gain during the race, a long racing
CLINICAL CORRELATION time, and body mass index extremes were associated with
hyponatremia, whereas female sex, composition of fluids
As she passed her coach and friends at the 15-mile mark ingested, and use of nonsteroidal anti-inflammatory
of the Boston Marathon, the 28-year-old runner smiled drugs were not. Mild cases can be managed by restricting
and waved cheerily. She looked good as she passed fluids until the onset of urination. Manifestations of hy-
Heartbreak Hill, about 6 miles from the finish. Two ponatremic encephalopathy indicate the need for emer-
miles later, she stopped to drink a cup of fluid. Another gent treatment with hypertonic solutions such as 3% saline
runner remembers her saying she felt dizzy and disori- (513 mmol/L).
ented. She began to falter and told a friend running next Na+ and Cl− account for most of the osmotic strength
to her that she felt rubber-legged, and then tumbled to of the serum. If Na+ levels are low, the serum will be hy-
the pavement. potonic and water will move into all the cells of the body
When she reached the hospital, she was unresponsive causing them to swell (edema). This can have serious ef-
with stable vital signs. After endotracheal intubation, a fects in the brain because it is in the closed space of the
blood serum sodium value was found to be very low at cranium and the swollen tissue will restrict blood flow
113 mmol/L. Computed tomography of the brain and and therefore oxygen supply. Swelling may cause hernia-
chest radiography showed diffuse cerebral and pulmo- tion of the brain through the tentorium and foramen
nary edema. She was given intravenous isotonic saline magnum, compressing the brainstem and causing respi-
(150 mmol/L) but never regained consciousness. Diffuse ratory arrest. Blood sodium level is influenced by salt
cerebral edema was found at postmortem examination. A and water intake, sweating, and urinary secretion and is
few days later, the newspapers reported she died from a regulated by the endocrine system. The recommenda-
condition called hyponatremic encephalopathy. tion for marathon runners is to drink only when thirsty.
CHAPTER 3 Cell Membranes and Transport Processes 31

CHAPTER SUMMARY 2. In an intestinal epithelial cell, glucose transport from the


intestinal lumen to the blood involves which of the following
■ A lipid bilayer surface membrane with embedded proteins processes?
separates and connects cells with the surrounding extracellular
A) secondary active transport
environment.
B) facilitated diffusion
■ Cell membrane lipid molecules are amphipathic, with C) active transport
hydrophobic groups facing the interior of the membrane and D) secondary active transport and facilitated diffusion
hydrophilic groups facing both aqueous interfaces. E) Active transport and secondary active transport
■ Cell membrane proteins carry out specific functions by acting 3. A solution is prepared by adding 10 g of NaCl (formula
as channels, pumps, transporters, receptors, or cell adhesion weight = 58.5) to 1 L of distilled water. An isotonic solution
molecules. is 300 mOsm. The prepared solution is
■ Cell membrane proteins are amphipathic, generally with one or A) very hypotonic (with less than 50% normal tonicity).
more hydrophobic TM helices. B) slightly hypotonic (about 10% low).
■ Ion channels are membrane proteins with a pore that selects for C) isotonic (within 1%).
the type of ion(s) that passes through the channel with their D) slightly hypertonic (about 10% high).
electrochemical gradient. E) very hypertonic (more than twice normal tonicity).
■ Mechanosensitive channels are generally cation-selective and 4. Drinking isotonic saline solution will decrease
have diverse structures. A) extracellular volume.
■ Voltage-sensitive channels have a 4-fold symmetry. Each of B) extracellular osmolarity.
the parts has six transmembrane helices, one of which carries C) intracellular volume.
multiple positively charged amino acids. D) intracellular osmolarity.
■ There are many different families of chemosensitive or E) none of the above.
ligand-gated channels corresponding to the different chemical
ligands.
■ Cell–cell channels connect the interior of one cell to the
interior of an adjacent cell by an aqueous path that permits
the passage of ions and other small molecules.
Rate of transport

■ Pumps move ions or other molecules against their gradients


at the expense of ATP.
■ Transporters move some ions or other molecules against their
gradients at the expense of having other ions move with their
gradients.
■ GPCRs initiate intracellular G-protein cascades under the
control of extracellular activators.
■ Some materials can simply diffuse with their concentration
gradients through the membrane lipids.
■ Many substances have specific facilitated diffusion mechanisms Concentration gradient
characterized by an affinity and a maximum transport velocity.
■ Osmotic pressure is proportional to the osmolarity or the total 5. The above diagram is typical for the concentration gradient
concentration of all solutes. dependence of
■ Tonicity describes a solution’s ability to prevent the shrinking A) the rate of secondary active transport.
or swelling of cells. B) the rate of primary active transport.
■ Substances may be transported through epithelial cell layers by C) the rate of transport by passive diffusion.
combinations of pumps and transporters arranged on opposite D) the rate of transport by facilitated diffusion.
sides of the cells. 6. Which one of the following is least likely to affect Na/K pump
activity?
A) cardiac glycosides
STUDY QUESTIONS B) second messengers (e.g., cAMP and diacylglycerol)
1. Cell membranes C) intracellular Na+ concentration
D) extracellular Mg2+ concentration
A) consist almost entirely of protein molecules.
E) extracellular K+ concentration
B) are impermeable to fat-soluble substances.
C) contain amphipathic phospholipid molecules.
D) are freely permeable to electrolytes but not to proteins.
E) have a stable composition throughout the life of the cell.
This page intentionally left blank
4
C H A P T E R

Channels and the Control


of Membrane Potential
David Landowne

O B J E C T I V E S

■ Describe how membrane potentials are measured and provide typical values
for different cells.
■ Discuss the relationship between the separation of charge across the
membrane and the membrane potential.
■ List the approximate concentrations of the major ions in the intracellular and
extracellular compartments.
■ Describe the three factors that control the movement of ions through
membranes.
■ Determine whether an ion will move into or out of cells given the membrane
potential and the concentration gradient of the ion.
■ Discuss how the membrane potential changes when ions flow across cell
membranes.
■ Explain the steps that occur during the generation of a Nernst potential.
■ Explain the steps that occur during the generation of a resting membrane
potential.
■ Discuss why the net flux of charge is 0 in the resting state even though ions are
moving through the membrane.
■ Discuss the role of the Na/K pump in the generation of the membrane potential.
■ Define single-channel recording and describe currents through single K channels.
■ Describe the two types of the spread of electrical information in nerve and
muscle cells.
■ Discuss why the cell membrane acts as a capacitor and what properties this
confers on nerve and muscle cells.
■ Discuss the difference between length (space) and time constants and the
relationship of these constants to nerve conduction.
■ Explain the steady-state and transient cable properties of nerve and muscle cells.

INTRODUCTION 10–100 mV. Concentration gradients of ions across the mem-


brane are the immediate supplier of the energy to create and
All living cells have an electrical potential difference across maintain the resting potential. The resting potential is neces-
their surface membranes. Cells act as miniature batteries; the sary for electrical excitability of nerve and muscle cells, sensory
battery cell is named after the biological cell. At rest, the inside reception, CNS computation, and to help regulate transfer of
of cells is negative to the outside by about 0.01–0.1 V or ions across the membrane.

33

Ch04_033-042.indd 33 11/26/10 9:41:01 AM


34 SECTION II Cell Physiology

0 mV
Microelectrode
−90 mV
In Out In Out
FIGURE 4–1 Membrane potentials are
Oscilloscope
measured with microelectrodes filled with
electrolyte solutions. (Modified with permission
from Landowne D: Cell Physiology. New York: Lange
Medical Books/McGraw-Hill, 2006.) Muscle

MEASURING MEMBRANE 150 mmol/L of cations and anions (Table 4–1) with exactly
balanced positive and negative charges except for the layer
POTENTIALS within about 1 nm from the surface of the membrane. The
Figure 4–1 shows how resting potentials are measured. A mus- bulk solutions on both sides are electrically neutral.
cle is secured to the bottom of a dish that is filled with an iso- The excess charges of opposite sign experience an attractive
tonic salt solution with an ionic composition similar to that of force for each other but are prevented from reaching each oth-
blood. A microelectrode with a fine tip pulled out of glass and er because they cannot easily leave the aqueous solutions and
filled with 3-M KCl is positioned over one of the muscle cells. enter the oily lipid membrane. Any charge within the mem-
A chlorided silver wire in the microelectrode is attached to brane also experiences this force, tending to pull positive
one terminal of a voltage-measuring device that displays a charges inward and push negative charges outward. The volt-
trace of voltage versus time. The other terminal is attached to age across the membrane is the electrical measurement of this
another chlorided silver wire placed in the dish; this is called electromotive force or this potential for movement of charges
the ground wire. When the microelectrode is in the solution, it if they happen to be within the membrane.
is at the same potential as the ground wire and the oscilloscope The voltage is directly proportional to the amount of charge
reads 0 mV. When the microelectrode is advanced a few mi- (Q) that is separated. The ratio of separated charge to the volt-
crometers into the muscle cell, the trace on the oscilloscope age is called the membrane capacitance:
abruptly jumps to about −90 mV and stays there as long as the C = __
Q
(1)
microelectrode remains in place. When the electrode is with- V
drawn, the trace returns to 0 mV. The experiment can be re- Electrical charge is measured in terms of coulomb (C); there
peated. If a second microelectrode is inserted, it measures the are 96,484 C/mol of charge (this is Faraday’s constant). The
same potential, showing that the electrodes are not somehow unit of capacitance is the farad (F); 1 C/1 V is 1 F. Capacitance
creating the potential. is the ability to store separated charges. Many small computers
When the microelectrode is inside the cell, the KCl is in use a capacitor to store enough charge to allow some minimal
contact with the cytoplasm that is in contact with the mem- function to remain for a short time while the battery is being
brane. The ground wire is in contact with the external solu-
tion, which is in contact with the outside of the membrane.
The potential difference is across the membrane; it is called the
membrane potential. The particular membrane potential
measured when the cell is at rest—that is, not active—is also
called the resting potential. Different cells have different
resting potentials. Skeletal and cardiac muscle cells have a rest-
ing potential of about −90 mV. Sensory and motor neurons
have a resting potential of about −70 mV; smooth muscle cells,
about −60 mV; and red blood cells, about −10 mV.

SEPARATION OF CHARGE
The resting membrane potential is a reflection of the separa-
tion of charges across the membrane. There are a few excess FIGURE 4–2 The separation of charge. Left: A single layer of
negative charges (about 1 pmol/cm2) on the inner surface and charges separated by the membrane. Right: Adding a representation
the same number of excess positive charges on the outer sur- of the mobile charges in the bulk solutions. (Modified with permission from
face (Figure 4–2). The solutions on the two sides contain about Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 4 Channels and the Control of Membrane Potential 35

TABLE 4–1 Concentrations of some ions of ent muscles for Na+, K+, and Cl− values. External Ca2+ is nor-
importance across a muscle cell membrane.a mally about 2.5 mM, but internal Ca2+ can change dramati-
cally with activity, increasing above 1 μM when the muscle is
Extracellular Intracellular contracting.
Ion concentration (mM) concentration Eion (mV) The cell membrane is permeable to all of the ions listed in
Cations Table 4–1 except A−; they can move through the membrane via
various channels. The active transport of Na+, K+, and Ca2+
Na+ 145 12 mM +65
maintains the gradients. If the cell membrane is damaged or
K +
4 155 mM −95 there is not an adequate supply of ATP, the cell will gain Na+
Ca 2+
2.5 100 nM +132 and Ca2+ and lose K+. The membrane is not very permeable to
ions; compared with water, their permeability is insignificant.
Anions
However, it is the control of this ionic permeability that regu-
Cl− 132 4 mM −90 lates the membrane potential and small (by chemical standards)
A− ~0 155 mM
movements of ions that change the membrane potential.

HCO3− 22 8 mM −26
a −
A represents impermeant anions inside the cell. Many are polyvalent; all together,
they contribute less than 155 mOsm to the osmotic pressure. There are also other
FACTORS THAT CONTROL
uncharged osmolytes in the cell.
ION MOVEMENTS
The movement of ions through the membrane is proportional
changed. The membrane stores the opposite charges by keep- to the net driving force on them. The net driving force is the
ing them separated. electrochemical gradient or the difference between the driv-
The membrane capacitance is about 1 μF/cm2. One pico- ing force due to the concentration gradient and the force due
mole of univalent ions carries about 100 nC of charge. Putting to the voltage gradient or membrane potential. Movement of
these values into equation (1) means separating 1 pmol/cm2 of charged particles is an electrical current. The current, I, car-
univalent ions will produce a 100-mV membrane potential. ried by a particular ion, x, is related to the driving force by the
following expression:
Ix = gx (V − Ex) (2)
GENERATION OF THE where Ex is the chemical driving force for ion x expressed as an
electrical potential; this is described more fully below. V is the
RESTING POTENTIAL membrane potential and (V − Ex) is the driving force on ion x.
The membrane separates two solutions with quite different The membrane conductance for ion x is gx. The overall mem-
ionic compositions. The extracellular concentrations are brane conductance for ion x is proportional to the number of
thought to represent the concentration of these ions in seawa- channels for that ion, N; the probability that a channel is open,
ter at the time the ancient ancestor left the sea. They are about Po; and the conductance of a single open channel, γ; or:
one third the concentration in seawater today. The generation
gx = NPoγ (3)
of the resting potential and all of the changes in potential (such
as the action potential and the synaptic potentials) depend on The conductance is proportional to the permeability of the
the concentration gradients of ions across the cell membrane. membrane (the ease with which ions move through it). The
Table 4–1 presents some typical values for a skeletal muscle. conductance is also proportional to the concentration of the
Both sides are electrically neutral; the sums of positive and conducting ion(s). In the absence of sodium ions, a sodium
negative charges are equal. The external solution has relatively channel may be permeable (if it is open), but it will not con-
high Na+ and Cl− concentrations and modest K+ and Ca2+ con- duct any current.
centrations, whereas the internal solution is high in K+ and The voltage gradient pushes or pulls an ion because the ion
low in Na+ and Cl− and very low in Ca2+; it has a high concen- is charged. The concentration gradient is a conjugate force;
tration of other anions (A−), such as phosphate groups on pro- ions tend to move from a high concentration to a low concen-
teins or nucleic acids and negatively charged amino acids on tration. More ions will hit an open channel from the side with
proteins. higher concentration than the side with lower concentration,
There is an inward concentration gradient for Na+ and Cl− so there will be a flow with the concentration gradient in pro-
and an outward concentration gradient for K+. The sodium portion to the gradient.
gradient is about 10-fold; Cl−, about 30-fold; and K+, about To determine the net flux of an ion through the membrane
40-fold. Table 4–1 indicates a 25,000-fold inward concentra- it is necessary to know the concentration gradient, the voltage
tion gradient for Ca2+. Exact numbers are given in this table to gradient (the membrane potential), and the conductance for
facilitate the calculation of examples later in this chapter. There the ion. Unless all three factors are known, it is not possible to
is a normal variation of about 10% in different people or differ- predict the flux of the ion. The two forces on the ion from the
36 SECTION II Cell Physiology

Cl− Na+ K+
C C C
V V V

Cl− Na+ K+
−90 mV −90 mV −90 mV

60 mV 132 60 mV 145 60 mV 4
ECI = log ENa = log EK = log
−1 4 +1 12 +1 155

ECI = −90 mV ENa = +65 mV EK = −95 mV

FIGURE 4–3 The driving force on ions crossing through the membrane, voltage gradients (V), and concentration gradients (C) for
the three most common ions in the solutions in the intracellular and extracellular fluids. (Modified with permission from Landowne D: Cell Physiology.
New York: Lange Medical Books/McGraw-Hill, 2006.)

voltage and concentration gradients may act in the same direc- For chloride, the concentration gradient is inward; Cl− would
tion or in opposite directions. like to move into the cell because there is a higher concentra-
tion outside. The –90-mV resting potential exerts an outward
force on the negatively charged chloride ions. These two are
THE NERNST EQUILIBRIUM equal and opposite, that is, (V – ECl) = –90 – (–90) = 0 mV, and
chloride ions are in electrochemical equilibrium.
POTENTIAL For sodium, the concentration gradient is also inward, but
For any particular concentration gradient, it is possible to pick the negative membrane potential exerts an inward force on the
a voltage gradient that is equal and opposite, so that the term positively charged Na+. Both forces are inward and sodium
in parentheses in Eq. (2) is zero and there is no net current. ions are far from equilibrium, that is, (V – ENa) = –90 – (+65) =
This is called the electrochemical equilibrium potential or –155 mV. If the membrane were permeable to Na+, it would
the Nernst potential and is given by: readily enter.
For potassium, the concentration gradient is outward while
RT __o C
Ex = ___ln
Fz e
(4) the force from the voltage gradient is inward. The magnitude
Ci
of the concentration gradient is slightly larger than that of the
where Ex is the Nernst potential (or the equilibrium potential voltage gradient, that is, (V – EK) = –90 – (–95) = + 5 mV.
or diffusion potential) for the ion, Co and Ci the concentra- Potassium ions are not at equilibrium; they have a tendency to
tions on the outside and inside of the cell, z the charge of the leave the cell.
ion or the valence, R the molar gas constant, T the absolute Chloride is the only ion in Table 4–1 that is at equilibrium.
temperature, and F the Faraday’s constant. RT is the thermal Cl− are distributed at or very near equilibrium in skeletal mus-
energy of the material at temperature T and RT/F is this ener- cle cells but not in most nerve cells.
gy expressed in electrical units. At room temperature, RT/F is
about 25 mV. At body temperature, 37°C, the equation can be
simplified to:
GENERATION OF THE
60mV Co
Ex = _____ __
z log10 C (5) NERNST POTENTIAL
i

with z = +1 for Na+ or K+, +2 for Ca2+, –1 for Cl−, and so on. The resting potential has its particular value because of the K+
The equilibrium potential for an ion is the potential at which and Na+ gradients and because the resting membrane is much
the net flux is zero. It can be calculated theoretically using the more permeable to K+ than to Na+. This is more easily under-
formula of equation (5) without knowledge of the actual mem- stood by first considering a membrane separating the same
brane potential. It is a way to express the concentration gradient that is permeable only to K+. Such a membrane could
gradient in electrical terms, so that the concentration gradient be constructed by reconstituting biological K channels into an
can be compared to the voltage gradient. artificial lipid bilayer (Figure 4–4).
The Nernst potentials for the various ions in Table 4–1 are When the solutions are first added to the compartments,
listed in the last column. Figure 4–3 compares three of these there is zero membrane potential. K+ will start to move with its
equilibrium potentials with a resting potential of –90 mV. concentration gradient and thereby move positive charge from
CHAPTER 4 Channels and the Control of Membrane Potential 37

electronically and then released. Such a condition can be ar-


ranged with the voltage-clamp apparatus in described Chapter
A B 6. In order to understand the process, it is necessary to know
4 mM 155 mM the concentration gradients listed in Table 4–1 and also that
the permeability of the membrane to K+ is 50–100 times great-
K+ Cl−
er than its permeability to Na+.
Starting at 0-mV membrane potential, K+ will start to move
K+ Cl− out of the cell while Na+ will start to move in, both moving
with their concentration gradients. However, more K+ will
move than Na+ because the permeability to K+ is much greater
FIGURE 4–4 K+ flowing with its concentration gradient
than the permeability to Na+, so a net positive charge will move
through an artificial bilayer that is permeable only to K+. Cl− is
impermeant and a separation of charge develops. (Modified with
out of the cell, making the inside of the cell negative with re-
permission from Landowne D: Cell Physiology. New York: Lange Medical Books/ spect to the outside.
McGraw-Hill, 2006.) The developing negative membrane potential opposes the
further efflux of K+ and acts to increase the influx of Na+. This
trend will continue, with the membrane potential becoming
compartment B to compartment A, leaving an excess negative more and more negative until three Na+ are entering through
charge on side B and producing an excess positive charge on the Na channels for every two K+ that are leaving through the
side A. This separation of charge means that there is now a K channels. At this point a steady state will be reached because
membrane potential with side B negative to side A (or, equiva- the Na/K pump is extruding three Na+ and taking up two K+
lently, side A positive with respect to side B). As side B becomes on each ATP-consuming cycle. There is no net flux in this
more negative, the further net flow of K from B to A will be steady state, so the membrane potential will not change as
reduced, until eventually sufficient charge will have been sepa- long as the ATP supply is adequate (Figure 4–5).
rated so that the flux due to the increasing electrical attraction It is important to realize that the major role of the pump is
is equal and opposite to the flux due to the concentration gra- indirect; the pump is very important for maintaining the gra-
dient. At this point electrochemical equilibrium will have been dients but contributes only a few millivolts directly to the
reached and the membrane potential will be equal to the Nernst membrane potential. If the “starting at 0 mV” experiment were
potential—in this example, –95 mV with side B negative to repeated with the pump blocked by ouabain (a cardiac glyco-
side A. This could also be expressed by saying that the Nernst side similar to digitalis) or the absence of ATP, the initial pro-
potential is +95 mV, with side A positive to side B. This is a cesses would be the same and the process would continue until
property of equation (5), because log A/B = –log B/A. the influx of sodium were equal to the efflux of potassium. At
Notice that less than 1 pmol/cm2 flux of positive charge is this point the membrane potential would stop becoming more
required to establish the membrane potential. The bulk con- negative and then, very slowly, start to move back toward 0
centrations of K+ on the two sides of the membrane have not mV as the concentrations on the two sides of the membrane
changed significantly. The change in concentrations is unde- changed over several hours.
tectable by ordinary chemical experiments. Using the figures in Table 4–1, it is possible to estimate the
immediate difference in membrane potential that can be at-
tributed to the running pump. When the membrane potential
THE RESTING POTENTIAL is –90 mV, there is a 5-mV net driving force on K+. If the mem-
The example above can be extended to explain the resting po- brane potential became 2.5 mV less negative to –87.5 mV, the
tential in a muscle cell by considering the situation that would driving force on K+ would be increased by 50%, so that three
occur if the membrane potential were artificially held at zero K+ would leave for every two that left at –90 mV. There would

Na K 3 Na 2 K

FIGURE 4–5 Ions flowing with their


concentration gradient through channels
and actively transported against their
concentration gradient by pumps. (Modified with
permission from Landowne D: Cell Physiology. New York:
ATP ADP Lange Medical Books/McGraw-Hill, 2006.)
38 SECTION II Cell Physiology

be a 2.5-mV decrease in the driving force on Na+, but this is Increased Ko depolarizes membranes because it reduces the
less than 2% of the 155-mV driving force, so it would make a K+ gradient across the membrane and makes EK closer to zero.
negligible change in the Na+ influx, and three Na+ would enter This reduces the tendency for K+ to leave the cell, so the bal-
for every three K+ leaving. Thus, about 87.5 mV of the resting ance is reached at a less negative potential. Increased Ko+ is a
potential comes from the gradients and an additional 2.5 mV dangerous, potentially lethal condition because excitable cells
comes directly from the pump. require the normal resting potential to remain excitable.
If the concentrations and the ionic conductances are known, Doubling the blood K+ level (hyperkalemia) is likely to com-
the membrane potential can be calculated using equation (4) to promise cardiac muscle function.
find the Nernst potentials and equation (2) to find the currents.
When the membrane potential is not changing, there is no net
Kir CHANNELS SUPPORT
current. If the pump is not running and the membrane only
conducts Na+ and K+, INa = –IK or gNa(V – ENa) = –gK(V – EK), THE RESTING POTENTIAL
which can be rearranged to solve for V:
Some cells, notably cardiac and skeletal muscle cells, have Kir
g E +g E channels that are open, thus conducting, at the resting poten-
V = _________
Na Na
g +g
K K
(6)
Na K
tial and are thought to be the major contributor to the resting
The membrane potential is the weighted average of the K conductance. These were named inward rectifiers when ex-
equilibrium potentials; the weighting is by their respective periments demonstrated that the inward current through
conductances. If gK >> gNa, the membrane potential will be them, when the membrane potential was hyperpolarized be-
near EK; if gNa >> gK, it will be near ENa, and if they are equal, yond EK, was larger than the outward current seen when the
it will be halfway between. If the membrane is permeable membrane was depolarized. It is perhaps an unfortunate name
only to these two ions and there is no external source of because, in normal life, the membranes never experience such
electrical current, the membrane potential will always be a large hyperpolarization. The important aspects of this chan-
between EK and ENa. These concepts will become more use- nel’s function are to be open for outward movement of potas-
ful when the conductances change, as seen in the next three sium near the resting potential and then to become noncon-
chapters. ducting when the cell is depolarized. This blocking in the
Because the resting membrane is preferentially permeable depolarized state will be seen to be important for cardiac mus-
to potassium, the resting potential is sensitive to the external cle action potentials, as described in Chapter 6.
potassium concentration (Figure 4–6). Increasing external K Kir is not a voltage-sensitive channel. The blocking comes
will bring the membrane potential closer to zero or depolarize about because Mg2+ or other polyvalent cations in the cyto-
the membrane. The resting membrane in its normal ionic en- plasm attempt to go through the channel when they are depo-
vironment is considered polarized. A change of potential in larized and get stuck, thus preventing K+ from using the chan-
the positive direction, toward 0 mV, is a depolarization. A nel. If the channels are studied under conditions without
change in the other direction, making the membrane potential polyvalent cations, they conduct K+ equally well in both
more negative, is a hyperpolarization. directions.

−20

−40
Membrane potential (mV)

−60

−80

−100
EK = 60 mV log [K]o/155
−120

−140

−160
1 10 100
[K]o mM (note log scale)

FIGURE 4–6 The observed membrane potential as a function of the external K+ concentration. The solid line is the theoretical
prediction for a membrane that is permeable only to K+. Notice the logarithmic concentration scale. (Modified with permission from Landowne
D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 4 Channels and the Control of Membrane Potential 39

GOLDMAN–HODGKIN–KATZ where K is a constant describing the material composition of


the membrane. If the area is larger, it will take a greater amount
EQUATION of charge to change the potential. The thinner the membrane,
If permeabilities are known, rather than conductances, a mem- the closer the charges are to each other and the more charges
brane potential can be calculated using the theoretical will have to be moved to change the potential. The capaci-
Goldman–Hodgkin–Katz (GHK) or constant field equation: tance of a typical membrane is about 1 μF/cm2; this value is
often used to estimate the size of a cell by measuring its
P Na + + P K + + P Cl −
V = 60mV log10_________________
Na o
+
K o
+
Cl i

(7) capacitance.
PNaNai + PKKi + PClClo
The membrane resistance is the reciprocal of the membrane
As in equation (6), the GHK equation simplifies to the Nernst conductance:
equation if only one permeability is greater than zero. The
1
GHK equation has been useful to describe experimental re- Rm = __
g (10)
m
sults when some of the concentrations are set to zero, which
makes the Nernst potentials in equation (6) meaningless. The longitudinal resistance is proportional to the length and
The relationship between permeability and conductance can inversely proportional to the cross-sectional area:
be set on a quantitative basis by considering the condition ρ × length
Rl = ________
Area
(11)
when the membrane potential is zero and then, after multiply-
ing the chemical flux by Faraday’s constant, equating equations where ρ is the resistivity of the cell contents.
(3–1) and (4–2) to obtain the electrical current. Thus, we have:
gxEx = PxF ΔCx (8)
PASSIVE PROPERTIES OF
CHANGES IN MEMBRANE A SMALL ROUND CELL
POTENTIAL When pulse of current is injected into a small round cell (which
can be assumed to have the same membrane potential over its
The membrane potential will change if current is injected into entire surface), the membrane potential does not change in-
the cell by opening channels that allow ions to flow with their stantaneously. Instead, it changes with an exponential time
electrochemical gradients. It takes time to change the mem- course with a characteristic time constant (τ), the time it takes
brane potential; it will not jump instantaneously to a new value. to discharge the change in voltage to 1/e = 37 percent of its
Many nerve and muscle cells are quite long, more than 1 m for value (or the time it takes to charge to 63 percent of its final
some nerve cells. The effect of a localized current will spread value) (Figure 4–7). Initially the injected charges are adding to
passively from the site of injection but may not change the po- the stored charges that were causing the original membrane
tential of the entire cell. These temporal and spatial effects are potential. Later, when the membrane potential has reached a
shared by electrical cables and are referred to as the cable prop- new steady state, current equal to the injected current is leaking
erties. They can be understood by considering the membrane back out through membrane channels. When the pulse is ter-
capacitance, the membrane resistance, and the longitudinal cy- minated, the excess stored charge leaks out through the chan-
toplasmic resistance between different parts of the cell. nels and the membrane potential decays exponentially to its
The passive spread by cable properties must be distin- original value.
guished from the active spread by action potentials. The pas- The time constant of these exponential changes is the prod-
sive effects occur without any change in the number of open uct of the cell’s membrane resistance and capacitance. Many
channels. If sufficient current enters a nerve axon and depolar- cells have time constants in the range of 1–20 milliseconds.
izes it above threshold, an action potential will be elicited and These time constants limit how rapidly the membrane poten-
will propagate without loss of amplitude over the entire length tial can change and permit temporal summation of synaptic
of the cell. The action potential is regenerated as it propagates. events in the central nervous system (see Chapter 7).
As the wave of opening sodium channels moves, energy is sup-
plied to the process from the Na+ gradient all along the axon.
In contrast, a smaller depolarization or a hyperpolarization PASSIVE PROPERTIES OF
that does not open Na channels will spread only a few millime-
ters, becoming progressively smaller when measured at a A LONG CYLINDRICAL CELL
greater distance from the stimulus. An extended cell or a tissue with cells that are electrically
The membrane capacitance is the ratio of the charge sepa- connected by gap junctions may have different membrane
rated to the membrane potential—equation (1). The capaci- potentials at different locations. If there is a local change in per-
tance is related to the membrane geometry by the following meability, current will flow into or out of the cell and the mem-
equation: brane potential will change at that location and, to a lesser extent,
K × area at nearby locations. With a prolonged steady current, which lasts
C = ________ (9)
Thickness much longer than the time constant described in the previous
40 SECTION II Cell Physiology

I V
value at the site of injection (Figure 4–8). Typical length con-
stants for nerve and muscle cells are 0.1–2.0 mm. A 10-μm cell is
approximately isopotential, but a 150-cm-long nerve cell re-
In quires an active propagation mechanism to be able to communi-
cate electrical activity from end to end.
C R
I V The voltage change declines because some of the injected
current leaks out of the cell and is not available to depolarize
the adjacent regions. The amount that leaks out is proportion-
Out
al to the voltage change, so the decline is exponential. The
A B length constant depends on the ratio of the membrane resis-
tance to the longitudinal axoplasmic resistance.
As the distance from the injection increases, the amplitude of
I
the transient response decreases and the rise time becomes lon-
ger and more sigmoidal (Figure 4–9). Initially most of the charge
entering the cell goes to the membrane immediately adjacent to
ΔV = IR[1 −exp(−t/τ)] the source; only later it is enough available to charge the distal
ΔV = IR exp(−t /τ)
V 63% 37% membrane. When the pulse is terminated, all responses decay at
τ τ the same rate. Synapses are distributed on the dendritic tree at
C
different distances from the cell body. The more distant syn-
apses will have less effect on the cell’s activity; the amplitude of
FIGURE 4–7 A spherical cell (A), its equivalent circuit (B), the effect will be lower and its time course will be slower.
and the voltage response to an injected pulse of current (C).
Passive spread is important for action potential propaga-
(Modified with permission from Landowne D: Cell Physiology. New York:
Lange Medical Books/McGraw-Hill, 2006.)
tion; it is the mechanism of connection between the active re-
gion and the adjacent resting region. Action potentials propa-
gate more rapidly in larger-diameter axons because they have
section, there will be a steady change in potential that is largest lower longitudinal resistance and longer length constants.
at the point of current entry and falls off exponentially with dis- The passive properties, membrane capacitance, membrane
tance with a characteristic length constant (λ) or space constant, resistance, and longitudinal resistance, are referred to as cable
the distance it takes for the potential to drop to 37 percent of its properties because they also determine the ability of under-

I V1 V2 V3

RI RI RI RI
In

I C Rm Rm Rm Rm

Out
B

1 ΔV(x) = ΔVo exp(−x/λ)

2
37% 3

λ Distance x

C
FIGURE 4–8 A long cell (A), its equivalent circuit (B), and the steady-state distribution of its membrane potential in response
to a steady injection of current (C). (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 4 Channels and the Control of Membrane Potential 41

I V1 V2 V3

1
2
3
V

FIGURE 4–9 The transient voltage responses at three distances from the site of an injected pulse of current. (Modified with permission
from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

water cables to transmit signals. The length constant for ■ Only a very small number of ions must be separated to produce
undersea cables is several kilometers; for nerve axons it rang- the membrane potential. This is negligible compared with the
es from about 0.1 to 20.0 mm depending on the diameter. concentrations available on both sides.
Undersea cables rely on repeater amplifiers for longer distanc- ■ The resting membrane potential is a steady state with ions
es; nerves use voltage-dependent sodium channels, as de- moving with their electrochemical gradient through channels
scribed in Chapter 6. and an equal number being pumped against their electrochem-
ical gradient at the expense of ATP.
When cell–cell junctions join cells, they can operate electri-
■ The GHK equation can be used to calculate the membrane
cally as if they were all one cell. Many of the cells in the heart
potential if the permeabilities to the various ions and their
are coupled and action potentials propagate from one cell to
concentrations are known.
another supported by the passive spread of depolarization via
■ When current flows through the membrane, the membrane
the cell–cell junctions. There are also cell–cell junctions be-
potential changes in time and in space, governed by the “cable
tween some neurons in the CNS.
properties.”
For some it is helpful to visualize a hydraulic analogy of
■ When a pulse of current is injected into a cell, there is a
these electrical phenomena. Electrical voltage is analogous to
characteristic time required for the membrane potential to
water pressure and electrical current to solution flow. The change.
long cell is similar to a leaky hose, with lower membrane resis-
■ When a steady current is injected into a long cell, the potential
tance corresponding to more leaks and lower longitudinal change is largest at the injection site and decreases characteris-
resistance corresponding to larger hose diameter. tically away from the site.

CHAPTER SUMMARY STUDY QUESTIONS


■ An electrical membrane potential is directly proportional to 1. If all the Na–K pumps in the membrane of a muscle cell were
the separation of positive and negative charges across the cell stopped, all of the following changes would be expected for the
membrane. The ratio of separated charge to voltage is the muscle cell except
membrane capacitance.
A) immediate loss of the ability of the cell to carry action
■ Cell membranes separate solutions with quite different ionic potentials
compositions. B) gradual decrease in internal K+ concentration
■ The movement of ions is directly proportional to the net C) gradual increase in internal Na+ concentration
driving force on the ions. The net driving force is the electro- D) gradual decrease in resting membrane potential (the
chemical gradient or the difference between the effect of the potential would become less negative)
membrane potential and the effect of chemical gradient. E) gradual increase in internal Cl− concentration.
■ The effect of the chemical gradient can be expressed by the
Nernst equilibrium potential.
42 SECTION II Cell Physiology

2. If the potassium ion concentration on the outside of a resting 4. A scientist is recording from the soma of a neuron with an
skeletal muscle cell is doubled to twice of the normal value by intracellular microelectrode to study synaptic inputs on the
adding K+ and Cl− in equal amounts, what would be the best dendrites. The letters a, b, and c below indicate the synaptic
estimate of the effect on the resting membrane potential? potentials recorded from three different synaptic inputs. For
A) hyperpolarize about 100 mV identical synaptic inputs to the dendrites, which synaptic
B) depolarize about 5 mV potential was generated by the synapse at a location on the
C) hyperpolarize about 15 mV dendrites closest to the soma?
D) depolarize about 20 mV
E) no measurable effect
3. The following cell in an organism called the Europa louse
was recovered from a moon of Jupiter with a space probe.
The intracellular and extracellular concentrations of all the
ions are given as follows: A) B) C)

Extracellular Intracellular
+
Rb = 100 mM Rb+ = 1 mM

SO42− = 50 mM SO42− = 0.5 mM

The cell membrane is permeable to Rb+ and not to SO42− or water.


What is the resting membrane potential? (The sign refers to the
potential inside of the cell.)
A) +30 mV
B) +60 mV
C) +120 mV
D) −30 mV
E) −60 mV
5
C H A P T E R

Sensory Generator
Potentials
David Landowne

O B J E C T I V E S

■ List eight sensations and the names of the specialized sensory receptor cells
responsible for generating these sensations.
■ Describe sensory adaptation in these receptors.
■ Draw a schematic cartoon of (a) a Pacinian corpuscle and its sensory ganglion
cell (including cell body and central process); (b) a cochlear hair cell and its
synapses; and (c) a photoreceptor and its synapses.
■ List three or more differences between ion channels that underlie action
potentials, resting potentials, and receptor potentials.

Animals have developed a wide variety of sensory organs ca- shock or sufficient pressure is applied to the eye, a person will
pable of monitoring chemicals, light, sound, and other me- report flashes of light, even if the room is dark.
chanical events in the external and internal environments. In Each cell also has a receptive field that is the region in stim-
all of these organs there are mechanisms to convert informa- ulus space that evokes a response in that cell. The receptive
tion about the environment into electrical signals within the field of a photoreceptor in the retina is a particular location in
nervous system. This chapter is concerned about the conver- the visual space in front of the eye and a range of colors to
sion process and some general properties of all receptors. More which that receptor is sensitive. The receptive field for a
details about the sensory organs and the systems that process somatosensory nerve in the skin is the area of skin that elicits
the nerve signals are provided in Section 4: Chapters 13, 15, a response. The receptive field for an olfactory neuron is the
16, and 17. range of chemicals it can detect. Cells in the CNS concerned
Transducers can convert one type of energy to another. with sensory information also have receptive fields. Different
The cells or portions of cells that perform the initial step of cells handle sensory information from the feet than that from
sensory transduction convert light or mechanical energy or the hands. The incoming information arrives on “labeled
the presence of specific chemical conditions into a change in lines”; the CNS processors know from whence it comes. There
the membrane potential called the receptor potential or sen- are several locations in the brain that have receptive fields in-
sory generator potential. In small sensory cells, this genera- cluding the same location in visual space. The receptive fields
tor potential directly controls the synaptic release process to of these higher-order cells are more complex, as signal pro-
be described in Chapter 7. In longer cells, the generator cessing has occurred comparing the output from one lower-
potential will initiate an action potential that propagates to a order cell with that of others.
distant presynaptic ending and then trigger the release pro- Mechanosensory transduction is direct, by mechanosensi-
cess. The information about the stimulus energy that was tive channels in the membrane. The sensory cell often has mol-
transduced into a generator potential is then encoded in the ecules or structures to focus the mechanical energy or filter out
frequency of action potentials. undesired mechanical disturbances, and there may be an elab-
Each sensory cell has an appropriate stimulus, called its orate organ—such as that comprising the outer, middle, and
adequate stimulus. The CNS interprets signals coming from inner ear—to deliver the desired mechanical energy to the ap-
this cell in terms of its adequate stimulus. The adequate stimu- propriate cell. In the end, a relatively nonspecific cation chan-
lus for photoreceptors in the eye is visible light. If an electric nel opens and both Na+ and K+ move with their concentration

43

Ch05_043-046.indd 43 11/26/10 9:41:42 AM


44 SECTION II Cell Physiology

disturbance of these cells by the appropriate sound also leads to


inward current carried by K+ through mechanosensitive chan-
nels on the cilia and depolarizes the cell. The sensory hair cells
are short and synapse with auditory nerve cells in the ear. The
hair cells do not have action potentials; they are short compared
to their length constant, so they can rely on passive spread to
open CaV channels to release transmitters.
Some taste chemosensation is supported directly by chemo-
sensitive channels, as in the glutamate receptors for the umami
FIGURE 5–1 The changes in membrane potential of a taste (the distinctive savory taste of glutamate); these are rela-
mechanosensory nerve ending to stimuli of three different tively nonselective cation channels that depolarize the cells.
amplitudes. (Modified with permission from Landowne D: Cell Physiology, New Others use channels even more directly; Na+ moving through
York: Lange Medical Books/McGraw-Hill, 2006.) epithelial sodium channels (ENaCs) depolarizes cells to pro-
vide the salty taste sensation. Odors are detected by G protein–
coupled receptors (GPCRs) whose G proteins activate adenylyl
gradients. In skin mechanoreceptors, such as the Pacinian cyclase, thus elevating levels of cyclic adenosine monophos-
corpuscle discussed below, there is a greater driving force on phate (cAMP). The cAMP opens a cyclic nucleotide–gated
Na+, so more Na+ than K+ moves and the cell depolarizes. The (CNG) nonspecific cation channel that depolarizes the cell.
number of mechanosensitive channels that open is propor- CNG channels are tetramers with six TM segments and are
tional to the amount the membrane is stretched by the stimu- structurally similar to KV channels but lack the latter’s exquisite
lus. A larger stimulus will open more channels and produce a selectivity for K ions and the voltage sensitivity.
larger depolarization (Figure 5–1). If the depolarization is large Light transduction also involves GPCRs with seven TM seg-
enough, action potentials will be initiated and will propagate ments: rhodopsin in the rods and three other opsins in the cones
toward the CNS. tuned for short, medium, and long (or blue, green, and red)
The situation is more complex in the ear, because the sensory wavelengths. The chromophore that absorbs the light is 11-cis
cells (called sensory hair cells, for the hairlike appearance of the retinal (MW 284). Absorption of a photon triggers conversion
modified cilia on their apical surface) are part of an epithelium of the retinal to the all-trans isomer, which causes a conforma-
that separates two different solutions. However, mechanical tional change in the opsin protein informing the G protein that

Disk membrane

Rhodopsin
(GPCR) Phosphodiesterase

β/γ
α
Transducin
(G protein)

cGMP GMP

hν cGMP

Na

FIGURE 5–2 The processes linking light absorption by rhodopsin and the closing of cyclic nucleotide–gated channels. Light induces
a conformational change in rhodopsin that causes the subunits of transducin to dissociate. The alpha subunit stimulates a phosphodiesterase
that degrades cGMP. In the absence of cGMP a channel that was allowing the entry of Na+ closes and the cell hyperpolarizes. (Modified with
permission from Landowne D: Cell Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 5 Sensory Generator Potentials 45

an event has taken place (Figure 5–2). The G protein is called


transducin; it was the first G protein to be identified and was
named before the family was well known. Transducin activates
a phosphodiesterase that hydrolyzes cyclic guanosine mono-
phosphate (cGMP). In the dark, there is a CNG channel that is FIGURE 5–3 Fast and slow sensory adaptation. The colored
open and carrying inward current. The channel closes when the bars indicate a steady level of stimulation. The rapidly adapting
cGMP level drops; when the light is on, the dark current de- receptor on the left adapts completely after two impulses have
creases and the cell hyperpolarizes. There is amplification along occurred. In the slowly adapting receptor on the right, the rate of
this chemical pathway, so one photon leads to the closure of firing declines less rapidly. (Modified with permission from Landowne D: Cell
many CNG channels. The hyperpolarization reduces a steady Physiology, New York: Lange Medical Books/McGraw-Hill, 2006.)
output of synaptic vesicles to pass the message on to the next cell
in the pathway to the brain.
The sensation of uncomfortably hot skin temperature has
cells that surrounds the nerve ending. When one side of the
been linked to the direct activation of a channel called VR1,
capsule is distorted by the stimulus, at first the distortion is
for vanilloid receptor. It is also known as the capsaicin recep-
transmitted to the nerve ending and the nerve depolarizes.
tor because it can be activated by the vanilloid capsaicin, the
Then the capsule balloons out to the sides, the forces on the
major piquant ingredient in hot peppers. VR1 is a member of
nerve are relieved, and the nerve stops firing. When the stimu-
the transient receptor potential (TRP) channel family; it has
lus is removed, the capsule rebounds to its original shape,
a six-TM architecture and is permeable to cations. Raising the
transiently pushing the sides of the nerve in the process. The
temperature into the range of 42°C (107.6°F), which many hu-
Pacinian corpuscle is tuned to provide maximum information
man observers identify as painfully hot, opens this channel,
about vibratory stimuli and to ignore steady pressure.
depolarizes the sensory ending, and initiates a train of action
Muscle spindle organs are sensory structures embedded in
potentials. Other members of the TRP family have been asso-
skeletal muscles, which provide information about the length
ciated with temperature sensation and other functions, though
of the muscle to the CNS (see Figure 2–3 and Chapter 14).
not all with pain.
Muscle spindles adapt rapidly to changes in length but also
Our everyday experience of our sense is not a direct repre-
continue to fire during a sustained stimulus. The firing rate
sentation of the stimuli but rather the result of processing
decreases slowly during the stimulus; muscle spindles are said
that occurs in the nervous system. We do not see the world as
to be slowly adapting (Figure 5–3).
flashes of light at different positions in our visual fields but
The nervous system responds to changes in the environ-
rather as objects and surroundings. Pain is an experience
ment, and by reducing the messages indicating that a stimulus
that can arise from a wide variety of stimuli without neces-
is still present, more attention can be given to any changes.
sarily telling us anything definite about the stimulus. A few
Adaptation takes place at many levels—accessory tissue before
specific nociceptors have been identified, but there are also
the receptor potential, the receptor potential itself, the encod-
many other receptors that may be associated with pain.
ing mechanism that initiates action potentials, and at many
Elevated K+ from damaged cells or the direct cutting of a
higher synapses where the incoming message is integrated
nerve cell can induce action potentials that may be interpret-
with other signals. Adaptation to light occurs by constricting
ed as pain. Acid-sensing ion channels (ASICs) in the ENaC
the pupils, by photobleaching the pigments, and by feedback
family respond to lactic acid released in the heart and depo-
regulation of the steps in the biochemical cascade.
larize nerves that provide the sensory pathway for the pain-
Many senses have some form of efferent control. The sympa-
ful experience of angina. P2X3 receptor channels, which can
thetic nervous system can release norepinephrine onto the Pa-
be activated by adenosine triphosphate (ATP) released by
cinian corpuscle, which will increase its sensitivity to mechani-
damaged cells, have been associated with pain from over-
cal stimuli. Muscle spindle organs (see Chapter 14) have efferent
stretched bladders, and P2X4 receptors have been associated
nerves (γ motor nerves) that set the range of lengths to which
with a neuropathic pain generated within the nervous system
the sensory nerve is most sensitive. There are also motor hair
without obvious outside stimuli.
cells in the ear that can selectively enhance the sensitivity of sen-
sory hair cells to particular sounds (see Chapter 16). There are
many controls on the eye to assure that the object of interest is
SENSORY ADAPTATION suitably focused on an appropriate portion of the retina even as
the head changes its position in space (see Chapter 15).
All senses except pain adapt; if they are presented with a
maintained stimulus, the response will diminish in time. The
Pacinian corpuscle adapts rapidly and responds to a sustained
stimulus with only one or two action potentials at the start CHAPTER SUMMARY
(Figure 5–3). When the stimulus is released, there is an off- ■ Each sensory cell has an adequate stimulus.
response and another action potential is initiated. Most of this ■ Touch, hearing, and other mechanosensation occur via
adaptation takes place in the onionlike capsule of accessory mechanosensitive channels.
46 SECTION II Cell Physiology

■ Taste is mediated by chemosensitive channels and odor by 2. Which of the following sensory cells has a hyperpolarizing
GPCRs and CNG channels. generator potential in response to its adequate stimulus?
■ Vision is also mediated by GPCRs—for example, A) Pacinian corpuscle nerve ending
rhodopsin—and CNG channels. B) muscle spindle nerve
■ Pain is mediated by ASICs and purine-activated channels. C) taste bud cell
■ All senses except pain adapt. D) retinal cone cell
E) olfactory nerve ending
3. Hair cells are the sensory receptor cells in the cochlea. They are
excited by the vibration of the hair bundle. Vibration of the hair
STUDY QUESTIONS bundle causes which one of the following events?
A) influx of K+ through mechanosensitive cation channels
10 10 in the tips of the cilia
Response

Response
impulse/s

impulse/s

B) influx of Ca2+ through cyclic nucleotide–gated (CNG)


5 5 channels in the tips of the cilia
C) long-lasting hyperpolarization of the hair cell
D) a train of action potentials propagated from the cilia to
10 30 50 10 30 50 the cell body of the hair cell
Seconds Seconds
A) B)
10 10
Response

Response
impulse/s

impulse/s

5 5

10 30 50 10 30 50
Seconds Seconds
C) D)

1. The graphs above show the frequency of action potentials (y-axis)


recorded from a primary sensory afferent fiber during sensory
stimulation. Which one of these graphs shows the response from
a typical sensory fiber (excluding pain fibers) to a constant
maintained stimulus applied beginning at 10 seconds and lasting
throughout the recording (i.e., until 50 seconds)?
6
C H A P T E R

Action Potentials
David Landowne

O B J E C T I V E S

■ Describe the activation of action potentials.


■ Explain the propagation of action potentials.
■ Describe the membrane currents underlying action potentials.
■ Describe the activity of channels producing action potentials.
■ Explain the membrane basis of the action potential threshold and
refractory period.
■ Explain actions of calcium, local anesthetics, and neurotoxins on
action potentials.
■ Describe the relationship between channel activity and cardiac muscle
contraction.
■ Describe the membrane basis of intrinsic cardiac pacemakers.
■ Describe the effects of acetylcholine and NE on cardiac action potentials.

ROLE OF VOLTAGE-SENSITIVE in nerve and skeletal muscles is on the order of 1 millisecond;


in cardiac ventricular muscle cells, their duration is several
SODIUM CHANNELS hundred milliseconds. In nerve and skeletal muscles, the un-
derlying permeability changes are a transient increase in so-
Action potentials are changes in membrane potential that dium permeability followed, after a delay, by an increase in
propagate along the surface of excitable cells. They are best potassium permeability due, respectively, to the activation of
known in nerve and muscle cells but also occur in some other sodium and potassium channels (Figure 6–1). Cardiac action
cells, including egg cells associated with fertilization. Unlike
some other changes in membrane potential, action potentials
are characterized as being “all-or-none”; they have a threshold
for excitation and a stereotyped duration. Immediately follow- 25 mV
ing an action potential, the excitable cell has a refractory pe- 10 mS/cm2
riod when it is more difficult or impossible to elicit a second
1 ms
action potential.
Like most changes in membrane potential, action potentials
are the result of changes in membrane permeability due to the
activity of channels, or proteins embedded in the membrane
lipid bilayer that facilitate the passive movement of specific
ions with their electrochemical gradients. An action potential FIGURE 6–1 An action potential (red trace) and the
is a change in membrane potential from a resting potential underlying changes in membrane conductance for Na+ (blue
of about –70 mV (the inside of the cell is negative) to about trace) and K+ (beige trace). (Modified with permission from Landowne D:
+30 mV and then back to the resting potential. Their duration Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

47

Ch06_047-058.indd 47 11/26/10 9:42:20 AM


48 SECTION II Cell Physiology

potentials are more complex and also involve the activation of Change of
calcium channels. channel
conformation
Action potentials are all-or-none and propagate because the
sodium channels are voltage-sensitive. Depolarization, the
reduction of the membrane potential, from –70 to 0 mV,
induces a conformational change within a few hundred micro-
seconds in the sodium channel protein, which leads to in in- Depolarization Increase of
crease in permeability to sodium ions. Sodium ions rush into of membrane sodium
potential permeability
the cell through these open voltage-dependent Na (NaV)
channels and bring positive charge with them, which further
depolarizes the cell, opening more NaV channels (Figure 6–2).
This positive feedback loop persists until all of the sodium
channels have opened. Once the loop is started, it continues to Entry of
completion. The depolarization spreads passively to adjacent sodium
into cell
regions of the membrane and activates nearby sodium chan-
nels. This wave of molecular conformational change and elec- FIGURE 6–2 The action potential’s positive feedback cycle.
trical activity propagates over the length or surface of the cell The cycle is started by a depolarization and continues until all of the
at velocities up to 120 m/s. Potential energy that is stored in sodium channels have been activated. (Modified with permission from
the sodium concentration gradient is sequentially used along Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

the propagation path. The propagation velocity is determined


by the rate of molecular change and the electrical properties of
the cell that control the spread of potential changes (cable through. In addition, the NaV channels are unable to open
properties). again until the membrane is repolarized to the resting po-
About 1 millisecond later, the sodium channels undergo a tential for a few milliseconds to allow recovery from inacti-
second conformational change and inactivate. In this third vation (Figure 6–3). This automatic closing of the sodium
conformation, they are closed and sodium no longer passes channels limits the duration of nerve and skeletal muscle

Open

Activated

Resting Inactivated
closed closed

FIGURE 6–3 Sodium channels can be in different functional states. A depolarization first causes the channel to change from the resting
state to the activated and open states and later to the inactivated state. Repolarization is required to go from the inactivated state back to the
resting state. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 6 Action Potentials 49

Vi
Vm Axon

Axial
wire

Vo

Vc I

FIGURE 6–4 A simplified voltage-clamp circuit for a squid giant axon. The membrane potential, Vm, is sensed as the difference
between the inside potential, Vi, and the outside potential, Vo. Vm is compared to the command potential, Vc, and, if they are different a current
flows through the axial wire and the cell membrane to make Vm equal to Vc. (Modified with permission from Landowne D: Cell Physiology. New York:
Lange Medical Books/McGraw-Hill, 2006.)

action potentials. Loss of the ability to open again produces measure the effect of a change in membrane potential in the
the refractory period. ionic permeabilities without any change to the membrane poten-
The outward movement of K+ carrying positive charge out of tial due to the movement of ions. Their technique was to include
the cell produces the repolarization (the falling phase of the the nerve membrane in a negative feedback circuit (Figure 6–4).
action potential). In some cells, voltage-dependent K channels A pair of electrodes measures the membrane potential;
(KV) channels—whose activation is slower than that of sodium this is then compared with a desired command potential. If
channels—facilitate repolarization. In mammalian myelinated the membrane potential is different from the command po-
axons, the repolarizing current passes through the (nonvoltage- tential, a current is made to flow through the membrane in
sensitive) potassium channels that produce the resting potential. a direction that reduces the difference. Thus, the voltage
The axons seem to be an exception; the presynaptic nerve termi- across the membrane is clamped at a desired value. When
nals and the cell bodies of most neurons have KV channels. the controlled voltage is a pulse from the resting potential to
0 mV, four different kinds of current can be identified
(Figure 6–5).
VOLTAGE CLAMPING The first is the charge movement necessary to change the
potential or change the charge on the membrane capacitance.
This understanding of the action potential mechanism comes Second, there is a small outward current called the gating
from the work of Alan Hodgkin and Andrew Huxley about current. Then there is an inward current that is replaced in a
50 years ago. Working with giant nerve axons isolated from few milliseconds by an outward current, which lasts as long as
squid, they were able to break the positive feedback loop and the pulse.

0 mV
“Voltage clamped”
membrane potential
−70 mV

Ic
IK
Ig Outward
INa Current with both
potassium and
sodium ions
Inward

FIGURE 6–5 The membrane currents (lower trace) in response to a voltage-clamp pulse (upper trace). Ic, capacity current; Ig, gating
current; INa, sodium current; IK, potassium current. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
50 SECTION II Cell Physiology

0 mV
“Voltage clamped”
membrane potential
−70 mV
IK

Outward
Current without
sodium ions
Inward

INa Current without


potassium ions

1 ms
Ig

Current without
either ions

FIGURE 6–6 The separation of the currents by changing the solutions. The labels have the same significance as Figure 6–5.
(Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

One can replace the contents of a segment of squid axon produce less inward Na current until, at about +60 mV, no net
with a simple salt solution and maintain functioning channels. current passes through the Na channels. Still larger pulses can
By changing the solutions bathing both sides of the membrane, drive outward Na current through the Na channels. The rever-
one can separate the currents carried by Na+ (INa) and K+ (IK) sal of the current occurs at the sodium equilibrium potential,
and also see the gating current (Ig) still present in the absence ENa. If the ratio of the sodium concentrations bathing both sides
of either ion (Figure 6–6). of the membrane is changed, this reversal potential also chang-
Notice that at 0 mV, the Na current is inward and the es. With modest depolarizations, the inward current increases
K current outward. The Na current activates or increases more because larger pulses open more sodium channels. However,
rapidly than the K current. It inactivates or decreases during the less negative potential decreases the inward driving force
the pulse, even though the membrane potential is kept at 0 mV, on the sodium ions; after most of the NaV channels have been
whereas the K current remains for the duration of the pulse. opened, still larger depolarizations decrease the Na current.
If the potential is pulsed to other depolarized potentials, all When the membrane potential exceeds the sodium equilibri-
four components of the current are present, although their am- um potential, Na is forced out of the cell through the open NaV
plitude and time course and, in the case of INa, direction may channels. In a free-running action potential, the membrane po-
change (Figure 6–7). The Na current becomes more inward tential never exceeds the sodium equilibrium potential and
between the resting potential and about 0 mV. Larger pulses there is always a net entry of Na into the cell.

+80 mV

1 mA/cm2 +60

1 ms +40

−20

+80 mV
+60
+40
0
−20
−70

FIGURE 6–7 The current’s responses (upper traces) to voltage steps of varying amplitude (lower traces). Capacity current transients
not shown. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 6 Action Potentials 51

The Hodgkin–Huxley equations are available in a com-


mercial computer program called Neuron. The Web site
(http://nerve.bsd.uchicago.edu/nerve1.html) has a JavaScript
rendition that will allow you to manipulate the equations
1 mA/cm2
with most modern Web browsers.
10 ms

0 mV
THRESHOLD
−70
The threshold arises because there are two different effects
FIGURE 6–8 The recovery from inactivation shown by a
of small depolarizations. On the one hand, depolarization
two-pulse experiment with different amounts of time at the
resting potential between pulses. Capacity current transients
will increase the probability that NaV channels open and
not shown. (Modified with permission from Landowne D: Cell Physiology. permit inward current, which will lead to further depolar-
New York: Lange Medical Books/McGraw-Hill, 2006.) ization. On the other hand, depolarization moves the
membrane potential further away from the potassium equi-
librium potential, increasing the net driving force on potas-
The Na current activates and inactivates more rapidly as the sium ions and thus producing an outward current through
size of the pulse is increased. If a second pulse is given imme- the resting potential potassium channels, which will lead to
diately after the first, the gating current and the sodium cur- repolarization.
rent during the second pulse are smaller than during the first If a sufficient number of sodium channels are opened so that
pulse (Figure 6–8). They both recover in parallel as the dura- the inward sodium current exceeds the outward potassium cur-
tion between pulses is increased. The rate of recovery from rent, the cell has exceeded threshold and will continue to depo-
inactivation is also voltage-dependent, as the channels recover larize until all of the available sodium channels have opened.
more rapidly at more hyperpolarized potentials. Treatments that reduce the sodium current—for example, re-
The K current increases and becomes more rapid as the ducing extracellular sodium concentration or reducing the
membrane potential is increased. Above about +20 mV, the number of NaV channels—will elevate the threshold.
increase in amplitude becomes proportional to the change in
potential, indicating that all of the channels are open and that
only the driving force continues to increase. REFRACTORY PERIODS
The gating current is a direct sign of the conformational
changes in the NaV channel proteins. These molecules con- During an action potential, most of the NaV channels activate
tain charged groups and dipoles that move or reorient when or open and then inactivate and close into a state that differs
the electrical field changes, specifically the S4 TM helices from their condition before the action potential. In order to
shown in Figures 3–3 and 3–4. This movement can be mea- recover from inactivation and be available to open again, the
sured as the gating current. As the pulse is made progressive- NaV channels must spend some time with the membrane po-
ly more positive and more sodium channels open, the ampli- tential near the resting potential. They will not recover if the
tude of the gating current grows and the currents become membrane remains depolarized.
more rapid. Above about +20 mV, these two changes are com- During this recovery, the axon is said to be refractory be-
plementary and the area under the gating current trace is cause it is resistant to stimulation. The refractory period is di-
constant, indicating that all of the channels are undergoing vided into two segments: an absolute refractory period when
conformational changes and are doing so more rapidly at no stimulus, however large, can elicit a second action potential,
more positive potentials. followed by a relative refractory period when the axon can be
The capacitance current increases linearly with the size of stimulated again but requires a larger stimulus to elicit the sec-
the pulse because it requires more charge to change the voltage ond response than was needed for the first (Figure 6–9).
by larger amounts. During the absolute refractory period, so few NaV channels
Hodgkin and Huxley separated the currents and showed have recovered that even if all of the recovered channels were
how the ionic currents were proportional to the driving opened, there would be insufficient sodium current to exceed
force on the ions. They created mathematical equations that the outward potassium current, which tends to restore and
emulated the amplitude and time course of the permeability maintain the resting potential. During the relative refractory
changes and showed that these equations could predict the period, a larger depolarization is required because a larger
amplitude and time course of action potentials as well as their fraction of the available NaV channels must be opened to ob-
threshold, conduction velocity, refractory period, and several tain the same number of channels opened in the first stimulus.
other features. Their concept of describing ionic current as the In addition, in many nerve and muscle cells, there are more
product of conductance times driving force is used to describe open potassium channels immediately following an action
most of the remaining electrophysiological phenomena in all potential, which also makes the cell more difficult to excite a
cells and tissues. second time.
52 SECTION II Cell Physiology

3
Absolute Relative

Relative threshold
2

0
0 2 4 ms 6
FIGURE 6–9 The absolute and relative refractory periods. The time axis begins with the occurrence of an action potential. During the
absolute refractory period no stimulus, however large, can elicit a second action potential. During the relative refractory period a second action
potential can be elicited but it requires a larger stimulus than that in the resting state. (Modified with permission from Landowne D: Cell Physiology. New York:
Lange Medical Books/McGraw-Hill, 2006.)

MYELINATION DISEASES
Vertebrate nervous systems present a specialization of ner- There are many diseases or conditions of reduced or excessive
vous function not seen in invertebrates, namely myelination excitation of cells. Perhaps the most familiar is the conduction
(Figure 6–10). Accessory cells wrap nerve axons with many of acute pain information, which is frequently treated with
layers of their own membrane, electrically insulating most of local anesthetics; these act by blocking the NaV channels. Some
the cell. NaV channels cluster in the regions between these forms of epilepsy and some cardiac arrhythmias are also treat-
wraps, in the nodes of Ranvier. The Na current enters the ed with NaV channel blockers. One type of long-QT (LQT)
cell only at these nodes; excitation “jumps” from node to syndrome, a cardiac arrhythmia, has been linked to a mutation
node in what is called saltatory conduction. The spread be- in one of the Na+ channel genes, and a hyperkalemic periodic
tween nodes is the same passive spread seen in unmyelinated paralysis (HyperKPP) has been linked to another. Other LQT
nerve cells, but it is more effective, that is, it produces a more syndromes have been associated with KV channels.
rapid conduction velocity. The myelin wraps increase the re- Hypocalcemia is associated with increased excitability of
sistance between the axoplasm and the surrounding media, nerves and skeletal muscle and may produce uncontrollable
which, in turn, increases the length constant for passive muscle contraction (tetany). Hypercalcemia renders nerves and
spread. The myelin also increases the effective thickness, muscles less excitable. Calcium binds to the membrane near the
which decreases the effective capacitance and reduces S4 voltage sensor (Figure 3–4) of the NaV channel and has an
the amount of charge required to change the potential. Both effect similar to hyperpolarization. The positive charge on the
effects speed conduction. calcium ion repels the positively charged S4 helix, making it

FIGURE 6–10 The effect of myelination on the longitudinal spread of current. In the upper diagram Na+ is shown entering
(colored arrow) at a node of Ranvier and the associated current loops are shown in black. In an unmyelinated nerve (lower diagram) the
same current loops occur but over a shorter distance; hence, the action potential propagates more slowly. (Modified with permission from
Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 6 Action Potentials 53

more difficult for the S4 to move outward and open the channel.
The result is that, in low calcium conditions, the sodium chan-
DRUGS AND TOXINS
nel opens in response to a smaller stimulus or even spontane- After the identification of these specific Na+ and K+ conduc-
ously at the resting potential. The calcium binding does not tances, they were shown to be molecularly separate because
change the resting potential as measured with electrodes in the they differ in pharmacology and respond differently to various
bulk compartments on both sides of the membrane. drugs. Tetrodotoxin (TTX), a poison found in the internal
There are demyelinating diseases, such as multiple sclero- organs of puffer fish, selectively blocks nerve NaV channels at
sis (MS), where myelin is lost and conduction can become nanomolar concentrations. Local anesthetics such as lido-
slower or fail altogether. MS is an autoimmune disease and is caine or benzocaine also block NaV channels. There is a great-
generally treated with synthetic corticosteroids such as pred- er diversity among KV channels and also among the drugs
nisone. The symptoms can be eased by providing air condi- that block them. Tetraethyl ammonium (TEA) ions and
tioning or moving to a cooler climate. Cooling helps, some- 4-aminopyridine are among the KV channel blockers. There
what paradoxically, because although it slows the opening of are also compounds that chronically activate NaV channels,
sodium channels and thereby slows the propagation velocity, such as veratridine, pyrethroid insecticides, and brevetoxin,
it also slows the inactivation of NaV channels and increases one of the red-tide toxins.
the duration of the action potentials; thus, the greater Na+
influx makes the propagation more reliable.
Reliability is often discussed in terms of the safety factor for EXTRACELLULAR RECORDINGS—
propagation. In healthy individuals, the 100-mV action poten-
tial that arrives at the next node of Ranvier is about five times
COMPOUND ACTION POTENTIALS
larger than the 20-mV depolarization required for initiating a Action potentials can be recorded with a pair of wires placed
new impulse at that node. In patients with MS, the action po- on the surface of a nerve bundle, typically about 1 cm apart.
tential reaching the next node may be diminished to near or When a nerve impulse passes these wires, a biphasic action
below the size needed to reinitiate the impulse. One effect of potential is seen on the display (Figure 6–11). This is a
cooling nerves is to increase the safety factor for propagation. differential recording of the same nerve impulse that would

++++−−−−+++++++++++++++ ++++−−−−++++
−−−−++++−−−−−−−−−−−−−−− −−−−++++−−−−
1 1

++++++++++−−−−+++++++++ ++++ ++++−−−−


−−−−−−−−−−++++−−−−−−−−− −−−− −−−−++++
2 2

+++++++++++−−−−++++++++ +++++++++++++
−−−−−−−−−−−++++−−−−−−−− −−−−−−−−−−−−−
3 3

++++++++++++++−−−−+++++
−−−−−−−−−−−−−−++++−−−−−
4

+++++++++++++++−−−−++++
−−−−−−−−−−−−−−−++++−−−−
5 FIGURE 6–11 Externally recorded
action potentials. Left: Biphasic action
2 2 potential recorded from an intact axon.
Right: Monophasic action potential recorded
near the site of a crush injury. The potential is
measured between the two circles above each
1 3 5 1 3 diagram. The numbers on the traces indicate
the timing of the associated diagram above.
The colored region inside the nerve cell is
propagating from left to right. (Modified with
permission from Landowne D: Cell Physiology. New York:
4 Lange Medical Books/McGraw-Hill, 2006.)
54 SECTION II Cell Physiology

B
δ
1 mV 1 μV

β 10 ms 50 ms
C
γ
δ

FIGURE 6–12 A compound action potential. Left: High sweep speed. Right: Lower sweep speed, higher vertical gain. The letters refer to
specific groups of axons within the nerve. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

appear as in Figure 6–1 if the recording were made with an Vertebrate peripheral axons are classified by their diameter
intracellular microelectrode. One deflection occurs as the im- (or conduction velocity or threshold to external stimulation).
pulse passes the first wire and the second occurs as it passes There are groups of nerve fibers with similar diameters. The
the second wire. They are in opposite directions because the groups of different diameters can be distinguished as separate
two wires lead to opposing inputs for the display. If the nerve elevations in the compound action potential (Figure 6–12).
is crushed between the electrodes so that the impulse does There is some correlation of function with diameter. For exam-
not reach the second electrode, the response becomes mono- ple, large myelinated motoneurons leading to skeletal muscles
phasic. are Aα fibers and small unmyelinated fibers carrying pain in-
This type of recording with external electrodes is used formation are C fibers. The larger fibers have faster conduction
clinically to test nerve integrity. A nerve bundle can also be velocities and lower thresholds to external electrical stimuli.
stimulated with another pair of wires on a remote stretch of
the same bundle. With appropriate equipment, stimulation
and recording can be made through the skin without CARDIAC ACTION POTENTIALS
dissecting out the nerve bundle. When a nerve bundle is
stimulated, more than one axon may be excited. The electri- The heart is a pump made up of excitable muscle cells. The
cal recording of the combination of the action potentials electrical activity of these cells controls their contraction. The
produced is called a compound action potential. The com- function of these cells will be discussed further in the context
pound action potential is also biphasic if the nerve is intact of function of the heart in Chapter 23. The overall control of
between the recording wires. the heart’s pattern of contraction is by the spread of action po-
Besides being biphasic, there are many differences between tentials through a special conducting system of modified heart
compound action potentials recorded with external electrodes muscle cells (Purkinje fibers) and through the atrial and ven-
and the single-cell action potential recorded with an electrode tricular muscle cells themselves (see Figure 23–3).
inside the cell and a reference electrode outside the cell. The There are two types of action potentials in the heart distin-
compound action potentials are much smaller, on the order of guished by their rate of depolarization and their conduction
1 mV, and there is no sign of the resting potential because both velocity. The fast action potentials, with a rapid rate of depo-
wires are outside the nerve. The compound action potential is larization and a rapid propagation velocity, are found in atrial
not all-or-none because a larger stimulus will bring more indi- and ventricular muscle cells and Purkinje fibers. The slow ac-
vidual axons above threshold and the compound action tion potentials are normally found in the sinoatrial (SA) node
potential’s amplitude is proportional to the number of axons and the atrioventricular (AV) node.
firing. The compound action potential becomes smaller and
longer at greater distances from the stimulating electrodes be-
cause the conduction velocity of the various axons is not CARDIAC MUSCLE
exactly the same and the action potentials disperse as they ACTION POTENTIALS
travel away from the stimulation site.
The threshold and conduction velocity of the various ax- In cardiac muscle action potentials, current from adjacent cells
ons within a nerve bundle vary with the diameter of the depolarizes the cell to a level where fast, voltage-dependent NaV
axons. Large axons have a lower threshold to stimulation by channels open and rapidly depolarize the membrane toward
external electrodes. (Of course, in life they are usually stimu- the sodium equilibrium potential (phase 0 in Figure 6–13).
lated more selectively by a specific receptor or synaptic These channels are similar to the sodium channels of nerve and
input.) The larger-diameter fibers have a lower threshold; skeletal muscle; they open in response to depolarization. They
more of the stimulating current flows through them because are also blocked by local anesthetics. After opening, they inac-
they have a lower internal resistance. Larger axons also have tivate quickly and the membrane potential starts to return.
a more rapid conduction velocity, again because of their low- However, the depolarization also opens voltage-activated
er internal resistance. L-type CaV channels that do not inactivate. This maintains the
CHAPTER 6 Action Potentials 55

1 The description above is a simplified view of cardiac muscle


2 action potentials. The complete story has several more K chan-
Vm nels and must account for differences among muscle action
50 mV potentials in different regions of the heart as well as age-related
0 changes. There are two KV channels that open transiently just
3 50 ms
after the NaV channels and produce the initial partial repolar-
ization (phase 1) from the peak to the plateau (IKto). There are
4 at least two different slow voltage-dependent K channels with
IK similar kinetics but distinct pharmacology (IKR and IKS). Some
cardiac muscle cells have T-type calcium channels. In all car-
diac cells, some current is carried by the sodium–calcium
ICa exchanger and by the Na/K pump.
The regional and age-related differences in the action po-
1 mA/cm2
tentials are functionally and clinically important. The ventric-
INa
ular muscle’s action potentials near the endocardial (inner)
FIGURE 6–13 A ventricular muscle cell action potential surface have a longer duration than those near the epicardial
(upper trace) and its underlying ionic currents. The INa and ICa (outer) surface. More work is done by the inner fibers, and
currents are inward and the IK current is outward. (Modified with they are more likely to be damaged in a heart attack. These
permission from Landowne D: Cell Physiology. New York: Lange Medical Books/ differences must arise because of a different balance of Na, Ca,
McGraw-Hill, 2006.)
and K channel activities. The interactions between the effects
of different channels are complex and are best explored with
computer models. Clearly more research is necessary to un-
action potential in the plateau phase (phase 2). Reducing derstand the details.
external Ca2+ concentration or adding drugs that block calcium
channels will reduce the plateau phase and also reduce the
strength of muscle contraction. Cardiac muscle, unlike skeletal SA AND AV NODE ACTION POTENTIALS
muscle, requires external Ca2+ for contraction (Figure 6–13).
Cardiac muscle cells also differ from nerve and skeletal The overall control of the heart’s pattern of contraction is nor-
muscle by lacking the fast KV channel for quick repolarization. mally initiated by action potentials that spontaneously arise
The potassium conductance system of the heart is rather com- 60–80 times/min from modified muscle cells in the SA node.
plex; at least five different components have been identified on Similar action potentials are also seen in the AV node, where
the basis of their kinetics and voltage dependence. Two of they regulate the activation of the ventricles. In the absence of
these are important to understand the plateau phase. During stimulation from the atria, the AV node’s cells spontaneously
the plateau phase, the conductance is less than that during produce about 40 action potentials/min; in healthy hearts, how-
diastole, the period between action potentials. This is because ever, the atrial cells drive them at the rate set by the SA node.
of the inward rectifier channel (Kir), which is responsible for The action potentials in the nodes lack the rapid upstroke
maintaining the resting potential and has a high conductance and do not have as pronounced a plateau phase as the cardiac
near and below the resting potential (at more negative poten- muscle action potentials. They are further characterized by the
tials); it does not conduct during the plateau phase when the slow depolarization between action potentials: the pacemaker
membrane is depolarized. potential. These cells fire rhythmically; they are never at rest
The Kir channel rectifies, allowing current to flow and main- and have no true resting potential.
tain the resting potential, but it does not allow much current to The upstroke of the action potential is produced by a slow
flow out during depolarization. The rectification is caused by inward current carried primarily by Ca2+ (Figure 6–14). There
Mg2+ or other polyvalent cations from the internal solution is an initial phase through T-type CaV channels and a major
moving into the channel and plugging it when the cell is depo- phase through L-type CaV channels. The T-type channels are
larized. The low conductance to K+ during the plateau phase transient and have a low threshold for opening, near –60 mV.
means that the modest conductance to Ca2+ through the CaV The L-type channels are long-lasting and have a higher thresh-
channels maintains the membrane potential at depolarized old, near –30 mV. The L-type channels are similar to the CaV
levels during the plateau. channels that maintain the plateau of the cardiac muscle ac-
Slow KV channels open very slowly during the action po- tion potentials; they are blocked by dihydropyridines. T-type
tential and are responsible for the downward slope during channels have a different pharmacology. Reducing external
the plateau phase. When the membrane potential falls below Ca2+ concentration or adding Ca2+ channel blockers reduces
a certain level, the CaV channels close and the repolarization the amplitude of the node’s action potentials. Outward K+ cur-
toward the potassium equilibrium potential accelerates rent gradually replaces the slow inward current and the cells
(phase 3). Since the membrane is no longer depolarized, the repolarize toward EK. As the potential passes –50 mV, an
KV channels close. inward hyperpolarization-activated current, If , appears,
56 SECTION II Cell Physiology

Vm
50 mV

50 ms

IK

If

1 mA/cm2
ICa

FIGURE 6–14 The SA node’s action potentials (upper trace) and their underlying currents. The If and ICa currents are inward and
the IK current is outward. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

competes with IK, and eventually begins to depolarize the cell depolarize between action potentials and can temporarily stop
again. If is carried mainly by sodium ions. When the potential the heart.
again passes –60 mV, the CaV channels are again activated and
the cycle is repeated.
NOREPINEPHRINE ALSO
INCREASES CONTRACTILITY
EFFECTS OF SYMPATHETIC
In the presence of NE, the plateau of the muscle action po-
AND PARASYMPATHETIC tentials is elevated and has a shorter duration (Figure 6–15).
INNERVATIONS This shortening of the action potential shortens the duration
of the muscle contraction, which is functionally important
The heart can beat spontaneously without neural input. In for the heart. At high heart rates, the time required to refill
healthy individuals, however, the autonomic nervous system the heart limits its performance. By reducing the time that
and circulating hormone levels regulate the heart rate and its muscle force is being generated (systole), more time is left
strength of contraction. The autonomic nervous system for filling (diastole). The shortening of the ventricular ac-
controls many internal organs through its two divisions, the tion potentials can be seen in the ECG as a shortening of the
sympathetic and the parasympathetic nervous systems. These QT interval.
release norepinephrine (NE) and acetylcholine (ACh), re- NE increases the amplitude of the plateau by causing the ac-
spectively, into the heart. The autonomic nervous system can tion potential to open more L-type Ca2+ channels. This drives
also cause the adrenal medulla to release epinephrine into the the membrane closer to the Ca equilibrium potential. The
blood. Epinephrine has effects on the heart similar to those of increased Ca influx leads to a greater strength of contraction
NE. Some of the details about the autonomic synapses and by a mechanism described in Chapter 10. NE shortens the
their pharmacology are described in the next chapter.
The cells in the SA and AV node cells have GPCRs that pro-
duce a stimulation (via Gαs) or inhibition (via Gαi) of adenylyl
cyclase, which, in turn, raises or lowers cAMP levels in re- NE elevates plateau
sponse to NE and ACh, respectively. The cAMP enhances the
activity of the If channels. The end result is that NE increases If NE shortens duration
and thus depolarizes the cells more rapidly and increases the
heart rate. ACh reduces If , slows the rate of depolarization, and
reduces the heart rate (see Figure 23–4). Changing If also leads 50 mV
to a speeding or slowing of conduction through the AV node. 50 ms
These effects are discussed further in terms of heart function
in Chapter 23.
High ACh levels lead to the opening of another potassium FIGURE 6–15 The effects of norepinephrine on ventricular
channel (KACh). (It is a G protein–activated inward rectifier muscle cells’ action potentials. (Modified with permission from Landowne
GIRK channel.) This channel further reduces the tendency to D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 6 Action Potentials 57

duration by making the KV channels open more rapidly. The


effects on the K and Ca2+ channels are mediated via cAMP It is caused by mutations of the NaV channel of skeletal
acting as a second messenger, stimulating protein kinase A muscle that make them slowly inactivating; the myotonia
(PKA) and phosphorylating the channels. This pathway also results from abnormal reopenings of the NaV channels.
enhances the calcium reuptake mechanism by phosphorylat- Moderate elevation of extracellular potassium favors ab-
ing phospholamban. This speeds up muscle relaxation. errant gating with persistent and prolonged reopenings.
The Na current through these channels may cause skeletal
muscle weakness by depolarizing the cells, thereby inacti-
ACETYLCHOLINE REDUCES vating normal NaV channels, which are then unable to
ATRIAL CONTRACTILITY generate action potentials. Patients with HyperKPP are at
increased risk for malignant hyperthermia induced by
The ACh-activated K channel (KACh) remains open during anesthesia during surgery. Other NaV channel mutations
the action potentials; in atrial muscle and Purkinje fibers, it in heart muscle are linked to sudden death syndromes.
makes the plateau phase shorter and lower. The atrial contrac- Attacks can be stopped by ingesting a high sugar load
tions are weaker. ACh receptors are relatively sparse on ven- or by thiazide diuretics, both of which reduce extracellu-
tricular muscle cells. lar potassium. They can be prevented by a diet low in po-
tassium and high in carbohydrates, and also with thiaz-
ides. The disease is a lifelong condition.

CLINICAL CORRELATION
Since early childhood, a 42-year-old woman experienced
stiffness of her muscles, particularly when loosening a
CHAPTER SUMMARY
tight handgrip or starting to walk. Cold exposure exacer- ■ Depolarization opens NaV channels, which allows Na+ to rush
in and produce further depolarization. This positive feedback
bated these symptoms. Outdoors on a cold and windy
loop produces the all-or-none quality and the propagation of
day, her face stiffened in a grimace and she could not open
action potentials.
her eyes or move them from side to side. These symptoms
■ K+ leaving the cell repolarizes the membrane potential and
disappeared within a few minutes after she had entered a
terminates action potentials.
warm room. When she ate ice cream, her throat stiffened
■ Voltage clamping, or negative feedback control of the
and she could not swallow. From age 16, she also had at-
membrane potential, facilitates understanding of the
tacks of generalized weakness unrelated to cold. Some- currents underlying the action potential.
times she woke up at night severely paralyzed. She was
■ The amplitude and direction of the sodium current vary with
more liable to have an attack when hungry. During preg- the amplitude of voltage-clamp steps in membrane potential.
nancy she had daily attacks of weakness; within a few days
■ Depolarizing steps first activate and then inactivate Na+
after delivery she improved. current. They also activate K+ current after a delay.
A neurologist performed diagnostic testing. The pa-
■ The gating current is a direct sign of the conformational
tient was given 60 mEq of potassium orally with a mixture changes in the sodium channel proteins.
of anions. Forty-five minutes later, she was so stiff that she
■ There is a threshold for action potential initiation.
could make no quick movements. About an hour later,
■ Following an action potential, excitable cells have an absolute
she noticed increasing weakness and had to lie down. The
refractory period when they will not produce a second action
paralytic attack reached its peak roughly half an hour lat- potential and then a relative refractory period when a larger
er. At that time, she could not lift her head, arms, or legs, stimulus is required to produce a second action potential.
nor could she move her limbs on the examination table. ■ Myelination increases conduction velocity by increasing the
Myotonia (difficulty of relaxing muscles) of her facial and length constant.
extraocular muscles was intense. Respiration was mildly ■ Hypocalcemia (low extracellular calcium) makes excitable cells
impaired. Reflexes were unchanged and sensation was more excitable.
normal. Improvement started half an hour later and was ■ Demyelinating diseases slow the conduction velocity and may
complete 3.5 hours from the start. Before, during, and af- block the propagation of action potentials.
ter, her serum potassium values were: 4.5, 7.3, and 3.9 ■ Action potentials appear differently when they are recorded
mEq/L (normal is 3.5–4.5 mEq/L). with a pair of wires placed on the outside of a nerve bundle.
This disease also affected her son, sister, mother, mater- Compound action potentials, the sum of many externally
nal aunt, and maternal grandfather. The inheritance was recorded action potentials, have properties that differ from
due to a single autosomal, dominant gene with probably those of single action potentials recorded with intracellular
complete penetrance. electrodes.
This patient was suffering from familial HyperKPP, ■ In the heart, action potentials arise automatically in the SA
which occurs in approximately 1 in every 200,000 people. node and then spread from cell to cell over the heart via gap
junctions.
58 SECTION II Cell Physiology

■ Cardiac muscle cells have KIR channels to maintain the resting 4. There is an inward current (If ) associated with pacemaker
potential, NaV channels for the upstroke of the action potential, activity in cells of the sinoatrial node. Stimulation of sympathetic
CaV channels for the plateau phase, and slow KV channels for nerves leading to the heart or application of norepinephrine
the repolarization. produces
■ SA node cells use CaV channels for the upstroke of the A) a decrease of If , a decrease in heart rate, and an increase in
action potential, KV channels for the repolarization, and strength of contraction.
a hyperpolarization-activated If channel to produce the slow B) a decrease of If , an increase in heart rate, and an increase
“pacemaker” depolarization between action potentials. in strength of contraction.
■ ACh and NE slow or speed the heart rate, respectively, C) an increase of If , an increase in heart rate, and an increase
via G protein–coupled receptors, which leads to a decrease in strength of contraction.
or increase in If. D) an increase of If , a decrease in heart rate, and a decrease in
■ NE increases the amplitude of the plateau and decreases the strength of contraction.
duration of ventricular muscle action potentials. E) an increase of If , an increase in heart rate, and a decrease
in strength of contraction.
5. Propagation of a nerve impulse does not require
STUDY QUESTIONS A) closure of potassium channels that maintain the
resting potential.
1. Hyperkalemia (high extracellular potassium concentration) can B) a conformational change in membrane proteins.
stop the heart because C) a membrane depolarization that opens Na+ channels.
A) potassium ions bind to sodium channels, preventing their D) current to enter the axon and flow within the axon.
activity. E) entry of sodium ions into the axon.
B) potassium ions stimulate the sodium–potassium pump and 6. The compound action potential recorded with a pair
thereby prevent cardiac action potentials. extracellular electrodes from an intact bundle of nerve fibers
C) the membrane potential of heart cells depolarizes and its A) propagates without change in size or shape.
sodium channels inactivate. B) is all-or-none. If a threshold is exceeded, further increase
D) potassium ions rush out through the inward rectifier. in stimulus does not increase the response.
E) potassium ions block the actin–myosin interaction C) has an amplitude of about 100 mV.
in the heart. D) is biphasic, showing both upward and downward
2. Myelination of axons deflections from the baseline.
A) reduces conduction velocity to provide more reliable E) is not blocked by tetrodotoxin (TTX).
transmission.
B) forces the nerve impulse to jump from node to node.
C) occurs in excess in multiple sclerosis (MS).
D) leads to an increase in effective membrane capacitance.
E) decreases the length constant for the passive spread of
membrane potential.
3. Consider the following three channels in ventricular muscle
cells: sodium channel (NaV), inward rectifier potassium channel
(Kir), and calcium channel (CaV). Choose the answer that best
describes which of these channels is open during the plateau
phase of the ventricular action potential.
A) all three
B) NaV and Kir only
C) CaV and Kir only
D) Kir only
E) CaV only
7
C H A P T E R

Synapses
David Landowne

O B J E C T I V E S

■ Describe the steps in chemical synaptic transmission.


■ Describe the biosynthesis and actions of acetylcholine, catecholamines
(dopamine, norepinephrine, epinephrine), serotonin, histamine, and
excitatory and inhibitory amino acids.
■ Describe the biosynthesis and actions of neuropeptides.
■ Describe the structure of the neuromuscular junction and the functions
of the various substructures.
■ Describe and explain the steps involved in neuromuscular transmission.
■ Describe the actions and explain the mechanisms for the effects of Ca2+
and Mg2+ on transmitter release.
■ Describe how acetylcholine interacts with receptors on the postsynaptic
membrane and the fate of the acetylcholine.
■ Describe the generation of the endplate potential and the effects and
mechanisms of action of acetylcholine esterase inhibitors and blockers
of acetylcholine receptors.
■ Describe facilitation and posttetanic potentiation of transmitter release and
how these processes can be used to explain certain features of myasthenia
gravis and recovery from receptor blockade.
■ Describe the structures and explain the functions of the various parts
of neurons.
■ Describe transport of materials up and down axons (orthograde and
retrograde axonal transport) including mechanisms and materials.
■ Calculate the time required for the regeneration of peripheral nerves.
■ Describe the differences and similarities between synaptic transmission
at a central synapse and at neuromuscular junctions.
■ Describe the generation of IPSPs and EPSPs by ionotropic and metabotropic
receptors.
■ Describe the integration of information and repetitive firing in neurons
and the concept of presynaptic inhibition.

INTRODUCTION synaptic cleft and binds to a receptor on the postsynaptic


cell. The postsynaptic receptor may be ionotropic, in
A synapse is a specialized region where a neuron communi- which case it will open a selective pore and allow ions to flow
cates with a target cell: another neuron, a muscle cell, or a to produce a postsynaptic potential (PSP), or it may be
gland cell. Most synapses are chemical; the presynaptic neu- metabotropic and inform a G protein to initiate a chemi-
ron releases a transmitter substance that diffuses across the cal cascade, which may include the opening or closing of

59

Ch07_059-078.indd 59 11/26/10 9:41:14 AM


60 SECTION II Cell Physiology

channels. A few synapses are electrical; current passes through Neurotransmitters can be chemically classified into five
cell–cell channels directly into the postsynaptic cell. Chemical groups (Figure 7–2). They are all hydrophilic and contain
synapses offer the possibility of amplification, signal inversion, groups that are charged at physiologic pH. Thus, they do not
and persistent effects; electrical synapses are faster and seem readily pass through lipid membranes and can be compart-
to be used when synchronization is more important than com- mentalized as needed.
putation (information processing).
Chemical synapses may be excitatory or inhibitory, depend-
ing on their effect on the postsynaptic cell. In the CNS, neu- ACETYLCHOLINE
rons receive both types of synapses and integrate the
information coming into them before sending the processed Acetylcholine (ACh) was the first recognized transmitter. It is
message on to another cell. Chemical synapses are major phar- used by spinal motoneurons to excite skeletal muscles; by the
maceutical targets. parasympathetic nerves to communicate with various end or-
gans, including the vagus nerve slowing pacemaker regions of
the heart; in sympathetic and parasympathetic ganglia; and in
PRESYNAPTIC PROCESSES various locations in the CNS. There are two classes of postsyn-
aptic ACh receptors (AChRs), which are named for other ag-
The presynaptic terminal must provide for the synthesis, pack- onists that can also bind to them. Nicotinic AChRs are at
aging, and release of the various transmitters (Figure 7–1). neuromuscular junctions, sympathetic and parasympathetic
The nonpeptide transmitters are concentrated inside vesicles ganglia, and in the CNS. Nicotinic AChRs are ionotropic re-
by specific H/transmitter cotransporters. A V-type H+ pump, ceptors or heteromeric pentamers (see Figure 3–5). They are
which consumes ATP, produces the H+ gradient. The trans- chemosensitive channels that are opened by nicotine and
mitter concentration inside the vesicle can be quite high, on blocked by curare. Muscarinic AChRs occur in the heart,
the order of 20,000 molecules in a 20-nm radius sphere, or smooth muscles, gland cells, and CNS. They are metabotropic
about 30 mM. 7-TM GPCRs that are activated by muscarine and blocked by
After release, transmitters are degraded or transported back atropine. nAChRs tend to excite the postsynaptic cell;
into the presynaptic terminal for reuse. The vesicular mem- mAChRs may be excitatory or inhibitory.
branes are also recycled. Some transmitters are small polypep- ACh is synthesized from acetyl-CoA and choline by the en-
tides that are synthesized on rough endoplasmic reticulum zyme choline acetyl transferase (CAT), found in the presyn-
near the nucleus, packaged by the Golgi apparatus, and then aptic cytoplasm. ACh is concentrated into vesicles by an H/
transported in vesicles the length of the axon by an active ACh cotransporter (Figure 7–3). A Ca2+-activated process re-
process called axoplasmic transport. This process also brings leases the vesicles. It is described below, after all of the trans-
other proteins to the presynaptic terminals. mitters have been discussed.

Fill

Dock

Fuse and release

Recycle

FIGURE 7–1 Synaptic vesicle docking, releasing of


contents, and recycling. (Modified with permission from Landowne
D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 7 Synapses 61

Cholinergic Biogenic Amines Amino Acids

O COOH
+ HO
N(CH3)3 NH2
O
HO HOOC NH2

Acetylcholine Dopamine Glutamic acid

HO
NH2
Peptidergic HO HOOC NH2
OH

Endorphin Norepinephrine γ-aminobutyric acid


Enkephalin (GABA)
Dynorphin
HO
Calcitonin gene N(CH3)
related peptide (CGRP) HO
Substance Y HOOC NH2
Substance P OH
Vasopressin (ADH) Epinephrine Glycine
Oxytocin
Cholecystokinin (CCK)
Vasoactive intestinal
peptide (VIP) HO NH2 Purinergic

ATP
NH

Serotonin Adenosine

NH2
HN
N
FIGURE 7–2 The neurotransmitters.
NH
N (Modified with permission from Landowne D: Cell Physiology.
Histamine New York: Lange Medical Books/McGraw-Hill, 2006.)

Poisoning by botulinum toxin (Botox) blocks ACh release gases are organofluorophosphates that irreversibly bind
and results in a failure of neuromuscular transmission. AChE but can be displaced by pyridine aldoxime methyl
Recently Botox injections have been used to treat dystonia, a iodide (PAM).
movement disorder characterized by involuntary muscle con-
tractions, and cosmetically to locally block facial muscles that
wrinkle the skin. Excessive doses or systemic toxin delivery AMINO ACIDS
from contamination during food canning can lead to death.
Black (or brown) widow spider venom (BWSV) also blocks GLUTAMATE
neuromuscular transmission. It makes the presynaptic mem-
branes permeable to Ca2+ and causes a massive release of vesi- Glutamate is the major excitatory neurotransmitter of the
cles, followed by a failure of transmission due to the lack of CNS. It is a nonessential amino acid, but, because it cannot
stored ACh. pass the blood–brain barrier, it must be synthesized in the
After release, ACh may be broken down into acetate and CNS. There are several synthetic pathways but none specific
choline by acetylcholinesterase (AChE) in the extracellu- for neurons. Ionotropic glutamate receptors, gluRs, are classi-
lar space. A Na/choline cotransporter recaptures most of fied as NMDA-type if the synthetic agonist N-methyl-d-
the choline; then ACh is resynthesized by CAT and repack- aspartate activates them or non-NMDA-type if it does not.
aged. AChE inhibitors or anticholinesterases are used for Both types are heteromeric tetramers (see Figure 3–6) and
medical purposes, as insecticides, and as nerve gases in permit the passage of Na+ and K+, but the NMDA gluRs also
chemical warfare. Their effect is to increase the amount and allow Ca2+ to enter the cell and have a special role in synaptic
duration of ACh interaction with the postsynaptic recep- plasticity, described later. There are also metabotropic gluRs
tors. The battlefield countermeasures are to block the post- (mgluRs). All of these are normally activated by glutamate
synaptic receptors on the heart with atropine. The nerve (Figure 7–4).
62 SECTION II Cell Physiology

Glia

Cholinesterase

H+ Muscarinic
GPCR
ATP
ACh
Nicotinic
Presynaptic channel

Choline
ACh Cholinesterase
Na
ATP
Postsynaptic

K Na
FIGURE 7–3 A generalized schematic acetylcholine synapse. The presynaptic terminal has transporters for choline reuptake and ACh
packaging. The postsynaptic membrane has ACh receptors and cholinesterase. The nearby glial cell membrane also has cholinesterase. ACh is
hydrolyzed by the esterases and the choline part is taken back by the presynaptic terminal. (Modified with permission from Landowne D: Cell Physiology.
New York: Lange Medical Books/McGraw-Hill, 2006.)

Glutamate is removed from the extracellular space by a Na/ membrane and nearby glial cell membranes. Inside the glia,
glu cotransporter, the excitatory amino acid transporter glutamate can be converted to glutamine, released, and taken
(EAAT), which also countertransports K+. EAATs are in the up by the presynaptic terminal by a Na/Cl-coupled cotrans-
presynaptic terminal membrane and also in the postsynaptic porter and finally reconverted into glutamate.

Na glu K
gln
Na gln
Glia

mgluR
H+ GPCR

ATP
Ionotropic
channel
Presynaptic

K
glu glu
ATP Na
Postsynaptic

K Na
FIGURE 7–4 A generalized schematic glutamate synapse. In addition to the reuptake and packaging transporters, glutamate synapses
have uptake transporters in the postsynaptic and glia cell membranes. There is also a glutamine (gln) pathway to move glutamate in the glial cell
back to the presynaptic terminal. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 7 Synapses 63

Na GABA Cl

Glia

GABAB
H+ GPCR
ATP

GABAA
Presynaptic channel

Cl
GABA GABA
ATP Na
Postsynaptic

K Na
FIGURE 7–5 A generalized schematic GABA synapse. This is similar to the glutamate scheme but has GABA receptors and transporters.
(Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

Excess extracellular glutamate kills neurons by allowing


excess Ca2+ into the cells, which can lead to necrosis (cell
BIOGENIC AMINES
death). This neurotoxicity is postulated to play a role in The catecholamines, serotonin, and histamine are all biogenic
ischemic stroke, amyotrophic lateral sclerosis (ALS), amines. The catecholamines are dopamine, norepinephrine
Huntington’s disease, Alzheimer’s disease, and possibly (NE), and epinephrine (EPI). Most of the effects produced by
some forms of epilepsy. Ischemia can raise extracellular these biogenic amines are via GPCRs, often without producing
glutamate by limiting oxidative metabolism, ATP, and so- PSPs. All are concentrated into vesicles and released by similar
dium gradients, thus the movement of glutamate away from mechanisms, but some are released by nerve swellings, which
the receptors. are in the vicinity of the receptors but not as closely apposed (see
Figure 7–19). Non-nerve cells also release EPI and histamine.

GABA AND GLYCINE


CATECHOLAMINES
Gamma-aminobutyric acid (GABA) and glycine are the
major inhibitory neurotransmitters in the CNS. Glutamate Dopamine and NE are found in the CNS. NE is the principal
decarboxylase (GAD) converts glutamate into GABA in the final transmitter of the sympathetic nervous system and EPI is
presynaptic terminal cytoplasm. GABA is packaged and made and released by the adrenal medulla. All three are syn-
released as other transmitters (Figure 7–5). There is a Na/ thesized by the same pathway, starting with the hydroxylation
GABA cotransporter that removes GABA from the synaptic of tyrosine to dihydroxyphenylalanine (DOPA), which is then
cleft. GABAA receptors and glycine receptors are pentam- decarboxylated to form dopamine. Adding a beta-hydroxyl
eric heteromers in the nAChR superfamily; they are perme- group forms NE and, in the adrenal medullary cells, a subse-
able to Cl– ions. GABAB receptors are GPCRs that activate quent transfer of an N-methyl group forms EPI. Tyrosine hy-
Kir (or GIRK) channels. droxylase (TH) is the rate-limiting enzyme. TH and DOPA
The CNS operates with a tonic level of inhibition that can be decarboxylase are in the presynaptic terminal cytoplasm.
shifted with various drugs. Muscimol, from the mushroom Dopamine is concentrated into vesicles, where dopamine
Amanita muscaria, is a potent GABAAR agonist. Common beta-hydroxylase (DBH) converts it to NE. NE is taken back
tranquilizers such as diazepam (Valium) and barbiturates into the presynaptic terminal by a Na/Cl-coupled cotrans-
such as phenobarbital enhance the opening of GABAARs. porter; there, it is broken down by monoamine oxidase
Picrotoxin, a potent convulsant, blocks GABAAR. Strychnine, (MAO) in mitochondria and by catecholamine-O-methyl
also a convulsant, blocks glyRs. Tetanus toxin produces a transferase (COMT) in the cytoplasm.
spastic paralysis by blocking the release mechanism for GABA Catecholamine receptors are GPCRs and are found in the
and glycine. CNS, smooth muscle, and heart. Adrenergic receptors respond
64 SECTION II Cell Physiology

to NE and/or EPI. There are two categories of adrenergic recep- N-methyl transferase. There are four different histamine re-
tors: alpha-adrenergic receptors have a higher affinity for NE ceptors, which are all GPCRs.
and beta-adrenergic receptors have a higher affinity for EPI.
However, there is cross-reactivity, and both receptors will re-
spond to higher concentrations of both agonists. In the cardio-
vascular system, the alpha receptors are primarily found on the
PURINES
smooth muscle cells that control the diameter of small blood ATP is contained in synaptic vesicles and released with NE in
vessels; NE acts to constrict these vessels. The beta receptors are sympathetic vasoconstrictor neurons. It induces constriction
primarily in the heart and can make it beat faster and harder. when applied directly to the smooth muscles. P2X ATP recep-
Muscle relaxation via adrenergic receptor activation occurs in tors are ion channels that permit the entry of Ca2+, and the
smooth muscle cells in the gut and the lungs. Some of these cells also have P2Y GPCRs. These receptors are also in the
functions are discussed at greater length in the next section. brain, as well as P1 receptors for adenosine.
Parkinson’s disease is characterized by the loss of dopamin-
ergic neurons; its treatment often includes DOPA, which can
partially relieve the symptoms. Drugs that block dopamine re-
ceptors have been used to treat schizophrenia; sometimes they
PEPTIDES
induce Parkinson-like tremors. Reserpine, an early tranquil- Neuropeptides are small polypeptides that are synthesized as
izer, inhibits dopamine transport into vesicles. Cocaine blocks larger inactive precursors (propeptides) and then cleaved out
the reuptake of catecholamines, prolonging their actions. by specific endopeptidases. As they are proteins, they are
Many over-the-counter remedies for nasal congestion, such as synthesized in the cell body and transported in vesicles to the
neosynephrine, activate catecholamine receptors. terminals. There is no reuptake mechanism. Peptides are less
concentrated than other neurotransmitters in vesicles but have
higher affinity for their receptors, which are GPCRs. Neuro-
SEROTONIN peptides are released from large dense-core vesicles, while
other neurotransmitters are secreted from smaller, clearer ves-
Serotonin, or 5-hydroxytryptamine (5-HT), is made from
icles. Neuropeptides often act in concert with classic neu-
tryptophan by hydroxylation and decarboxylation. 5-HT re-
rotransmitters.
ceptors function in the gut in secretion and peristalsis, medi-
Not much is known about the function of most neuropeptides
ate platelet aggregation and smooth muscle contraction, and
in the CNS except the opiate peptides, endorphin, enkephalin,
are distributed throughout the limbic system of the brain. Se-
and dynorphin, which are involved in the regulation of pain
rotonin was initially identified as a substance in blood serum
perception. Three opiate receptors have been identified, initially
that constricted blood vessels, hence the name.
as the sites that bind synthetic opiates such as morphine.
Tryptophan hydroxylase is the rate-limiting step of 5-HT
There are many nonopiod peptides released from neurons.
synthesis; in the CNS tryptophan hydroxylase is present only
The calcitonin gene–related peptide (CGRP) and sub-
in serotonergic neurons. 5-HT is deactivated by reuptake and
stance Y are involved in maintaining blood pressure. Antidi-
then broken down by MAO in mitochondria. Most 5-HT re-
uretic hormone (ADH, also called vasopressin) helps control
ceptors are GPCRs; 5-HT3 receptors are ion channels.
water reuptake in the kidney. Oxytocin, luteinizing hormone
Selective serotonin reuptake inhibitors such as fluoxetine
(LH), and follicle-stimulating hormone (FSH) are involved
hydrochloride (Prozac) are commonly prescribed as antide-
in reproduction. Cholecystokinin (CCK), gastrin, and vaso-
pressants. Lysergic acid diethylamide (LSD) and psilocin,
active intestinal peptide (VIP) facilitate digestion. All of these
the active metabolite of psilocybin, are hallucinogens that
and many more have been identified as potential neurotrans-
activate 5-HT receptors.
mitters in the CNS.

HISTAMINE
SYNAPTIC RELEASE
Most histamine in the body is released from mast cells (part of
the immune system) in response to antigens or tissue injury. The details of the synaptic release process are currently under
Histamine also is a regulator of acid secretion in the gut and active investigation. It is clear that the process is triggered by
acts as a neurotransmitter in the central nervous system. His- an increase in cytoplasmic Ca2+ levels. At many synapses, when
tamine release is associated with allergic reactions; it initiates a presynaptic action potential arrives, the Ca2+ enters the ter-
inflammatory responses, dilates blood vessels and increases minal through CaV channels. In some small sensory cells there
capillary permeability, decreases heart rate, and contracts is no action potential and the sensory generator potential
smooth muscles in the lung. Enterochromaffin-like cells in the opens the CaV channels.
gastric mucosa also release histamine, which promotes acid Synaptic vesicles cycle through loading with transmitters,
production. Histamine is made from histidine, stored in docking at an active zone or release site, fusion with the sur-
vesicles, and released; it is then broken down by histamine face membrane and release of contents, endocytotic recovery,
CHAPTER 7 Synapses 65

Na
Dock

Ca

Fuse and release

Recycle

FIGURE 7–6 The channels involved in synaptic release. NaV channels depolarize the ending and CaV channels permit Ca2+ influx to trigger
release. There are also KV channels that repolarize the membrane and thereby limit Ca2+ influx. (Modified with permission from Landowne D: Cell Physiology.
New York: Lange Medical Books/McGraw-Hill, 2006.)

and then loading again. In Figure 7–6, each step in the vesicu- Docking and fusion involves the SNARE or soluble N-
lar cycle is illustrated by a shift in the position of the vesicle. In ethylmaleimide-sensitive factor attachment protein (SNAP)—
reality, however, there is little movement in the attached states. receptor proteins that are present on both membranes before
In many synapses, the release site is across from a postsynaptic fusion and associate into tight core complexes during fusion.
area containing the channels that are sensitive to the transmit- Figure 7–7 shows the vesicular v-SNARE synaptobrevin bind-
ter. In the neuromuscular junction (see Figure 7–9), CaV chan- ing the target t-SNARE syntaxin and SNAP-25. Synaptobrevin
nels are adjacent to the release site, so that internal Ca2+ need is the substrate of the endopeptidases contained in botulinum
only be elevated locally to cause release. and tetanus toxins.

Synaptotagmin

Synaptobrevin
v-SNARE
Ca

Syntaxin
t-SNARE
FIGURE 7–7 A possible mechanism of vesicle fusion. SNARE proteins dock the vesicle and Ca2+ binds to synaptotagmin to cause fusion.
(Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
66 SECTION II Cell Physiology

Ca-stimulated fusion requires the Ca2+-binding protein syn- membrane. During development, it can also deliver cell adhe-
aptotagmin, which is in the vesicular membrane and binds sion molecules that recognize or induce targets. Retrograde
Ca2+. A proposed model suggests that Ca2+ allows the synap- transport can return damaged proteins for the endolytic path-
totagmin to bind the surface membrane and pull the two lipid way and bring information about signaling events back to the
layers together. cell body.
The recycling process returns the lipids and proteins to the Retrograde transport is part of the pathophysiology of several
vesicle pool. The vesicle is reformed as a clathrin-coated pit. diseases including polio, rabies, tetanus, and herpes simplex.
The clathrin molecules have the shape of a triskelion, or three The herpesvirus enters peripheral nerve terminals and then trav-
bent legs. The clathrin forms a closed surface covered with els back to the cell body, where it replicates or enters latency. It
pentagons and pinches the recovered vesicle off the surface. can later return to the nerve ending by orthograde transport and
make itself available for contact transmission to another person.
The tetanus toxin is transported retrogradely in motoneurons to
AXOPLASMIC TRANSPORT the dendrites and then transsynaptically to GABA- and glycine-
releasing terminals, where it inhibits synaptic release.
All of the proteins in the presynaptic terminal are synthesized in Axoplasmic transport is important for the regeneration of
the cell body and transported perhaps 1 m before they are use- nerves following injury in the peripheral nervous system. Un-
ful. In addition, the neuron has mechanisms that transport some der usual circumstances, nerves in the CNS do not regenerate,
materials in the reverse or retrograde direction back to the cell although current researchers are hopeful that this will change
body. Some of the mechanisms used for this transport are used in the future. If a peripheral nerve axon is cut or crushed, the
in other cells to deliver material to the periphery of the cell and distal portion will die and go through a characteristic Walle-
also for the movement of chromosomes during mitosis. rian degeneration as the axon is resorbed over a few weeks.
Axoplasmic transport is distinguished by the direction into Within a few days, the cell body undergoes the axon reaction,
orthograde and retrograde. Orthograde transport can be fur- often called chromatolysis, because of a change in staining
ther divided into fast (100–400 mm per day or 1–5 μm/s) and when it is studied histologically. The nucleolus enlarges, the
slow (0.5–4 mm per day). Fast transport is for vesicles and mi- rough endoplasmic reticulum, or ER (Nissl substance), dis-
tochondria; slow transport is for soluble enzymes and those perses, and the nucleus is displaced. Genes have been activat-
that make up the cytoskeleton. Retrograde transport is only of ed, RNA transcribed, and proteins synthesized. The longer the
the fast type. distance from the injury to the cell body, the greater the la-
Fast axoplasmic transport involves molecular motors that tency, indicating that retrograde transport is involved in the
hydrolyze ATP and walk along microtubules, long hollow cyl- signaling to initiate the axonal reaction.
inders 25 nm in diameter. Two different classes of motors are At the site of injury, the end that is coupled to the cell body
used, kinesins for orthograde transport and dyneins for retro- will reseal in hours and buds or sprouts will appear in a day or
grade. Microtubules are polarized, and these motors can sense two. The cut tip swells with mitochondria and smooth ER. The
the polarity and move by 8-nm steps in the appropriate direc- sprouts grow out as thin fibers. If the regeneration is success-
tion. The motors have two “feet,” or sites of interaction with ful, one of the new fibers finds its way down the sheath of the
the microtubules, and exhibit processivity, or the ability to distal degenerating nerve and reinnervates a postsynaptic tar-
function repetitively without dissociating from their substrate, get. The fiber will then increase in diameter and become remy-
the microtubule. Accessory molecules are used to attach the elinated. The rate of fiber growth is about 1 mm per day, in the
payload to the motor (Figure 7–8). range of slow axonal transport. This is the number to use in
Fast orthograde transport delivers the membrane proteins estimating recovery times.
needed in the terminal for both the vesicles and the terminal In addition to the microtubule-based systems, intracellular
transport can also occur via myosin motors traveling along ac-
Orthograde
tin filaments. The interaction is similar to that described in the
section Chapters 8 and 9 except that the actin stays fixed and
individual myosin molecules process along it. There are adapt-
er molecules that attach the payload to the myosin.
Kinesin

Microtubule
POSTSYNAPTIC PROCESSES
Dynein
There are several different postsynaptic receptors for each
Retrograde transmitter; they are distinguished by their amino acid se-
FIGURE 7–8 Axoplasmic transport. Kinesin motors carry quences and, in some cases, pharmacology. Different regions
vesicles toward the nerve terminals. Dynein motors carry different of the nervous system have characteristic receptors; sometimes
vesicles toward the cell body. (Modified with permission from Landowne an individual postsynaptic cell will have multiple receptor
D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.) types. The ionotropic receptors are excitatory or inhibitory ac-
CHAPTER 7 Synapses 67

cording to their ionic selectivity. The metabotropic receptors for that ion (equation (4) in Chapter 4). If the channel is perme-
may indirectly cause channels to open or close and may also able to multiple ions, its reversal potential is the weighted average
modulate the activity of the cells in other ways. of the Nernst potentials for its ions (equation (6) in Chapter 4).
The PSPs are called excitatory postsynaptic potentials nAChR channels and GluR channels are approximately
(EPSPs), if their effect is to make the postsynaptic cell more equally permeable to Na+ and K+, and their reversal potential is
likely to respond with an action potential, or inhibitory post- about –10 mV; when activated, they make EPSPs. GABAAR
synaptic potentials (IPSPs), if they make the postsynaptic cell and glyR are Cl channels; their reversal potential is about –80
less likely to fire an action potential. Each channel has a selec- mV. The cardiac mAChR, through a G protein, activates a Kir
tivity pattern and allows different ions to flow through with dif- channel (KACh) that has a reversal potential about –90 mV. Both
fering ease. This means that each channel will have a reversal Cl− channels and K+ channels make IPSPs. If for some reason
potential: a potential at which there will be no net flow of ions the cell happens to be more negative than –80 mV, opening Cl
through the channel. If the membrane potential is more posi- channels will depolarize the cell but still work to keep other
tive than the reversal potential, net current will flow out of the channels from further depolarizing the cell to threshold.
cell, tending to hyperpolarize it. If the membrane is less positive
or more negative, current will flow in and tend to depolarize the
cell. The current that flows through channels drives the mem- THE NEUROMUSCULAR
brane potential toward the reversal potential for that channel.
Most neurons in the CNS receive a constantly fluctuating in- JUNCTION—A SPECIALIZED
put from a variety of synapses and their membrane potential is SYNAPSE
always changing. If a synapse opens channels having a reversal
potential more positive than the threshold for action potentials, Because of its easy accessibility, the neuromuscular (or myo-
they will produce an EPSP. If the reversal potential is more nega- neural) junction (Figure 7–9) is the best-studied synapse; it is
tive than the threshold, an IPSP will result. If a channel is perme- the source of much of what is known about synapses. This sec-
able to a single ion, its reversal potential is the Nernst potential tion describes the functioning of this synapse, bringing together

Myelin

Axon

Schwann cell

Mitochondria

Presynaptic
Synaptic membrane
vesicles
Synaptic
cleft Postsynaptic
membrane

Synaptic Muscle
fold nucleus

Myofibrils

FIGURE 7–9 The neuromuscular junction. A myelinated nerve (gray) ends on a specialized region of a skeletal muscle (colored). (Modified
with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
68 SECTION II Cell Physiology

and illustrating many of the ideas introduced more abstractly


above. The neuromuscular junction is of considerable clinical
interest. Myasthenia gravis is a disease that incapacitates the
neuromuscular junction; there are other diseases and several
drugs and toxins that target the junction. The neuromuscular
junction provides a convenient assay for the anesthesiologist
gauging recovery from muscle immobilization after surgery.
A single motoneuron controls between 3 and 1,000 muscle
cells. Each muscle cell receives input from one motoneuron.
The motoneuron and all of its muscle cells function together as
a motor unit. In healthy people, an action potential in the mo- FIGURE 7–10 An endplate potential and action potential
toneuron will produce a large EPSP in all of its muscle cells, at the neuromuscular junction (left) and 2 cm away (right).
large enough to greatly exceed the threshold of the muscle Dashed lines indicate responses in low Ca2+ (see text). (Modified with
cells and produce action potentials and contraction. The CNS permission from Landowne D: Cell Physiology. New York: Lange Medical Books/
regulates movement by choosing which motor units to acti- McGraw-Hill, 2006.)
vate. Smaller motor units produce finer movements.
At the nerve ending, the axon loses its myelin and spreads eral centimeters away from the neuromuscular junction, the
out to form the motor endplate, named for its anatomic ap- potential shown in the right trace will be seen. If the concen-
pearance. The nerve terminals contain many mitochondria tration of Ca2+ in the bath is decreased or the concentration of
and many 40-nm-diameter synaptic vesicles that contain ACh. Mg2+ is increased and the nerve is stimulated again, the poten-
The nerve terminal is separated from the muscle by a 50-nm tial at the neuromuscular junction will change as shown in the
gap, the synaptic cleft, which contains a basal lamina. The dashed trace. Under these conditions there will be no change
muscle membrane contains AChRs and also AChE. In trans- in the membrane potential several centimeters away from the
mission electron micrographs, both presynaptic and postsyn- junction.
aptic membranes appear thickened, indicating the presence of The solid trace on the left shows an action potential super-
channels and other proteins. imposed on an endplate potential (EPP). There is an initial
Neuromuscular transmission can be described as a 10-step depolarization due to a net entry of positive charge through
process: (1) an action potential enters the presynaptic termi- AChRs that were activated by the released ACh. When the po-
nal; (2) the nerve terminal is depolarized; (3) depolarization tential reached about –50 mV, an action potential was initiat-
opens CaV channels; (4) Ca2+ enters the cell, moving with its ed. In normal Ca2+ the EPP is two or three times larger than
electrochemical gradient; (5) Ca2+ acts on a release site, prob- necessary to depolarize the muscle membrane to threshold.
ably synaptotagmin, causing synaptic vesicles to fuse with the The pure action potential is seen in the trace on the right; it
presynaptic membrane; (6) about 200 vesicles release their can be recorded by stimulating one end of the muscle electri-
ACh into the synaptic cleft; (7) the ACh in the cleft either (a) cally or by placing the recording electrode a few centimeters
diffuses away out of the cleft, (b) is hydrolyzed by AChE into away from the endplate. The dashed trace on the left shows an
acetate and choline, or (c) interacts with AChRs on the post- EPP with reduced amplitude. The EPP is not visible a few cen-
synaptic membrane; (8) the activated AChRs are very perme- timeters away from the endplate (right). A reduction in extra-
able to Na+ and K+ and slightly permeable to Ca2+; hence, a net cellular Ca2+ reduces the release of ACh and thus reduces the
influx of positive charge into the muscle cell depolarizes the EPP. An increase in Mg2+ reduces transmitter release by reduc-
muscle membrane in the endplate region; (9) when the muscle ing Ca2+ entry through CaV channels. These opposing effects of
membrane is depolarized to threshold, an action potential is Ca2+ and Mg2+ have been seen on all chemical synapses that
elicited, which propagates in both directions to the ends of the have been examined; this is now considered one of the tests for
muscle cell (the link between muscle excitation and contrac- identifying a chemical synapse.
tion is discussed in the next section); and finally (10) choline Ca2+ and Mg2+ concentrations have different effects on the
is recycled into the nerve terminal, Ca2+ is pumped out of the excitability or the threshold for action potentials on the nerve
nerve terminal, and vesicles are recycled and refilled. and muscle cells. The reduction of Ca2+ makes the cells more
excitable or have a more negative threshold, or requires a small-
er depolarization to reach threshold for an action potential.
RECORDING THE ENDPLATE POTENTIAL This is an effect on the NaV channels; in low Ca2+, NaV channels
will open at more negative potentials. Ca2+ and Mg2+ have a
If a microelectrode is inserted into a muscle fiber near the neu- synergistic action on NaV channels; they have opposing actions
romuscular junction, a resting potential of about –90 mV will on neuromuscular transmission. Clinically, the effects of hy-
be measured. If the nerve is stimulated and the muscle is pre- pocalcemia are hyperexcitability and spontaneous action po-
vented from contracting by extreme stretching the membrane tentials in nerve and muscle. These effects are seen when there
potential will be seen to change, as shown in the solid trace on is still enough Ca2+ to support sufficient ACh release, so that
the left in Figure 7–10. If, instead, the electrode is placed sev- every nerve action potential leads to a muscle action potential.
CHAPTER 7 Synapses 69

200,000 vesicles are released, which is equal to the number

1 mV
seen by the electron microscope in an unstimulated neuro-
10 ms muscular junction. After BWSV treatment, no vesicles are
visible. BWSV paralyzes by depleting the nerve terminals of
synaptic vesicles. It can be deadly if the nerve endings control-
ling breathing are compromised.

TRANSMITTER–RECEPTOR
INTERACTION
The nicotinic AChR at the neuromuscular junction has five
subunits, each with four TM segments. Two of the subunits are
FIGURE 7–11 Some miniature endplate potentials (MEPPs)
seen by stimulating a neuromuscular junction bathed in low
called alpha subunits and bind ACh at the α–γ and α–δ inter-
Ca2+ four times. The second MEPP on the bottom trace was faces near the top of the molecule, about 5 nm from the center
spontaneous. (Modified with permission from Landowne D: Cell Physiology. of the membrane. The channel then undergoes a conforma-
New York: Lange Medical Books/McGraw-Hill, 2006.) tional change that is transmitted through the molecule to open
the pore, most likely by causing the M2 TM segments to move
out away from the axis of the pore, making it larger. The open
In this low-Ca2+ and high-Mg2+ case, the EPP is not large pore allows Na+ and K+ and, to a lesser extent, Ca2+ to pass. The
enough to reach threshold and elicit an action potential. Action pore stays open about 1 millisecond and about 20,000 ions
potentials are actively propagated; the EPP spreads passively pass at a rate of 2 × 107/s, which is equivalent to about 3 pA. If
and will not be visible a few centimeters from the neuromus- a single AChR is captured in a patch of membrane and main-
cular junction. These two potentials are produced by the activ- tained with a –90 mV potential, application of ACh will cause
ity of different channels that have differing pharmacology. the channel to open and close several times, each opening ap-
Curare will block AChRs and the EPP without affecting the pearing as a 3-pA current pulse of varying duration averaging
action potential seen following direct electrical stimulation of about 1 millisecond. A single quantum opens about 2,000
the muscle. A toxin from a cone snail (μ-conotoxin) will block channels; 200 quanta open about 400,000. A neuromuscular
the muscle action potential but not the EPP. The μ-conotoxin junction has many more channels, about 20 million; thus, only
blocks muscle NaV channels but not nerve NaV channels, which a small fraction is used at any one time.
are a different gene product. The number of open channels is proportional to the concen-
If the Ca2+/Mg2+ ratio is sufficiently low, the response to tration of ACh squared and the effective number of receptors.
stimulation will appear as in Figure 7–11. Each trace repre- A kinetic scheme for the reaction is shown in Figure 7–12. The
sents the response to a stimulation that is repeated every 5 sec- receptor can open with one or two ACh molecules bound; it
onds. Three of the traces show a small EPP; in the third trial stays open about 10 times longer with two bound. It is the con-
there was no response. The first response is about 1 mV high; centration of R • 2ACh that is proportional to the concentra-
the second and fourth responses are about 0.5 mV. When this tion of ACh squared:
experiment was repeated many times, the responses were Number of open channels = k[R][ACh]2 (1)
found to be quantized with a unit response of about 0.5 mV.
That is, there were many 0.5-, 1-, and 1.5-mV responses but
very few with amplitudes in between. In addition, there are
DESENSITIZATION
sometimes spontaneous 0.5-mV responses without any stimu- If a single AChR is exposed to continuous ACh for several
lation; one of these was caught on the fourth trace. These min- minutes, its response will slow and openings will become
iature endplate potentials (MEPPs) represent the postsynap- less frequent. If ACh is added to the bath containing a
tic response to the release of one, two, or three quanta of ACh. neuromuscular junction, the muscle membrane potential will
Each quantum is the contents of a single vesicle. The exact
number of vesicles released on any particular stimulation can-
not be known; only the average number or the mean quantal
R R • ACh R • 2ACh Closed
content can be predicted. The EPP in normal Ca2+/Mg2+ con-
ditions is the response to about 200 quanta.
The average rate of spontaneous MEPPs is about 1 vesicle/s. R∗ • ACh R∗ • 2ACh Open
In a normal EPP, the 200 vesicles are released within 1 milli- FIGURE 7–12 A kinetic scheme of the reaction between
second, which means that stimulation increased the rate of acetylcholine and the nicotinic acetylcholine receptor. The receptor
release by 200,000-fold. If BWSV is applied to a neuromuscu- (R) can bind two ACh molecules. Once ACh is bound the receptors can
lar junction, the MEPP frequency increases to a few hundred open (R*) and allow the passage of ions. (Modified with permission from
per second for about 30 minutes and then stops. In total about Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
70 SECTION II Cell Physiology

10 μM ACh The Bungarus snake paralyzes its prey with α-bungarotoxin,


which binds AChRs irreversibly and prevents their opening.
Bungarotoxin has been fluorescently labeled and used experi-
10 mV mentally to identify and locate nAChRs.
1 min

MYASTHENIA GRAVIS
FIGURE 7–13 Desensitization of acetylcholine receptors Myasthenia gravis is a disease associated with muscle weakness
(AChRs). With prolonged ACh exposure the AChRs first open and and fatigability on exertion. It is an autoimmune disease that
then enter a desensitized and closed state where they no longer leads to the destruction of AChRs. Patients may have only
respond to ACh. (Modified with permission from Landowne D: Cell Physiology. 10–30% of the number of AChRs found in healthy individuals.
New York: Lange Medical Books/McGraw-Hill, 2006.)
Treatment with anticholinesterases increases the amount of
available ACh, which makes it more likely that the remaining
depolarize but the response will reach a peak and then decline, AChRs will be activated (equation (1)). There is a danger of
as shown in Figure 7–13. This decline is called desensitiza- giving too much anticholinesterase, which can lead to desensiti-
tion; the AChR molecule has entered an inactivated state from zation of the AChRs and further weakness. If this weakness is
which it does not open. This is functionally somewhat similar misinterpreted as insufficient anticholinesterase therapy, a tragic
to the inactivation of NaV channels except that the time course, positive feedback loop leading to myasthenic crisis can ensue.
the agent that causes the inactivation, and the molecular basis
in the channels are completely different. Desensitization prob-
ably does not occur with normal use of neuromuscular junc- LAMBERT–EATON SYNDROME
tions but may become a problem when drugs are used that
block AChE. A patient with desensitized AChRs may be para- The Lambert–Eaton syndrome is seen with an autoimmune
lyzed and unable to breathe due to a lack of functional disease that reduces the number of CaV channels in the presyn-
AChRs. aptic terminal. With prolonged effort, these patients gain
strength, the opposite of myasthenic patients. Prolonging the
presynaptic action potentials with drugs that block KV chan-
SOME DRUGS THAT ACT AT THE nels, such as diaminopyridine, may alleviate some of the symp-
NEUROMUSCULAR JUNCTION toms. The prolonged depolarization opens the remaining CaV
channels for a longer time, allowing more Ca2+ entry and there-
d-Tubocurare is a classic neuromuscular blocking agent, fore more release. If the experiment shown in Figure 7–11 is
originally discovered as an arrow poison from South America. performed on these neuromuscular junctions, they will be
Curare binds AChRs reversibly and prevents ACh from open- found to have a lower quantal content, that is, they release a
ing the channels. After application of curare, the EPP becomes lower number of vesicles per stimulus. This is in contrast to
smaller; if there is sufficient curare, the EPP becomes so small myasthenia gravis, which will show the normal quantal content
that it no longer elicits an action potential, similar to the but a smaller MEPP, the depolarization for each quantum.
dashed response in Figure 7–10, and the junction is effectively
blocked. Higher doses of curare can eliminate the EPP. Curare
reduces the EPP by reducing the number of receptors available REPETITIVE STIMULATION
to respond to ACh. Curare or a related drug is often used dur-
ing surgery to immobilize muscles; it can also facilitate tra- The amount of transmitter released by a synapse is not con-
cheal intubation and mechanical ventilation. stant from impulse to impulse but depends on the past history
Anticholinesterases such as neostigmine and physostigmine of activity. If the nerve leading to a neuromuscular junction is
combine with AChE and prevent hydrolysis of ACh, which leads stimulated once every 10 seconds or slower, it will consistently
to a larger EPP. Neostigmine is used to speed recovery from the release about 200 vesicles. If the stimulation rate is abruptly
effects of curare and to reduce the symptoms of myasthenia changed to 50/s, which is roughly the rate used by the CNS to
gravis. There are dangers associated with neostigmine; an excess cause normal muscle contraction, the amount released per im-
of ACh can lead to desensitization of the remaining receptors. pulse will increase in the first half second and then decrease
Also, the body uses ACh to slow the heart and release saliva; (Figure 7–14). The increase, called facilitation, is related to a
both of these effects may be enhanced by physostigmine. buildup in residual calcium in the nerve terminal. The de-
Botulism is a potentially fatal food poisoning caused by the crease, called depression, is thought to reflect a depletion of
anaerobic bacterium Clostridium botulinum. Some of the tox- vesicles at the release sites.
ins released by this organism are endopeptidases, which are This variation does not affect the functioning of the neuro-
taken up by nerve cells and cleave synaptobrevin, thus muscular junction in a healthy individual. Each of those
preventing transmitter release. The purified toxins are used nerve impulses releases sufficient ACh to produce an EPP
clinically to prevent unwanted neuromuscular transmission. large enough to fire a muscle action potential. However, the
CHAPTER 7 Synapses 71

tetanus, the release increased and decreased, as in Figure 7–14.


Facilitation After the tetanus, as the synapse recovered from depression, a
1 posttetanic potentiation (PTP) was seen that lasted for sev-
eral minutes. PTP is also related to an increase in residual Ca2+
X Normal

concentration in the nerve terminal, but it has a slower onset


Depression and slower decline than facilitation.
PTP is used as a diagnostic procedure following surgical
procedures when curare or other neuromuscular blocking
agents have been used to prevent unwanted motion. The anes-
40 80 120 thesiologist will give the patient cholinesterase inhibitors, but
Impulses she or he wants to know when just enough inhibitor has been
FIGURE 7–14 Facilitation and depression of synaptic given to avoid giving too much and desensitizing the AChRs.
transmission at the neuromuscular junction. With repetitive The anesthesiologist will repeat the experiment shown in
stimulation the amount of transmitter released by each stimulus Figure 7–15, stimulating the thenar branch of the patient’s me-
changes, first increasing and later decreasing. (Modified with dian nerve and feeling the strength of contraction of the thenar
permission from Landowne D: Cell Physiology. New York: Lange Medical Books/ muscles. Two shocks are given before the tetanus and then one
McGraw-Hill, 2006.) 30 seconds later. Under deep curare, none of these will pro-
duce a palpable contraction. As more ACh is made available
by blocking the esterase, the stimulus following the tetanus
myasthenic person may have functional neuromuscular trans-
will give a larger response than the two before the tetanus be-
mission only early in the task and experience weakness as the
cause it will be the first one with an EPP large enough to excite
depression occurs with a prolonged effort and the amount of
the muscle. The endpoint is when enough esterase has been
ACh released falls below what is necessary to trigger a muscle
given that all three responses are the same because all three
action potential. An anticholinesterase with a short duration
EPPs are above threshold for muscle activation.
of action, edrophonium chloride (Tensilon), is often used as a
test for myasthenia gravis in patients who show rapid weaken-
ing when asked to perform a sustained contraction.
AUTONOMIC SYNAPSES
POSTTETANIC POTENTIATION The autonomic nervous system (ANS) has two divisions,
both with two synapses outside the CNS (Figure 7–16). The
When the 50/s stimulus is stopped, there is an increase in the synapse closer to the CNS is referred to as the ganglionic syn-
amount of transmitter that can be released by a single nerve apse; the nerves leading into and out of the ganglia are called
impulse (Figure 7–15). The nerve was stimulated once every preganglionic and postganglionic. The sympathetic ganglia
30 seconds before and after the tetanic stimulation. During the lie in a chain adjacent to the spinal column; the parasympa-
thetic ganglia are close to the end organs where the second
synapse occurs. The second synapses are onto smooth muscles
PTP or cardiac cells or gland cells. Many tissues receive both sym-
pathetic and parasympathetic innervations.
The primary transmitter in the ganglionic synapse of both
divisions is ACh; the receptors are nicotinic nAChRs that are
heteromeric pentamers of related but different gene products
1
than the nAChR of the skeletal muscle. The ganglionic recep-
x Normal

tors are less sensitive to curare and more easily blocked by


hexamethonium. The primary postganglionic transmitter in
the sympathetic nervous system is NE, and there are two cat-
egories of GPCRs on the postsynaptic cells called alpha- and
beta-adrenergic receptors. The primary postganglionic recep-
tor in the parasympathetic division is ACh and the receptors
1 2 3 4 are muscarinic mAChRs, which are also GPCRs but generally
Minutes with different G proteins than the NE receptors.
The ganglionic synapses are usually described as behaving
FIGURE 7–15 Posttetanic potentiation (PTP) of synaptic
transmission at the neuromuscular junction. After the end of a
more or less like the neuromuscular junction. However, the
period of repetitive stimulation the amount of transmitter released situation is more complicated; the postsynaptic neurons have
by subsequent infrequent stimuli is increased for several minutes. dendrites with more than one presynaptic nerve ending on
(Modified with permission from Landowne D: Cell Physiology. New York: Lange them. Different subpopulations of presynaptic and postsynap-
Medical Books/McGraw-Hill, 2006.) tic cells have been distinguished by looking at the peptide
72 SECTION II Cell Physiology

CNS
Parasympathetic ACh ACh
Target
Preganglionic

ACh Sympathetic NE
Target
Postganglionic

ACh
Epinephrine

Motor ACh
Muscle

FIGURE 7–16 A schematic view of the efferent fibers of the autonomic nervous system and the motoneurons. From top to bottom:
parasympathetic, sympathetic, adrenal medulla, motoneuron. (Modified with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/
McGraw-Hill, 2006.)

transmitters that are in these cells along with their classic ACh and NE from the ANS, so the blocking of one set of recep-
transmitters. Postganglionic cells also have mAChRs that pro- tors may produce effects similar to activating the other.
duce a slow EPSP by closing a K channel. There are also small,
intensely fluorescent cells in the ganglia that are innervated by
preganglionic fibers and release NE or dopamine. All in all, it CENTRAL NERVOUS
seems that some computation must be carried out in the gan- SYSTEM SYNAPSES
glia, more than the simple passthrough circuit seen at the neu-
romuscular junction. The human CNS has billions of neurons with trillions of syn-
The synapses between the postganglionic cells and the end or- apses between them. A single neuron may have thousands of
gans are different from those in the neuromuscular junction. The both excitatory and inhibitory inputs; some larger neurons
presynaptic processes are similar, but postganglionic cells make may have over 100,000 endings on them. In order to accom-
“en passant” synapses on tissue targets over a length of axon. modate this convergence of synaptic inputs, most neurons
Synaptic vesicles are stored in varicosities of the nerve, which have a dendritic tree that greatly expands the area available for
continues on to other varicosities before reaching its terminal. synaptic contact. The cell body (soma) and the initial region of
Activation of mAChRs by the ANS increases GI tone and the axon (axon hillock) integrate the incoming synaptic sig-
motility, increases urinary bladder tone and motility, increases nals and determine when and how often the neuron will fire
salivation and sweating, constricts bronchioles, and decreases action potentials (Figure 7–17). The axon carries the output of
heart rate and blood pressure. The ANS activates α-, β1-, and the neuron to the next group of neurons or to skeletal muscle
β2-adrenergic receptors, and α-adrenergic receptors raise cells if it is a motoneuron. Usually only a single axon leaves
blood pressure. β1-Adrenergic receptors increase heart rate the cell body, but it later branches to allow the neuron to syn-
and strength of contraction and blood pressure. β2-Adrenergic apse with many other cells. This divergence of information
receptors dilate bronchioles in the lungs. The mechanism of combined with the convergence of many inputs onto a neuron
the effects on cardiac and smooth muscle is discussed in the gives the CNS much of its computational power.
next section. Each neuron in the CNS acts as one or more small comput-
Many agonist and antagonist drugs have been used to control ers. While each cell performs its computations in milliseconds,
these processes, some with more specificity than others. Thus, millions of times slower than the central processing unit of a
there are specific α agonists and β blockers. Amphetamines and modern computer, the billions of neurons operating in parallel
cocaine have an indirect adrenergic effect by stimulating NE make the CNS shine in comparison. The CNS is capable of
release. Some compounds, such as ephedrine, have both direct creating every thought in recorded history while simultane-
and indirect adrenergic effects. Atropine is the archetypical ously regulating both walking and chewing gum. The synapses
mAChR antagonist; its effects are the opposite of those attrib- make this possible. Learning and memory are accomplished
uted to ACh above. At many sites there is a tonic release of both by the modification of synapses.
CHAPTER 7 Synapses 73

Axons converge
Dendrites on neuron

Synaptic inputs
cover most of the
surface of the
soma and dendrites

Soma

Axon hillock or
initial segment
Synapses
Axon diverge
FIGURE 7–17 The convergence and divergence of
synapses in the CNS. (Modified with permission from Landowne D:
Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

There are two general types of synapses in the CNS, elec- There are two general types of chemical synapses in the
trical and chemical. Electrical synapses operate by direct CNS, excitatory and inhibitory. Excitatory synapses generate
electrical current flow from the presynaptic neuron into the EPSPs that depolarize the membrane toward threshold. In-
postsynaptic neuron through gap-junction channels between hibitory synapses generate IPSPs that either hyperpolarize the
the membranes of the two cells (Figure 7–18). Neurotrans- membrane or resist depolarization to threshold. Each of these
mitters are not involved and electrical synapses can have less types can be further divided into chemosensitive ion channels
synaptic delay than chemical synapses. However, unlike (or ionotropic receptors) and G protein–linked ion channels
chemical synapses, electrical synapses cannot amplify the (or metabotropic receptors). Chemosensitive ion channels
signal, nor can they reverse the direction of current flow. Gap typically give rise to fast synaptic events that last a few milli-
junctions, which work as electrical synapses and allow action seconds; G protein–linked ion channels may produce effects
potentials to flow selectively from one cell to another, also for hundreds of milliseconds.
connect cells in the heart and some types of smooth muscle.
INTEGRATION OF SYNAPTIC CURRENTS
Excitatory and inhibitory synapses inject current (positive or
negative) into cells. These currents flow into the cell body and
are summed. The PSPs passively spread to the spike initiation
site or the part of the cell with the lowest threshold because of
the cable properties of the cell. More distal synapses will be
decremented compared to those near the site. The cell pro-
duces the spike initiation site by controlling the local density
of NaV channels. Often the spike initiation site is the axon hill-
ock near the start of the axon (see Figure 7–17) or at the first
node of Ranvier.
Because the PSPs last for several to many milliseconds, they
can add together even though they do not occur synchronous-
ly; this is called temporal summation. The effects of synapses
FIGURE 7–18 An electrical synapse. Current passes directly at different locations on the same postsynaptic cell can also
from the presynaptic cell to the postsynaptic cell through specialized add up; this is called spatial summation. Spatial summation is
cell–cell channels. (Modified with permission from Landowne D: Cell Physiology. weighted inversely by the distance from the synapse to the ini-
New York: Lange Medical Books/McGraw-Hill, 2006.) tiation site of the action potential.
74 SECTION II Cell Physiology

Synaptic channels may be called AMPA, quisqualate, or kainate chan-


cleft nels, according to which of these nonphysiologic agonists
Postsynaptic opens them. Non-NMDA channels typically generate fast
spine on EPSPs lasting about 5 milliseconds.
Axon dendrite
When they are activated by glutamate, non-NMDA chan-
nels allow Na+ and K+ to flow through their pores. Each ion
Presynaptic moves in the direction that will tend to bring the membrane
nerve terminal potential to its Nernst equilibrium potential. Because both are
moving, the membrane potential tends to approach the aver-
Synaptic age of the two equilibrium potentials, which is about –10 mV.
Postsynaptic
vesicles
density This potential, where the two ionic currents are equal, is called
the reversal potential for the channel. When these channels
FIGURE 7–19 A CNS synapse. These are less elaborate than open at potentials more negative than the reversal potential,
the synapse at the neuromuscular junction (Figure 6–9). (Modified with the tendency for Na+ to enter the cell will dominate and the
permission from Landowne D: Cell Physiology. New York: Lange Medical Books/ membrane will depolarize toward the reversal potential. If the
McGraw-Hill, 2006.) starting potential were more positive than the reversal poten-
tial, the K+ ions would dominate and the cell would hyperpo-
larize toward the reversal potential.
Figure 7–19 is a schematic drawing of a chemical synapse in NMDA receptor channels generate EPSPs lasting hundreds
the CNS. The presynaptic terminal is about 1 μm in diameter of milliseconds. Open NMDA channels allow Na+ and K+ and
and contains mitochondria and synaptic vesicles filled with also Ca2+ to pass through their pores. In the presence of gluta-
neurotransmitter. Depolarization of the terminal opens CaV mate, NMDA channels open only if the postsynaptic cell is
channels, and Ca2+ flows with its electrochemical gradient to also depolarized by some other means. This dual control of
act on synaptotagmin and trigger the fusion of a few vesicles Ca2+ entry has a key role in learning, as discussed below.
with the presynaptic membrane in order to exocytose the neu- GABA is the major inhibitory transmitter in the brain.
rotransmitter. The membrane is then recycled and the vesicles Glycine is an inhibitory transmitter in the brainstem and spi-
are refilled. The postsynaptic receptors are often on protru- nal cord. GABA opens GABAA channels directly, which allows
sions from dendrites, called spines, although synapses are also Cl− ions to pass through their pores. GABA can also cause in-
found on the dendritic shaft, the neuronal cell body, and other hibition through GABAB receptors, which are GPCRs that
synaptic endings. lead to the opening of K channels. The reversal potential for
CNS synapses share many features with the neuromuscular GABAA channels is at the Nernst potential for Cl−, about –80
junction but differ in several important respects. CNS synaps- mV. If the membrane is more positive than ECl, Cl− will enter
es are much smaller and release far fewer vesicles, typically less the cell and make the membrane potential more negative,
than 5 per impulse, compared to about 200 at the motor end- which will make it less likely to initiate an action potential.
plate. In the CNS, synaptic clefts are narrower, about 20 nm, Benzodiazepines such as diazepam (Valium) and barbitu-
and cadherins and other cell adhesion molecules span the gap. rates enhance the open probability of activated GABAARs.
ACh is the transmitter at the neuromuscular junction; there is Both have been used as sedatives and anticonvulsants. General
a wide variety of transmitters in the CNS. The EPP is always anesthetics such as ether, chloroform, and halothane increase
excitatory and large enough to bring the muscle membrane to the duration of IPSPs and decrease the amplitude and duration
threshold; synapses in the CNS are excitatory or inhibitory of EPSPs.
and threshold is reached by the combination of hundreds of
EPSPs.
There are some exceptional CNS synapses. In the cerebel- CNS—MODULATORY
lum, a climbing fiber axon may make dozens of synapses on a NEUROTRANSMITTERS
Purkinje cell. In the calyx of Held, in the auditory pathway, the
presynaptic ending forms a cap with fingerlike stalks that en- In the CNS, ACh, NE, dopamine, and serotonin primarily act
velop the postsynaptic neuron, covering about 40% of its soma. as diffuse modulators of activity, acting over timeframes that
At both of these synapses a single presynaptic impulse releases are long compared to action potentials, as opposed to being
hundreds of quanta, and the resulting EPSP is large enough to involved in specific discrete tasks. Each of these neurotrans-
trigger a postsynaptic action potential. mitters has its own set of neurons and targets; some of these
Glutamate is the principal excitatory neurotransmitter in neurons may influence more than 100,000 postsynaptic neu-
the CNS. There are several postsynaptic glutamate receptors, rons. The postsynaptic receptors are metabotropic and alter
both channels and GPCRs. The channels can be grouped into the responsiveness of the postsynaptic neurons through sec-
two major types, NMDA and non-NMDA channels, accord- ond messenger pathways. There are also ionotropic nAChRs
ing to their sensitivity to the synthetic agonist N-methyl-d- in the CNS, but there are 10–100 times more mAChRs. The
aspartate. Both types respond to glutamate. The non-NMDA ACh and NE modulatory systems are part of the ascending
CHAPTER 7 Synapses 75

Axon

Neuron C

FIGURE 7–20 Presynaptic inhibition can occur by a synapse (A and B) on another synaptic ending. (Modified with permission from Landowne
D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)

reticular activating system that arouses the forebrain in re- cellular space by a nonvesicular process. It binds to presyn-
sponse to stimuli. In some general ways, the modulatory sys- aptic cannabinoid receptors (CB1), which are GPCRs, and
tems play a role in the CNS similar to the role played by the can alter the subsequent release of traditional neurotrans-
ANS in the rest of the body. mitters.

PRESYNAPTIC INHIBITION REPETITIVE FIRING OF NERVE CELLS


Some CNS synapses act directly on other synaptic endings If an axon or a muscle cell is subjected to a maintained depo-
rather than on dendrites or cell bodies (Figure 7–20). Termi- larization, it will respond with one or perhaps two action po-
nal A releases GABA on to terminal B, activating Cl channels tentials and then stop firing because the NaV channels enter
that tend to hyperpolarize terminal B. If an action potential the inactivated state and require a brief period near the resting
arrives in B while the Cl channels are open, the action poten- potential to recover. Many CNS cells and the slowly adapting
tial’s amplitude will be reduced, so that it will open fewer CaV sensory nerve endings will respond to a sustained depolariza-
channels and therefore fewer vesicles will be released by termi- tion with a train of action potentials at about 50/s. This is made
nal B, and it will have a smaller effect on neuron C. possible by CaV channels and Ca-activated K channels. The ac-
tion potential depolarization opens the CaV channels and the
Ca2+ that enters opens the Ca-activated K channels by binding
RETROGRADE FREELY DIFFUSIBLE to the intracellular portion of the molecule. The Ca-activated
CHEMICAL TRANSMITTERS K channel then allows K+ to leave and the membrane potential
to approach EK for a long-lasting hyperpolarization, long
In addition to the classic transmitters that are released from enough for the NaV channels to recover from inactivation
vesicles and bind to receptors, there are chemical messengers (Figure 7–21). The balance between the sustained stimulus
in the CNS with a different mode of operation. Nitric oxide and the rate that Ca2+ is removed from the Ca-activated K
(NO) is not stored but rather produced when needed. It can channels determines the firing rate.
freely diffuse across cell membranes from the inside of one cell
(typically a postsynaptic cell body) to the inside of other cells
(typically presynaptic endings), where it alters some chemical LEARNING, MEMORY, AND
reactions. NO may spread to several presynaptic endings in SYNAPTIC PLASTICITY
the vicinity. It is removed from the tissue by binding to hemo-
globin. The cellular basis of learning and memory is a functional re-
Anandamide, an endogenous cannabinoid, is also pro- modeling of synaptic connections, often called synaptic plas-
duced as needed in postsynaptic cells and reaches the extra- ticity. This includes both explicit or declarative memory when
76 SECTION II Cell Physiology

+40 experiment is similar to the PTP demonstration shown in


Figure 7–15; the synapse is tested infrequently, subjected to
high-frequency stimulation, and then tested infrequently again.
0 Unlike PTP, which disappears in a few minutes, with LTP, the
potentiation remains for many hours or days (Figure 7–22).
mV

Also unlike PTP, LTP is primarily a postsynaptic event. It is


not necessary to provide the high-frequency stimulation to the
presynaptic terminals; simple depolarization of the postsynap-
−60 tic cell paired with the presynaptic stimulation will induce LTP.
−70
This response to pairs of inputs made LTP a candidate basis for
associative learning. There are two types of glutamate receptors
0 200 400 ms
on the postsynaptic membranes: AMPA (non-NMDA) and
Sustained excitatory synaptic input
NMDA receptors. During low-frequency unpaired stimulation,
only the AMPA receptors are activated; the NMDA receptors
FIGURE 7–21 Repetitive firing of a motoneuron. Unlike axons, are plugged by external Mg2+ ions. The AMPA receptor chan-
many nerve cells respond repetitively to a sustained input. (Modified
nels are permeable to Na+ and K+; near the resting potential,
with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/
McGraw-Hill, 2006.)
Na+ movement into the cell is favored. When the postsynaptic
membrane is depolarized, either by high-frequency synaptic
input or by injecting current into the postsynaptic cell, the Mg2+
is driven off the NMDA receptors and they respond to gluta-
the person can recall and describe some fact or past event and mate and allow Na+ and Ca2+ to enter the cell. The elevated Ca2+
implicit or procedural memory, as in a learned motor skill. activates a series of biochemical events that lead to the insertion
Memory is often subdivided into short-term, minutes to of more AMPA receptors into the postsynaptic membrane.
hours, and long-term, days to a lifetime. Short-term memory LTP has been associated with learning in rats using a water
formation involves the modification of existing proteins, often maze. Rats with surgically removed hippocampi do not learn
by phosphorylation. Long-term changes involve gene activa- the maze, neither do rats that have been treated with a specific
tion, protein synthesis, and membrane rearrangement, includ- antagonist for NMDA receptor channels. There are other ex-
ing the formation and/or resorption of presynaptic terminals amples of synaptic plasticity in other regions of the brain and
and postsynaptic spines. In a few studies, the volume of cere- there may well be additional mechanisms, including retro-
bral cortex dedicated to a task has been shown to increase with grade action of NO or anandamide.
specific training.
The most intensively studied cellular learning phenomenon
is long-term potentiation (LTP) in hippocampal synapses.
The hippocampus is required for the formation of new long- CLINICAL CORRELATION
term memories. If both hippocampi are compromised, the per-
son will live continuously in the present with no recollection of About a month before coming to the hospital, a 56-year-
events after the damage. In the hippocampus, LTP occurs at old woman noticed she was unable to hold her shopping
glutamate synapses between presynaptic CA3 cells and post- bag and that her head fell forward when she knelt to tie
synaptic CA1 cells. LTP and the related long-term depression her shoes. Two weeks later she had to remain in bed and
(LTD) also occur in other locations in the CNS. The classic had difficulty sitting up. Her jaw began to droop, she had

3
mV

0
10 20 30 40 50 60
Minutes
FIGURE 7–22 Long-term potentiation. A brief conditioning stimulus (blue bar) causes a long-term increase in synaptic efficacy. (Modified
with permission from Landowne D: Cell Physiology. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 7 Synapses 77

■ GABA and glycine are the major inhibitory neurotransmitters


to hold it up with her hand, and her left eyelid began to in the CNS.
droop. Her speech became indistinct when she was ex- ■ Several biogenic amines are important neurotransmitters.
cited, swallowing was difficult and fluid sometimes re- NE is released by sympathetic nerves to control the heart
gurgitated through her nose. A few days after admission and vascular smooth muscle.
to the hospital, she developed weakness in the middle ■ Neuropeptides are small proteins released as neurotransmitters.
and ring fingers of both hands that was increased by ex- ■ Synaptic release involves many proteins and is controlled by
citement and lessened by rest. There was no muscle wast- CaV channels, which are opened when an action potential
ing and tendon reflexes were all present. Her masseter invades the presynaptic terminal.
muscles showed a decremental response to tetanic elec- ■ Axons have a microtubule-based transport system to move
trical stimulation. materials from the cell body to the presynaptic terminal
In the hospital, she was injected with 1 mg of physostig- (orthograde transport) and in the other direction (retrograde
mine. About 1 hour later, her left eyelid elevated, her arm transport).
movements were stronger, her jaw drooped less, swallow- ■ PSPs are excitatory (EPSPs) if they make the postsynaptic cell
ing was improved, and she reported feeling “less heavy.” more likely to initiate an action potential and inhibitory
The effect wore off gradually in 2–4 hours. With 1.3 mg (IPSPs) if they make it less likely.
the improvement was greater and lasted 4–5 hours. Still ■ Neuromuscular transmission is a well-studied example of
greater improvements, lasting 6–7 hours, followed an in- synaptic transmission.
jection of 1.5 mg but the patient felt faint as if “something ■ Hypocalcemia reduces the number of vesicles released when an
were going to happen.” action potential invades the presynaptic terminal.
The diagnosis is myasthenia gravis. It is an autoim- ■ At the neuromuscular junction, the number of open channels is
mune disease that affects about 1 in 5,000 people. The im- proportional to the concentration of ACh squared times the
mune system produces antibodies to the nicotinic AChR effective number of AChR channels.
of the neuromuscular junction and neuromuscular trans- ■ Several clinically important drugs act at the neuromuscular
mission is impaired. With fewer receptors the effects of junction.
depression of synaptic transmission lead to a failure of ■ The number of vesicles released per action potential depends
neuromuscular transmission during sustained effort. This on the rate and pattern of arrival of the action potentials.
fatigability is a characteristic of the disease. ■ The ANS has two synapses outside the central nervous system.
Physostigmine is an inhibitor of AChE. In its presence The first is cholinergic; the second is either adrenergic or
cholinergic.
more of the released ACh can interact with the receptor
■ In general, CNS synapses are similar to the neuromuscular
and transmission is more reliable. AChE inhibitors are
junction, but they differ in many important ways.
used to relieve the symptoms of myasthenia gravis. Im-
■ In the CNS, several transmitters act through G protein–cou-
munosuppressants, for example, the synthetic corticoster-
pled receptors to modulate the activity of the brain.
oid prednisone, are used to reduce antibody production.
■ In order to fire repetitively, nerve cells use Ca-activated K
In some cases thymectomy (surgical removal of the thy-
channels to hyperpolarize the cell and allow the NaV channels
mus) is performed to suppress the immune system.
to recover from their inactivation.
Edrophonium chloride (Tensilon) is a short-acting
■ Learning and memory involve changes in synaptic
AChE inhibitor that has been used to assist in diagnosis.
efficacy.
Electrical stimulation and testing for circulating antibod-
ies are also used diagnostically.

STUDY QUESTIONS
CHAPTER SUMMARY 1. Ca2+ ions are needed in the extracellular solution for synaptic
■ Synapses may be chemical or electrical. Chemical synapses transmission because
may be excitatory or inhibitory. A) Ca2+ ions enter the presynaptic nerve terminal with depolar-
■ In chemical synapses, the presynaptic terminal packages a ization and trigger synaptic vesicles to release their contents
neurotransmitter into vesicles. When the synapse is activated, into the synaptic cleft.
the vesicle’s contents are released and then a recycling process B) Ca2+ ions are required to activate glycogen metabolism in
recovers some of the released transmitter and vesicular the presynaptic cell.
components. C) Ca2+ ions must enter the postsynaptic cell to depolarize it.
D) Ca2+ ions prevent Mg2+ ions from releasing the transmitter
■ ACh is the neurotransmitter at the neuromuscular junction.
in the absence of nerve impulses.
It is also an important component of the autonomic and central
E) Ca2+ ions inhibit the acetylcholine esterase, enabling
nervous system synapses.
the released acetylcholine to reach the postsynaptic
■ Glutamate is the major excitatory neurotransmitter in the CNS. membrane.
78 SECTION II Cell Physiology

2. Inhibitory postsynaptic potentials can arise from all of the 5. Which of the following ions is countertransported to energize
following except neurotransmitter transport into presynaptic vesicles?
A) increased permeability of the nerve membrane A) Na+
to Cl− ion. B) K+
B) direct application of GABA to neurons. C) H+
C) increased permeability of the nerve membrane to K+ ion. D) Cl−
D) increased permeability of the cell membrane to Na+ ion. E) Ca2+
3. Electrical and chemical synapses differ in that 6. A branch of a 26-year-old man’s ulnar nerve was crushed in his
A) electrical synapses have a longer synaptic delay than left forearm, severing axons at a point about about 6 in (15 cm)
chemical synapses. from the skin on the medial part of the palm, where cutaneous
B) chemical synapses can amplify a signal while electrical sensation was lost. About how long will it likely take before the
synapses cannot. patient begins to feel stimuli in that part of the palm?
C) chemical synapses do not have a synaptic cleft while A) 1 day
electrical synapses do have a synaptic cleft. B) 10 days
D) electrical synapses use agonist-activated channels and C) 100 days
chemical synapses do not. D) 1,000 days
E) electrical synapses are found only in invertebrate animals E) never, since peripheral axons do not regenerate
while chemical synapses are found in all animals. 7. Treatments for nerve gas poisoning target which of the following
4. Which one of the following does not contribute to the integration proteins?
of synaptic potentials by neurons? A) acetylcholinesterase (AChE) and choline acetyltransferase
A) convergence of many synaptic inputs on one neuron, (CAT)
allowing spatial summation B) AChE and nicotinic acetylcholine receptors
B) the presence of EPSPs having amplitudes that exceed the C) muscarinic and nicotinic acetylcholine receptors
threshold for generation of an action potential in the D) muscarinic acetylcholine receptors and AChE
neuron E) CAT and synaptic choline transporters
C) temporal summation of synaptic potentials in neurons due
to the time constant of the neurons
D) the flow of currents from the distal regions of the dendrites
to the soma due to the length constants of the dendrites
E) inhibitory synaptic inputs
SECTION III MUSCLE PHYSIOLOGY

8
C H A P T E R

Overview of Muscle
Function
Kathleen H. McDonough

O B J E C T I V E S

■ Explain the differences in skeletal, cardiac, and smooth muscle with


reference to appearance, contractile proteins, calcium-binding proteins,
and neural input.
■ Describe how the sarcolemma and the sarcoplasmic reticulum are involved
in muscle contraction.
■ Explain the energy source for contraction.
■ Describe the role of the following proteins: actin, myosin, dihydropyridine
receptors, ryanodine receptors, calmodulin, and troponin.

GENERAL COMPARISON the overriding unifier is that cytosolic calcium concentra-


tions must increase in order for contraction to occur and
Muscle is the largest organ system of the body. It consists of cytosolic calcium concentrations must decrease for relax-
three different types based on factors such as morphology, cel- ation to occur. Since, in smooth muscle, there is usually
lular signaling pathways, ways to alter strength of contraction, some moderate level of contraction, cytosolic calcium must
contraction pattern (cyclic vs. graded), and the role of the ner- increase for the strength of the contraction to increase and
vous system in muscle function. The three types of muscle are must decrease for the strength of the contraction to de-
skeletal, cardiac, and smooth muscle. Skeletal muscle is, for crease. Thus, the organelles such as the sarcolemma (SL)
the most part, attached to bone and represents approximately and the sarcoplasmic reticulum (SR) that contain proteins
40% of the body mass of a typical healthy person. Cardiac that effect calcium fluxes are highly organized and efficient
muscle is the main component of the heart and contracts in a in muscle. Calcium levels in resting cardiac muscle, for ex-
cyclic fashion for the lifetime of the individual. Smooth muscle ample, are only 0.1 μM and can increase 100-fold during
is the major component of the organs of the gastrointestinal excitation. Removal of calcium for relaxation to occur is
tract, bladder, uterus, airways, and blood vessels—in general, therefore critical. Processes involved in calcium movement
smooth muscle makes up the walls of hollow structures in the are important in the understanding of muscle contraction in
body except the heart. all three types of muscle.
The requirement for calcium to initiate contraction is Muscle contraction is supported by hydrolysis of adenos-
uniform in all muscle. Mechanisms for increasing calcium ine triphosphate (ATP). ATP is produced mainly by
for contraction may vary from muscle type to muscle type mitochondrial oxidative phosphorylation from substrates sup-
and removal of calcium for relaxation also varies. However, plied by glycogen or triacylglycerol stores in the tissue or by

79

Ch08_079-082.indd 79 11/26/10 9:41:53 AM


80 SECTION III Muscle Physiology

blood-borne substrates such as glucose and fatty acids. Glyco- lar smooth muscle in the blood vessels is completely relaxed.
lysis also produces ATP but not as efficiently as oxidative In smooth muscle, gradations of contraction are regulated and
phosphorylation. In some types of skeletal muscle, glycogen affected by many different influences, depending on the loca-
supplies glucose for glycolysis that provides energy for rapid, tion and function of the smooth muscle.
short-term contraction. Energy is additionally provided by
creatine phosphate (CP), which can rapidly supply a source of
CALCIUM
high-energy phosphate bonds for resynthesis of ATP from
ADP. The ADP is produced by ATPase enzymes located in the Although calcium is uniformly required for muscle to con-
muscle cell. CP is not used directly by the ATPase but is used tract or increase strength of contraction, the calcium-binding
for rapid regeneration of ATP at the site of ATP use. ATP is proteins in the three types of muscle differ, as do the sources
used for both contraction and relaxation of muscle. The myo- of calcium. Troponin is the calcium-binding protein that ini-
sin ATPase hydrolyzes ATP to provide the energy for the slid- tiates contraction in skeletal and cardiac muscle. Calmodulin
ing of the actin filament over the myosin filament; ATP also binds calcium in smooth muscle and initiates increases in
provides the energy for removal of calcium from the cytosol by strength of contraction. Actin and myosin form the cross-
calcium ATPases so that relaxation can take place. bridges in all three types of muscle. The source of the calcium
that initiates contraction is different in the three muscle types.
Calcium released from the SR through ryanodine receptors
DIFFERENCES IN SKELETAL, or channels raises the cytosolic calcium concentration and
CARDIAC, AND SMOOTH MUSCLE contraction begins in skeletal muscle (see Chapter 9). In car-
diac muscle, the calcium that binds to troponin comes
Skeletal and cardiac muscle are both striated in appearance from both the SR and the extracellular space through SL volt-
due to the orderly arrangement of the contractile proteins age-gated calcium channels (dihydropyridine receptors).
actin and myosin. In smooth muscle, the contractile pro- In addition, it is the calcium entering the cell through the
teins actin and myosin are responsible for contraction but calcium channels that activates the release of calcium from
they are not arranged in such an organized pattern; there- the SR through the ryanodine receptors or channels (see
fore, striations do not appear. Skeletal muscle is the only Chapter 10). In smooth muscle, calcium can enter the cytosol
muscle type that is voluntary—you decide when to contract from extracellular fluid via the voltage-gated calcium chan-
skeletal muscle for the most part and the muscle has to be nels in the SL and from the SR via receptors activated by sig-
activated by neurons regulated by the central nervous naling molecules from SL receptor pathways. Smooth muscle
system. also has other receptors on the SL and SR for calcium mobili-
Cardiac muscle is involuntary; it contracts spontaneously. zation that will be discussed in Chapter 11. In all three muscle
Action potentials are generated by specialized cells within the types, increases in cytosolic calcium initiate cycling of cross-
heart itself; thus, hearts can be transplanted from one person bridge attachments between actin and myosin.
to another and the heart functions adequately even without
neural input in the transplanted heart of the recipient. The
beating rate of the heart as well as the strength of contraction CONTRACTION PERIOD
of the cardiac muscle cells can, however, be modulated by the
The time course of muscle contraction is different in skeletal,
autonomic nervous system (ANS; see Chapter 19). The sym-
cardiac, and smooth muscle. Skeletal muscle contractions take
pathetic nervous system (SNS) component of the ANS will
several milliseconds to occur, cardiac muscle contractions take
increase the heart rate, whereas the parasympathetic nervous
hundreds of milliseconds, while smooth muscle is much slow-
system (PNS) will decrease the heart rate.
er and can take up to minutes for contractions to occur. This
Smooth muscle is also involuntary. It has the potential to
difference in contraction time is mainly due to the rate of ATP
contract from many different types of stimuli but does not re-
hydrolysis occurring in the myosin head. Fast rates of ATP hy-
quire neural input for contraction to occur. Even changes in
drolysis by the myosin ATPase result in more rapid contrac-
the resting membrane potential and stretch of the muscle can
tions such as in skeletal muscle. Muscles with slower rates of
change the strength of contraction. Smooth muscle does not
ATP hydrolysis exhibit slower contractions such as in smooth
usually exhibit contractions followed by complete relaxation
muscle. Signaling pathways that cause increases in calcium in
as do skeletal and cardiac muscle but rather demonstrates in-
the cytosol can contribute to the delay between the signal and
creased or decreased strength of contraction. For example, if
the contraction.
all of the vascular smooth muscle making up the blood vessels
to the systemic organs were to relax completely, the individual
would go into shock; blood pressure would decrease to dan- CHANGES IN STRENGTH
gerously low levels. This occurs under pathological conditions OF CONTRACTION
such as severe brain injury causing withdrawal of all neural
control of vascular smooth muscle and resulting in neurogenic The strength of muscle contraction can be altered in all three
shock. Blood pressure cannot be maintained if all of the vascu- muscle types but by different means. Phosphorylation of
CHAPTER 8 Overview of Muscle Function 81

proteins results in stronger contractions in both cardiac mus- TABLE 8–1 Comparison of skeletal, cardiac,
cle (Chapter 10) and smooth muscle (Chapter 11), whereas and smooth muscle.
skeletal muscle achieves stronger contractions by recruiting
more muscle cells or activating muscle cells with a higher fre- Skeletal Cardiac Smooth
quency of nerve firing (Chapter 9).
Appearance Striated Striated Nonstriated

Sarcoplasmic Most Less Least


SIMILARITIES IN SKELETAL, reticulum

CARDIAC, AND SMOOTH Voluntary Yes No No

Calcium- Troponin Troponin Calmodulin


MUSCLE binding
protein
As stated above, actin and myosin are the contractile proteins
involved in crossbridge cycling in all three types of muscle al- Source of SR SR and SL SL and SR
calcium
though the anatomic distribution of the actin and myosin is
different in smooth muscle compared to skeletal and cardiac Innervation Motor SNS; PNS SNS; PNS
neuron
muscle (nonstriated vs. striated, respectively). The myosin
head contains a binding site for actin. This site is blocked when Contraction Milliseconds 100 ms 100 ms –
calcium levels are low but open when calcium binds to tro- length minutes
ponin (skeletal and cardiac muscle) or calmodulin (smooth Strength of Recruitment Phosphorylation; Phosphorylation;
muscle). With binding of actin and myosin, the ATPase site contraction length–tension length–tension
located on the myosin head can release energy from the ATP Metabolism Oxidative, Oxidative Oxidative
to allow cycling of the crossbridges, that is, actin sliding across glycolytic
myosin. Velocity of Rapid Less rapid Slow
All three types of muscle demonstrate the property of in- ATPase
creasing strength of contraction by increasing the precontrac- reaction
tion (resting) length of the muscle. This phenomenon is
termed the length–tension relationship. Since skeletal mus-
cle is attached to bones by tendons, variations in resting cell ■ Muscle can increase strength of contraction by increasing the
length are very limited and the muscle is usually operating at length of the muscle prior to the contraction—length–tension
the peak of the length–tension relationship. In the heart, rest- relationship.
ing muscle cell length is usually not at the optimum length, so ■ The energy for contraction is released from ATP by the
there is reserve, that is, stronger contractions can be elicited myosin ATPase.
when the resting length is increased prior to the contraction. ■ Skeletal muscle requires neural input from a motor neuron to
Smooth muscle also demonstrates the length–tension rela- initiate contraction (voluntary muscle).
tionship but other influences on the muscle can override the ■ Cardiac and smooth muscle can contract without neural input
effects of increased cell length. For example, in certain types but the strength of contraction can be altered by input from the
of vascular smooth muscle, when the cell is stretched, the cell ANS—sympathetic and parasympathetic branches of the ANS
responds with an increased level of contraction. This phe- (involuntary muscle).
nomenon is called the myogenic response. In the gastrointes-
tinal tract, this occurs with the presence of food in the stomach STUDY QUESTIONS
and small intestines. Hollow organs with specialized functions
such as the bladder and uterus can be “stretched” but contrac- 1. Which of the following statements about muscle is true?
tion is not stimulated due to other influences on muscle A) The calcium source for skeletal muscle contraction is solely
function. calcium entering the cell through the dihydropryidine
A comparison of skeletal, cardiac, and smooth muscle is receptors.
B) The calcium source for smooth muscle contraction is solely
given in Table 8–1. More details on the individual muscle types
calcium entering the cell through the dihydropyridine
will be given in the next three chapters. receptors.
C) The calcium source for cardiac muscle contraction is solely
CHAPTER SUMMARY calcium entering the cell through the dihydropryidine
receptors.
■ There are three types of muscle in the body classified by D) The calcium source for skeletal muscle contraction is
morphology, function, and cellular mechanisms of solely calcium entering the cytosol through the ryanodine
contraction—skeletal, cardiac, and smooth. receptors.
■ All types of muscle require calcium to initiate contraction. E) The calcium source for cardiac muscle contraction is
■ SL and SR have specialized functions that increase cytosolic solely calcium entering the cytosol through the ryanodine
calcium for contraction and remove calcium for relaxation. receptors.
82 SECTION III Muscle Physiology

2. Which of the following statements about muscle is true? 4. Which of the following statements about muscle is true?
A) Both smooth and cardiac muscle remain partially A) In all three muscle types (cardiac, skeletal, and smooth)
contracted at all times. all cells contract as a unit.
B) Both cardiac and skeletal muscle contraction is initiated by B) All three muscle types are innervated by the autonomic
calcium binding to troponin. nervous system.
C) Both cardiac and smooth muscle must have action C) In all three muscle types, calcium is involved in contraction.
potentials to initiate contraction. D) In all three muscle types, dihydropryidine antagonists or
D) Both cardiac and smooth muscle initiate contraction by blockers increase strength of contraction.
calcium binding to troponin.
3. Which of the following statements about muscle is true?
A) Skeletal muscle can increase strength of contraction by
recruiting more motor units.
B) Cardiac muscle can increase strength of contraction by
recruiting more muscle cells.
C) Smooth muscle cannot change strength of contraction.
D) Cardiac muscle cannot change strength of contraction.
9
C H A P T E R

Skeletal Muscle
Structure and Function
Kathleen H. McDonough

O B J E C T I V E S

■ Describe the processes that take place at the neuromuscular junction.


■ Explain excitation–contraction coupling in skeletal muscle.
■ Describe the role of the proteins that are involved in contraction.
■ Describe what happens during an isometric contraction.
■ Describe what happens during an isotonic contraction. How does the load
affect shortening and velocity of shortening?
■ Explain how muscle fiber strength of contraction can be increased by
summation and tetanus.
■ Explain the length–tension relationship in skeletal muscle.
■ Explain the motor unit.
■ Explain how whole muscle strength of contraction can be increased by
recruitment of motor units.
■ Explain the force–velocity relationship in skeletal muscle; explain the basis for
the Vmax.
■ Describe the three different types of skeletal muscle fibers and the bases for
their differences. State when these fibers are recruited.

STRUCTURE over the myosin filament. The A band remains the same length
(myosin does not shorten) but the I band becomes smaller as
Skeletal muscle is distinctive because of its anatomic actin is pulled over the myosin. The sliding of the actin over
structure—striations due to the regular pattern of sarcomeres the myosin, with energy provided by the myosin ATPase that
that are composed of the orderly positioning of the actin and is located on the myosin head, is the molecular basis of skeletal
myosin proteins. Figure 9–1 shows sarcomeres composed of muscle contraction (Figure 9–2). Activation of the complex
parallel alignments of thick filaments (i.e., myosin) and thin occurs when the calcium concentration in the cytosol increases
filaments (i.e., actin, tropomyosin, and troponin). Myosin and binds to the calcium-binding site on the troponin. Tro-
makes up the A band. Actin, along with the two other proteins, ponin has three components designated as TnT that attaches it
tropomyosin and troponin, makes up the I band (portion of to tropomyosin, TnI that inhibits interactions between actin
the sarcomere where actin does not overlap with myosin). Part and myosin, and TnC that binds the calcium. When calcium
of the actin filament overlaps with the myosin filament, thus binds to TnC, there is a conformational change in the troponin/
allowing interaction of these two proteins to initiate contrac- tropomyosin position, removing the hindrance by TnI and tro-
tion. The degree of overlap of thick and thin filaments is pomyosin and allowing the actin and myosin head to interact,
important in determining the amount of force that skeletal thereby hydrolyzing ATP to supply the energy for the
muscle, and cardiac muscle, can generate. The Z lines repre- contraction—sliding of the actin over the myosin or cross-
sent the borders of the sarcomere and, during shortening, the bridge cycling. The cross-bridges will continue to cycle, that
Z lines come closer to each other as the actin filament is pulled is, myosin heads will continue to bind to adjacent sites on actin

83

Ch09_083-092.indd 83 11/26/10 9:41:31 AM


84 SECTION III Muscle Physiology

(a) Sarcomere

I band A band

(b) H zone

Z line Z line
Titin Thin filament M line Thick filament

FIGURE 9–1 (a) Magnified section of a sarcomere within a skeletal muscle cell showing the pattern of striation due to the
orientation of the actin and myosin filaments. (b) Drawing of the components of the sarcomeres from Z band to Z band showing the
structural protein titin and the thick filaments (myosin) and the thin filaments (actin, tropomyosin, and troponin). (Reproduced with permission
from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

Thick filament Cross-bridge

Thin filament
(a)
Actin binding sites

ATP binding sites

Light chains
Tropomyosin
Heavy chains Cross-bridge

Troponin Actin Myosin


(b)

FIGURE 9–2 (a) Drawing of the thick filament with the myosin heads or cross-bridges extending from the thick filament. Also shown
is the twisted structure of the thin filaments. (b) Magnification of the myosin and actin showing the three components of the thin filament—
actin, tropomyosin, and troponin—and the heavy and light chains of the myosin. Note the actin- and ATP-binding sites on the myosin. The
actin-binding sites are blocked by tropomyosin when calcium levels in the cytosol are low. With calcium binding to troponin, tropomyosin is
moved away and the binding site for actin is available. The energy for sliding the actin filament across the myosin filament is provided by the
ATP hydrolyzed by the myosin ATPase that is located on the myosin head. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human
Physiology, 11th ed. McGraw-Hill, 2008.)
CHAPTER 9 Skeletal Muscle Structure and Function 85

and slide the actin further over the myosin, until the contrac- fibers. Duchenne muscular dystrophy is one type of dystro-
tion is terminated by removal of calcium. phy in which there is a complete absence of the dystrophin
Cross-bridge cycling results in either tension development protein resulting in rapid decline in skeletal muscle function
or shortening or a combination of the two, depending on the and early death.
load on the muscle. If the load is too great, there will be
an isometric contraction in which there is tension develop-
ment but no shortening of the muscle. If the load is less, there
will be an isotonic contraction in which the muscle short-
NEUROMUSCULAR JUNCTION
ens after developing tension (discussed in more detail in Skeletal muscle cells or fibers generally extend from one ten-
Chapter 10). Other proteins are involved in maintaining the don to the other tendon that attaches the muscle to the bones.
precise structure of the sarcomeres. Titin, a large structural Skeletal muscle is classified as voluntary muscle since its
protein in the skeletal muscle cell, extends from the Z line to contraction is mandated by the central nervous system—we
the center of the sarcomere, stabilizing the structure. Another can contract muscles at will. Thus, the innervation of skeletal
large protein complex consists of dystrophin and several gly- muscle is essential for activation of contraction. Each fiber is
coproteins. This complex is instrumental in attaching the activated by one motor neuron, whereas one motor neuron
sarcomere, in particular, actin, to the sarcolemma (SL) and can innervate a number of muscle fibers forming a motor
the extracellular matrix, again for maintaining structure and unit. When one motor neuron is activated, all of the fibers
stability of the sarcomeres. The gene that codes for the dys- innervated by that motor neuron will contract. The motor
trophin complex is large and subject to mutations resulting neurons from the spinal cord or brainstem, in response to
in disorders of skeletal muscle termed muscular dystrophy. action potentials traveling down the axon toward the skeletal
One symptom of this disease is progressive muscle weakness muscle cell, release the neurotransmitter acetylcholine as
due to the loss of the proper structural integrity of the muscle shown in Figure 9–3 at the neuromuscular junction.

1 Motor neuron
action potential

Acetylcholine
vesicle
2 Ca2+ enters 8 Propagated action
voltage-gated potential in muscle
channels plasma membrane

Voltage-gated
3 Acetylcholine Na+ channels
release
+ + + + + + + +
+
– – – – + 4 Acetylcholine binding – – – –
+ – opens ion channels 5 Na+ entry – +
+
– + + + + + +
+ +
– – – – – – –

9 Acetylcholine 7 Muscle fiber
Acetylcholine receptor
degradation action potential
Acetylcholinesterase initiation

Motor end plate 6 Local current between


depolarized end plate and
adjacent muscle plasma
membrane

FIGURE 9–3 The neuromuscular junction is the specialized part of the muscle cell—motor end plate—at which the motor
neuron releases the neurotransmitter acetylcholine to activate the muscle cell or fiber. Events at the junction are listed in chronological
order. Note that each muscle fiber receives impulses from only one motor neuron and all of the fibers receiving input from that motor neuron
make up the motor unit and will contract in synchrony. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed.
McGraw-Hill, 2008.)
86 SECTION III Muscle Physiology

The amount of acetylcholine released is proportional to the Sarcolemma T tubule


frequency of action potentials.
Acetylcholine diffuses across the synaptic cleft and binds to
a cholinergic receptor—the nicotinic receptor—on the mus-
cle cell membrane (SL). The part of the muscle SL that is asso-
ciated with the neuromuscular junction is called the motor
end plate. Within the cleft is the enzyme acetylcholine
esterase that can hydrolyze unbound acetylcholine and thereby
limit the activation of the muscle cell membrane nicotinic Ca ATPase
receptors. This receptor is a channel that allows sodium and
potassium flux. The predominant ion movement is sodium
entering the muscle cell causing partial depolarization of the Sarcoplasmic Ryanodine Dihydropyridine
cell membrane in the synaptic cleft—an end plate potential reticulum receptor, receptor
(see Chapter 7). Since motor neurons cause only depolariza- cisterna channel
tion of the postsynaptic membrane and not necessarily an FIGURE 9–4 The connection between the sarcolemmal (SL)
action potential, the change is similar to an excitatory post- T-tubules and the cisternae of the sarcoplasmic reticulum (SR) is
synaptic potential (EPSP) occurring in neurons. the mechanism for the coupling of the action potential traveling
along the SL to the release of calcium from the SR. The connection
between the calcium channels (dihydropyridine receptors) on the SL
EXCITATION–CONTRACTION and the ryanodine receptors on the SR is altered by the action
COUPLING potential allowing opening of the SR calcium channels and release of
calcium into the cytosol.
This depolarization is conducted to the SL outside of the neu-
romuscular junction and if strong enough will induce an action and continuous activation of the skeletal muscle. Eventually
potential. The action potential is transmitted along the SL, into the cells can no longer generate action potentials because the
the T-tubules, which are invaginations of the SL that allow the cells remain depolarized and sodium ion channels that would
cell membrane to come in close contact with an intracellular normally open and initiate depolarization are inactivated.
membrane system called the sarcoplasmic reticulum (SR; Muscle weakness ensues and since the diaphragm contains
Figure 9–4). In skeletal muscle, the T-tubule makes contact skeletal muscle, respiratory failure leads to death. Autoim-
with two components (cisternae or lateral sacs) of the SR mune diseases such as myasthenia gravis can result in pro-
forming a triad. Calcium is released from ryanodine recep- duction of antibodies to the nicotinic cholinergic receptor.
tors located on the cisternae when the T-tubule is depolarized Binding of antibodies to the receptors results in impaired sig-
during an action potential. Dihydropyridine (DHP) recep- naling or communication between the motor neuron and the
tors, also known as calcium channels, on the SL cause a con- skeletal muscle fibers. Contractions are impaired and with
formational change in the ryanodine receptors causing them time the entire structure of the motor end plate deteriorates.
to open and allowing calcium to diffuse from the SR into the Degeneration of the motor neuron, which is required to initi-
cytosol. Contraction is initiated when calcium levels in the ate contraction, results in diseases such as amyotrophic lateral
cytosol reach a critical level and bind to TnC. In skeletal muscle, sclerosis (ALS or Lou Gehrig’s disease). The motor neuron
all of the calcium used for contraction is released from the SR. shrinks and degenerates, leading to denervation of the muscle
For relaxation to occur, calcium must be returned to the SR. As cells and resulting in impaired ability of skeletal muscle to
seen in Figure 9–4, the SR consists of longitudinal components contract. Eventually the muscle cells atrophy. An early symp-
as well as the cisternal component. The longitudinal portion of tom of ALS is muscle weakness.
the SR contains the calcium ATPase enzyme, referred to as
SERCA. SERCA has a high Vmax and, using the energy from
ATP, pumps calcium against its concentration gradient into FUNCTION
the SR. Proteins such as calsequestrin within the SR bind cal-
cium, providing a storage function in the SR but also maintain- TYPES OF CONTRACTIONS
ing an optimal free calcium concentration so that the calcium
gradient for pumping calcium out of cytosol and back into the Contraction can occur in two modalities: isometric and iso-
SR is not excessive. This process in which an action potential tonic and combinations thereof. Isometric, as its name
leads to increased calcium leading to contraction is termed implies, refers to contractions in which the length (metric) of
excitation–contraction coupling. the muscle stays the same (iso) but tension or force increases.
Due to the complexity of the neuromuscular junction, many Figure 9–5 shows the schematic of the apparatus for measur-
disease states can occur when there is dysfunction. For exam- ing the output of isometric contractions. A thin skeletal mus-
ple, nerve gases inhibit the acetylcholine esterase, thereby cle strip is suspended between a force transducer and an
resulting in continuous activation of the nicotinic receptors immovable bar. Since the muscle is tethered at both ends,
CHAPTER 9 Skeletal Muscle Structure and Function 87

Isometric contractions Force


transducer
Isotonic contractions

Muscle 100

75
Stimulator
Muscle Stimulator
50
length

25

0
Force transducer Load (preload)
Afterload
FIGURE 9–5 Isolated muscle preparation to study isometric
muscle contractions. The muscle is not allowed to shorten. The
FIGURE 9–6 Isolated muscle preparation to study isotonic
contractions. The passive tension is set by the preload, and the
passive tension on the muscle as a function of resting muscle length
muscle length is measured. A bar is place under the muscle so that
is measured with a force transducer and then the muscle is stimulated
when the afterload is added, the muscle does not lengthen (does
to contract.
not sense the afterload). On stimulation, the bar is removed and the
muscle develops tension to just match the afterload. During the
remainder of the contraction, the tension remains constant and the
when the muscle is stimulated, cross-bridge cycling results muscle shortens. The length of the muscle and the rate of shortening
only in tension (dyne/cm) or force (dyne) development. The are measured.
length of the muscle does not change during contraction.
Isotonic refers to contractions in which the tension (tonic)
stays the same but the length changes. Prior to the shorten-
ing, however, the muscle must increase tension or force to
exceed the load it is lifting or contracting against; thus, the
B C A B C
contraction consists of tension development followed by
Shortening

shortening. Figure 9–6 shows the apparatus for measuring CE


CE
isotonic contractions. The change in muscle length (short- CE
ening) can be measured after the muscle is stimulated.
There are two loads on the muscle: (1) the preload that sets
SE
the resting muscle length and (2) the afterload that the
Tension

SE
muscle does not sense until after the contraction begins. In Load SE
the protocol, the preload is added to the muscle strip, the L
passive or resting length is established, and then the hori- Time L L
zontal bar is placed under the muscle such that addition of Stimulation L = Load

the more weight, the afterload, does not allow the muscle to
FIGURE 9–7 A model of skeletal muscle consists of the
lengthen anymore. When the stimulator excites the muscle, contractile element (CE) made up of the thick and thin filaments,
the bar is removed and the muscle contraction results in the and the series elastic (SE) component that consists of
muscle generating force to equal the afterload and then noncontractile components of the muscle. Phase A is the muscle
shortening. at rest. Using the model to represent an isotonic contraction, after
Figure 9–7 shows a model for skeletal muscle contraction. stimulation, the muscle develops tension (phase B) and stretches the
Muscle consists of the contractile element (CE; the contractile series elastic component, that is, tension is developed (to match
proteins actin and myosin) and a series elastic component. The the load) but the whole muscle does not shorten. Note the contractile
load can be considered the weight the muscle must lift in an element is shortening, that is, cross-bridges are cycling and actin
isotonic contraction. Note that prior to shortening, the CE is filament is being pulled over the myosin, but the whole muscle is not
shortening. At point C, the tension developed by the contractile
getting smaller, that is, the cross-bridges are cycling and pulling
element stretching the series elastic component just exceeds the
actin across myosin, but the entire muscle is not shortening—
afterload and during the remainder of the contraction, the cross-
the cross-bridge cycling is generating tension (Figure 9–7, B). bridge cycling is actually shortening the whole muscle. The load
When the tension or force matches the load (afterload), the on the muscle determines how much tension the muscle will have to
remainder of the cross-bridge cycling (contraction) results in develop in order to shorten and lift the load. (Reproduced with permission
shortening of the muscle (Figure 9–7, C). Note that the afterload from Sonnenblick EH: The Myocardial Cell: Structure, Function and Modification. Briller
determines how much tension the muscle will have to generate SA, Conn HL (editors). University of Pennsylvania Press, 1966.)
88 SECTION III Muscle Physiology

prior to shortening. A heavier load will require more tension relationship will also be discussed in Chapter 10 with reference
development, and a lighter load will require less tension devel- to cardiac muscle contraction.
opment. With a heavier load and more tension to develop, the
muscle will exhibit less shortening. With a lighter load, and less
tension to develop, the muscle will exhibit more shortening. REGULATION OF CONTRACTION—
When afterload is plotted on the x-axis and shortening LENGTH–TENSION
velocity is plotted on the y-axis, an inverse relationship is dem-
onstrated—the force–velocity curve (Figure 9–8). Where the The type of contraction, isometric versus isotonic, is deter-
curve intersects with the x-axis, there is no shortening (zero mined by the loading conditions on the muscle. If the muscle
velocity of shortening)—this is an isometric contraction— is not allowed to shorten, tension development is the total out-
maximum force is developed but there is no shortening. If come of cross-bridge cycling resulting in an isometric contrac-
afterload is decreased—red circle—less force must be devel- tion. For example, pulling on an immoveable object results in
oped and some shortening occurs and therefore the velocity of an isometric contraction—the muscle is developing tension
shortening can be depicted. If the afterload is decreased again, but cannot shorten. The amount of force generated during
still less force is developed and even more shortening occurs that contraction (twitch) is determined by the amount of cal-
and velocity of shortening increases. At the y-axis intercept, cium released from the SR. Under normal circumstances, the
there is the maximum velocity of shortening—Vmax. Note the amount of calcium released in response to an action potential
dashed line connecting the curve to the y-axis—this denotes is maximal in skeletal muscle fibers.
that the intercept is an extrapolation of the curve—one cannot The length (preload) of the muscle prior to the contraction
study a contraction in a muscle with zero load; therefore, Vmax also affects the strength of the contraction. The length of the
is an estimate of the maximum velocity of shortening. One muscle fibers prior to contraction determines how much
other fact to note is that shortening and velocity of shortening overlap there will be between actin and myosin and, thus,
are changing in the same direction—increases in shortening how many cross-bridges can be formed. Since the energy for
occur with increases in shortening velocity. The force–velocity the contraction is released by the myosin ATPase activity,
altering the number of cross-bridges that interact alters the
amount of myosin ATPase that is activated and thus the
amount of ATP that will be hydrolyzed to provide the energy
Vmax
for contraction and relaxation. This has a significant effect
on the strength of the contraction. As seen in Figure 9–9,
length of the muscle (preload) affects developed tension,
passive tension, and total tension. In skeletal muscle, pas-
Velocity sive tension is low until the Po point at which it begins to
increase substantially. Total tension increases as a function of
muscle length as does active or developed tension. Active
tension is the tension that is developed during contraction by

Po Total
Force or load
FIGURE 9–8 The force–velocity curve is generated from the
study of isolated muscle during isotonic contractions. To Active or developed
Tension,
generate a typical curve, the preload on the muscle is held constant,
dynes/cm
that is, the resting length is the same for each contraction studied,
but the afterload is varied. At the intercept of the x-axis, the greatest
afterload, there is no shortening—this represents a maximal Passive
isometric contraction (Po). As the load decreases—the red point, less
tension must be developed to match the afterload and therefore
some shortening can occur. With more shortening there is a greater Po
initial velocity of shortening that is plotted on the y-axis. With a
Length, mm
further decrease in afterload to the light red point, there is even less
tension developed and even more shortening can occur, so a greater FIGURE 9–9 The relationship between the length of muscle
velocity of shortening occurs. The green point represents an even (set by the preload on the isolated muscle) and the tension that
lighter afterload and therefore an even greater velocity of can be measured is shown. The active or developed tension is the
shortening. The curve is extrapolated to the intercept of the y-axis difference between the total tension and the passive tension. It is the
that yields the maximum velocity of shortening (Vmax). This is a tension that the muscle produces during the contraction. At Po the
theoretical point because the muscle cannot be studied under muscle is at the optimum length to give the greatest tension—
conditions of zero load. maximum isometric tension.
CHAPTER 9 Skeletal Muscle Structure and Function 89

cross-bridge cycling and is therefore the difference between Tetanic contraction


total tension and passive tension. Passive tension is due to the
structural properties of skeletal muscle. Skeletal muscle dem- Summation
onstrates the length–tension relationship but in the body,
A B C D
since most of the skeletal muscle is attached to the bone by
tendons, the optimum length is generally set by anatomy.
E

REGULATION OF STRENGTH OF Tension


Twitch
CONTRACTION IN SKELETAL MUSCLE—
RECRUITMENT, SUMMATION, AND
TETANUS
The physiological way for intact skeletal muscle to increase Single 300 msec 120 40–50 60 stimuli 1
tension is via changes in the stimulation pattern by the motor stimulus delay msec msec per sec msec
neurons. Spatial recruitment refers to increased numbers of delay delay delay
motor neurons firing, and, therefore, more motor units con- FIGURE 9–10 Muscle tension developed during
tracting. Temporal recruitment refers to increased number of contractions performed under different patterns of stimulation
action potentials in a motor neuron thereby affecting the con- is shown. (A) With a single stimulus, one twitch occurs. (B) With two
traction of the muscle fibers within that motor unit. Within a stimuli that are 300 milliseconds apart, two identical twitches occur.
muscle, usually only a small percent of the muscle cells or (C) When the second stimulus occurs prior to complete relaxation
fibers will contract at any one time but the contraction of each from the first twitch, the second contraction shows greater tension
fiber will be maximal. All of the muscle fibers innervated development. (D) When the two stimuli are approximately 40–50
by the same motor neuron will contract at the same time. The milliseconds apart, there appears to be only one contraction but the
tension is 2–3-fold greater than that with one stimulus. When the
strength of the contraction of the entire muscle increases if
muscle is stimulated with a rapid burst of stimuli (60 stimuli/s),
more motor neurons are activated and therefore more muscle
tetanus occurs—the greatest amount of tension is developed and
fibers are stimulated to contract—spatial recruitment. The there is no relaxation. During this stimulation pattern, the release
order of recruitment of motor units will be discussed below of calcium with each stimulus outstrips the calcium uptake
with the presentation of muscle fiber types. mechanisms, so cytosolic calcium remains elevated and relaxation
Temporal recruitment results from increasing the number does not occur. Depending on the muscle type, tension will remain
of action potentials in the motor neuron. In Figure 9–10, curve elevated until the stimulation ends or until fatigue develops and the
A shows the contraction or twitch in response to one stimulus. muscle can no longer maintain tension. (E) When the two stimuli are
More rapid firing (more action potentials per second) repeti- approximately 1 millisecond apart, the contraction looks identical
tively releases acetylcholine to activate nicotinic receptors giv- to the contraction given after one stimulus—the muscle cannot
ing more action potentials in the muscle membrane. If two respond to the second stimulus because it is in the refractory
period—the muscle cell membrane is not responsive to a normal
stimuli are far enough apart (e.g., 300-millisecond delay as
stimulus.
shown for B in Figure 9–10), two separate identical contrac-
tions occur. When the two stimuli are approximately 40–50
milliseconds apart, the muscle contraction appears to be one become identical to the single stimulus–initiated contrac-
twitch but the strength of the contraction is greater than that tion—all fibers will have become refractory to the second
generated by a single stimulus (curve D, Figure 9–10). This stimulus—responding only to the first stimulus. This gener-
response is called summation. The mechanism for summa- ally occurs with stimuli that are 1–2 milliseconds apart (E).
tion is that the second contraction begins prior to the begin- The maximum tension that can be developed occurs during
ning of relaxation of the first contraction. Therefore, the total tetanic contractions (Figure 9–10). The basis for this increase
contraction is measurable tension development with no energy in tension is that there are so many action potentials (e.g., 60/s)
spent in overcoming the series elastic component or the resis- that the calcium release mechanisms occurring with every
tance to contraction by all of the “noncontractile elements” action potential outstrip the calcium uptake mechanisms;
present in the muscle. If the stimuli are more than 40–50 mil- thus, cytosolic calcium levels remain elevated continuously
liseconds apart (curve C), the first contraction begins to wane and the muscle does not relax in between stimuli. The
prior to initiation of the second contraction giving a biphasic cross-bridges continue to cycle either until stimulation stops
appearance to the contractions. The optimum delay between and cytosolic calcium concentrations decrease or until the
two stimuli may vary in different types of skeletal muscle but cells fatigue. Both summation and tetanus (tetanic contrac-
the capacity to increase force by summation is present in all tions) are examples of temporal recruitment—the same fibers
skeletal muscle. As the delay between the stimuli becomes are stimulated to contract by the same motor neurons but the
smaller and smaller, the contraction becomes weaker and frequency of stimulation by the neuron alters the muscle
weaker until eventually the “summated” contraction will response. In summary, strength of contraction of an intact
90 SECTION III Muscle Physiology

muscle made up of many different motor units can be increased neurons that are also the smallest in diameter. This makes both
by: (1) increasing the number of motor neurons activated, the neurons and the fibers easy to activate. These small fibers
thereby increasing the number of motor units contracting; and that are recruited earliest have a high oxidative capacity and
(2) increasing the frequency of action potentials of the motor can perform work for long periods of time without fatiguing.
neuron, thereby eliciting summation or tetanus of those mus- Adequate blood flow and high concentrations of mitochondria
cle fibers in the motor unit. for oxidative metabolism allow these fibers to contract for
hours. These fibers have been called slow twitch because their
myosin ATPase activity is low. These fibers also contain myo-
FIBER TYPES globin, a heme-containing protein that binds oxygen and can
therefore serve as an oxygen store that can be used when oxida-
As mentioned above, the strength of contraction of skeletal tive phosphorylation is occurring at elevated rates to support
muscle can be increased by spatial recruitment. Spatial elevated rates of contraction. These type I fibers were classified
recruitment occurs when more motor neurons participate in a as “red” fibers in the past because the high concentration of
contraction, thus “recruiting” more motor units, that is, more myoglobin gives color to the muscle fibers.
muscle fibers to contract. Type II fibers have a higher myosin ATPase activity and
There are three basic types of muscle fibers in skeletal therefore a faster rate of contraction. There are two subtypes in
muscle—type I, type IIa, and type IIb (Table 9–1). These were this group—type IIa are fibers with a fast twitch and both oxi-
formerly called red and white muscle for the color imbued by dative and glycolytic capacity, and type IIb are fibers with a
the presence of myoglobin and many mitochondria in red fast twitch but rely almost entirely on glycolysis for ATP pro-
muscle and little myoglobin in white muscle. Since the red duction. The type IIb fibers have high concentrations of the
muscle has many mitochondria, it has the capacity, by oxida- enzymes involved in glycolysis. These fibers have the largest
tive phosphorylation production of ATP, to sustain contrac- diameter and are recruited last. They are innervated by motor
tions over long periods of time. neurons with large diameters that require a greater stimulus in
The fiber types have different diameters as do the motor order to generate an action potential, thereby making them
neurons that innervate them. the last to be recruited. They are more likely to fatigue than the
The pattern of spatial recruitment is governed by the size of other types of fibers due to the dependence on glycogen as a
the muscle fibers with smallest fibers being most easily recruited substrate for ATP to provide the energy for contraction. The
(earliest recruitment), and largest fibers recruited last. The type supply of glycogen is limited and since they have a relatively
I fibers have the smallest diameter and are innervated by motor sparse blood supply, glucose may not be as readily available. If

TABLE 9–1 Comparison of skeletal muscle fiber (cell) types.


Type I Type IIa Type IIb

Metabolism Oxidative Oxidative/glycolytic Glycolytic

Twitch Slow Intermediate Fast

Mitochondria Abundant Intermediate Few

Myoglobin Abundant Abundant Few

Color Red Red White

Glycogen Little Intermediate Abundant

Myosin ATPase rate of Lowest Fastest Fastest


hydrolysis of ATP

Speed of contraction Slowest Intermediate Fastest

Blood flow Great Intermediate Low

Fatigue Not readily Intermediate Rapid onset

Force Least Intermediate Greatest

Size of motor neuron Smallest Intermediate Largest

Size of fiber Smallest Intermediate Largest

Recruitment First Second Last

Total tension Least Intermediate Greatest


CHAPTER 9 Skeletal Muscle Structure and Function 91

glycolysis results in lactic acid production because oxygen is


not readily available, the cells will fatigue in a matter of min- (eyelid drooping) occurred with repeated, rapid eye
utes and decrease tension development in spite of repeated movements. The physician suspects myasthenia gravis
motor neuron firing. These fibers were formerly classified as and orders tests to confirm the diagnosis.
“white” fibers because they had lower levels of mitochondria, Myasthenia gravis is an autoimmune disease in which
myoglobin, and blood flow—all of which lend the red color to the immune system produces antibodies to the nicotinic
the type I fibers. The type IIa fibers are of intermediate size receptor. Initially small motor units, especially in ocular
and therefore are recruited after the type I slow fibers are acti- muscles for eye movement, demonstrate the defect. Rapid
vated. Type IIa fibers can use both glycolysis and oxidative motor neuron firing for rapid muscle contractions for eye
phosphorylation for their energy supply and therefore also movement eventually leads to release of less acetylcholine
exhibit an intermediate time course for fatigue to occur. The (production lags behind release). In normal individuals,
ATPase activity and therefore speed of contraction is fast as in there are adequate receptors to compensate for the
the type IIb fibers and the time for fatigue to occur is interme- decreased amount of acetylcholine released. With myas-
diate. The capacity for oxidative phosphorylation to provide thenia gravis, antibodies bound to the receptors prevent
some of the ATP for contraction prolongs the time of sustained the acetylcholine binding, thus leading to impaired
contraction before fatigue occurs. muscle contractions. Rest can replenish the acetylcho-
In general, the diameter of the muscle fiber is indicative of line stores. Antibodies bound to the nicotinic receptor
the amount of actin and myosin in the fiber. Therefore, larger- seem to trigger an immune response and degeneration
diameter fibers have more actin and myosin, more cross- of the muscle motor end plate. Eventually, with more
bridges that can form, and can develop more tension. Smaller antibody production, more muscle units become
fibers develop less tension due to lower amounts of actin and involved and can eventually lead to large muscle weak-
myosin but again can contract for prolonged period due to ness including impaired respiratory muscle function.
their abundant blood supply and oxidative capacity. Treatments to decrease antibody production, often
Most muscles in the body are made up of combinations of exacerbated by the thymus gland, include removal of
the three muscle fiber types with a predominance of either fast the thymus and treatment with immunosuppressive
or slow fibers. Muscles involved in maintaining posture must drugs such as corticosteroids. Cholinesterase inhibitors
have long-lasting capacity to contract and not to fatigue, so are also used since they inhibit the enzyme that hydro-
these have more of the type I, slow-twitch fibers. Of course, in lyzes acetylcholine at the neuromuscular junction,
maintaining posture, not all muscle fibers will be contracted at thereby maintaining higher concentrations of acetyl-
any one time but different motor units will take over contrac- choline and greater stimulation of the motor end plate.
tion cyclically. Muscles that are involved in rapid changes such
as eye movements are predominantly type IIb fibers—fast
contractions that are not sustained for long periods of time. CHAPTER SUMMARY
Many muscles have intermediate amounts of the different fiber
■ Skeletal muscle cells are also called fibers.
types. For example, people who do prolonged activities such as
■ Skeletal muscle cells have a specialized section of SL called the
endurance running have slower-twitch, oxidative fibers in
motor end plate where the motor neuron forms a synapse with
their muscle responsible for running. Sprinters have more the muscle. Acetylcholine is the neurotransmitter and nicotinic
fast-twitch, glycolytic fibers that are best for bursts of activity receptors on the SL bind the acetylcholine and increase sodium
but not for sustained activity. influx, causing partial depolarization, and eventually an action
potential in the adjacent SL.
■ The action potential travels down the invaginations of the SL
(T-tubules) and, via the dihydropyridine receptors, causes the
CLINICAL CORRELATION ryanodine receptors to open and release calcium.
A 45-year-old woman notices that she has been feeling ■ Troponin binds calcium and begins the contraction process.
unusually tired after work for the past month. She also ■ The motor neuron can innervate more than one skeletal
notices that her left eyelid begins to droop at the end of muscle fiber—the motor neuron and the fibers it innervates
the day. Gradually she is noticing that the eyelid begins to are called a motor unit. All of these muscle cells contract at the
droop even by the end of the work day if it has been a same time.
particularly stressful day. She is also experiencing more ■ Contraction can be either isometric or isotonic.
and more severe fatigue but both of these problems are ■ In isotonic contractions, the load determines how much
gone after a good night’s sleep. She is concerned and tension or force the muscle must develop before the shortening
makes an appointment with her physician. On physical phase of the contraction can occur.
examination, her physician notes that all measured vari- ■ Skeletal muscle can increase strength of contraction by
ables were in the normal range except for movement of recruiting more motor units (spatial recruitment).
the left eye. Lateral movement was impaired and ptosis ■ The motor units with the smallest diameter of the neurons
and fibers are recruited most readily (first). These are the
92 SECTION III Muscle Physiology

type I fibers that are highly oxidative and have a low 2. Which of the following statements about muscle contraction is
myosin ATPase rate and therefore exhibit a slower contrac- true for skeletal muscle?
tion time. A) All cells have pacemaker potential.
■ Type II fibers have a high myosin ATPase rate and therefore a B) The strength of contraction is correlated with the degree of
faster contraction time. Type IIa are both glycolytic and phosphorylation of the myosin light chains.
oxidative and are recruited second. Type IIb are mainly C) The strength of contraction is increased by recruiting more
glycolytic and recruited last (fibers and neurons have the motor units.
greatest diameter). D) All muscle cells have a high oxidative capacity due to the
■ Type II fibers fatigue more quickly than do type I fibers, with abundant presence of mitochondria and myoglobin.
type IIb fibers fatiguing most rapidly—in a few minutes after 3. Which of the following statements about muscle contraction is
repeated stimulation. true for skeletal muscle?
■ Skeletal muscle can also increase strength of contraction by A) In the body, strength of contraction is altered physiologi-
more rapid firing of the motor neuron—summation and cally by changing the resting cell length from 25% up to
tetanus. 100% of the maximum length.
B) Strength of contraction is altered physiologically by altering
the frequency of motor neuron firing.
STUDY QUESTIONS C) Tetanus cannot occur because the muscle action potential
keeps the cell refractory to stimuli that are closer than 1
1. Which of the following statements about skeletal muscle second apart.
contraction is correct? D) Muscle contraction consists only of tension development.
A) Acetylcholine release at the neuromuscular junction
initiates an action potential in the motor end plate.
B) Acetylcholine binds to a nicotinic receptor on the postsyn-
aptic membrane.
C) The depolarization in skeletal muscle results from influx of
calcium through voltage-gated calcium channels (dihydro-
pyridine receptors).
D) Depolarization of the muscle fiber is not essential for
skeletal muscle contraction.
E) Norepinephrine activating adrenergic receptors causes
increased strength of contraction.
10
C H A P T E R

Cardiac Muscle Structure


and Function
Kathleen H. McDonough

O B J E C T I V E S

■ Explain the process of excitation–contraction coupling in cardiac muscle and


how it differs from that of skeletal muscle.
■ Explain the effects of changes in resting cell length on muscle tension
development, that is, the length–tension relationship.
■ Describe the sequence of events in isotonic contractions—tension
development and shortening.
■ Describe the effects of afterload on isotonic contractions in cardiac muscle.
■ Explain the effects of changes in resting cell length on isotonic contractions
at different afterloads, that is, explain the force–velocity curve.
■ Describe the effects of increased contractility on the force–velocity curve.
■ Explain the terms preload, afterload, contractility, force, and tension.

INTRODUCTION tials rapidly. Cardiac muscle cells have gap junctions through
which cells communicate information about membrane poten-
Cardiac muscle, like skeletal muscle, is striated due to the orderly tial—that is, if one cell depolarizes, the adjacent cells will also
structure of the actin and myosin filaments and the accessory depolarize due to communication through the gap junctions.
proteins that stabilize the sarcomere. Like type I skeletal muscle, Thus, all cardiac myocytes in the atria contract together and
cardiac muscle appears to be red in color due to the high content then all of the myocytes in the ventricle contract together
of mitochondria and myoglobin and its blood supply. The heart (Chapter 23). Because of this unified contraction of the ventri-
uses large amounts of ATP in beating 60–100 times/min (dur- cles (or the atria), the heart is said to be a functional syncytium.
ing normal resting conditions) for the lifetime of the normal Since all ventricular muscle cells contract together, there is no
adult and oxidative phosphorylation is the main source of that type of spatial recruitment in the heart. The heart relies on other
ATP, thus the high myoglobin concentration and large mito- mechanisms to increase strength of contraction.
chondrial content. There are estimates that the myocardial ATP
pool turns over every 10 seconds. The heart is able to use any
substrate provided to it in the blood and uptake is dependent on EXCITATION–CONTRACTION
the concentration of those substrates such as glucose, pyruvate,
lactate, free fatty acids, and ketone bodies. Normally fatty acid
COUPLING
oxidation provides 60–90% of the ATP used by the adult heart. Cardiac muscle cells contract when calcium levels in the cells
Like skeletal muscle, calcium is essential for contraction and is increase from approximately 10−7 M (0.1 μM) to 10−6 to 10−5 M
provided by excitation–contraction coupling. Although cardiac (1–10 μM). The level of calcium present in the cytosol to initi-
muscle can contract spontaneously due to pacemaker activity ate contraction has a profound effect on the strength of the con-
in the sinoatrial (SA) node, the individual muscle cells (myo- traction (contractility). Excitation–contraction coupling in
cytes) normally contract only when an action potential is initi- cardiac muscle varies somewhat from the process in skeletal
ated by the conduction system present in the heart and muscle. The anatomy of the sarcolemma (SL)–sarcoplasmic
transmitted through cells specialized to conduct action poten- reticulum (SR) interaction is different—diads are formed

93

Ch10_093-098.indd 93 11/26/10 9:48:54 AM


94 SECTION III Muscle Physiology

rather than triads as in skeletal muscle. There is less SR in car- potentials per minute) can increase calcium availability for
diac muscle, so the calcium release process relies on entry of contraction, thereby increasing the amount of tension that can
calcium into the cardiac cell through the voltage-gated chan- be generated. This phenomenon is called the staircase phe-
nels (dihydropyridine receptors). These open when the mem- nomenon or treppe. Physiologically heart rate is altered by
brane potential reaches approximately −40 mV. These calcium autonomic nervous system (ANS) modulation of SA node
channels are also called “slow” or L-type calcium channels firing rate and, as will be seen later, the sympathetic nervous
because they open more slowly than sodium channels and system (SNS) component of the ANS not only increases heart
remain open longer, generally about 200–300 milliseconds. rate, but also increases contractility. Therefore, the physiologic
Therefore, the action potential in cardiac ventricular cells is role of treppe is difficult to assess independent of SNS modu-
much longer than the action potential in skeletal muscle lation of heart rate and contractility.
in which the calcium channels do not actually open (see There are two additional variations in contraction that occur
Chapter 9). Calcium entry through SL calcium channels is in cardiac muscle that do not occur in skeletal muscle. Phos-
essential for contraction to occur. Absence of calcium in the phorylation of contractile proteins alters the strength of con-
extracellular fluid would prevent the heart from contracting. traction in the heart. The heart is very responsive to the
The process of excitation–contraction coupling is initiated by SNS— the “fight or flight” component of the ANS. With acti-
the pacemaker cells in the SA node that spontaneously generate vation of the SNS, beta-adrenergic receptors on the cardiac
action potentials (termed slow action potentials because they muscle cells are activated and an intracellular signaling scheme
lack fast sodium channels and depolarization is due to calcium results in production of cAMP and activation of protein
entry through the slow calcium channels). Action potentials are kinase A. Phosphorylation of proteins follows. Several pro-
transmitted through the atrial conduction fibers across the teins involved in contraction are phosphorylated and their
atrioventricular valves and finally to the conduction system in activity is altered. SL calcium channels are phosphorylated and
the ventricles. All ventricular muscle cells depolarize at the same allow more calcium to enter the cell and the strength of con-
time due to the rapid influx of sodium down its electrochemical traction is increased (contractility is enhanced). A protein
gradient (higher concentration of sodium outside of the cell and called phospholamban normally inhibits the SR calcium
negative membrane potential on the inside of the SL) through ATPase; when phospholamban is phosphorylated, it exerts
SL fast sodium channels. When the membrane potential less inhibition of the ATPase, so calcium uptake is enhanced.
reaches ~−40 mV, the slow calcium channels open, allowing cal- Phosphorylation of the calcium channels does not seem to
cium to diffuse down its concentration gradient into the cytosol. occur in skeletal muscle in which the maximum amount of
Some of this calcium causes opening of ryanodine channels calcium is released during each action potential and therefore
(receptors) on the SR and calcium diffuses out of the SR down cannot be increased. Remember that skeletal muscle has two
its concentration gradient. Some of the calcium from the SL SR cisternae in conjunction with the T-tubule, whereas car-
binds to troponin as does all of the calcium released from the diac muscle has only one cisterna associated with the T-tubule.
SR. Calcium binding to troponin results in a similar type of Skeletal muscle does not seem to have a functional phospho-
interaction of actin and myosin and cross-bridge cycling to that lamban, so the calcium ATPase activity is always operating at
which occurs in skeletal muscle. Relaxation occurs when the its maximal capacity. Increasing the amount of calcium enter-
calcium concentration in the cytosol is lowered by the calcium ing the cytosol is an important mechanism for increasing
ATPase on the longitudinal part of the SR pumping calcium strength of contraction (contractility); removing calcium
back into the SR. Calsequestrin is also present in cardiac muscle faster for relaxation is an important mechanism when the
to serve as a “sink” for calcium. When calcium levels decrease, heart rate increases with SNS stimulation and there is less time
calcium diffuses from the troponin and the cells relax. during the contraction-relaxation cycle.
Two other proteins are involved in removing calcium from
the cardiac cell. Since calcium enters the cell with each action
potential, there must be mechanisms to remove calcium or the CONTRACTION—LENGTH–
cell calcium content would increase with each heartbeat. The
SL contains a calcium ATPase that has a high affinity for cal- TENSION, ISOMETRIC
cium and can therefore pump calcium out of the cell probably CONTRACTIONS
even during diastole. The other protein is the sodium–calcium
exchanger. The exchanger operates on the basis of the sodium The strength of contraction in cardiac muscle can be altered by
ion gradient. The sodium ion concentration is greater outside changes in the initial or resting length of the muscle cells (pre-
of the cell than in the cell. Via the exchanger, sodium ion enters load) similar to the phenomenon in skeletal muscle. Cardiac
the cell and calcium ion is removed from the cell. Three sodium muscle, unlike skeletal muscle, can have physiologic changes in
ions enter for every one calcium ion leaving the cell. Manipula- the length of the muscle cells. For example, when the volume in
tion of the sodium gradient can have significant effects on cal- the ventricle at the end of diastole (the relaxation phase of the
cium extrusion from the cell and thus affect contraction. cardiac cycle) is changed, the muscle cell length is changed in
Since calcium levels change during each action potential, the same direction. Increased ventricular end-diastolic volume
there is some evidence that increases in heart rate (more action results in increased ventricular muscle cell length prior to the
CHAPTER 10 Cardiac Muscle Structure and Function 95

ere lengths, the opposing thin filaments may overlap with each
other and interfere with interaction with myosin. At long sar-
comere lengths, overlap may be insufficient for optimal cross-
bridge formation.
More cross-bridge interaction leads to a stronger contraction.
Tension, dynes/cm

Two other factors may contribute to the length–tension phe-


Active or developed nomenon in cardiac muscle. The second mechanism may result
from a length-dependent change in calcium sensitivity of the
myofilaments. For a similar cytosolic calcium concentration, a
Passive – cardiac less stretched muscle develops less force than does a more
muscle stretched (longer) cardiac muscle preparation. This change in
Passive – skeletal calcium sensitivity occurs immediately after a change in length
muscle with no delay. The sensitivity of the contractile proteins, specifi-
cally troponin C, seems to increase at greater resting lengths.
Finally, there is some evidence that the amount of calcium
released from the SR is greater at longer resting lengths. How
Po much these two factors contribute to the greater tension devel-
Length, mm opment is open to speculation since studies to demonstrate
FIGURE 10–1 The length–tension relationship in cardiac these two effects of length on calcium dynamics are generally
muscle is slightly different from that in skeletal muscle— performed in isolated cells or organelles. In summary, the heart
primarily due to the presence of passive tension at shorter usually operates at lower than maximal preloads and therefore
lengths. This is in part due to the anatomic differences in structure of has reserve—increasing muscle length can have a profound
skeletal muscle (all of the fibers in parallel) and cardiac muscle (fibers effect on strength of contraction that allows the heart to meet
exist in a basket weave-type pattern) as well as the properties of the the demands of increased work such as occurs during exercise.
noncontractile components in skeletal muscle versus cardiac muscle.
Note that in skeletal muscle, the fibers are usually operating at the
blue point—resting length is optimum because most skeletal muscle
is held in place by the bones and resting length cannot vary greatly. CONTRACTION—
Cardiac muscle normally operates at lower (red point) than optimum
length and therefore has reserve capacity to increase tension
FORCE–VELOCITY,
development, that is, have stronger contractions, when resting length ISOTONIC CONTRACTIONS
is increased. In the intact heart, cardiac cell resting length is set by the
volume in the ventricle at the end of diastole (the relaxed state of The effects of altered preload on heart function can also be
cardiac muscle). observed with isotonic contractions that represent a better
match to physiologic contractions of the heart as a pump. The
left ventricle must develop tension (pressure) to match the
onset of contraction. The heart normally operates at lower than afterload (aortic pressure) in order to open the aortic valve
maximal cell length or preload (Figure 10–1, red circle), and then allow the shortening phase of the contraction to
whereas skeletal muscle usually works at maximal preloads pump blood (stroke volume) into the aorta. Recall from the
(blue circle). Note also that the passive tension properties of the discussion of skeletal muscle that there is an inverse relation-
heart differ from those of skeletal muscle. Skeletal muscle does ship between afterload and velocity of shortening and there-
not increase passive tension until the muscle cell length is close fore between afterload and shortening. Greater afterload
to the length that gives the maximum active tension. Cardiac results in less shortening. Using isotonic contractions, the
muscle has passive tension even at low cell lengths. These dif- effects of increased preload, that is, more cross-bridge cycling,
ferences are due to the anatomic arrangement of the muscle on the force–velocity curve can be analyzed. When shorten-
cells with the noncontractile components in the muscle. Skeletal ing and velocity of shortening are measured as a function of
muscle is more distensible than cardiac muscle. In Figure 10–1, afterload, higher afterloads result in less shortening (see
the effects of increases in preload are shown through isometric Figures 9–8 and 10–2, black curve). If the preload is increased
contractions—that is, greater tension is developed from greater from length 1 (L1) to length 2 (L2) and the same afterloaded
resting cell length. The principle for the length–tension rela- contractions are studied from the higher preload, velocity of
tionship, as in skeletal muscle, is that the change in cell and shortening (and shortening) is greater for each afterload. If
sarcomere length alters the degree of overlap of the actin and more cross-bridges can interact, there is more myosin ATPase
myosin filaments and therefore increases the potential for activity and therefore more energy from ATP hydrolysis avail-
cross-bridges to form. Changes in the resting length of the able for the contraction. The cross-bridges develop the greater
whole muscle are associated with proportional changes in the tension needed to match the greater afterload and more
individual sarcomere length. Maximum tension development energy is available for more shortening and a greater velocity
occurs at sarcomere lengths of 2.2–2.3 μm. At shorter sarcom- of shortening to occur (blue curve, labeled L2). Note that the
96 SECTION III Muscle Physiology

maximum isometric tension (x-axis intercept) is increased (extrapolated intercept on the y-axis) increase. This increased
but the Vmax is not increased compared to the Vmax at L1. contractility should be compared to the curve generated at the
Preload shifts only the maximum tension, not the maximum same preload (L1), the black curve. Generally, increases in
velocity of shortening. contractility result in more rapid contractions such that
indexes of speed of tension development or maximum velocity
of shortening (Vmax) are used to indicate increases in contrac-
INCREASES IN STRENGTH tility. The figure demonstrates that there are two ways to
increase the velocity of shortening at the same afterload (the
OF CONTRACTION IN three points in the figure), one is by increasing preload (blue
CARDIAC MUSCLE curve), and the other is by increasing contractility (red curve).
The mechanisms by which the contractions are stronger are,
As stated above, cardiac muscle can increase strength of con- however, different. More optimum overlap of the actin and
traction when the preload is increased as demonstrated by the myosin filaments mediates the preload effect, whereas more
length–tension relationship. Another way that cardiac muscle cytosolic calcium to induce more rapid cross-bridge cycling
can increase strength of contraction is by increases in cytosolic mediates the contractility effect.
calcium resulting in increased contractility. Contractility The effects of increasing contractility can also be demon-
increases the velocity of cross-bridge cycling; therefore, strated by looking at the length–tension relationship. In Figure
increasing contractility can alter shortening and the velocity of 10–3, sympathetic nerve stimulation to the heart results in a
shortening. In comparing the force–velocity curve during shift of the length–tension relationship upward and to the left.
enhanced contractility (red curve in Figure 10–2), one can This indicates that for any given resting length of cardiac mus-
notice that increasing contractility causes the entire force– cle, the tension that can be developed is greater as a result of SNS
velocity curve to shift to the right—both the maximum iso- stimulation. The mechanism is the increase in calcium that
metric tension (intercept on the x-axis) and the Vmax results from the activation of beta-adrenergic receptors with the

Vmax

L1 ↑Contractility
Velocity

↑Preload or
length - L2
L1

Po
Force or load
FIGURE 10–2 The force–velocity curve in cardiac muscle can be altered by changes in resting cell length and by changes in
contractility. L1 represents the shorter resting cell length and L2 represents a greater resting cell length. In comparing the muscle at the same
afterload (black point vs. blue point), the muscle can shorten more if the contraction starts from a greater preload or resting cell length (L2). In
both contractions, the tension developed is set by the afterload. Note that the curve for increased preload intercepts the x-axis further to the
right—greater resting length allows for greater maximum isometric tension (the length–tension relationship). If the muscle is studied at L1, and
a drug that increases contractility is given, the entire force–velocity curve shifts upward and to the right from the black curve to the red
curve—both Po and Vmax are increased. More calcium results in stronger contractions and a greater velocity of contraction, that is, a greater
velocity of cross-bridge cycling. In comparing the black point to the red point, when contractility is greater, the muscle can develop the same
tension to match the load and there is more capacity to shorten and a greater velocity of shortening. Changes in Vmax therefore indicate changes
in contractility. Changes in Po can result from changes in preload or from changes in contractility.
CHAPTER 10 Cardiac Muscle Structure and Function 97

↑SNS STUDY QUESTIONS


1. Which of the following statements about cardiac muscle
contraction is correct?
Developed tension

A) Acetylcholine release at the neuromuscular junction


initiates an action potential in the postsynaptic membrane.
B) Acetylcholine binds to a nicotinic receptor on the
postsynaptic membrane.
C) Depolarization of the muscle fiber is not essential for
cardiac muscle contraction.
D) Norepinephrine activating adrenergic receptors causes
increased strength of contraction.
2. Which of the following statements about muscle contraction is
Length true for cardiac muscle?
FIGURE 10–3 The effects of changes in contractility on A) All cells in the heart contract at their own rate.
the length–tension relationship are shown. With sympathetic B) The strength of contraction is independent of the degree of
stimulation (SNS) of cardiac muscle, contractility increases and phosphorylation of cellular proteins.
developed tension is greater at each resting cell length. C) The strength of contraction is increased by recruiting more
motor units.
D) All muscle cells have a high oxidative capacity due to the
consequent production of cyclic AMP and activation of phos- abundant presence of mitochondria and myoglobin.
phorylation of the SL calcium channels by protein kinase A. The 3. Which of the following statements about muscle contraction is
importance of these two mechanisms will become very obvious true for cardiac muscle?
in discussions of the cardiovascular system in Section 5. A) Strength of contraction is altered physiologically by
changing the resting cell length from approximately
25% up to 100% of the maximum length.
CHAPTER SUMMARY B) Strength of contraction is altered physiologically by altering
the frequency of motor neuron firing.
■ Excitation–contraction coupling in cardiac muscle is similar
C) Tetanus occurs because the muscle action potential keeps
to that in skeletal muscle except that calcium must enter the
the cell refractory to stimuli that are closer than 1 second
cardiac muscle cell through the voltage-gated calcium channels
apart.
to cause release of calcium from the SR ryanodine channels.
D) Muscle contraction consists only of tension development.
■ In cardiac muscle, strength of contraction can be altered by
4. Which of the following statements about muscle contraction is
changes in resting cell length (length–tension) and by changes
true for cardiac muscle?
in contractility.
A) The Po (maximum isometric tension) is altered by both
■ Indices of contractility, such as the maximum velocity of
contractility and the length–tension relationship.
shortening (Vmax), are increased by beta-adrenergic agonists.
B) On the force–velocity curve the Vmax is altered by both
■ Since the heart has cells with pacemaker potential, innerva- contractility and the length–tension relationship.
tion of the heart is not required for contraction to occur. C) Afterload determines how many cross-bridges can interact
Ventricular cells contract at the same time (a functional during a contraction.
syncytium) due to the conduction system and gap junctions D) Preload determines the phosphorylation state of the
between the cardiac cells. myosin light chain.
■ Sympathetic and parasympathetic nerves modulate the
intrinsic beating rate.
■ Sympathetic nerves modulate the strength of contraction
(contractility) of cardiac cells.
This page intentionally left blank
11
C H A P T E R

Smooth Muscle Structure


and Function
Kathleen H. McDonough

O B J E C T I V E S

■ Explain the contraction process in smooth muscle and compare it to


those of skeletal and cardiac muscle.
■ Describe how smooth muscle can be activated to induce a contraction
or to change the strength of a contraction.
■ Explain the relationship between vascular smooth muscle membrane
potential, voltage-gated calcium channels, and strength of contraction.
■ Describe the difference between multiunit and unitary smooth muscle.
■ Explain the following terms and their role in smooth muscle function:
calmodulin, myosin light chain kinase, and myosin light chain phosphatase.

INTRODUCTION CONTRACTION
Smooth muscle makes up the walls of most of the hollow The general contractile process is uniform in all types of
organs of the body except the heart. As such, the function and smooth muscle. An increase in calcium in the cytosol results
control of contraction of the smooth muscle will vary depend- in binding of calcium to a calcium-binding protein, calmod-
ing on the organ in which it is located and the function of that ulin (Figure 11–1). This complex will bind to, and activate,
organ or organ system. For example, smooth muscle in the myosin light chain kinase (MLCK) that, in turn, phosphory-
gastrointestinal tract will be activated not only by mechanical lates the myosin light chain located on the myosin head. In
stimulation by the presence of food in the GI tract, but also by smooth muscle, the myosin light chain must be phosphory-
its neural and hormonal input. Smooth muscle in the uterus lated in order for the actin and myosin to form crossbridges
will respond differently during development of an embryo/ and initiate the crossbridge cycling or contraction. Relaxation
fetus than during the normal menstrual cycle. Hormones and or decreased tension development requires dephosphoryla-
neural input will even change the morphology of smooth tion of the myosin light chain by myosin light chain
muscle during pregnancy, making the uterus work as a unit phosphatase. The balance of phosphorylation and dephos-
rather than as independent muscle cells in the nonpregnant phorylation is important in regulating tension development
uterus. in smooth muscle since the kinase and the phosphatase are
The myosin ATPase activity in smooth muscle has a much always active. Increasing cytosolic calcium tips the balance
slower rate of hydrolysis of ATP (10–100 times lower than that toward more kinase activity and therefore more tension
of skeletal muscle); therefore, contractions are much slower development. Lower calcium levels tip the balance toward
and sometimes the mode of contraction results in increases less kinase and therefore more phosphatase activity and less
and decreases in the strength of contraction rather than com- tension development.
plete relaxation after a contraction as occurs in skeletal and There are other mechanisms to increase and decrease the
cardiac muscle. activity of the kinase and the phosphatase. For example,

99

Ch11_099-104.indd 99 11/26/10 9:49:20 AM


100 SECTION III Muscle Physiology

Smooth muscle cell


Contractions

Myosin light
Inactive MLCK ATP chain phosphatase

↑Cytosolic Active ~P myosin Myosin


Ca calmodulin
Ca2+ MLCK light chain light chain

Cross bridge cycling Relaxation


FIGURE 11–1 Scheme of steps in smooth muscle contraction. As in other types of muscle, calcium initiates the contraction. Calcium binds
to calmodulin that activates the myosin light chain kinase to phosphorylate the myosin light chain. On phosphorylation the myosin can interact
with the actin resulting in crossbridge cycling. Phosphatases can dephosphorylate the myosin light chain leading to relaxation or less tension
development. The balance of kinase and phosphatase activities determines the level of tension development in smooth muscle. Phosphorylation
of both the kinase and the phosphatase leads to a decrease in their activity—one resulting in weaker contraction and one in stronger contraction.

phosphorylation of the MLCK enzyme decreases its activity, would theoretically utilize large amounts of ATP. The latch state
thereby decreasing phosphorylation of myosin and resulting seems to occur because the crossbridges do not dissociate very
in more relaxation. This occurs when a specific receptor, the rapidly in spite of the fact that the myosin light chain is dephos-
beta2-adrenergic receptor, on the vascular smooth muscle phorylated; thereby, energy expenditure is minimized. The exact
and bronchiolar smooth muscle sarcolemma (SL) is acti- mechanism by which the latch state occurs is unknown. The
vated and increases intracellular cAMP levels. Subsequent physiologic significance, however, is remarkable—maintenance
activation of protein kinase A phosphorylates the MLCK of tension with very little energy expenditure.
and decreases its activity. Nitric oxide causes a similar relax-
ation of smooth muscle although the kinase that phosphory-
lates the MLCK is protein kinase G that is activated by cyclic VASCULAR VERSUS VISCERAL;
GMP. Regulation of the phosphatase is also important.
For example, phosphorylation of the myosin light chain MULTIUNIT VERSUS UNITARY
phosphatase decreases its activity, resulting in less dephos-
Smooth muscle can be divided into visceral and vascular
phorylation and therefore more phosphorylation of the myo-
muscle—visceral muscle making up the walls of most of the
sin light chains and more contraction. The Rho kinase
hollow organs and vascular making up the walls of blood
pathway leads to phosphorylation of the phosphatase. There
vessels. Vascular smooth muscle and to some extent, visceral
are several other types of regulation of the MLC kinase and
smooth muscle, can also be divided into two cell types—
phosphatase that alter the contraction properties of smooth
multiunit and unitary (Table 11–1). These two types of
muscle and are more specific for each organ’s particular func-
muscle cells have unique features that contribute to the vari-
tion and therefore will be discussed in the organ-specific sec-
ety of functions of smooth muscle. Multiunit smooth muscle
tions of this book.

ENERGY FOR CONTRACTION TABLE 11-1 Comparison of smooth muscle cell types.
AND RELAXATION Multiunit Unitary

The ATP used in contraction and relaxation in smooth muscle is Functional Individual units Syncytium
produced primarily by oxidative phosphorylation. The sub- Innervation Yes Little
strates such as glucose and fatty acids are provided in the blood
Gap junctions Few Yes
and mitochondrial oxidative processes produce adequate energy
for the slower contractions that occur in smooth muscle due to Response to stretch Little Yes
the lower rate of the myosin ATPase enzyme. An interesting Response to SNS Yes Little
adaptation of smooth muscle ensures that sustained contractions
Control of Central or neural Local factors
can occur at a lower-than-predicted ATP utilization. Smooth contraction factors
muscle can maintain tension by a phenomenon termed the latch
state. This is thought to be important in sphincter muscles where Examples Airway smooth muscle Small blood
vessels
tension development must occur for long periods of time that
CHAPTER 11 Smooth Muscle Structure and Function 101

Autonomic
nerve fiber

Varicosity

Sheet of
cells

Mitochondrion

Synaptic
vesicles

Varicosities

FIGURE 11–2 Pattern of innervation of smooth muscle. Note that the nerve has multiple branches and varicosities on each of the branches.
Neurotransmitter is released at the varicosities and diffuses to the smooth muscle. Binding to the appropriate receptor will result in the neural
modulation of smooth muscle contraction. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

consists of cells that act as independent units—they are an action potential, activation of receptors that initiate an
innervated and can respond strongly to nerves of the sympa- intracellular signaling network, activation of receptors that
thetic and parasympathetic nervous systems. These types are ion channels, and stretch, by itself. The exact stimuli
of cells have very few gap junctions and therefore activation that alter smooth muscle contraction may differ in various
of one cell does not necessarily lead to activation of cells in organs and even in two different types of muscle—unitary and
juxtaposition to that activated cell. Other cells receiving the multiunit. In Figure 11–3, the effects of sympathetic and para-
same neural input will respond but only because the nerve is sympathetic nerve stimulation on the membrane potential of
releasing neurotransmitters from varicosities (Figure 11–2) visceral smooth muscle are shown. Acetylcholine, the neu-
that release neurotransmitter near the muscle cell membrane. rotransmitter of the parasympathetic nervous system, gener-
Note that the same nerve will release neurotransmitter onto ally causes the membrane potential to become less negative
many cells. Released neurotransmitter diffuses to the muscle and for spikes (action potentials) to occur—generating more
cell membrane and binds to appropriate receptors—there is contractile activity. Sympathetic stimulation generally results
no specialized motor end plate on the muscle cell membrane, in the opposite—more negative membrane potential and
just the presence of receptors. Both sympathetic and para- resultant decreased contractile activity and relaxation. Note
sympathetic nerves can innervate the same smooth muscle that stretch is also conducive to more action potential spikes,
causing opposite effects on the cells as described below. and, subsequently, more contraction; food in the gut induces
Unitary muscle, on the other hand, has many gap junctions increased contractile activity of the gut.
(as described in Chapter 3), so activation of one cell leads Many other stimuli lead to activation or relaxation of smooth
rapidly to activation of cells juxtaposed to that cell. Thus, the muscle. Figure 11–4 shows a smooth muscle cell and some of
cells contract as a “unit.” These cells generally have little the many mechanisms involved in eliciting contraction and
innervation and exhibit a response to stretch, that is, cells relaxation. Calcium can be provided by both influx of calcium
will increase tension in response to stretch, a property that from the extracellular fluid through the L-type voltage-gated
will be discussed in more detail in Sections 5 and 8. Table 11–1 calcium channels on the SL and release of calcium from the
presents a list of the properties of multiunit versus unitary sarcoplasmic reticulum (SR). The SR, which is less abundant
smooth muscle. than in skeletal and cardiac muscle, can, nevertheless, release
calcium through activation of IP3 receptors. The IP3 receptors
are channels similar to the ryanodine receptors in the other
METHODS OF STIMULATION two muscle types and when the channel is open, calcium dif-
fuses down its concentration gradient into the cytosol to initi-
Smooth muscle can be stimulated to contract or alter the ate contraction. IP3 is a product of receptor-mediated activation
strength of a contraction by many different stimuli—action of phospholipase C (PLC) that hydrolyzes phosphatidylinos-
potentials, changes in membrane potential that do not achieve itol (PIP2). Diacyglycerol, the other product, activates protein
102 SECTION III Muscle Physiology

0 Acetylcholine, parasympathetic bind acetylcholine, and specific endothelin receptors that


stimulation, cold, stretch bind endothelin 1.
Influx of calcium through the voltage-gated calcium chan-
nels can be modulated by the resting membrane potential
that is primarily a function of K+ movement. Opening of
mV

K channels (by calcium) or closing of K channels (by ATP)


alters the membrane potential. Hyperpolarization of the cell
(by opening the K channels) results in closure of the voltage-
Membrane potential
−50 gated calcium channels and relaxation or dilation if the cell is
a vascular smooth muscle cell. Depolarization of the cell (not
necessarily enough to generate an action potential) causes
Epinephrine, sympathetic opening of the voltage-gated calcium channels leading to
stimulation contraction (constriction of smooth muscle if the cell is vas-
cular smooth muscle). Notice also the presence of storage-
operated channels (SOC) on the SL. These channels allow
entry of calcium into smooth muscle in order to replenish SR
calcium stores. The mechanisms sensing decreased SR cal-
cium levels and the communication between the SR and the
FIGURE 11–3 Examples of stimuli such as the SNS and the SL are not clear but do seem to be effective in maintaining
PNS on smooth muscle membrane potential and action potential
adequate calcium stores in the smooth muscle cell. Finally,
generation. At more negative membrane potentials, as with
epinephrine or SNS stimulation, there are no action potentials and
receptor-operated channels exist on smooth muscle cells.
muscle becomes more relaxed. At less negative action potentials, action These receptors are literally channels that allow ion move-
potentials occur and the muscle is more likely to have more tension or ments. Purinergic channels represent this type of control—
tone. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: ATP, which is a purine, opens this type of channel allowing
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) calcium entry into the cell and promoting contraction. In the
renal vascular smooth muscle, adenosine, which is normally
considered a vasodilator, binds to ROC allowing calcium
kinase C. There are several different receptors on smooth entry and contraction.
muscle that are linked to the PLC pathway resulting in stronger Not shown in Figure 11–4 is the modulation of smooth
contractions. Some of these include alpha-adrenergic recep- muscle contraction that occurs by products from the other
tors that bind norepinephrine, muscarinic receptors that cells associated with smooth muscle. For example, endothelial

Smooth muscle cell

Receptor

PLC

IP3 SR
PIP2 IP3
receptor Ca2+
+ release
DAG ROC
PKC SOC
~P proteins

Ca2+
(opens K
ATP channel)
Voltage gated
(closes K
calcium channel
channel) Channels
FIGURE 11–4 A smooth muscle cell with some of the many influences on contraction. Contractions can be initiated by action potentials,
by receptors that couple to phospholipase C, and by alterations in the open state of the voltage-gated calcium channels that are sensitive to the
membrane potential as controlled primarily by potassium movements across the membrane. SOC are the storage-operated channels that open
when SR calcium stores are low. ROC are receptor-operated channels—primarily responsive to agents such as adenosine and ATP. Calcium release
from the SR or calcium entry through voltage-gated calcium channel leads to the calmodulin binding and ultimately contraction.
CHAPTER 11 Smooth Muscle Structure and Function 103

cells that line the blood vessels release several factors that ■ The myosin ATPase activity is lowest in smooth muscle,
modulate vascular smooth muscle force development. As resulting in the slowest contractions or changes in tension
stated above, acetylcholine, which activates muscarinic recep- development.
tors on vascular smooth muscle, can cause contraction by a ■ Multiunit smooth muscle cells are innervated, have few gap
PLC mechanism. However, acetylcholine binding to muscar- junctions, and contract individually.
inic receptors on endothelial cells causes the production of ■ Unitary smooth muscle cells can respond to stretch and have
nitric oxide that diffuses to the vascular smooth muscle cell, gap junctions that enable them to contract as a “unit.”
and activates guanylate cyclase to produce cGMP. The cGMP
activates protein kinase G that phosphorylates MLCK and
decreases contraction. Thus, the site at which acetylcholine STUDY QUESTIONS
binds to the receptor (smooth muscle vs. endothelial cell) 1. Which of the following statements about smooth muscle is true?
determines the response. In the body, due to the anatomy of A) Phosphorylation of myosin light chains is required for
the endothelial cells and the vascular smooth muscle cells and contraction.
the presence of acetylcholine esterase, acetylcholine from the B) Inhibition of myosin light chain kinase increases strength
parasympathetic varicosities would predominantly release of contraction.
acetylcholine that would bind to muscarinic receptors on C) Inhibition of myosin light chain phosphatase decreases
endothelial cells resulting in relaxation or dilation. contraction.
Other biological responses of smooth muscle to stimulation D) Stimulation of the smooth muscle cells by nitric oxide
are also site specific. For example, visceral smooth muscle in will increase contraction.
the gastrointestinal tract becomes quiescent with sympathetic 2. Which of the following statements about smooth muscle
nerve stimulation (Figure 11–3), whereas vascular smooth contraction is correct?
muscle in the blood vessels of the gastrointestinal tract devel- A) Acetylcholine release at the neuromuscular junction
ops stronger contraction when stimulated by sympathetic initiates an action potential in the postsynaptic membrane.
nerves. This site-specific response is due to the type of recep- B) Acetylcholine binds to a nicotinic receptor on the
postsynaptic membrane.
tors on the cells—beta-adrenergic receptors cause relaxation
C) Depolarization of the muscle fiber is not essential for
in response to sympathetic stimulation in visceral smooth smooth muscle contraction.
muscle, whereas alpha-adrenergic receptors cause stronger D) Norepinephrine activating adrenergic receptors always
contraction in response to sympathetic stimulation in vascular causes increased strength of contraction in all smooth
smooth muscle. muscle cells.
In summary, smooth muscle is the most diverse muscle in 3. Which of the following statements about muscle contraction is
the body. Its functions are dependent to a great extent on the true for smooth muscle?
tissue in which they are found. Therefore, more specific A) All cells have pacemaker potential.
detail about smooth muscle function will be presented in B) The strength of contraction is correlated with the degree
Sections 5, 7–9. of phosphorylation of the myosin light chains.
C) The strength of contraction is increased by recruiting
more motor units.
CHAPTER SUMMARY D) All muscle cells have a high myosin ATPase velocity and
therefore a rapid contraction.
■ In smooth muscle, the calcium-binding protein is calmodulin
rather than troponin as in skeletal and cardiac muscle. 4. Which of the following statements about muscle contraction is
true for smooth muscle?
■ The calcium–calmodulin complex activates MLCK.
A) Strength of contraction cannot be altered physiologically by
■ Contraction is dependent on MLCK phosphorylating the
changing the resting cell length from 25% up to 100% of the
myosin light chain allowing for binding of the myosin to
maximum length.
the actin.
B) Strength of contraction is altered physiologically by altering
■ MLC phosphatase removes the phosphate from the myosin the frequency of motor neuron firing.
light chain resulting in decreased strength of contraction. C) Strength of contraction can be changed by changing the
■ Many different stimuli can induce contraction or increase the balance of the myosin light chain kinase and phosphatase
strength of contraction of smooth muscle—voltage-gated activities.
calcium channels, voltage-gated potassium channels, receptor- D) Muscle contraction occurs as contractions followed by
operated channels, SOC, and receptor-mediated pharmacom- complete relaxation of the cell.
echanical coupling. Even stretch can activate smooth muscle
contraction.
This page intentionally left blank
SECTION IV CNS/NEURAL PHYSIOLOGY

12
C H A P T E R

Introduction to the
Nervous System
Susan M. Barman

O B J E C T I V E S

■ Name the various types of glia and their functions.


■ Name the parts of a neuron and their functions.
■ Describe the role of myelin in nerve conduction.
■ List the types of nerve fibers found in the mammalian nervous system.
■ Describe the general organization of thalamic, cortical, and reticular
formation neurons.
■ Describe the function of neurotrophins.
■ Compare peripheral and central nerve regeneration.

INTRODUCTION these cells are now recognized for their role in communica-
tion within the CNS in partnership with neurons. Unlike neu-
The nervous system can be divided into two parts: the central rons, glial cells continue to undergo cell division in adulthood
nervous system (CNS), which is composed of the brain and spi- and their ability to proliferate is particularly noticeable after
nal cord, and the peripheral nervous system, which is com- brain injury.
posed of nerves that connect the CNS to muscles, glands, and There are two major types of glia, microglia and macroglia.
sense organs. Neurons are the basic building blocks of the ner- Microglia are scavenger cells that resemble tissue macrophages
vous system. The human brain contains about 1011 (100 billion) and remove debris resulting from injury, infection, and disease.
neurons. It also contains 10–50 times this number of glial cells Microglia arise from macrophages outside of the CNS and are
or glia. The CNS is a complex organ; it has been calculated that physiologically and embryologically unrelated to other neural
40% of the human genes participate, at least to a degree, in its cell types.
formation. There are three types of macroglia: oligodendrocytes,
Schwann cells, and astrocytes (Figure 12–1). Oligodendro-
CELLULAR ELEMENTS IN THE CNS cytes and Schwann cells are involved in myelin formation
around axons in the CNS and peripheral nervous system,
GLIAL CELLS respectively. Astrocytes, which are found throughout the
brain, are of two subtypes. Fibrous astrocytes, which con-
The word glia is Greek for glue; for many years, glia were tain many intermediate filaments, are found primarily in
thought to function merely as connective tissue. However, white matter. Protoplasmic astrocytes are found in gray

105

Ch12_105-114.indd 105 11/26/10 9:49:36 AM


106 SECTION IV CNS/Neural Physiology

A Oligodendrocyte B Schwann cell C Astrocyte

Oligodendrocyte Perineural
in white matter oligodendrocytes
Nodes of Ranvier Capillary

End-foot

Neuron
Layers
of myelin

Axons
Schwann End-foot Fibrous
cell astrocyte
Nucleus

Inner
tongue Axon
Neuron

FIGURE 12–1 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) Schwann cells provide myelin to the peripheral nervous
system. Each cell forms a segment of myelin sheath about 1 mm long; the sheath assumes its form as the inner tongue of the Schwann cell turns
around the axon several times, wrapping in concentric layers. Intervals between segments of myelin are the nodes of Ranvier. C) Astrocytes are
the most common glia in the CNS and are characterized 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. (Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM
[editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

matter and have a granular cytoplasm. Both types of astro- ending in a number of synaptic knobs that are also called ter-
cytes send processes to blood vessels, where they induce minal buttons or boutons. They contain granules or vesicles
capillaries to form the tight junctions making up the that store the synaptic transmitters secreted by the nerves.
blood–brain barrier. The blood–brain barrier prevents the Based on the number of processes that emanate from the cell
diffusion of large or hydrophilic molecules (e.g., proteins) body, neurons can be classified as unipolar, bipolar, and mul-
into the cerebrospinal fluid and brain, while allowing tipolar (Figure 12–3).
diffusion of small molecules. The astrocytes also send pro- The axons of many neurons are myelinated, that is, they
cesses that envelop synapses and the surface of nerve cells. acquire a sheath of myelin, a protein–lipid complex that is
Protoplasmic astrocytes have a membrane potential that var- wrapped around the axon (Figure 12–2). In the peripheral
ies with the external K+ concentration but do not generate nervous system, myelin forms when a Schwann cell wraps its
propagated potentials. They help maintain the appropriate membrane around an axon. This can occur up to 100 times,
concentration of ions and neurotransmitters by taking up K+ resulting in many layers of myelin around an axon
and the neurotransmitters glutamate and γ-aminobutyrate (Figure 12–1). The myelin is then compacted when the ex-
(GABA). tracellular portions of a membrane protein called protein
zero (P0) lock to the extracellular portions of P0 in the appos-
ing membrane. Various mutations in the gene for P0 cause
NEURONS peripheral neuropathies. The myelin sheath envelops the
axon except at its ending and at the nodes of Ranvier, peri-
Neurons in the mammalian CNS come in many different odic 1-μm constrictions that are about 1 mm apart (Figure
shapes and sizes. Most have the same parts as the typical spi- 12–2). The insulating function of myelin is critical for salta-
nal motor neuron illustrated in Figure 12–2. The cell body tory conduction of action potentials (see Chapter 6). Some
(soma) contains the nucleus and is the metabolic center of the neurons have axons that are unmyelinated, that is, they are
neuron. Dendrites extend outward from the cell body and simply surrounded by Schwann cells without the wrapping
arborize extensively. Particularly in the cerebral and cerebel- of the Schwann cell membrane that produces myelin around
lar cortex, the dendrites have small knobby projections called the axon.
dendritic spines. A typical neuron has a long fibrous axon Within the CNS, the cells that form the myelin are oligoden-
that originates from a thickened area of the cell body, the drocytes (Figure 12–1). Unlike the Schwann cell, which forms
axon hillock. The first portion of the axon is called the initial the myelin on a single neuron, oligodendrocytes emit multiple
segment. The axon divides into presynaptic terminals, each processes that form myelin on many neighboring axons. In
CHAPTER 12 Introduction to the Nervous System 107

Cell body
(soma)

Initial segment
of axon Node of Ranvier Schwann cell

Axon hillock
Nucleus Terminal buttons

Dendrites

FIGURE 12–2 Motor neuron with a myelinated axon. A motor 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 ending and at the nodes of Ranvier. Terminal buttons (boutons) are located at
the terminal endings. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

multiple sclerosis (MS), a crippling autoimmune disease, and pressure, while smaller axons subserve pain and
patchy destruction of myelin occurs in the CNS. The loss of temperature sensations and autonomic function. Dorsal root
myelin is associated with delayed or blocked conduction in the C fibers conduct some impulses generated by touch and oth-
demyelinated axons. er cutaneous receptors in addition to impulses generated by
pain and temperature receptors. A numerical system (Ia, Ib,
II, III, IV) has also been used to classify sensory fibers. A
comparison of the number system and the letter system is
PERIPHERAL NERVOUS SYSTEM shown in Table 12–3.
In addition to variations in speed of conduction and fiber
The peripheral nervous system transmits information from
diameter, the various classes of fibers in peripheral nerves
the CNS to the effector organs throughout the body. It
differ in their sensitivity to hypoxia and anesthetics (Table
contains 12 pairs of cranial nerves and 31 pairs of spinal
12–4). This fact has clinical as well as physiological signifi-
nerves. The cranial nerves have rather well-defined sensory
cance. For example, local anesthetics depress transmission in
and motor functions (Table 12–1). Many of these functions
group C fibers before they affect group A touch fibers. Con-
are described individually in more detail in later chapters in
versely, pressure on a nerve can cause loss of conduction in
this section. Spinal nerves are named on the basis of the ver-
large-diameter motor, touch, and pressure fibers while pain
tebral level from which the nerve exits (cervical, thoracic,
sensation remains relatively intact. This is sometimes seen in
lumbar, sacral, and coccygeal). These nerves include motor
individuals who sleep with their arms under their heads for
and sensory fibers of muscles, skin, and glands throughout
long periods, causing compression of the nerves in the arms.
the body.
Because of the association of deep sleep with alcoholic in-
toxication, the syndrome is most common on weekends and
has acquired the interesting name Saturday night or Sunday
NERVE FIBER TYPES AND FUNCTION morning paralysis.
Axonal conduction velocity is the speed by which an action
potential travels along the axon. In general, there is a direct
relationship between the diameter of a given nerve fiber and ORGANIZATION OF THE
its speed of conduction. Nerve conduction tests are often used
by neurologists in the diagnosis of some diseases. THALAMUS, CEREBRAL CORTEX,
Axonal conduction velocity and other characteristics have & RETICULAR FORMATION
led to the classification of nerve fibers as shown in Table
12–2. Mammalian nerve fibers are divided into three major The thalamus is a large collection of neuronal groups within
groups (A, B, and C); the A group is further subdivided into the diencephalon; it participates in sensory, motor, and limbic
α, β, γ, and δ fibers. In Table 12–2, the various fiber types are functions that will be described in later chapters in this section.
listed with their diameters, electrical characteristics, and Virtually all information that reaches the cerebral cortex is
functions. Large axons are concerned primarily with propri- first processed by the thalamus, leading to its being called the
oceptive sensation, somatic motor function, conscious touch, “gateway” to the cortex.
108 SECTION IV CNS/Neural Physiology

A Unipolar cell B Bipolar cell C Pseudo-unipolar cell

Dendrites Peripheral axon


Dendrite to skin and
muscle

Cell body

Axon
Single bifurcated
Cell body process

Axon Central
axon
Cell body
Axon terminals
Invertebrate neuron Bipolar cell of retina 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 Pyramidal cell of Purkinje cell of cerebellum


spinal cord hippocampus

FIGURE 12–3 Different 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 two specialized processes: a dendrite that carries
information to the cell and an axon that transmits information from the cell. C) Some sensory neurons are in a subclass of bipolar cells called
pseudounipolar 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 dendrites. 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. (Adapted from Ramon Y Cajal:
Histology. 10th ed. Baltimore: Wood, 1933.)

The thalamus is divided into nuclei that project diffusely to project to the motor cortex. The anterior nuclei receive affer-
wide regions of the neocortex and nuclei that project to spe- ents from the mamillary bodies and project to the limbic cor-
cific portions of the neocortex and limbic system. The nuclei tex (memory and emotion). Most thalamic neurons are excit-
that project to wide regions of the neocortex are the midline atory and release glutamate. The thalamic reticular nucleus
and intralaminar nuclei. The nuclei that project to specific neurons are inhibitory and release GABA; they modulate the
areas include the specific sensory relay nuclei and the nuclei responses of other thalamic neurons to input coming from
concerned with efferent control mechanisms. The specific the cortex.
sensory relay nuclei include the medial and lateral geniculate The neocortex is arranged in six layers (Figure 12–4). The
bodies, which relay auditory and visual impulses to the audi- most common neuronal type is the pyramidal cell with an
tory and visual cortices, and the ventral posterior lateral extensive vertical dendritic tree (Figure 12–5) that may ex-
(VPL) and ventral posteromedial, which relay somatosensory tend to the cortical surface. Their cell bodies can be found in
information to the postcentral gyrus. The ventral anterior all cortical layers except layer I. The axons of these cells give
and ventral lateral nuclei are concerned with motor function; off recurrent collaterals that turn back and synapse on the su-
they receive input from the basal ganglia and cerebellum and perficial portions of the dendritic trees. Afferents from the
CHAPTER 12 Introduction to the Nervous System 109

TABLE 12–1 Functions of cranial nerves.


Cranial Nerve Type Function

I. Olfactory Sensory Smell

II. Optic Sensory Vision

III. Occulomotor Motor Upward, downward, and medial eye movements; pupil diameter; lens shape

IV. Trochlear Motor Downward and lateral eye movement

V. Trigeminal Motor Chewing


Sensory Proprioception from skin and muscle of the face

VI. Abducens Motor Lateral eye movements

VII. Facial Motor Facial expression; salivary gland secretions


Sensory Sensation from skin of external ear canal; taste from anterior two thirds of the tongue

VIII. Vestibulocochlear Sensory Hearing; sense of motion

IX. Glossopharyngeal Motor Swallowing; parotid salivary gland secretions


Sensory Taste from posterior one third of the tongue; baroreceptor and chemoreceptors
X. Vagus Motor Skeletal muscles of larynx and pharynx; smooth muscle and glands in pharynx,
larynx, thorax, and abdomen
Sensory Receptors in thorax and abdomen; taste from posterior tongue and oral cavity

XI. Accessory Motor Skeletal muscles in the neck

XII. Hypoglossal Motor Skeletal muscle of tongue

specific nuclei of the thalamus terminate primarily in corti- (basket cells and chandelier cells) release GABA. Basket cells
cal layer IV, and the nonspecific afferents are distributed to account for most inhibitory synapses on the pyramidal soma
layers I–IV. and dendrites. Chandelier cells are a powerful source of inhi-
Pyramidal neurons are the only projection neurons of the bition of pyramidal neurons because they terminate on the
cortex, and they are excitatory neurons that release glutamate. initial segment of the pyramidal cell axon. Their terminal bou-
The other cortical cell types are local circuit neurons tons form short vertical rows that resemble candlesticks, thus
(interneurons) that are classified based on their shape, pattern accounting for their name. Spiny stellate cells, excitatory
of projection, and neurotransmitter. Inhibitory interneurons interneurons that release glutamate, are located primarily in

TABLE 12–2 Classification of mammalian nerve fibers.a


Fiber Conduction Spike Duration Absolute Refractory
Fiber Type Function Diameter (μm) Velocity (m/s) (milliseconds) Period (milliseconds)

α Proprioception; 12–20 70–120


somatic motor

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

Dorsal root Pain, temperature, some 0.4–1.2 0.5–2 2 2


mechanoreception

Sympathetic Postganglionic sympathetic 0.3–1.3 0.7–2.3 2 2


a
A and B fibers are myelinated; C fibers are unmyelinated.
110 SECTION IV CNS/Neural Physiology

TABLE 12–3 Numerical classification sometimes Pial surface Golgi stain Nissl stain Weigert stain
used for sensory fibers. Molecular
I layer
Number Origin Fiber Type
External
II granule
Ia Muscle spindle, annulospiral ending Aα cell layer

Ib Golgi tendon organ Aα

II Muscle spindle, flower-spray ending; Aβ External


touch, pressure III pyramidal
cell layer
III Pain and cold receptors; some Aδ
touch receptors

IV Pain, temperature, and Dorsal root C Internal


other receptors IV granule
cell layer

Internal
V pyramidal
cell layer

layer IV and are a major recipient of sensory information aris-


ing from the thalamus.
In addition to being organized into layers, the cortex is also
organized into columns. Neurons within a column have simi-
VI Multiform
lar response properties, suggesting they comprise a local layer
processing network (e.g., orientation and ocular dominance
columns in the visual cortex, as described in Chapter 15).
The reticular formation is a complex neural network in the
central core of the medulla and midbrain. It is made up of
White matter
various neural clusters and ascending and descending fibers. It
contains the cell bodies and fibers of many of the serotonergic,
noradrenergic, adrenergic, and cholinergic systems. Some of
the descending fibers in it inhibit transmission in sensory and FIGURE 12–4 Structure of the cerebral cortex. The cortical
motor pathways in the spinal cord; various reticular areas and layers are indicated by the numbers. Golgi stain shows neuronal cell
the pathways from them are concerned with spasticity (in- bodies and dendrites, Nissl stain shows cell bodies, and Weigert
myelin sheath stain shows myelinated nerve fibers. (Modified with
creased resistance to passive stretch of a muscle) and adjust-
permission from Ranson SW, Clark SL: The Anatomy of the Nervous System, 10th ed.
ment of stretch reflexes.
Saunders, 1959.)
The reticular activating system (RAS) is a complex
polysynaptic pathway arising from the brain stem reticular
formation with projections to the intralaminar and reticular
nuclei of the thalamus that, in turn, project diffusely and non- by different sensory stimuli. The system is therefore nonspe-
specifically to wide regions of the cortex (Figure 12–6). cific, whereas the classic sensory pathways are specific in that
Collaterals funnel into it not only from the long ascending the fibers in them are activated by only one type of sensory
sensory tracts, but also from the trigeminal, auditory, visual, stimulation. The RAS is concerned with consciousness and
and olfactory systems. The complexity of the neural network sleep (see Chapter 20).
and the degree of convergence in it abolish modality specific-
ity, and most reticular neurons are activated with equal facility
NEUROTROPHINS: TROPHIC
TABLE 12–4 Relative susceptibility of mammalian A, SUPPORT OF NEURONS
B, and C nerve fibers to conduction block produced by Proteins necessary for survival and growth of neurons are
various agents. called neurotrophins. Many are products of the muscles or
other structures that the neurons innervate, but others are
Most Least produced by astrocytes. These proteins bind to receptors at the
Susceptibility To Susceptible Intermediate Susceptible
endings of a neuron. They are internalized and then transport-
Hypoxia B A C ed by retrograde transport to the neuronal cell body, where
Pressure A B C they foster the production of proteins associated with neu-
ronal development, growth, and survival. Other neurotrophins
Local anesthetics C B A
are produced in neurons and transported in an anterograde
CHAPTER 12 Introduction to the Nervous System 111

neurons. It is found in many different tissues. NGF is picked


up by neurons and transported in retrograde fashion from the
endings of the neurons to their cell bodies. It is also present in
the brain and appears to be responsible for the growth and
B maintenance of cholinergic neurons in the basal forebrain
and striatum.
C
A
D

AXONAL DEGENERATION &


REGENERATION
B
When a motor nerve to skeletal muscle is cut and allowed to
degenerate, the muscle will become extremely sensitive to ace-
tylcholine, the transmitter normally released at the nerve end-
ing. This denervation supersensitivity (or hypersensitivity)
Axon of the postsynaptic structure to the transmitter previously
secreted by the axon endings is largely due to the synthesis or
activation of more receptors. This and other reactions trig-
FIGURE 12–5 Neocortical pyramidal cell, showing the gered by damage to an axon are summarized in Figure 12–7.
distribution of neurons that terminate on it. A) Nonspecific Orthograde degeneration (Wallerian degeneration) and ret-
afferents from the reticular formation and the thalamus; B) recurrent rograde degeneration of the axon stump to the nearest collat-
collaterals of pyramidal cell axons; C) commissural fibers from mirror eral (sustaining collateral) occur commonly when a nerve is
image sites in the contralateral hemisphere; D) specific afferents from
cut. Also, a series of changes occur in the cell body including a
thalamic sensory relay nuclei. (Modified with permission from Chow KL,
decrease in Nissl substance (chromatolysis).
Leiman AL: The structural and functional organization of the neocortex. Neurosci
Res Program Bull 1970;8(2):157–220.)
Peripheral nerve damage is often reversible. Although the
axon will degenerate distal to the damage, connective elements
of the distal stump often survive. Axonal sprouting occurs
fashion to the nerve ending, where they maintain the integrity
of the postsynaptic neuron.
The first neurotrophin to be characterized was nerve growth Axon branch Receptor
factor (NGF), a protein that is necessary for the growth and (sustaining collateral)
maintenance of sympathetic neurons and some sensory

Retrograde Receptor
degeneration hypersensitive
Cortex Site of injury

X
Retrograde Regenerative Orthograde
reaction: sprouting (wallerian)
chromatolysis degeneration

Intralaminar nuclei FIGURE 12–7 Summary of changes occurring in a neuron and


of thalamus the structure it innervates when its axon is crushed or cut at the
Midbrain reticular formation point marked X. Hypersensitivity of the postsynaptic structure to the
transmitter previously secreted by the axon occurs largely due to the
FIGURE 12–6 Diagram showing the ascending reticular synthesis or activation of more receptors. There is both orthograde
system in the human midbrain, its projections to the intralaminar (Wallerian) degeneration from the point of damage to the terminal and
nuclei of the thalamus, and the output from the intralaminar retrograde degeneration of the axon stump to the nearest collateral
nuclei to many parts of the cerebral cortex. Activation of these (sustaining collateral). Changes also occur in the cell body, including
areas is shown by PET scans when subjects shift from a relaxed awake chromatolysis. The nerve starts to regrow, with multiple small branches
state to an attention-demanding task. (Reproduced with permission from projecting along the path the axon previously followed (regenerative
Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, sprouting). (Reproduced with permission from Barrett KE, Barman SM, Boitano S,
23rd ed. McGraw-Hill Medical, 2009.) Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
112 SECTION IV CNS/Neural Physiology

from the proximal stump, growing toward the nerve ending.


This results from growth-promoting factors secreted by The woman is diagnosed with multiple sclerosis (MS), an
Schwann cells that attract axons toward the distal stump. autoimmune disease that affects over 3 million people
Adhesion molecules of the immunoglobulin superfamily worldwide, usually striking people between the ages of 20
promote axon growth along cell membranes and extracellular and 50 and affecting women about twice as often as men. In
matrices. Inhibitory molecules in the perineurium, the connec- MS, antibodies and white blood cells in the immune system
tive tissue sheath that covers a nerve bundle, assure that the re- attack myelin, causing inflammation and injury to the sheath
generating axons grow in a correct trajectory. Denervated distal and eventually the nerves that it surrounds. Loss of myelin
stumps are able to increase the production of neurotrophins leads to leakage of K+ through voltage-gated channels, hy-
that promote growth. Once the regenerated axon reaches its tar- perpolarization, and failure to conduct action potentials.
get, a new functional connection is formed. Regeneration allows Deficits may include muscle weakness, fatigue, diminished
for considerable, although not full, recovery. For example, fine coordination, slurred speech, blurred or hazy vision, blad-
motor control may be permanently impaired because some mo- der dysfunction, and sensory disturbances. Symptoms are
tor neurons are guided to an inappropriate muscle fiber. often exacerbated by increased body temperature or ambi-
Recovery of peripheral nerves from damage far surpasses ent temperature. In most cases, transient episodes appear
that of CNS pathways. The proximal stump of a damaged axon suddenly, last a few weeks or months, and then gradually
in the CNS will form short sprouts, but distant stump recovery disappear. Subsequent episodes can appear years later, and
is rare, and the damaged axons are unlikely to form new eventually full recovery does not occur. Others have a pro-
synapses. This is because CNS neurons do not have the growth- gressive form of the disease in which there are no periods of
promoting chemicals needed for regeneration. In fact, CNS remission. Diagnoses of MS is generally delayed until there
myelin is a potent inhibitor of axonal growth. Also, follow- are multiple episodes with deficits separated in time and
ing CNS injury, several events—astrocytic proliferation, space. Nerve conduction tests can detect slowed conduction
activation of microglia, scar formation, inflammation, and in motor and sensory pathways. Cerebral spinal fluid analy-
invasion of immune cells—provide an inappropriate environ- sis can detect the presence of oligoclonal bands indicative
ment for regeneration. Thus, treatment of brain and spinal of an abnormal immune reaction against myelin. The most
cord injuries often focuses on rehabilitation rather than re- definitive assessment is MRI to detect multiple scarred (scle-
versing the nerve damage. Researchers are trying to identify rotic) areas in the brain. Although there is no cure for MS,
ways to initiate and maintain axonal growth, to direct regener- some drugs such as beta-interferon and corticosteroids
ating axons to reconnect with their target neurons, and to re- that suppress the immune response can reduce the severity
constitute original neuronal circuitry. and slow the progression of the disease.

CHAPTER SUMMARY
CLINICAL CORRELATION ■ Glia are abundant in the CNS. Microglia are scavenger cells.
A 27-year-old school teacher awakens one morning with Macroglia include oligodendrocytes, Schwann cells, and
astrocytes. The first two are involved in myelin formation.
rather severe pain in her left eye and blurry vision (optic
Astrocytes help maintain the appropriate concentration of ions
neuritis). She is out of the country on a summer vacation and neurotransmitters in the CNS.
and decides to wait until she returns home to see her phy-
■ Neurons are composed of a cell body (soma) that is the
sician. Over the next couple of days, the pain and visual metabolic center of the neuron, dendrites that extend outward
loss worsen. But by the time she returns home 10 days lat- from the cell body and arborize extensively, and a long fibrous
er, the symptoms have abated enough that she decides it is axon that originates from a somewhat thickened area of the
not necessary to see her physician. About 8 months later cell body, the axon hillock.
she develops a sudden onset of weakness in her right leg ■ Axons of many neurons acquire a sheath of myelin, a protein–
after a difficult day in the classroom. She decides to relax in lipid complex that is wrapped around the axon. Myelin is
a hot bath, but this exacerbates the symptoms. Her prob- an effective insulator, and depolarization in myelinated axons
lem rapidly progresses to the point where she cannot walk. jumps from one node of Ranvier to the next (saltatory
Three days later, she is seen by her physician and also re- conduction).
ports the incident that occurred while on her summer va- ■ Nerve fibers are divided into different categories based on their
cation. A brain magnetic resonance imaging (MRI) and a axonal diameter, conduction velocity, and function.
visual evoked potential test are ordered. About 1 week ■ Thalamic nuclei that project to wide regions of the neocortex
later, she notes significant improvement but she is notified are the midline and intralaminar nuclei and those that project
by her physician that the MRI showed multiple periven- to specific areas include the specific sensory relay nuclei.
tricular white matter lesions, and the visual evoked poten- ■ The neocortex is arranged in six layers; the most common
tial test showed a delayed response (slowed conduction). neuronal type is the pyramidal cell whose cell bodies are
located in all layers except layer I.
CHAPTER 12 Introduction to the Nervous System 113

■ Neurotrophins are produced by astrocytes and transported by 3. A man falls into a deep sleep with one arm under his head.
retrograde transport to the neuronal cell body, where they This arm is paralyzed when he awakens, but it tingles, and pain
foster the production of proteins associated with neuronal sensation in it is still intact. The reason for the loss of motor
development, growth, and survival. function without loss of pain sensation is that in the nerves to
■ After a peripheral nerve is damaged, Schwann cells secrete a his arm
growth-promoting factor that attracts the proximal stump of A) A fibers are more susceptible to hypoxia than B fibers.
the axon toward the distal stump, allowing for regeneration. B) A fibers are more sensitive to pressure than C fibers.
In the CNS, neural regeneration is impaired by factors such C) C fibers are more sensitive to pressure than A fibers.
as astrocytic proliferation, scar formation, and inflammation. D) motor nerves are more affected by sleep than sensory
nerves.
E) sensory nerves are nearer the bone than motor nerves and
STUDY QUESTIONS hence are less affected by pressure.
4. The thalamus
1. The distance from one stimulating electrode to recording
A) is organized into six layers.
electrode is 4.5 cm. When the axon is stimulated, the latent
B) does not relay auditory or visual information to
period is 1.5 milliseconds. What is the conduction velocity
the neocortex.
of the axon?
C) is a component of the reticular activating system.
A) 15 m/s D) contains neurons that project diffusely throughout
B) 30 m/s the neocortex.
C) 40 m/s E) is a component of the brain stem.
D) 67.5 m/s
5. Which of the following is not true about the neocortex?
E) This cannot be determined from the information given.
A) It is organized into six layers.
2. Which of the following has the slowest conduction velocity?
B) The most common neuronal type is the pyramidal cell.
A) Aα fibers C) It receives direct input from the thalamus.
B) Aβ fibers D) It contains a group of inhibitory interneurons called
C) Aγ fibers basket cells.
D) B fibers E) It contains a group of excitatory interneurons called
E) C fibers chandelier cells.
This page intentionally left blank
13
C H A P T E R

General Sensory
Systems: Touch, Pain,
and Temperature
Susan M. Barman

O B J E C T I V E S

■ List common senses and their receptors.


■ Explain the terms hyperalgesia and allodynia.
■ Explain sensory coding.
■ Compare the pathway that mediates sensory input from touch,
proprioceptive, and vibratory senses to that mediating information
from pain and thermoreceptors.
■ Describe mechanisms to modulate transmission in pain pathways.

INTRODUCTION cells that surround them, forming a sense organ. Touch and
pressure are sensed by four types of mechanoreceptors
Information about the internal and external environment acti- (Figure 13–1). Meissner’s corpuscles are dendrites encapsu-
vates the central nervous system (CNS) via sensory receptors. lated in connective tissue and respond to changes in texture
These receptors are transducers that convert various forms of and slow vibrations. Merkel cells are expanded dendritic end-
energy into action potentials in neurons. The characteristics of ings, and they respond to sustained pressure and touch.
some of these receptors and the way they generate impulses in Ruffini corpuscles are enlarged dendritic endings with elon-
afferent neurons were considered in Chapter 5. Cutaneous gated capsules, and they respond to sustained pressure.
receptors for touch and pressure are mechanoreceptors. Pacinian corpuscles consist of unmyelinated dendritic end-
Potentially harmful stimuli such as pain, extreme heat, and ings of a sensory nerve fiber, encapsulated by concentric
extreme cold are mediated by nociceptors. Chemoreceptors lamellae of connective tissue that give the organ the appear-
are stimulated by a change in the chemical composition of the ance of a cocktail onion. These receptors respond to deep pres-
environment in which they are located. These include recep- sure and fast vibration.
tors for taste and smell as well as visceral receptors such as
those sensitive to changes in the plasma level of O2, pH, and
osmolality. Photoreceptors are those in the rods and cones in
NOCICEPTORS AND
the retina that respond to light. This chapter will focus primar- THERMORECEPTORS
ily on cutaneous receptors and transmission in somatosensory
Pain and temperature sensations arise from unmyelinated den-
pathways mediating touch and proprioception (dorsal
drites of sensory neurons located around hair follicles through-
column–medial lemniscus pathway) and pain and tempera-
out the glabrous and hairy skin, as well as in deep tissue.
ture (spinothalamic tract).
Impulses from nociceptors (pain) are transmitted via two fiber
types. One system comprises thinly myelinated Aδ fibers that
SENSORY RECEPTORS conduct at rates of 12–30 m/s. The other is unmyelinated
C fibers that conduct at low rates of 0.5–2 m/s. Thermorecep-
CUTANEOUS MECHANORECEPTORS tors also span the following two fiber types: cold receptors are
on dendritic endings of Aδ fibers and C fibers, whereas warm
Sensory receptors can be specialized dendritic endings of receptors are on C fibers. Mechanical nociceptors respond to
afferent nerve fibers and are often associated with nonneural strong pressure. Thermal nociceptors are activated by skin

115

Ch13_115-124.indd 115 11/26/10 9:49:50 AM


116 SECTION IV CNS/Neural Physiology

A Modality Touch

Receptors

Meissner’s Merkel Pacinian Ruffini


corpuscle cells corpuscle endings

B Location

Receptive
field

C Intensity and time course

Neural
spike train

Stimulus

FIGURE 13–1 Sensory systems encode four elementary attributes of stimuli: modality, location (receptive field), intensity, and
duration (timing). A) The human hand has four types of mechanoreceptors; their combined activation 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 stimulus 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 response to touch. Merkel cells and Meissner’s corpuscles provide the most precise
localization as they have the smallest receptive fields and are most sensitive to pressure applied by a small probe. C) Stimulus intensity is
signaled by firing rates of individual receptors; duration of stimulus is signaled by time course of firing. The spike trains indicate action potentials
elicited by pressure from a small probe at the center of each receptive field. Meissner’s and Pacinian corpuscles adapt rapidly; the others adapt
slowly. (Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

temperatures above 45°C or by severe cold. Chemically sensi- from an injury and is often refractory to common analgesic
tive nociceptors respond to various agents such as bradykinin, agents, including nonsteroidal anti-inflammatory drugs
histamine, high acidity, and environmental irritants. Polymodal (NSAIDs) and opiates. It can result from nerve injury includ-
nociceptors respond to combinations of these stimuli. ing diabetic neuropathy, toxin-induced nerve damage, and
Pain is defined by the International Association for the ischemia.
Study of Pain (IASP) as “an unpleasant sensory and emo- Pain is often accompanied by hyperalgesia, an exaggerated
tional experience associated with actual or potential tissue response to a noxious stimulus, and allodynia, a sensation of
damage….” This is different from nociception, which the pain in response to an innocuous stimulus. An example of the
IASP defines as the unconscious activity induced by a harm- latter is the painful sensation from a warm shower when the
ful stimulus applied to sense receptors. skin is damaged by sunburn.
Pain can be classified as physiological (or acute) pain and Hyperalgesia and allodynia signify increased sensitivity of
pathological (or chronic) pain, which includes inflamma- nociceptive afferent fibers. Figure 13–2 shows how chemicals
tory pain and neuropathic pain. Acute pain typically has a released at the site of injury can further activate nociceptors
sudden onset and recedes during the healing process. It can be leading to inflammatory pain. Injured cells release chemicals
considered “good pain” because it serves an important protec- such as K+ that depolarize nerve terminals, making nocicep-
tive mechanism. The withdrawal reflex is an example of this tors more responsive. Injured cells also release bradykinin and
protective role of pain (see Chapter 14). Chronic pain can be substance P, which can further sensitize nociceptive termi-
considered “bad pain” because it persists long after recovery nals. Histamine is released from mast cells, serotonin (5-HT)
CHAPTER 13 General Sensory Systems: Touch, Pain, and Temperature 117

Mast cell

CGRP
Substance P

Histamine Dorsal root


Bradykinin ganglion neuron
Lesion 5-HT
Prostaglandin
K+
CGRP
Substance P

Blood
vessel
Spinal cord

FIGURE 13–2 In response to tissue injury, chemical mediators can sensitize and activate nociceptors. These factors contribute to
hyperalgesia and allodynia. Tissue injury releases bradykinin and prostaglandins that sensitize or activate nociceptors, which in turn releases
substance P and calcitonin gene-related peptide (CGRP). Substance P acts on mast cells to cause degranulation and release histamine, which
activates nociceptors. It causes plasma extravasation and CGRP dilates blood vessels; the resulting edema causes additional release of
bradykinin. Serotonin (5-HT) is released from platelets and activates nociceptors. (Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM
[editors]: Principles of Neural Science. McGraw-Hill, 2000.)

from platelets, calcitonin gene-related peptide (CGRP) from Modality is the type of energy transmitted by the stimu-
nerve terminals, and prostaglandins from cell membranes, all lus. The particular form of energy to which a receptor is
contributing to the inflammatory process and activating most sensitive is called its adequate stimulus. Location is
or sensitizing the nociceptors. Some released substances act by the site on the body or space where the stimulus originated.
releasing another one (e.g., bradykinin activates both Aδ and A sensory unit is a single sensory axon and all its peripheral
C fibers and increases synthesis and release of prostaglandins). branches; the receptive field of a sensory unit is the spatial
Prostaglandin E2 (a cyclooxygenase metabolite of arachidonic distribution from which a stimulus produces a response in
acid) is released from damaged cells and produces hyperalge- that unit (Figure 13–1). One of the most important mecha-
sia. This is why aspirin and other NSAIDs (inhibitors of nisms that enable localization of a stimulus site is lateral
cyclooxygenase) alleviate pain. inhibition. Activity arising from sensory neurons whose
receptors are at the peripheral edge of the stimulus is inhib-
ited compared to that from the sensory neurons at the cen-
SENSORY RECEPTORS IN ter of the stimulus. Thus, lateral inhibition enhances the
SKELETAL MUSCLES & JOINTS contrast between the center and periphery of a stimulated
area and increases the ability of the brain to localize a sen-
Skeletal muscles contain receptors called muscle spindles and sory input.
Golgi tendon organs that are important for proprioception. Lateral inhibition underlies the neurological assessment
They play major roles in motor control and are described in called the two-point discrimination test, which is used to test
Chapter 14. Muscles also contain nociceptors that respond to the integrity of the dorsal column (medial lemniscus) system,
pressure and the release of metabolites during ischemia. Limb the central pathway for touch and proprioception. In this pro-
joints also contain mechanoreceptors (Pacinian and Ruffini cedure, the two points on a pair of calipers are simultaneously
corpuscles) and nociceptors. positioned on the skin and one determines the minimum dis-
tance between the two caliper points that can be perceived as
separate points of stimulation. This is called the two-point
SENSORY CODING discrimination threshold and is a measure of tactile acuity. If
the distance is very small, each caliper point is touching the
Converting a receptor stimulus to a recognizable sensation is receptive field of only one sensory neuron. If the distance
termed sensory coding. All sensory systems code for four between stimulation points is less than this threshold, only
elementary attributes of a stimulus: modality, location, inten- one point of stimulation can be felt. The magnitude of two-
sity, and duration. These attributes of sensory coding are point discrimination thresholds varies from place to place on
shown for the modality of touch in Figure 13–1. the body and is smallest where touch receptors are most abun-
118 SECTION IV CNS/Neural Physiology

dant. Stimulus points on the back, for instance, must be sepa- To dorsal columns
rated by at least 65 mm before they can be distinguished as
separate, whereas on the fingertips, two stimuli are recognized Mechanoreceptors Aβ
if they are separated by as little as 2 mm. Mechanoreceptors
Intensity is signaled by the response amplitude or fre- Nociceptors Aδ
quency of action potential generation. Duration refers to the Cold receptors
time from start to end of a response in the receptor. If a stim- I Nociceptors
ulus of constant strength is applied to a receptor, the fre- II Thermoreceptors C
quency of the action potentials in its sensory nerve declines III
Mechanoreceptors
over time. This phenomenon is known as adaptation or IV
desensitization. The degree to which adaptation occurs var- V
ies from one sense to another. Receptors can be classified VI
into rapidly adapting (phasic) receptors and slowly adapt- VII
ing (tonic) receptors. This is illustrated for different types of
touch receptors in Figure 13–1.

FIGURE 13–3 Schematic representation of the terminations


SOMATOSENSORY PATHWAYS of the three types of primary afferent neurons in the various
layers of the dorsal horn of the spinal cord. (Reproduced with
DORSAL HORN permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of
Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
The dorsal horn in the spinal cord is divided on the basis of
histologic characteristics into laminae I–VII, with I being
the most superficial and VII the deepest. Lamina II and part fibers from the sacral cord are positioned most medially and
of lamina III make up the substantia gelatinosa, the area those from the cervical cord most laterally. This arrangement
near the top of each dorsal horn. Three types of primary continues in the medulla with lower body (e.g., foot) represen-
afferent fibers (with cell bodies in the dorsal root ganglia) tation in the gracilis nucleus and upper body (e.g., finger)
mediate cutaneous sensation: (1) large myelinated Aα and representation in cuneate nucleus. The medial lemniscus is
Aβ fibers that transmit impulses generated by mechanical organized dorsal to ventral, representing neck to foot.
stimuli; (2) small myelinated Aδ fibers that transmit impulses Somatotopic organization continues through the thalamus
from cold receptors, nociceptors, or mechanoreceptors; and and cortex. VPL thalamic neurons carrying sensory informa-
(3) small unmyelinated C fibers that are concerned primar- tion project in a highly specific way to the two somatic sensory
ily with pain and temperature. The orderly distribution of areas of the cortex (Figure 13–5): somatic sensory area I (SI)
these fibers in various layers of the dorsal horn is shown in in the postcentral gyrus and somatic sensory area II (SII) in
Figure 13–3. the wall of the Sylvian fissure. In addition, SI projects to SII. SI
corresponds to Brodmann’s areas 3, 2, and 1.
The arrangement of projections to SI is such that the parts of
DORSAL COLUMN PATHWAY the body are represented in order along the postcentral gyrus,
with the legs on top and the head at the foot of the gyrus
The principal direct pathways to the cerebral cortex for (Figure 13–5). Not only is there detailed localization of the
touch, vibratory sense, and proprioception (position sense) fibers from the various parts of the body in the postcentral
are shown in Figure 13–4. Fibers mediating these sensations gyrus, but also the size of the cortical receiving area for
ascend ipsilaterally in the dorsal columns to the medulla, impulses from a particular part of the body is proportionate to
where they synapse in the gracilis and cuneate nuclei. The the use of the part. The relative sizes of the cortical receiving
second-order neurons from these nuclei cross the midline areas are shown dramatically in Figure 13–6, in which the
and ascend in the medial lemniscus to end in the contralat- proportions of the homunculus are distorted to correspond to
eral ventral posterior lateral (VPL) nucleus and related the size of the cortical receiving areas for each. Note that the
specific sensory relay nuclei of the thalamus. This ascending cortical areas for sensation from the trunk and back are small,
system is called the dorsal column or medial lemniscal sys- whereas very large areas are concerned with impulses from the
tem. The fibers within the dorsal column pathway are joined hand and the parts of the mouth concerned with speech.
in the brain stem by fibers mediating sensation from the SII is located in the superior wall of the Sylvian fissure, the
head. Touch and proprioception are relayed mostly via the fissure that separates the temporal from the frontal and pari-
main sensory and mesencephalic nuclei of the trigeminal etal lobes. The head is represented at the inferior end of the
nerve. postcentral gyrus, and the feet at the bottom of the Sylvian fis-
Within the dorsal columns, fibers arising from different lev- sure. The representation of the body parts is not as complete
els of the cord are somatotopically organized. Specifically, or detailed as it is in the postcentral gyrus.
CHAPTER 13 General Sensory Systems: Touch, Pain, and Temperature 119

Postcentral
gyrus

Axons of
third-order
neurons

Thalamus

Cerebral
cortex

Medial lemniscal tract


(axons of second-order
neurons)

Medulla oblongata

Fasciculus cuneatus
Lateral spinothalamic tract
(axons of first-order
(axons of second-order neurons)
sensory neurons)

Joint stretch receptor


(proprioceptor)
Pain receptor
Spinal cord
Axons of first-order
Fasciculus gracilis neurons (not part of
(axons of first-order spinothalamic tract)
sensory neurons)

Temperature
receptor
(a) Touch receptor (b)

FIGURE 13–4 Ascending tracts carrying sensory information from peripheral receptors to the cerebral cortex. a) Dorsal column
pathway mediating touch, vibratory sense, and proprioception. b) Ventrolateral spinothalamic tract mediating pain and temperature.
(Reproduced with permission from Fox SI: Human Physiology. McGraw-Hill, 2008.)

Sl
Posterior VENTROLATERAL
8 6 4
3 1 2 parietal SPINOTHALAMIC TRACT
9 cortex
Trunk
Fibers from nociceptors and thermoreceptors synapse on neu-
Hand rons in the dorsal horn (Figure 13–3). The axons from these
neurons cross the midline and ascend in the ventrolateral
Sll Face
Tongue quadrant of the spinal cord, where they form the lateral
spinothalamic tract (Figure 13–4). Fibers within this tract
Auditory
synapse in the VPL nuclei. Other dorsal horn neurons that
al
receive nociceptive input synapse in the reticular formation of
su
Vi the brain stem (spinoreticular pathway) and then project to
the centrolateral nucleus of the thalamus.
FIGURE 13–5 Brain areas concerned with somatic sensation, Positron emission tomographic (PET) and functional
and some of the cortical receiving areas for other sensory
magnetic resonance imaging (fMRI) studies in normal
modalities in the human brain. The numbers are those of
Brodmann’s cortical areas. The primary auditory area is actually
humans indicate that pain activates cortical areas SI, SII, and
located in the Sylvian fissure on the top of the superior temporal the cingulate gyrus on the side opposite the stimulus. Also,
gyrus and is not normally visible in a lateral view of the cortex. the mediofrontal cortex and insular cortex are activated.
(Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks These technologies were important in distinguishing
H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) two components of pain pathways. From VPL nuclei in the
120 SECTION IV CNS/Neural Physiology

afferent terminals that also synapse in the dorsal horn. This is


called the gate-control hypothesis.

Neck
Head

Hip
Trunk
Shoulder
Arm
MORPHINE & ENKEPHALINS

Leg
Elbow
Forea
Wris d
Han
ot

Litt g
Fo

t
Ri ddle

rm
le
M i dex b
n e s
To One of the most effective analgesic agents is morphine. The
In um

.
Gen
Th

E
No ye receptors that bind morphine and the endogenous mor-
s
Fa e
ce phines, the opioid peptides, are found in the midbrain, brain
Upp
er li
p stem, and spinal cord. There are at least three sites at which
Lips opioids can act to produce analgesia: peripherally, at the site
Lower lip of an injury; in the dorsal horn, where nociceptive fibers syn-
Teeth, gums, and jaw apse on dorsal root ganglion cells; and at more rostral sites in
Tongue the brain stem.
Pharynx
Figure 13–7 shows various modes of action of opiates to
Intra- decrease transmission in pain pathways. Opioid receptors are
abdominal located on dorsal root ganglion cells and on afferent nerve
fibers. In the periphery, inflammation causes the production
of opioid peptides by immune cells, and these may act on the
receptors in the afferent nerve fibers to reduce the pain that
FIGURE 13–6 Sensory homunculus, drawn overlying a would otherwise be felt. The opioid receptors in the dorsal
coronal section through the postcentral gyrus. Gen., genitalia. horn region may act presynaptically to decrease release of sub-
(Reproduced with permission from Penfield W, Rasmussen G: The Cerebral Cortex stance P.
of Man. Macmillan, 1950.) Injections of morphine into the periaqueductal gray matter
(PAG) of the midbrain relieve pain by activating descending
pathways that produce inhibition of primary afferent transmis-
thalamus, fibers project to SI and SII. This is called the neo- sion in the dorsal horn. This activation may occur via projections
spinothalamic tract, and it is responsible for the immediate from the PAG to the raphe magnus nucleus and descending
awareness of the painful sensation and the awareness of the serotonergic fibers from this nucleus mediate the inhibition.
location of the noxious stimulus. The pathway that includes Acupuncture at a location distant from the site of a pain may
synapses in the brain stem reticular formation and centrolat- act by releasing endorphins in the brain; acupuncture at the
eral thalamic nucleus projects to the frontal lobe, limbic sys- site of the pain appears to act primarily in the same way as
tem, and insula. This is called the paleospinothalamic tract, touching or shaking (gate-control mechanism).
and it mediates the emotional response to pain.
In the CNS, visceral sensation travels along the same path-
ways as somatic sensation in the spinothalamic tracts and NEUROLOGICAL EXAM
thalamic radiations, and the cortical receiving areas for vis- The sensory component of a neurological exam includes an
ceral sensation are intermixed with the somatic receiving assessment of various sensory modalities including touch,
areas. This likely contributes to the phenomenon called proprioception, vibratory sense, and pain. The integrity of the
referred pain. Irritation of a visceral organ produces pain pain pathway is assessed by stimulating the skin with a pin and
that is felt not at that site but in a somatic structure. Such asking the patient if the stimulus is perceived as sharp. To test
pain is said to be referred to the somatic structure. Perhaps for proprioception, a physician holds the patient’s finger (toe,
the best-known example is referral of cardiac pain to the hand, or foot) and, with the subject’s eyes closed, asks whether
inner aspect of the left arm. the digit is being moved up or down. Vibratory sensibility is
tested by applying a vibrating (128-Hz) tuning fork to the skin
on the fingertip, tip of the toe, or bony prominences of the
MODULATION OF toes. The normal response is a “buzzing” sensation. The sensa-
PAIN TRANSMISSION tion is most marked over bones. A pattern of rhythmic pres-
sure stimuli is interpreted as vibration. The impulses
Many people have learned from practical experience that responsible for the vibrating sensation are carried in the dorsal
touching or shaking an injured area decreases the pain due to columns. Degeneration of this part of the spinal cord occurs in
the injury. The relief may result from inhibition of pain path- poorly controlled diabetes mellitus, pernicious anemia, and
ways in the dorsal horn gate by stimulation of large-diameter vitamin B12 deficiencies. Elevation of the threshold for vibra-
touch–pressure afferents. Figure 13–3 shows that collaterals tory stimuli is an early symptom of this degeneration. Vibratory
from these myelinated afferent fibers synapse in the dorsal sensation and proprioception are closely related; when one is
horn. These collaterals may modify the input from nociceptive diminished, so is the other.
CHAPTER 13 General Sensory Systems: Touch, Pain, and Temperature 121

A Norepinephrine
Nociceptor Serotonin

ENK

Projection
neuron

B1 Sensory input B2 Sensory input + opiates/opioids

Control Control
Opiate
Nociceptor Morphine

Glutamate
Neuropeptides

Glutamate Neuropeptides

Enkephalin Enkephalin
Morphine

Ca2+ Ca2+

No input + opiates
No input Enkephalin

Sensory input Sensory input + opiates


Control Control
Projection Enkephalin
neuron

FIGURE 13–7 Local circuit interneurons in the superficial dorsal horn of the spinal cord integrate descending and afferent
pathways. A) Possible interactions of nociceptive afferent fibers, interneurons, and descending fibers in the dorsal horn. Nociceptive fibers
terminate on second-order spinothalamic projection neurons. Enkephalin (ENK)-containing interneurons exert both presynaptic and
postsynaptic inhibitory actions. Serotonergic and noradrenergic neurons in the brain stem activate opioid interneurons and suppress the
activity of spinothalamic projection neurons. B1) Activation of nociceptors releases glutamate and neuropeptides from sensory terminals,
depolarizing and activating projection neurons. B2) Opiates decrease Ca2+ influx leading to a decrease in the duration of nociceptor action
potentials and a decreased release of transmitter. Also, they hyperpolarize the membrane of dorsal horn neurons by activating K+ conductance
and decrease the amplitude of the EPSP (see Chapter 7) produced by stimulation of nociceptors. (Reproduced with permission from Kandel ER,
Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

Stereognosis is the perception of the form and nature of occurs in the absence of any detectable defect in touch and
an object without looking at it. Healthy subjects can readily pressure sensation when there is a lesion in the parietal lobe
identify objects such as keys and coins of various denomina- posterior to the postcentral gyrus. Stereoagnosis can also be
tions. This ability depends on relatively intact touch and expressed by the failure to identify an object by sight (visual
pressure sensation and is compromised when the dorsal col- agnosia), the inability to identify sounds or words (audi-
umns are damaged. The inability to identify an object by tory agnosia) or color (color agnosia), or the inability to
touch is called tactile agnosia. Impaired stereognosis is an identify the location or position of an extremity (position
early sign of damage to the cerebral cortex and sometimes agnosia).
122 SECTION IV CNS/Neural Physiology

■ Nociceptors and thermoreceptors are free nerve endings on


CLINICAL CORRELATION unmyelinated or lightly myelinated fibers in hairy and glabrous
skin and deep tissues.
A 55-year-old female executive at a large corporation ■ Hyperalgesia is an exaggerated response to a noxious stimulus;
developed a burning sensation in the palm of her right allodynia is a sensation of pain in response to an innocuous
hand about 6 months ago. She also noticed tingling and stimulus.
numbness in her right thumb, index finger, and middle ■ Converting a receptor stimulus to a recognizable sensation
finger. These symptoms first developed after spending is termed sensory coding. All sensory systems code for four
many long hours at the computer preparing annual report elementary attributes of a stimulus: modality, location,
documents for the corporation. Initially, symptoms were intensity, and duration.
most prominent at night, interrupting her sleep. The ■ Discriminative touch, proprioception, and vibratory sensations
problem has recently intensified, and she now has pain in are relayed via the dorsal column (medial lemniscus) pathway
to SI. Pain and temperature sensations are mediated via the
her right wrist and difficulty picking up small objects
ventrolateral spinothalmic tract to SI.
from her desk. She visited her doctor because working on
■ Descending pathways from the mesencephalic PAG inhibit
the computer has become increasingly more difficult.
transmission in nociceptive pathways. This descending
Her physician performed several simple diagnostic tests. pathway includes a synapse in the raphe nucleus and the
When the doctor pressed on her median nerve in the wrist, release of endogenous opiates.
she experienced a shocklike sensation (Tinel’s sign). When ■ Morphine is an effective antinociceptive agent that binds to
the doctor had her hold her forearms upright by pointing endogenous opiate receptors in the midbrain, brain stem, and
the fingers down and pressing the backs of the hands spinal cord.
together, within 1 minute she felt tingling and increasing
numbness in her fingers (Phalen’s sign). Nerve conduction
tests indicated slowed conduction in the median nerve. STUDY QUESTIONS
She was diagnosed with carpal tunnel syndrome,
1. Pacinian corpuscles are
which is due to compression (possibly due to inflamma-
A) a type of thermoreceptor.
tion) of the median nerve that passes through the tun-
B) usually innervated by Aδ nerve fibers.
nel. It is more prevalent in women than men and is
C) rapidly adapting touch receptors.
diagnosed primarily in individuals who use their wrists D) slowly adapting touch receptors.
in repetitive activities (computer operators, cashiers, E) nociceptors.
musicians, painters). About 3% of women and 2% of 2. Adaptation to a sensory stimulus produces
men are likely to be diagnosed with this syndrome dur-
A) a diminished sensation when other types of sensory stimuli
ing their lifetime. The median nerve provides sensory are withdrawn.
information from the thumb, index, and ring fingers, B) a more intense sensation when a given stimulus is applied
and the nine tendons that flex the fingers. The syndrome repeatedly.
is characterized by pain, paresthesia, and weakness in C) a sensation localized to the hand when the nerves of the
the distribution of the median nerve. Wrist or hand pain brachial plexus are stimulated.
or numbness and tingling of the fingers (except the little D) a diminished sensation when a given stimulus is applied
finger which is not innervated by the median nerve) are repeatedly over time.
often the first symptoms. Patients sometimes report E) a decreased firing rate in the sensory nerve from the
weakness in the hand and a tendency to drop things. receptor when one’s attention is directed to another
matter.
Symptoms often first appear at night rather than during
activity. Wrist splinting, NSAIDs, or corticosteroids are 3. Sensory systems code for which of the following attributes
of a stimulus?
frequently the therapy of choice. If pain persists follow-
ing these treatments, surgery may be required. 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
CHAPTER SUMMARY 4. Thermoreceptors
■ Sensory receptors are commonly classified as mechanoreceptors, A) are activated only by severe cold or severe heat.
nociceptors, chemoreceptors, or photoreceptors. B) are located on superficial layers of the skin.
■ Touch and pressure are sensed by four types of C) are a subtype of nociceptor.
mechanoreceptors: Meissner’s corpuscles (respond to D) are on dendritic endings of Aδ fibers and C fibers.
changes in texture and slow vibrations), Merkel cells E) All of the above.
(respond to sustained pressure and touch), Ruffini corpuscles
(respond to sustained pressure), and Pacinian corpuscles
(respond to deep pressure and fast vibrations).
CHAPTER 13 General Sensory Systems: Touch, Pain, and Temperature 123

5. A 50-year-old woman undergoes a neurological exam that


indicates loss of pain and temperature sensitivity, vibratory sense,
and proprioception in both legs. These symptoms could be
explained by
A) a tumor on the medial lemniscal pathway in the sacral
spinal cord.
B) a peripheral neuropathy.
C) a large tumor in the sacral dorsal horn.
D) a large tumor affecting the posterior paracentral gyri.
E) a large tumor in the ventral posterolateral and
posteromedial thalamic nuclei.
This page intentionally left blank
14
C H A P T E R

Spinal Reflexes
Susan M. Barman

O B J E C T I V E S

■ Describe the components of a reflex arc.


■ Describe the muscle spindles and their role in the stretch reflex.
■ Describe the functions of the Golgi tendon organs as part of a
feedback system that maintains muscle force.
■ Define reciprocal innervation, inverse stretch reflex, and clonus.
■ Describe the short- and long-term effects of spinal cord injury on
spinal reflexes.

INTRODUCTION The simplest reflex arc is one with a single synapse between
the afferent and efferent neurons. Such arcs are monosynaptic,
The basic unit of integrated reflex activity is the reflex arc. and reflexes occurring in them are called monosynaptic
This arc consists of a sense organ, an afferent neuron, synapses reflexes. Reflex arcs in which one or more interneuron is
within a central integrating station, an efferent neuron, and an interposed between the afferent and efferent neurons are called
effector organ. The afferent neurons enter the central nervous polysynaptic reflexes. There can be anywhere from two to
system (CNS) via the spinal dorsal roots or cranial nerves and hundreds of synapses in a polysynaptic reflex arc.
have their cell bodies in the dorsal root ganglia or in the As will be evident from the description below, reflex activity
homologous ganglia for the cranial nerves. The efferent fibers is stereotyped and specific in terms of both the stimulus and
leave the CNS via the spinal ventral roots or corresponding the response; a particular stimulus elicits a particular response.
motor cranial nerves. The fact that reflex responses are stereotyped does not
Activity in the reflex arc starts in a sensory receptor with a exclude the possibility of their being modified by experience.
generator potential whose magnitude is proportional to the Reflexes are adaptable and can be modified to perform motor
strength of the stimulus (Figure 14–1). This generates all-or- tasks and maintain balance. Descending inputs from higher
none action potentials in the afferent nerve, the number of brain regions play an important role in modulating and adapt-
action potentials being proportional to the size of the genera- ing spinal reflexes.
tor potential. In the CNS, the responses are again graded in
terms of excitatory postsynaptic potentials (EPSPs) and
inhibitory postsynaptic potentials (IPSPs) at the synaptic MONOSYNAPTIC REFLEX:
junctions (see Chapter 7). All-or-none responses are generated
in the efferent nerve. When these reach the effector organ, they
THE STRETCH REFLEX
again set up a graded response. When the effector is smooth When a skeletal muscle with an intact nerve supply is stretched,
muscle, responses summate to produce action potentials in the it contracts. This response is called the stretch reflex. The stim-
smooth muscle, but when the effector is skeletal muscle, the ulus that initiates the reflex is stretch of the muscle, and the
graded response is adequate to produce action potentials that response is contraction of the same muscle. The sense organ
bring about muscle contraction. Activity within the reflex arc (receptor) is a small encapsulated spindlelike or fusiform-
is modified by the multiple inputs converging on the efferent shaped structure called the muscle spindle, located within the
neurons or at any synaptic station within the reflex loop. fleshy part of the muscle. The impulses originating from the

125

Ch14_125-132.indd 125 11/26/10 9:49:10 AM


126 SECTION IV CNS/Neural Physiology

Sense Afferent Synapse Efferent Neuromuscular Muscle


organ neuron neuron junction

Generator Action EPSPs Action Endplate Action


potential potentials (and IPSPs) potentials potentials potentials
FIGURE 14–1 Reflex arc. At the receptor and in the CNS a nonpropagated, graded response occurs that is proportionate to the magnitude
of the stimulus. The response at the neuromuscular junction is also graded, though under normal conditions it is always large enough to
produce a response in skeletal muscle. On the other hand, in the portions of the arc specialized for transmission (afferent and efferent axons,
muscle membrane), the responses are all-or-none action potentials. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s
Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

spindle are transmitted to the CNS by fast sensory fibers the ends of the spindle capsule attached to the tendons at either
(group Ia) that pass directly to the motor neurons that supply end of the muscle. They do not contribute to the overall con-
the same muscle. tractile force of the muscle, but rather serve a purely sensory
The stretch reflex is the best-known and studied monosyn- function. There are two types of intrafusal fibers in mamma-
aptic reflex and is typified by the knee-jerk reflex. Tapping lian muscle spindles. The first type contains many nuclei in a
the patellar tendon elicits the knee jerk, a stretch reflex of the dilated central area and is called a nuclear bag fiber (Figure
quadriceps femoris muscle, because the tap on the tendon 14–2B). There are two subtypes of nuclear bag fibers, dynamic
stretches the muscle. The knee-jerk reflex is an example of a and static. Typically, there are two or three nuclear bag fibers
deep tendon reflex in a neurological exam. Absence of the per spindle. The second intrafusal fiber type, the nuclear
knee jerk can signify an abnormality anywhere within the chain fiber, is thinner and shorter and lacks a definite bag.
reflex arc, including the muscle spindle, the Ia afferent nerve Each spindle has about five nuclear chain fibers.
fibers, or the motor neurons to the quadriceps muscle. The There are two kinds of sensory endings in each spindle, a sin-
most common cause is a peripheral neuropathy from such gle primary (group Ia) ending and up to eight secondary
things as diabetes mellitus, alcoholism, and toxins. A hyper- (group II) endings. The Ia afferent fiber wraps around the center
active reflex can signify an interruption of inhibitory corti- of the dynamic and static nuclear bag fibers and nuclear chain
cospinal and other descending pathways that influence the fibers. Group II sensory fibers are located adjacent to the centers
reflex arc. of the static nuclear bag and nuclear chain fibers; these fibers do
not innervate the dynamic nuclear bag fibers. Ia afferent fibers
are very sensitive to the velocity of the change in muscle length
STRUCTURE OF MUSCLE SPINDLES during a stretch (dynamic response); thus, they provide infor-
mation about the speed of movements and allow for quick cor-
Figure 14–2A illustrates the composition of a muscle spindle rective movements. The steady-state (tonic) activity of group Ia
and its innervation. Each muscle spindle has three essential and II afferent fibers provides information on steady-state length
elements: (1) a group of specialized intrafusal muscle fibers of the muscle (static response). The top trace in Figure 14–2C
with contractile polar ends and a noncontractile center, shows the dynamic and static components of activity in a Ia affer-
(2) large-diameter myelinated afferent nerves (types Ia and II) ent fiber during muscle stretch. Note that they discharge most
originating in the central portion of the intrafusal fibers, and rapidly while the muscle is being stretched (shaded area of
(3) small-diameter myelinated efferent nerves supplying the graphs) and less rapidly during sustained stretch.
polar contractile regions of the intrafusal fibers. It is important The spindles have their own efferent motor nerve supply
to understand the relationship of these elements to each other called γ-motor neurons. They are small-diameter (3–6 μm)
and to the skeletal muscle itself to appreciate the role of this fibers and constitute about 30% of the fibers in the ventral
sense organ in signaling changes in the length of the muscle in roots. There are two types of γ-motor neurons: dynamic,
which it is located. Changes in muscle length are associated which supply the dynamic nuclear bag fibers, and static,
with changes in joint angle; thus, muscle spindles provide which supply the static nuclear bag fibers and the nuclear
information on position (i.e., proprioception). chain fibers. Activation of dynamic γ-motor neurons increases
The intrafusal fibers are located in parallel to the extra- the dynamic sensitivity of the group Ia afferent endings.
fusal fibers (the regular contractile units of the muscle) with Activation of the static γ-motor neurons increases the tonic
CHAPTER 14 Spinal Reflexes 127

A Muscle spindle B Intrafusal fibers of the muscle spindle C Response of Ia sensory fiber to selective
activation of motor neurons
Static nuclear 200
bag fiber
Dynamic nuclear

Imp/s
bag fiber Dynamic response

Intrafusal Steady-state response


muscle 0 Stretch alone
fibers Nuclear
chain fiber
200
Capsule

Imp/s
II
Sensory Ia
endings
0
Stimulate static gamma fiber
Static
Afferent
200
axons

Imp/s
Efferent
axons Dynamic

0
Stimulate dynamic gamma fiber
Gamma
motor 6

Stretch
endings
0

0.2 s

FIGURE 14–2 Mammalian muscle spindle. A) Diagrammatic representation of the main components of mammalian muscle spindle
including intrafusal muscle fibers, afferent sensory fiber endings, and efferent motor fibers (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 innervates nuclear chain and static bag fibers to form a secondary sensory ending. Dynamic
γ-motor neurons innervate dynamic bag fibers; static γ-motor neurons innervate combinations of chain and static bag fibers. C) Comparison of
discharge pattern of Ia afferent activity during stretch alone and during stimulation of static or dynamic γ-motor neurons. Without γ-stimulation,
Ia afferent fibers show a small dynamic response to muscle stretch and a modest increase in steady-state firing. When static γ-motor neurons are
activated, the steady-state response increases and the dynamic response decreases. When dynamic γ-motor neurons are activated, the dynamic
response is markedly increased but the steady-state response gradually returns to its original level. (Reproduced with permission from Kandel ER, Schwartz
JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

level of activity in both group Ia and II afferent endings, is the time taken for the reflex activity to traverse the spinal
decreases the dynamic sensitivity of group Ia afferent fibers, cord. In humans, the central delay for the knee jerk is 0.6–0.9
and can prevent silencing of Ia afferent fibers during muscle millisecond. Since the minimal synaptic delay is 0.5 millisec-
stretch (Figure 14–2C). ond, only one synapse could have been traversed.
Muscle spindles also make connections that cause muscle con-
traction via polysynaptic pathways, and the afferent fibers
CENTRAL CONNECTIONS OF involved are probably those from the secondary group II
AFFERENT FIBERS endings.

Group Ia afferent fibers end directly on α-motor neurons sup-


plying the extrafusal fibers of the same muscle (Figure 14–3). FUNCTION OF MUSCLE SPINDLES
The time between the application of the stimulus and the
response is called the reaction time. In humans, the reaction When the muscle spindle is stretched, its sensory endings
time for a stretch reflex such as the knee jerk is 19–24 millisec- are distorted and receptor potentials are generated. These
onds. Because the conduction velocities of the afferent and in turn set up action potentials in the sensory fibers at a
efferent fiber types are known and the distance from the mus- frequency proportional to the degree of stretching. Because
cle to the spinal cord can be measured, it is possible to calcu- the spindle is in parallel with the extrafusal fibers, when the
late how much of the reaction time was taken up by conduction muscle is passively stretched, the spindles are also stretched,
to and from the spinal cord. When this value is subtracted referred to as “loading the spindle.” This initiates reflex
from the reaction time, the remainder, called the central delay, contraction of the extrafusal fibers in the skeletal muscle. On
128 SECTION IV CNS/Neural Physiology

Dorsal root Spindle


Tendon Extrafusal fiber
Sensory nerve
Interneuron releasing
inhibitory mediator

Motor neuron Impulses in


Ib fiber sensory nerve
from I a fiber Muscle at rest
Golgi from
tendon muscle
organ spindle Ventral root

Motor endplate
on extrafusal fiber
Muscle stretched

FIGURE 14–3 Diagram illustrating the pathways responsible


for the stretch reflex and the inverse stretch reflex. Stretch
stimulates the muscle spindle, which activates Ia afferent fibers that
excite the motor neuron. It also stimulates the Golgi tendon organ,
which activates Ib afferent 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. (Reproduced with permission from Barrett KE, Barman SM, Muscle contracted
Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill
Medical, 2009.)

the other hand, the muscle spindle afferent fibers character-


istically stop firing when the muscle is made to contract by
electrical stimulation of the α-motor neurons to the extra-
fusal fibers because the muscle shortens while the spindle is
unloaded (Figure 14–4). Thus, the spindle and its reflex con- Increased γ efferent discharge
nections constitute a feedback device that operates to main-
tain 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.
When a stretch reflex occurs, the muscles that antagonize
the action of the muscle involved (antagonists) relax. This
phenomenon is due to reciprocal innervation. Impulses in Increased γ efferent
the Ia afferent fibers from the muscle spindles of the protago- discharge—muscle stretched
nist muscle cause postsynaptic inhibition of the α-motor FIGURE 14–4 Effect of various conditions on muscle spindle
neurons to the antagonists. The pathway mediating this effect discharge. When the whole muscle is stretched, the muscle spindle
is bisynaptic. A collateral from each Ia afferent fiber passes in is also stretched and its sensory endings are activated at a
the spinal cord to an inhibitory interneuron that synapses on a frequency proportional to the degree of stretching (“loading the
motor neuron supplying the antagonist muscles. spindle”). Spindle afferent fibers stop firing when the muscle
contracts (“unloading the spindle”). Stimulation of γ-motor neurons
EFFECTS OF γ-MOTOR causes the contractile ends of the intrafusal fibers to shorten. This
stretches the nuclear bag region, initiating impulses in sensory fibers.
NEURON DISCHARGE If the whole muscle is stretched during stimulation of the γ-motor
neurons, the rate of discharge in sensory fibers is further increased.
Stimulation of γ-motor neurons causes the contractile ends of (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H:
the intrafusal fibers to shorten and therefore stretches the Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
CHAPTER 14 Spinal Reflexes 129

nuclear bag portion of the spindles, deforming the endings tory interneurons that in turn terminate directly on the
and initiating impulses in the Ia afferent fibers (Figure 14–4). α-motor neurons (Figure 14–3). They also make excitatory
This can lead to reflex contraction of the muscle. Thus, skeletal connections with α-motor neurons supplying antagonists to
muscle can be made to contract via stimulation of the α-motor the muscle.
neurons that innervate the extrafusal fibers or the γ-motor Unlike the spindles, the Golgi tendon organs are in series
neurons that initiate contraction indirectly via the stretch with the muscle fibers. Thus, they are stimulated by both pas-
reflex. Increased γ-motor neuron activity increases spindle sive stretch and active contraction of the muscle. The thresh-
sensitivity during stretch. old of the Golgi tendon organs is low. The degree of stimulation
In response to descending excitatory input to spinal motor by passive stretch is not great because the more elastic muscle
circuits, both α- and γ-motor neurons are activated. Because fibers take up much of the stretch, and this is why it takes a
of this “α–γ coactivation,” intrafusal and extrafusal fibers strong stretch to produce relaxation. However, discharge is
shorten together, and spindle afferent activity can occur regularly produced by contraction of the muscle, and the Golgi
throughout the period of muscle contraction. In this way, the tendon organ thus functions as a transducer in a feedback
spindle remains capable of responding to stretch and adjusting circuit that regulates muscle force in a fashion analogous to
reflexly α-motor neuron discharge. the spindle feedback circuit that regulates muscle length.
Input from various brain regions to the γ-motor neurons The importance of the primary endings in the spindles and
influences the sensitivity of the muscle spindles. Thus, the the Golgi tendon organs in regulating the velocity of the muscle
threshold of the stretch reflexes in various parts of the body contraction, muscle length, and muscle force is illustrated by
can be adjusted and shifted to meet the needs of postural con- the fact that that section of the afferent nerves to an arm causes
trol. Anxiety causes an increased γ-motor neuron discharge, the limb to hang loosely in a semi-paralyzed state. The organi-
which may explain the appearance of hyperactive tendon zation of the system is shown in Figure 14–6.
reflexes in anxious patients. Also, unexpected movement is
associated with a greater discharge.
MUSCLE TONE
INVERSE STRETCH REFLEX The resistance of a muscle to stretch is often referred to as its
tone. If the motor nerve to a skeletal muscle is cut, the muscle
Up to a point, the harder a muscle is stretched, the stronger is offers very little resistance and is said to be flaccid. A hypertonic
the reflex contraction. However, when the tension becomes (spastic) muscle is one in which the resistance to stretch is high
great enough, contraction suddenly ceases and the muscle because of hyperactive stretch reflexes. Somewhere between the
relaxes. This relaxation in response to strong stretch is called states of flaccidity and spasticity is the ill-defined area of normal
the inverse stretch reflex. tone. The muscles are generally hypotonic when the rate of
The receptor for the inverse stretch reflex is in the Golgi γ-motor neuron discharge is low and hypertonic when it is high.
tendon organ (Figure 14–5). This organ consists of a netlike When the muscles are hypertonic, the sequence of moderate
collection of knobby nerve endings among the fascicles of a stretch → muscle contraction, and strong stretch → muscle
tendon. There are 3–25 muscle fibers per tendon organ. The relaxation is seen. Passive flexion of the elbow, for example,
sensory fibers from the Golgi tendon organs form the Ib group meets immediate resistance as a result of the stretch reflex in
of myelinated, rapidly conducting nerve fibers. Stimulation of the triceps muscle. Further stretch activates the inverse stretch
these Ib afferent fibers leads to the production of IPSPs on the reflex. The resistance to flexion suddenly collapses, and the
α-motor neurons that supply the muscle from which the fibers arm flexes. Continued passive flexion stretches the muscle
arise. The Ib afferent fibers end in the spinal cord on inhibi- again, and the sequence may be repeated. This sequence of

Nerve fiber
Tendon bundles

Organ of Golgi, showing Muscular fibers


ramification of nerve fibrils
FIGURE 14–5 Golgi tendon organ. (Reproduced with permission from Gray H [editor]: Gray’s Anatomy of the Human Body, 29th ed. Lea & Febiger, 1973.)
130 SECTION IV CNS/Neural Physiology

Interneuronal
control
signal Inter- Force feedback Tendon
neurons organs
External
Internal forces
disturbances
α Control − Muscular + Muscle
signal + Efferent signal force length
α Muscle Load

+
Length and velocity
Length and velocity
feedback
γ-Dynamic Spindles
control
signal
γd
γ-Static
control
signal
γs

FIGURE 14–6 Block diagram of peripheral motor control system. Non-neural feedback from muscle (“Length and velocity”) that limits
length and velocity via the inherent mechanical properties of muscle. γd, dynamic γ-motor neurons; γs, static γ-motor neurons. (Reproduced with
permission from Houk J: Medical Physiology, 13th ed. In: Mountcastle VB (editor). Mosby, 1974.)

resistance followed by give when a limb is moved passively is body part stimulated is flexed and withdrawn from the stimu-
known as the clasp-knife effect because of its resemblance to lus. When a strong stimulus is applied to a limb, the response
the closing of a pocket knife. includes not only flexion and withdrawal of that limb, but also
Clonus is the occurrence of regular, repetitive, rhythmic extension of the opposite limb. This crossed extensor response
contractions of a muscle subjected to sudden, maintained is part of the withdrawal reflex.
stretch. Sustained clonus with five or more beats is considered Flexor responses can be produced by innocuous stimula-
abnormal. During a neurological exam, ankle clonus can be tion of the skin or by stretch of the muscle, but strong flexor
initiated by brisk, maintained dorsiflexion of the foot, and the responses with withdrawal are initiated only by stimuli that
response is rhythmic plantar flexion at the ankle. are noxious or at least potentially harmful (nociceptive
Clonus may also occur after disruption of descending cor- stimuli). Flexion of the stimulated limb gets it away from the
tical input to a spinal glycinergic inhibitory interneuron called source of irritation, and extension of the other limb supports
the Renshaw cell. This cell receives excitatory input from the body.
α-motor neurons via axon collaterals (and in turn the As the strength of a noxious stimulus is increased, the reac-
Renshaw cell inhibits the motor neuron). In addition, cortical tion time is shortened. Spatial and temporal facilitation
fibers activating ankle flexors contact Renshaw cells (as well occurs at synapses in the polysynaptic pathway. Stronger stim-
as inhibitory interneurons activated by Ia afferent fibers) that uli produce more action potentials per second in the active
inhibit the antagonistic ankle extensors. This circuitry pre- branches and cause more branches to become active; summa-
vents reflex stimulation of the extensors when flexors are tion of the EPSPs to the firing level therefore occurs more
active. Therefore, when the descending cortical fibers rapidly.
are damaged (upper motor neuron lesion), the inhibition of Another characteristic of the withdrawal response is the
antagonists is absent. The result is repetitive, sequential fact that supramaximal stimulation of any of the sensory
contraction of ankle flexors and extensors (clonus). Clonus nerves from a limb never produces as strong a contraction of
may be seen in patients with amyotrophic lateral sclerosis, the flexor muscles as that elicited by direct electrical stimula-
stroke, multiple sclerosis, spinal cord damage, and hepatic tion of the muscles themselves. This indicates that the affer-
encephalopathy. ent inputs fractionate the α-motor neuron pool, that is, each
input goes to only part of the motor neuron pool for the flex-
ors of that particular extremity. On the other hand, if all the
POLYSYNAPTIC REFLEXES: sensory inputs are dissected out and stimulated one after the
THE WITHDRAWAL REFLEX other, the sum of the tension developed by stimulation of each
is greater than that produced by direct electrical stimulation
The withdrawal reflex is a typical polysynaptic reflex that of the muscle or stimulation of all inputs at once. This indi-
occurs in response to a painful stimulation of the skin or sub- cates that the various afferent inputs share some of the motor
cutaneous tissues and muscle. The response is flexor muscle neurons and that occlusion occurs when all inputs are stimu-
contraction and inhibition of extensor muscles, so that the lated at once.
CHAPTER 14 Spinal Reflexes 131

SPINAL INTEGRATION ties and the right leg, but he was unable to detect the nox-
The spinal α-motor neurons that supply the extrafusal fibers ious stimuli applied to the left leg. He also lost the
in skeletal muscles are the efferent side of many reflex arcs. All sensation of touch and vibration on his right leg but sen-
neural influences affecting muscular contraction ultimately sation was normal on his left leg and upper limbs. There
funnel through them to the muscles, and they are therefore was little, if any, spontaneous movement in the right leg,
called the final common pathway. The surface of the average although all other limbs appeared to have normal move-
α-motor neuron and its dendrites accommodates about 10,000 ment. A magnetic resonance image showed that the right
synaptic knobs, allowing for numerous synaptic inputs. At side of his spinal cord was severely damaged at the 10th
least five inputs go from the same spinal segment to a typical thoracic level.
spinal motor neuron. In addition to these, there are excitatory It has been estimated that the worldwide annual inci-
and inhibitory inputs, generally relayed via interneurons, from dence of sustaining SCI is between 10 and 83 per million
other levels of the spinal cord and multiple long-descending of the population. Leading causes are vehicle accidents,
tracts from the brain. All of these pathways converge on and violence, and sports injuries. Approximately 52% of SCI
determine the activity in the final common pathway. cases result in quadriplegia and about 42% lead to para-
plegia. The mean age of patients who sustain an SCI is
33 years old, and males outnumber females nearly 4 to 1.
SPINAL CORD INJURY This patient’s injury led to a hemisection of the spinal
cord at the 10th thoracic level. Such an injury causes a
A key component of neurological exams includes an assess- characteristic clinical picture that reflects damage to
ment of the integrity of spinal reflexes. Abnormalities in the ascending sensory (dorsal column pathway, ventrolateral
reflexes often point to the location of a spinal cord injury spinothalamic tract) and descending motor (corticospi-
(SCI). The deficits after SCI vary, of course, depending on the nal tract) pathways, which is called the Brown-Séquard
level and severity of the injury. Transection of the spinal cord syndrome. The lesion to fasciculus gracilus or fasciculus
is followed by a period of spinal shock during which all spinal cuneatus leads to ipsilateral loss of discriminative touch,
reflex responses are profoundly depressed. Subsequently, vibration, and proprioception below the level of lesion.
reflex responses return and become hyperactive. In humans, The loss of the spinothalamic tract leads to contralateral
spinal shock usually lasts for a minimum of 2 weeks. loss of pain and temperature sensation beginning one or
Cessation of tonic bombardment of spinal neurons by two segments below the lesion. Damage to the corticospi-
excitatory impulses in descending pathways undoubtedly nal tract produces weakness and spasticity in certain
plays a role in spinal shock. The recovery of reflex excitability muscle groups on the same side of the body. Although a
may be due to the development of denervation hypersensitiv- precise spinal hemisection is rare, the syndrome is fairly
ity to the mediators released by the remaining spinal excit- common because it can be caused by spinal cord tumor,
atory endings (see Chapter 12). Another possibility is the trauma, degenerative disc disease, and ischemia.
sprouting of collaterals from existing neurons, with the for-
mation of additional excitatory endings on interneurons and
motor neurons.
The first reflex response to appear as spinal shock wears off CHAPTER SUMMARY
is often a slight contraction of the leg flexors and adductors in ■ A reflex arc consists of a sense organ, an afferent neuron,
response to a noxious stimulus. In some patients, the knee- one or more synapses within a central integrating station,
jerk reflex recovers first. Once the spinal reflexes begin to an efferent neuron, and an effector response.
reappear after spinal shock, their threshold steadily drops. ■ A muscle spindle is a group of specialized intrafusal muscle
fibers with contractile polar ends and a noncontractile center
that is located in parallel to the extrafusal muscle fibers and is
innervated by types Ia and II afferent fibers and γ-motor
CLINICAL CORRELATION neurons. Muscle stretch activates the muscle spindle to initiate
reflex contraction of the extrafusal muscle fibers in the same
A 21-year-old male medical student was stabbed in the muscle (stretch reflex).
back during a robbery attempt. A bystander called 911 ■ A Golgi tendon organ is a netlike collection of knobby nerve
and, when the paramedics arrived, the student could not endings among the fascicles of a tendon that is located in
move his right leg. He was rushed to the emergency room series with extrafusal muscle fibers and innervated by type
of the local hospital. In addition to examination and treat- Ib afferent fibers. They are stimulated by both passive stretch
ment of the injury site, he was given a neurological exam. and active contraction of the muscle to relax the muscle
The muscle spindle reflex was normal in both arms and (inverse stretch reflex) and function as a transducer to
the left leg but was hyperactive in the right leg. He had regulate muscle force.
normal sensation to pinprick and pinch in upper extremi- ■ A collateral from an Ia afferent branches to terminate on an
inhibitory interneuron that synapses on an antagonistic muscle
132 SECTION IV CNS/Neural Physiology

(reciprocal innervation) to relax that muscle when the agonist 3. Which of the following is not characteristic of a reflex?
contracts. Clonus is the occurrence of regular, rhythmic A) modification by impulses from various parts of the CNS
contractions of a muscle subjected to sudden, maintained stretch. B) may involve simultaneous contraction of some muscles
■ Spinal cord transection is followed by a period of spinal and relaxation of others
shock during which all reflexes are profoundly depressed. C) chronically suppressed after spinal cord transection
This is followed by a period of hyperactive reflexes. D) always involves transmission across at least one synapse
E) frequently occurs without conscious perception
4. Withdrawal reflexes are not
STUDY QUESTIONS A) initiated by nociceptive stimuli.
B) an example of a polysynaptic reflex.
1. The inverse stretch reflex C) prolonged if the stimulus is strong.
A) has a lower threshold than that of the stretch reflex. D) an example of a flexor reflex.
B) is a monosynaptic reflex. E) accompanied by the same response on both sides of
C) is a relaxation of a muscle in response to a strong stretch the body.
of the muscle.
D) has the muscle spindle as its receptor.
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 afferent fibers
takes place.
B) contraction of the muscle is prolonged.
C) the muscle will not contract.
D) the muscle will not relax.
E) the number of impulses in spindle Ia afferent fibers is
greater than when α discharge alone is increased.
15
C H A P T E R

Special Senses I: Vision


Susan M. Barman

O B J E C T I V E S

■ Describe the various parts of the eye and list the functions of each.
■ Explain how light rays in the environment are brought to a focus on the retina
and the role of accommodation in this process.
■ Define the following terms: hyperopia, myopia, astigmatism, presbyopia,
and strabismus.
■ Describe the electrical responses produced by rods and cones and explain
how these responses are produced.
■ Trace the neural pathways that transmit visual information from the rods and
cones to the visual cortex.
■ Name the four types of eye movements and the function of each.

INTRODUCTION junction. In front of the lens is the pigmented and opaque


iris, the colored portion of the eye, which contains circular
The eyes are complex sense organs. Within its protective cas- muscle fibers that constrict and radial fibers that dilate the
ing, each eye has a layer of receptors, a lens system that focuses pupil. Variations in the diameter of the pupil can produce
light on these receptors, and a system of nerves that conducts up to a 5-fold change in the amount of light reaching the
impulses from the receptors to the brain. The way these com- retina.
ponents operate to set up conscious visual images is the sub- The space between the lens and the retina is filled primar-
ject of this chapter. ily with a clear gelatinous material called the vitreous
humor. Aqueous humor, a clear liquid that nourishes the
cornea and lens, is produced in the ciliary body by diffusion
ANATOMY OF THE EYE and active transport from plasma. It flows through the pupil
and fills the anterior chamber of the eye. It is normally reab-
The principal structures of the eye are shown in Figure 15–1. sorbed through the canal of Schlemm, a venous channel
The outer protective layer of the eyeball, the sclera, is modi- at the junction between the iris and the cornea (anterior
fied anteriorly to form the transparent cornea, through which chamber angle). Obstruction of this outlet leads to increased
light rays enter the eye. Inside the sclera is the choroid, a layer intraocular pressure. One cause of increased pressure
that contains many of the blood vessels that nourish the struc- is decreased permeability through the trabecular mesh-
tures in the eyeball. Lining the posterior two thirds of the work, the tissue around the base of the cornea that drains
choroid is the retina, the neural tissue containing the recep- the aqueous humor from the eye (open-angle glaucoma),
tor cells. and another is forward movement of the iris, obliterating
The crystalline lens is a transparent structure held in the angle (angle-closure glaucoma). Glaucoma can be
place by a circular lens suspensary ligament (zonule) that treated with β-adrenergic blocking drugs or carbonic anhy-
is attached to the thickened anterior part of the choroid, drase inhibitors, both of which decrease the production of
the ciliary body. The ciliary body contains circular and lon- aqueous humor, or with cholinergic agonists, which increase
gitudinal muscle fibers that attach near the corneoscleral aqueous outflow.

133

Ch15_133-146.indd 133 11/26/10 9:42:37 AM


134 SECTION IV CNS/Neural Physiology

Superior rectus muscle

Sclera Conjunctiva

Choroid Ciliary body


Posterior
Retina chamber Anterior
Anterior cavity
Fovea centralis chamber

Central artery Cornea


Central vein Pupil
Lens
Iris
Posterior chamber
Optic nerve Zonular fibers of
suspensory ligament

Vitreous chamber
(posterior cavity)
Inferior rectus muscle

FIGURE 15–1 The internal anatomy of the eye. (Reproduced with permission from Fox SI: Human Physiology. McGraw-Hill, 2008.)

The eye is well protected from injury by the bony walls of terior pole of the globe. This region is visible through the oph-
the orbit. The cornea is moistened and kept clear by tears thalmoscope as the optic disk. There are no visual receptors
that course from the lacrimal gland in the upper portion of over the disk, and consequently it is a blind spot.
each orbit across the surface of the eye to empty via the Near the posterior pole of the eye is a yellowish pigmented
lacrimal duct into the nose. Blinking helps keep the cornea spot, the macula lutea. This marks the location of the fovea
moist. centralis, a thinned-out, rod-free portion of the retina. In it,
the cones are densely packed, and each synapses to a single
bipolar cell, which, in turn, synapses on a single ganglion
cell, providing a direct pathway to the brain. There are very
RETINA few overlying cells and no blood vessels; thus, the fovea is the
The retina extends anteriorly almost to the ciliary body. It is point where visual acuity is greatest. When attention is
organized into 10 layers and contains rods and cones, which attracted to or fixed on an object, the eyes are normally
are the visual receptors, plus four types of neurons: bipolar moved so that light rays coming from the object fall on
cells, ganglion cells, horizontal cells, and amacrine cells the fovea.
(Figure 15–2). Rods and cones, which are next to the chor-
oid, synapse with bipolar cells, and bipolar cells synapse with VISUAL RECEPTORS IN THE RETINA
ganglion cells. The axons of ganglion cells converge and leave
the eye as the optic nerve. Horizontal cells connect receptor Rods are responsible for vision in low light (night vision)
cells to the other receptor cells in the outer plexiform layer. and provide only black and white vision. Cones are respon-
Amacrine cells connect ganglion cells to one another in the sible for color vision. Each rod and cone is divided into an
inner plexiform layer via processes of varying length and outer segment, an inner segment that includes a nuclear
patterns. Gap junctions also connect retinal neurons to one region, and a synaptic zone (Figure 15–3). The outer seg-
another. ments are modified cilia and are made up of regular stacks
The receptor layer of the retina rests on the pigment epithe- of flattened saccules or disks composed of membrane. These
lium next to the choroid, so light rays must pass through the saccules and disks contain the photosensitive compounds
ganglion cell and bipolar cell layers to reach the rods and that react to light, initiating action potentials in the visual
cones. The pigment epithelium absorbs light rays, preventing pathways. The inner segments are rich in mitochondria. The
the reflection of rays back through the retina. Such reflection rods are named for the thin, rodlike appearance of their
would produce blurring of the visual images. outer segments. Cones generally have thick inner segments
The optic nerve leaves the eye and the retinal blood vessels and conical outer segments, although their morphology var-
enter it at a point 3 mm medial to and slightly above the pos- ies from place to place in the retina. In cones, the saccules
CHAPTER 15 Special Senses I: Vision 135

Pigment epithelium

Rod and cone

Outer segments

Inner segments

Outer nuclear layer C C C C


R R R R
R

Outer plexiform layer

H
FB
Inner nuclear layer MB
RB FB
MB RB
A
A

Inner plexiform layer

Ganglion cell layer MG DG MG


DG

Optic nerve fibers

FIGURE 15–2 Neural components of the extrafoveal portion of the retina. Direction of light is from the bottom to the top of the figure.
C, cone; R, rod; MB, RB, and FB, midget, rod, and flat bipolar cells; DG and MG, diffuse and midget ganglion cells; H, horizontal cells; A, amacrine
cells. (Modified with permission from Dowling JE, Boycott BB: Organization of the primate retina: electron microscopy. Proc R Soc Lond B 1966;166:80–111.)

are formed in the outer segments by infoldings of the cell in the visual cortex, the number of neurons concerned with
membrane, but in rods the disks are separated from the cell vision is 1,000 times the number of fibers in the optic nerves.
membrane.
In the extrafoveal portions of the retina, rods predominate
(Figure 15–4), and there is a good deal of convergence. Flat THE IMAGE-FORMING
bipolar cells (Figure 15–2) make synaptic contact with several MECHANISM
cones, and rod bipolar cells make synaptic contact with several
rods. Because there are approximately 6 million cones and The eyes convert energy in the visible spectrum into action
120 million rods in each human eye but only 1.2 million nerve potentials in the optic nerve. The images of objects in the envi-
fibers in each optic nerve, the overall convergence of receptors ronment are focused on the retina. The light rays striking the
through bipolar cells on ganglion cells is about 105:1. However, retina generate potentials in the rods and cones. Impulses ini-
there is divergence from this point on. There are twice as many tiated in the retina are conducted to the cerebral cortex, where
fibers in the geniculocalcarine tracts as in the optic nerves, and they produce the sensation of vision.
136 SECTION IV CNS/Neural Physiology

Rod Cone at the anterior surface of the cornea (Figure 15–5). The retinal
Plasma membrane image is inverted. The connections of the retinal receptors are
such that from birth any inverted image on the retina is viewed
right side up and projected to the visual field on the side
30 nm Outer
segment opposite to the retinal area stimulated. This perception is pres-
Disks Sacs ent in infants and is innate.

Ciliary neck

Mitochondria
Inner
COMMON DEFECTS OF THE
segment IMAGE-FORMING MECHANISM
Nucleus
In some individuals, the eyeball is shorter than normal and the
parallel rays of light are brought to a focus behind the retina.
Synaptic
terminal This abnormality is called hyperopia or farsightedness (Figure
15–6). Sustained accommodation (focusing due to contrac-
FIGURE 15–3 Schematic diagram of a rod and a cone. tion of the ciliary muscle), even when viewing distant objects,
Each rod and cone is divided into an outer segment, an inner
can partially compensate for the defect, but the prolonged
segment with a nuclear region, and a synaptic zone. The saccules
and disks in the outer segment contain photosensitive compounds muscular effort is tiring and may cause headaches and blur-
that react to light to initiate action potentials in the visual ring of vision. The defect can be corrected by using glasses
pathways. (Reproduced with permission from Lamb TD: Electrical responses with convex lenses, which aid the refractive power of the eye in
of photoreceptors. In: Recent Advances in Physiology, No.10. Baker PF [editor]. shortening the focal distance.
Churchill Livingstone, 1984.) In myopia (nearsightedness), the anteroposterior diameter
of the eyeball is too long (Figure 15–6). The shape of the eye
appears to be determined in part by the refraction presented to
Light rays are bent when they pass from a medium of one den- it. In young adult humans, the extensive close work involved
sity into a medium of a different density, except when they strike in activities such as studying accelerates the development of
perpendicular to the interface (Figure 15–5). The bending of myopia. This defect can be corrected by glasses with biconcave
light rays is called refraction and is the mechanism that allows lenses, which make parallel light rays diverge slightly before
one to focus an accurate image onto the retina. Parallel light rays they strike the eye.
striking a biconvex lens are refracted to a point behind the lens. Astigmatism is a common condition in which the curvature
In the eye, light is actually refracted at the anterior surface of of the cornea is not uniform (Figure 15–6). When the curva-
the cornea and at the anterior and posterior surfaces of the ture in one meridian is different from that in others, light rays
lens. The process of refraction can be represented diagram- in that meridian are refracted to a different focus, so that part
matically by drawing the rays of light as if all refraction occurs of the retinal image is blurred. Astigmatism can usually be

2000
Cones
Number of rods or cones in an area

Rods
1600
of 0.0069 mm2

1200
Blind spot

800

400

0
100° 80° 60° 40° 20° 0° 20° 40° 60° 80°
Nasal retina Fovea Temporal retina
Distance from the fovea

FIGURE 15–4 Rod and cone density along the horizontal meridian through the human retina. A plot of the relative acuity of vision in
the various parts of the light-adapted eye would parallel the cone density curve; a similar plot of relative acuity of the dark-adapted eye would
parallel the rod density curve. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill
Medical, 2009.)
CHAPTER 15 Special Senses I: Vision 137

(a)

Glass Air
Refraction
Point source
of light
No refraction

Refraction

(b)

b' a

a' b

FIGURE 15–5 Focusing point sources of light. a) When diverging light rays enter a dense medium at an angle to its convex surface,
refraction bends them inward. b) Refraction of light by the lens system. For simplicity, refraction is shown only at the corneal surface (site
of greatest refraction) although it also occurs in the lens and elsewhere. Incoming light from a (above) and b (below) is bent in opposite
directions, resulting in b′ being above a′ on the retina. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed.
McGraw-Hill, 2008.)

corrected with cylindrical lenses placed in such a way that they The problem of bringing diverging rays from close objects to a
equalize the refraction in all meridians. focus on the retina can be solved by increasing the curvature of
Strabismus is a misalignment of the eyes usually due to the lens, a process called accommodation. At rest, the lens is
problems with eye muscles and one of the most common eye held under tension by the lens ligaments and is pulled into a
problems in children, affecting about 4% of children under 6 flattened shape. The ciliary muscle contracts when the gaze is
years of age. It is characterized by one or both eyes turning directed at a near object. This decreases the distance between
inward (crossed-eyes), outward (wall eyes), upward, or down- the edges of the ciliary body and relaxes the lens ligaments, so
ward. Strabismus is also commonly called “wandering eye” or that the lens springs into a more convex shape (Figure 15–7).
“crossed-eyes.” It occurs when visual images do not fall on cor- The degree to which the lens curvature can be increased is
responding retinal points. When visual images chronically fall limited, and light rays from an object very near the individual
on noncorresponding points in the two retinas in young chil- cannot be brought to a focus on the retina, even with the
dren, one is eventually suppressed (suppression scotoma). greatest of effort. The nearest point to the eye at which an
object can be brought into clear focus by accommodation is
called the near point of vision. Due to increasing hardness of
ACCOMMODATION the lens, the near point recedes throughout life, slowly at first
and then rapidly with advancing age, from 9 cm at age 10 to
When the ciliary muscle is relaxed, parallel light rays striking 83 cm at age 60. By the time a healthy individual reaches age
the optically normal (emmetropic) eye are brought to a focus 40–45, the loss of accommodation is usually sufficient to
on the retina. As long as this relaxation is maintained, rays from make reading and close work difficult. This condition, which
objects closer than 6 m from the observer are brought to a focus is known as presbyopia, can be corrected by wearing glasses
behind the retina, and consequently the objects appear blurred. with convex lenses.
138 SECTION IV CNS/Neural Physiology

(a)

Normal sight (faraway object is clear)

Nearsighted (eyeball too long)

FIGURE 15–7 Accommodation. Solid lines represent the shape


Nearsightedness corrected of the lens, iris, and ciliary body at rest; dashed lines represent the
shape during accommodation. Ciliary muscles contract when gaze is
directed at a near object, which decreases the distance between the
edges of the ciliary body and relaxes the lens ligaments, and the lens
becomes more convex. (Reproduced with permission from Barrett KE, Barman
(b) SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill
Medical, 2009.)
Normal sight (near object is clear)

segment maintains ionic equilibrium. Release of synaptic


transmitter is steady in the dark. When light strikes the outer
Farsighted (eyeball too short) segment, the reactions that are initiated close some of the Na+
channels, and the result is a hyperpolarizing receptor poten-
tial. The hyperpolarization reduces the release of synaptic
transmitter, and this generates a signal in the bipolar cells that
ultimately leads to action potentials in ganglion cells. The
action potentials are transmitted to the brain.
Farsightedness corrected

FIGURE 15–6 Common defects of the optic system of the eye.


a and b) In hyperopia (farsightedness), the eyeball is too short and Na+
light rays come to a focus behind the retina. A biconvex lens corrects
this by adding to the refractive power of the lens of the eye. In myopia
(nearsightedness), the eyeball is too long and light rays focus in front
of the retina. Placing a biconcave lens in front of the eye causes the
light rays to diverge slightly before striking the eye, so that they are
brought to a focus on the retina. (Reproduced with permission from Widmaier
EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
K+ K+

THE PHOTORECEPTOR
MECHANISM Na+ Na+
Dark Light
IONIC BASIS OF PHOTORECEPTOR
FIGURE 15–8 Effect of light on current flow in visual
POTENTIALS receptors. In the dark, Na+ channels in the outer segment are
held open by cGMP. Light leads to increased conversion of cGMP
Na+ channels in the outer segments of the rods and cones are to 5′-GMP, and some of the channels close. This produces
open in the dark, so current flows from the inner to the outer hyperpolarization of the synaptic terminal of the photoreceptor.
segment (Figure 15–8). Current also flows to the synaptic end- (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H:
ing of the photoreceptor. The Na+, K+-ATPase in the inner Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
CHAPTER 15 Special Senses I: Vision 139

Outer segment membrane


Incident light

cGMP
Structural change in the phospho-
retinene1 of photopigment Rhodopsin Transducin diesterase

Conformational change Disk


of photopigment

Activation of transducin GTP cGMP


5'-GMP
Na+
Light
Activation of
phosphodiesterase
cGMP-gated
channel ECF
Decreased
Rod outer segment
intracellular cGMP
FIGURE 15–10 Initial steps in phototransduction in rods.
Light activates rhodopsin, which activates transducin to bind GTP.
Closure of Na+ channels
This activates phosphodiesterase, which catalyzes the conversion of
cGMP to 5′-GMP. The resulting decrease in the cytoplasmic cGMP
Hyperpolarization concentration causes cGMP-gated ion channels to close. (Reproduced
with permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of
Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
Decreased release of
synaptic transmitter

Cone receptors subserve color vision and respond maxi-


Response in bipolar cells
and other neural elements mally to light at wavelengths of 440, 535, and 565 nm. The
cone opsin resembles rhodopsin. The cell membrane of cones
FIGURE 15–9 Sequence of events involved in is invaginated to form the saccules, but the cones have no sep-
phototransduction in rods and cones. (Reproduced with permission arate intracellular disks like those in rods. The details of the
from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical
responses of cones to light are similar to those in rods.
Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

PROCESSING OF VISUAL
PHOTOSENSITIVE COMPOUNDS INFORMATION IN THE RETINA
The photosensitive compounds in the rods and cones of the A characteristic of the bipolar and ganglion cells is that they
eyes are made up of a protein called an opsin and retinal, respond best to a small, circular stimulus and that, within their
the aldehyde of vitamin A. The photosensitive pigment in the receptive field, an annulus of light around the center (surround
rods is called rhodopsin, one of the many receptors coupled to illumination) inhibits the response to the central spot (Figure
G proteins. Its opsin is called scotopsin. Rhodopsin has a peak 15–11). The center can be excitatory with an inhibitory sur-
sensitivity to light at a wavelength of 505 nm. round (an “on-center” cell) or inhibitory with an excitatory
Figure 15–9 summarizes the sequence of events in photore- surround (an “off-center” cell). The inhibition of the center
ceptors by which incident light produces a signal in the next response by the surround is probably due to inhibitory feed-
succeeding neural unit in the retina. Light activates rhodopsin back from one photoreceptor to another mediated via horizontal
that then activates the associated heterotrimeric G protein, cells. The inhibition of the response to central illumination by
transducin (Figure 15–10). The G protein exchanges GDP for an increase in surrounding illumination is an example of lat-
GTP, and the α-subunit separates. This subunit remains active eral inhibition in which activation of a particular neural unit is
until its intrinsic GTPase activity hydrolyzes the GTP. The associated with inhibition of the activity of nearby units. It is a
α-subunit activates cGMP phosphodiesterase, which converts general phenomenon in sensory systems and helps to sharpen
cGMP to 5′-GMP. cGMP normally acts directly on Na+ chan- the edges of a stimulus and improve discrimination.
nels to maintain them in the open position, so the decline in
the cytoplasmic cGMP concentration causes some Na+ chan-
nels to close. This produces the hyperpolarizing potential. This
cascade of reactions occurs very rapidly and amplifies the light
VISUAL PATHWAYS
signal. The amplification helps explain the remarkable sensi- The axons of the retinal ganglion cells pass caudally in the
tivity of rod photoreceptors; these receptors are capable of pro- optic nerve and optic tract to end in the lateral geniculate
ducing a detectable response to as little as one photon of light. body in the thalamus (Figure 15–12). The fibers from each
140 SECTION IV CNS/Neural Physiology

On-center field Off-center field


Light Light

Central
illumination

Surround
illumination

FIGURE 15–11 Responses of retinal ganglion cells to light on the portions of their receptive fields indicated in white. Beside each
receptive field diagram is a diagram of the ganglion cell response, indicated by extracellularly recorded action potentials. Note that in three of
the four situations, there is increased discharge when the light is turned off. (Adapted with permission from Kuffler SW: Discharge patterns and functional
organizations of mammalian retina, J Neurophysiol. 1953;16(1):37–68.)

Temporal Nasal
field field LEFT RIGHT

A
LEFT RIGHT
EYE EYE

B
Ganglion
cell B Optic
nerve
A C
Optic chiasm

Pretectal
C region D
Optic
tract

Lateral
geniculate
body
Geniculocalcarine
tract

Occipital cortex
FIGURE 15–12 Visual pathways. Transection of the pathways at the locations indicated by the letters causes the visual field defects
shown in the diagrams on the right. The fibers from the nasal half of each retina decussate in the optic chiasm, so fibers in the optic tracts are
those from the temporal half of one retina and the nasal half of the other. A lesion that interrupts one optic nerve causes blindness in that eye
(A). A lesion in one optic tract causes blindness in half of the visual field (C) and is called homonymous (same side of both visual fields)
hemianopia (half-blindness). Lesions affecting the optic chiasm destroy fibers from both nasal hemiretinas and produce a heteronymous
(opposite sides of the visual fields) hemianopia (B). Occipital lesions may spare the fibers from the macula (as in D) because of the separation
in the brain of these fibers from the others subserving vision (see Figure 15–13). (Reproduced with permission from Barrett KE, Barman SM, Boitano S,
Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
CHAPTER 15 Special Senses I: Vision 141

Upper peripheral
quadrant of retina

Upper Optic
quadrant nerves
of macula Optic
chiasm
Lower
quadrant
of macula
Lower peripheral Lateral geniculate
quadrant of retina Optic nucleus
tracts C Dorsal
FIGURE 15–13 Medial view of the human right cerebral I
hemisphere showing projection of the retina on the primary
C
visual cortex (Brodmann’s area 17; also known as V1) in the
occipital cortex around the calcarine fissure. The geniculocalcarine I
fibers from the medial half of the lateral geniculate terminate on the I
superior lip of the calcarine fissure, and those from the lateral half C
6
terminate on the inferior lip. The fibers from the lateral geniculate 5
Ventral 4
body that relay macular vision separate from those that relay 1 2
3
peripheral vision and end more posteriorly on the lips of the calcarine
fissure. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Magnocellular Parvocellular
pathway pathway
Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

nasal hemiretina decussate in the optic chiasm. In the genic- Primary visual cortex
ulate body, the fibers from the nasal half of one retina and (area 17)
the temporal half of the other synapse on the cells whose FIGURE 15–14 Ganglion cell projections from the right
axons form the geniculocalcarine tract. This tract passes to hemiretina of each eye to the right lateral geniculate body and from
the occipital lobe of the cerebral cortex. The effects of lesions this nucleus to the right primary visual cortex. Note the six layers of
in these pathways on visual function are discussed below. the geniculate. P ganglion cells project to layers 3–6, and M ganglion
The primary visual receiving area (primary visual cortex, cells project to layers 1 and 2. The ipsilateral (I) and contralateral (C) eyes
Brodmann’s area 17; also known as V1) is located princi- project to alternate layers. Not shown are the interlaminar area cells,
which project via a separate component of the P pathway to blobs in
pally on the sides of the calcarine fissure (Figure 15–13).
the visual cortex. (Modified with permission from Kandel ER, Schwartz JH, Jessell TM
The organization of the primary visual cortex is discussed [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
below.
The axons of retinal ganglion cells project a detailed spatial
representation of the retina on the lateral geniculate body.
carries signals for detection of movement, depth, and flicker.
Each geniculate body contains six well-defined layers (Figure
The parvocellular pathway, from layers 3–6, carries signals for
15–14). Layers 3–6 have small parvocellular cells, and layers
color vision, texture, shape, and fine detail.
1 and 2 have large magnocellular cells. On each side, layers 1,
Cells in the interlaminar region of the lateral geniculate
4, and 6 receive input from the contralateral eye; layers 2, 3,
nucleus also receive input from P ganglion cells, probably via
and 5 receive input from the ipsilateral eye. In each layer, there
dendrites of interlaminar cells that penetrate the parvocellular
is a precise point-for-point representation of the retina, and all
layers. They project via a separate component of the P pathway
six layers are in register so that along a line perpendicular to
to the “blobs” in the visual cortex. These are clusters of cells
the layers, the receptive fields of the cells in each layer are
about 0.2 mm in diameter that, unlike the neighboring cells,
almost identical. Only 10–20% of the input to the lateral genic-
contain a high concentration of the mitochondrial enzyme
ulate nucleus comes from the retina; major inputs also come
cytochrome oxidase.
from the visual cortex and other brain regions. The feedback
pathway from the visual cortex is involved in visual processing
related to the perception of orientation and motion. PRIMARY VISUAL CORTEX
There are two kinds of retinal ganglion cells: large magno or
M cells, which are concerned with movement and stereopsis, The lateral geniculate body projects a point-for-point repre-
and small parvo or P cells, which are concerned with color, sentation on the primary visual cortex (Figure 15–13). Like
texture, and shape. The M and P ganglion cells project to the the rest of the neocortex, the visual cortex has six layers. The
magnocellular and parvocellular portions of the lateral genic- axons from the lateral geniculate nucleus that form the mag-
ulate, respectively. From the lateral geniculate nucleus, a mag- nocellular pathway end in layer 4. Many of the axons that form
nocellular pathway and a parvocellular pathway project to the the parvocellular pathway also end in layer 4; however, the
visual cortex. The magnocellular pathway, from layers 1 and 2, axons from the interlaminar region end in layers 2 and 3.
142 SECTION IV CNS/Neural Physiology

Layers 2 and 3 of the cortex contain blobs. They are arranged so that the fibers in the optic tracts are those from the tempo-
in a mosaic in the visual cortex and are concerned with color ral half of one retina and the nasal half of the other. Since
vision. However, the parvocellular pathway also carries color- each optic tract subserves half of the field of vision, a lesion of
opponent data to the deep part of layer 4. one optic nerve causes blindness in that eye, but a lesion in one
Like the ganglion cells, the lateral geniculate neurons and optic tract causes blindness in half of the visual field. This
the neurons in layer 4 of the visual cortex respond to stimuli in defect is classified as a homonymous (same side of both visual
their receptive fields with on-centers and inhibitory surrounds fields) hemianopia (half-blindness). Lesions affecting the
or off-centers and excitatory surrounds. A bar of light cover- optic chiasm (e.g., pituitary tumors) cause disruption of the
ing the center is an effective stimulus for them because it stim- fibers from both nasal hemiretinas and produce a heterony-
ulates the entire center and relatively little of the surround. mous (opposite sides of the visual fields) hemianopia. Because
However, the bar has no preferred orientation and, as a stimu- the fibers from the maculas are located posteriorly in the optic
lus, is equally effective at any angle. chiasm, hemianopic scotomas develop before vision in the two
The responses of the neurons in other layers of the visual cor- hemiretinas is completely lost. Selective visual field defects are
tex are strikingly different. Simple cells respond to bars of light, further classified as bitemporal, binasal, and right or left.
lines, or edges, but only when they have a particular orientation. The optic nerve fibers from the upper retinal quadrants
When a bar of light is rotated as little as 10° from the preferred subserving vision in the lower half of the visual field terminate
orientation, the firing rate of the simple cell is usually decreased, in the medial half of the lateral geniculate body, whereas the
and if the stimulus is rotated much more, the response disap- fibers from the lower retinal quadrants terminate in the lateral
pears. Complex cells, which resemble simple cells in requiring half. The geniculocalcarine fibers from the medial half of the
a preferred orientation of a linear stimulus, are less dependent lateral geniculate terminate on the superior lip of the calcarine
on the location of a stimulus in the visual field than the simple fissure, and those from the lateral half terminate on the infe-
cells and the cells in layer 4. They often respond maximally rior lip. The fibers from the lateral geniculate body that sub-
when a linear stimulus is moved laterally without a change in its serve macular vision separate from those that subserve
orientation. They may receive input from the simple cells. peripheral vision and end more posteriorly on the lips of the
The visual cortex is arranged in vertical columns that are calcarine fissure (Figure 15–13). Because of this anatomic
concerned with orientation (orientation columns). Each is arrangement, occipital lobe lesions may produce discrete
about 1 mm in diameter. However, the orientation preferences quadrantic visual field defects (upper and lower quadrants of
of neighboring columns differ in a systematic way; as one each half visual field). Macular sparing (i.e., loss of peripheral
moves from column to column across the cortex, sequential vision with intact macular vision) is also common with occip-
changes occur in orientation preference of 5°–10°. Thus, it ital lesions (Figure 15–12) because the macular representation
seems likely that for each ganglion cell receptive field in the is separate from that of the peripheral fields and very large
visual field, there is a collection of columns in a small area of relative to that of the peripheral fields. Therefore, occipital
visual cortex representing the possible preferred orientations lesions must extend considerable distances to destroy macular
at small intervals throughout the full 360°. The simple and as well as peripheral vision. Bilateral destruction of the occipi-
complex cells have been called feature detectors because they tal cortex in humans causes subjective blindness.
respond to and analyze certain features of the stimulus.
Another feature of the visual cortex is the presence of ocular
dominance columns. The geniculate cells and the cells in layer
4 receive input from only one eye, and the layer 4 cells alter-
COLOR VISION
nate with cells receiving input from the other eye. Colors have three attributes: hue, intensity, and saturation
About half the simple and complex cells receive an input (degree of freedom from dilution with white). For any color there
from both eyes. The inputs are identical or nearly so in terms is a complementary color that, when properly mixed with it,
of the portion of the visual field involved and the preferred produces a sensation of white. Black is the sensation produced by
orientation. However, they differ in strength, so that between the absence of light, but it is probably a positive sensation because
the cells to which the input comes totally from the ipsilateral the blind eye does not “see black;” rather, it “sees nothing.”
or the contralateral eye, there is a spectrum of cells influenced The sensation of white, any spectral color, and even the
to different degrees by both eyes. extraspectral color, purple, can be produced by mixing various
proportions of red light (wavelength 723–647 nm), green light
(575–492 nm), and blue light (492–450 nm). Red, green, and
EFFECT OF LESIONS IN blue are therefore called the primary colors. Also, the color
THE OPTIC PATHWAYS perceived depends in part on the color of other objects in the
visual field. Thus, for example, a red object is seen as red if the
Lesions along the neural pathways from the eyes to the brain field is illuminated with green or blue light, but as pale pink or
can be localized with a high degree of accuracy by the effects white if the field is illuminated with red light.
they produce in the visual fields (Figure 15–12). The fibers Color is mediated by ganglion cells that subtract or add input
from the nasal half of each retina decussate in the optic chiasm, from one type of cone to input from another type. Processing in
CHAPTER 15 Special Senses I: Vision 143

the ganglion cells and the lateral geniculate nucleus produces second-order neurons project to the ipsilateral and contralat-
impulses that pass along three types of neural pathways that eral Edinger–Westphal nucleus. The third-order neurons
project to V1: a red-green pathway that signals differences pass from this nucleus to the ciliary ganglion in the oculomo-
between L- and M-cone responses, a blue-yellow pathway that tor nerve, and the fourth-order neurons pass from this gan-
signals differences between S-cone and the sum of L- and glion to the ciliary body.
M-cone responses, and a luminance pathway that signals the
sum of L- and M-cone responses. These pathways project to the
blobs and the deep portion of layer 4 of V1. From the blobs and EYE MOVEMENTS
layer 4, color information is projected to V8. However, it is not
known how V8 converts color input into the sensation of color. The eye is moved within the orbit by six ocular muscles
Color blindness is most often an inherited condition in which (Figure 15–15). These are innervated by the oculomotor, tro-
individuals are unable to distinguish certain colors. The most chlear, and abducens (cranial) nerves. Because the oblique
common type is a red-green color vision deficit, a genetically muscles pull medially, their actions vary with the position of
sex-linked condition that occurs in about 8% of males and 0.4% the eye. When the eye is turned nasally, the inferior oblique
of females. Blue-yellow color vision deficits are less common elevates it and the superior oblique depresses it. When it is
and show no gender selectivity. Color blindness is usually due to turned laterally, the superior rectus elevates it and the inferior
an inherited absence of cones for specific colors. It can also rectus depresses it.
occur in individuals with lesions of area V8 of the visual cortex. Because much of the visual field is binocular, a very high
order of coordination of the movements of the two eyes is nec-
essary if visual images are to fall at all times on corresponding
PUPILLARY LIGHT REFLEX points in the two retinas and diplopia (double vision) is to be
avoided.
When light is directed into one eye, the pupil constricts There are four types of eye movements, each controlled by a
(pupillary light reflex). The optic nerve fibers that carry the different neural system but sharing the same final common
impulses initiating these pupillary responses leave the optic path, the motor neurons that supply the external ocular mus-
nerves near the lateral geniculate bodies. On each side, they cles. Saccades, sudden jerky movements, occur as the gaze
enter the midbrain via the brachium of the superior colliculus shifts from one object to another. They bring new objects of
and terminate in the pretectal nucleus. From this nucleus, the interest onto the fovea and reduce adaptation in the visual

FIGURE 15–15 Extraocular muscles subserving six cardinal positions of gaze. The eye is adducted by the medial rectus and
abducted by the lateral rectus. The adducted eye is elevated by the inferior oblique and depressed by the superior oblique; the abducted
eye is elevated by the superior rectus and depressed by the inferior rectus. (Reproduced with permission from Squire LR, et al [editors]: Fundamental
Neuroscience, 3rd ed. Academic Press, 2008.)
144 SECTION IV CNS/Neural Physiology

pathway that would occur if gaze were fixed on a single object ■ The bending of light rays (refraction) allows one to focus an
for long periods. Smooth pursuit movements are tracking accurate image onto the retina. Light is refracted at the anterior
movements of the eyes as they follow moving objects. Vestibu- surface of the cornea and at the anterior and posterior surfaces
lar movements, adjustments that occur in response to stimuli of the lens. To bring diverging rays from close objects to a focus
on the retina, the curvature of the lens is increased, a process
initiated in the semicircular canals, maintain visual fixation as
called accommodation.
the head moves. Convergence movements bring the visual
■ In hyperopia (farsightedness), the eyeball is too short and light
axes toward each other as attention is focused on objects near
rays come to a focus behind the retina. In myopia (nearsighted-
the observer. The similarity to a human-made tracking system
ness), the anteroposterior diameter of the eyeball is too long.
on an unstable platform such as a ship is apparent: saccadic Astigmatism is a common condition in which the curvature of
movements seek out visual targets, pursuit movements follow the cornea is not uniform. Presbyopia is the loss of accommo-
them as they move about, and vestibular movements stabilize dation for near vision. Strabismus is a misalignment of the eyes
the tracking device as the platform on which the device is usually due to problems with eye muscles.
mounted (i.e., the head) moves about. Saccades are pro- ■ Na+ channels in the outer segments of the rods and cones are
grammed in the frontal cortex and the superior colliculi and open in the dark, so current flows from the inner to the outer
pursuit movements in the cerebellum. segment. When light strikes the outer segment, some of the Na+
channels are closed and the cells are hyperpolarized.
■ Neurons in layer 4 of the visual cortex respond to stimuli in
their receptive fields with on-centers and inhibitory surrounds
CLINICAL CORRELATION or off-centers and excitatory surrounds. Neurons in other
layers are called simple cells if they respond to bars of light,
A 33-year-old salesman was driving his car in a rural area lines, or edges, but only when they have a particular orienta-
and did not notice the bicyclist riding on the sidewalk to tion. Complex cells also require a preferred orientation of a
the right of his car. He made a right-hand turn at the cor- linear stimulus but are less dependent on the location of a
ner and accidentally hit the young woman. Luckily she stimulus in the visual field.
was not hurt, but he recalled that a few days ago as he was ■ The visual pathway is from the rods and cones to bipolar cells
walking along a sidewalk, he did not notice a dog to ganglion cells, and then via the optic tract to the thalamic
approaching him from the left. He was now aware that he lateral geniculate body to the occipital lobe of the cerebral
cortex. The fibers from each nasal hemiretina decussate in the
has reduced peripheral vision. He made an appointment
optic chiasm; the fibers from the nasal half of one retina and
with an ophthalmologist for a visual field exam. The the temporal half of the other synapse on the cells whose axons
results showed that he had reduced vision on the tempo- form the geniculocalcarine tract.
ral half of the visual field of both eyes. When asked ■ Saccades (sudden jerky movements) occur as the gaze shifts
whether he had noticed an increase incidence of head- from one object to another, and they reduce adaptation in the
aches recently, he responded positively. He had thought visual pathway that would occur if gaze were fixed on a single
the headaches were due to recent stress of his job. object for long periods. Smooth pursuit movements are
The results of the visual field exam indicated bitempo- tracking movements of the eyes as they follow moving objects.
ral hemianopia (a type of heteronymous hemianopia). Vestibular movements occur in response to stimuli in the
This signifies damage to the optic chiasm that carries semicircular canals to maintain visual fixation as the head
axons of ganglion cells from the nasal halves of the retina moves. Convergence movements bring the visual axes toward
that transmits visual information from the temporal visual each other as attention is focused on objects near the observer.
fields. A common cause for this defect is a pituitary tumor
such as a pituitary adenoma. The pituitary gland is located
ventral to the optic chiasm. When the tumor grows, it
STUDY QUESTIONS
presses on the optic chiasm. Headaches and decreased 1. A visual exam in an 80-year-old man shows he has a reduced
libido often occur as a result of the tumor that is typically ability to see objects in the upper and lower quadrants of the left
benign. A magnetic resonance image of the pituitary gland visual fields of both eyes but some vision remains in the central
in this patient revealed a tumor pressing on the optic chi- regions of the visual field. The diagnosis is
asm. Medical treatment caused the tumor to shrink and A) central scotoma.
his visual fields were restored to normal. (See Chapter 62 B) heteronymous hemianopia with macular sparing.
C) lesion of the optic chiasm.
for more details about tumors of the anterior pituitary.)
D) homonymous hemianopia with macular sparing.
E) retinopathy.
2. Visual accommodation involves
A) increased tension on the lens ligaments.
CHAPTER SUMMARY B) a decrease in the curvature of the lens.
■ The major parts of the eye are the sclera (protective covering), C) relaxation of the sphincter muscle of the iris.
cornea (transfer light rays), choroids (nourishment), retina D) contraction of the ciliary muscle.
(receptor cells), lens, and iris. E) increased intraocular pressure.
CHAPTER 15 Special Senses I: Vision 145

3. The fovea of the eye 6. Vitamin A is a precursor for the synthesis of


A) has the lowest light threshold. A) transducin.
B) is the region of highest visual acuity. B) retinals.
C) contains only cones. C) the pigment of the iris.
D) contains only rods. D) scotopsin.
E) is situated over the head of the optic nerve. E) aqueous humor.
4. Which of the following parts of the eye has the greatest 7. Which of the following is not involved in color vision?
concentration of rods? A) activation of a pathway that signals differences between
A) ciliary body S-cone responses and the sum of L- and M-cone responses
B) iris B) geniculate layers 3–6
C) optic disk C) P pathway
D) fovea D) area V3A of visual cortex
E) extrafoveal region E) area V8 of visual cortex
5. The correct sequence of events involved in phototransduction in
rods and cones in response to light is
A) activation of transducin, decreased release of glutamate,
structural changes in rhodopsin, closure of Na+ channels,
and decrease in intracellular cGMP.
B) decreased release of glutamate, activation of transducin,
closure of Na+ channels, decrease in intracellular cGMP, and
structural changes in rhodopsin.
C) structural changes in rhodopsin, decrease in intracellular
cGMP, decreased release of glutamate, closure of Na+
channels, and activation of transducin.
D) structural changes in rhodopsin, activation of transducin,
decrease in intracellular cGMP, closure of Na+ channels, and
decreased release of glutamate.
E) activation of transducin, structural changes in rhodopsin,
closure of Na+ channels, decrease in intracellular cGMP, and
decreased release of glutamate.
This page intentionally left blank
16
C H A P T E R

Special Senses II:


Hearing and Equilibrium
Susan M. Barman

O B J E C T I V E S

■ Describe the components and functions of the external, middle, and


inner ear.
■ Describe the way that movements of molecules in the air are converted into
impulses generated in hair cells in the cochlea.
■ Trace the path of auditory impulses in the neural pathways from the cochlear
hair cells to the auditory cortex, and discuss the function of the auditory
cortex.
■ Explain how pitch and loudness are coded in the auditory pathways.
■ Describe the various forms of deafness and tests for their diagnosis.
■ Explain how the receptors in the semicircular canals detect rotational
acceleration and how the receptors in the saccule and utricle detect linear
acceleration.
■ List the major sensory inputs that provide the information synthesized in the
brain into the sense of position in space.

INTRODUCTION air-filled cavity in the temporal bone that opens via the
auditory (Eustachian) tube into the nasopharynx and
Receptors for hearing and equilibrium are housed in the ear. through the nasopharynx to the exterior. The tube is usually
The external ear, middle ear, and cochlea of the inner ear are closed, but during swallowing, chewing, and yawning it
concerned with hearing. The semicircular canals, utricle, and opens, equalizing air pressure on the two sides of the ear-
saccule of the inner ear are concerned with equilibrium. drum. The three auditory ossicles (malleus, incus, and
Receptors in the semicircular canals (hair cells) detect rota- stapes) are in the middle ear (Figure 16–2). The manubrium
tional acceleration, receptors in the utricle detect linear accel- (handle of the malleus) is attached to the back of the tym-
eration in the horizontal direction, and receptors in the saccule panic membrane. Its head is attached to the wall of the mid-
detect linear acceleration in the vertical direction. dle ear, and its short process is attached to the incus, which
articulates with the head of the stapes. The foot plate of the
stapes is attached by an annular ligament to the walls of the
ANATOMY OF THE EAR oval window. Two small skeletal muscles (tensor tympani
and stapedius) are located in the middle ear. Contraction of
EXTERNAL AND MIDDLE EAR the former pulls the manubrium of the malleus medially and
decreases the vibrations of the tympanic membrane; con-
The external ear funnels sound waves to the external traction of the latter pulls the foot plate of the stapes out of
auditory meatus (Figure 16–1). Sound waves pass inward to the oval window.
the tympanic membrane (eardrum). The middle ear is an

147

Ch16_147-158.indd 147 11/26/10 9:45:37 AM


148 SECTION IV CNS/Neural Physiology

FIGURE 16–1 Structures of the outer, middle, and inner portions of the human ear. For clarity, the cochlea has been turned slightly
and the middle ear muscles have been omitted. (Reproduced with permission from Fox SI: Human Physiology. McGraw-Hill, 2008.)

FIGURE 16–2 The medial view of the middle ear. The locations of auditory muscles attached to the middle ear ossicles are indicated.
(Reproduced with permission from Fox SI: Human Physiology. McGraw-Hill, 2008.)
CHAPTER 16 Special Senses II: Hearing and Equilibrium 149

INNER EAR AND COCHLEA The scala media is continuous with the membranous laby-
rinth and does not communicate with the other two scalae.
The inner ear (labyrinth) is made up of two parts, one within The organ of Corti contains the auditory receptors (hair
the other. The bony labyrinth is a series of channels in the cells) whose processes pierce the reticular lamina that is sup-
temporal bone. Inside these channels, surrounded by a fluid ported by the pillar cells or rods of Corti (Figure 16–4). The
(perilymph) is the membranous labyrinth (Figure 16–3) that hair cells are arranged in four rows: three rows of outer hair
is filled with a K+-rich fluid (endolymph). There is no com- cells lateral to the tunnel formed by the rods of Corti, and one
munication between the spaces filled with endolymph and row of inner hair cells medial to the tunnel. Covering the rows
those filled with perilymph. of hair cells is the tectorial membrane in which the tips of the
The cochlear portion of the labyrinth is a coiled tube that, in hairs of the outer cells are embedded. The cell bodies of
humans, is 35-mm long and makes approximately 2.75 turns. the sensory neurons are located in the spiral ganglion within
The basilar membrane and Reissner’s membrane divide it the modiolus; ~95% of these sensory neurons innervate inner
into three chambers or scalae (Figure 16–4). The upper scala hair cells, ~5% innervate outer hair cells, and each sensory
vestibuli and the lower scala tympani contain perilymph and neuron innervates several outer hair cells. By contrast, most
communicate with each other at the apex of the cochlea via a efferent fibers in the auditory nerve terminate on the outer
small opening (helicotrema). At the base of the cochlea, the hair cells. The axons of afferent neurons that innervate hair
scala vestibuli ends at the oval window, which is closed by the cells form the auditory (cochlear) division of the eighth cranial
footplate of the stapes. The scala tympani end at the round nerve.
window, a foramen on the medial wall of the middle ear that The semicircular canals are oriented in the three planes.
is closed by the flexible secondary tympanic membrane. Inside the bony canals, the membranous canals are suspended

Cupula

Semicircular canal

VIII

Tectorial membrane

Basilar
membrane

Otolithic membrane
Cochlea

Sacculus
FIGURE 16–3 Schematic of the human inner ear showing the membranous labyrinth with enlargements of the structures in which
hair cells are embedded. The membranous labyrinth is suspended in perilymph and filled with K+-rich endolymph that bathes the receptors.
Hair cells (darkened for emphasis) are in different arrays characteristic of the receptor organs. The semicircular canals are sensitive to angular
acceleration that deflects the gelatinous cupula and associated hair cells. In the cochlea, hair cells spiral along the basilar membrane in the organ
of Corti. Airborne sounds set the eardrum in motion, which is conveyed to the cochlea by bones of the middle ear. This flexes the membrane up
and down. Hair cells in the organ of Corti are stimulated by shearing motion. The otolithic organs (saccule and utricle) are sensitive to linear
acceleration in vertical and horizontal planes. Hair cells are attached to the otolithic membrane. VIII, eighth cranial nerve, with auditory and
vestibular divisions. (Adapted with permission from Hudspeth AJ. How the ear’s works work. Nature. 1989;341:397. Copyright 1989 by Macmillan Magazines.)
150 SECTION IV CNS/Neural Physiology

AUDITORY RECEPTORS:
Scala Stria HAIR CELLS
vestibuli vascularis
Reissner’s Spiral The hair cells in the organ of Corti signal hearing, the hair cells
membrane ligament in the utricle signal horizontal acceleration, the hair cells in the
Tectorial
membrane Scala saccule signal vertical acceleration, and a patch in each of the
Limbus media three semicircular canals signals rotational acceleration. These
hair cells have a common structure (Figure 16–5). Each is
Spiral embedded in an epithelium made up of supporting cells, with
prominence the basal end in close contact with afferent neurons. Project-
Spiral ing from the apical end are 30–150 rod-shaped processes or
ganglion Scala Spiral
tympani ligament hairs. Except in the cochlea, one of these, the kinocilium, is a
Modiolus
true but nonmotile cilium with nine pairs of microtubules
around its circumference and a central pair of microtubules. It
Spiral Basilar
lamina Organ
membrane is one of the largest processes and has a clubbed end. The
of Corti
kinocilium is lost from the hair cells of the cochlea in adults;
however, the other processes (stereocilia) are found in all hair
Reticular Outer cells. They have cores composed of parallel filaments of actin
lamina hair cells DCs that is coated with isoforms of myosin. Within the clump of
processes on each cell there is an orderly structure. Along an
axis toward the kinocilium, the stereocilia increase progres-
Tectorial
membrane sively in height; along the perpendicular axis, all stereocilia are
Inner the same height.
hair cell Arch
IPC
Tunnel
ELECTRICAL RESPONSES
Nerve
fibers Habenula Pillar cell Basilar
The resting membrane potential of the hair cells is about
perforata (rod of Corti) membrane –60 mV. When the stereocilia are pushed toward the kinocil-
Spiral ium, the membrane potential is decreased to about –50 mV.
lamina The hair processes provide a mechanism to generate changes
FIGURE 16–4 Top: Cross-section of the cochlea, showing the in membrane potential proportional to the direction and dis-
organ of Corti and the three scalae of the cochlea. Bottom: tance the hair moves. When the bundle of processes is pushed
Structure of the organ of Corti, as it appears in the basal turn of the in the opposite direction, the cell is hyperpolarized. Displac-
cochlea. DC, outer phalangeal cells (Deiters’ cells) supporting outer ing the processes in a direction perpendicular to this axis pro-
hair cells; IPC, inner phalangeal cell supporting inner hair cell.
vides no change in membrane potential, and displacing the
(Reproduced with permission from Pickels JO: An Introduction to the Physiology of
processes in directions that are intermediate between these
Hearing, 2nd ed. Academic Press, 1988.)
two directions produces depolarization or hyperpolarization
that is proportionate to the degree to which the direction is
in perilymph. A receptor structure (crista ampullaris) is toward or away from the kinocilium.
located in the expanded end (ampulla) of each of the mem-
branous canals. Each crista consists of hair cells and support-
ing (sustentacular) cells surmounted by a gelatinous partition GENESIS OF ACTION POTENTIALS
(cupula) that closes off the ampulla (Figure 16–3). The pro- IN AFFERENT NERVE FIBERS
cesses of the hair cells are embedded in the cupula, and the
bases of the hair cells contact the afferent fibers of the vestibu- Very fine processes called tip links (Figure 16–6) tie the tip of
lar division of the eighth cranial nerve. each stereocilium to the side of its higher neighbor, and at the
Within each membranous labyrinth is an otolithic organ junction are mechanosensitive cation channels. If shorter ste-
(macula). Another macula is located on the wall of the saccule reocilia are pushed toward higher ones, the open time of the
in a semivertical position. The maculae contain supporting channels increases. K+ and Ca2+ enter via the channel and pro-
cells and hair cells, surmounted by an otolithic membrane in duce depolarization. A molecular motor in the higher neigh-
which are embedded crystals of calcium carbonate, the oto- bor then may move the channel toward the base, releasing
liths (Figure 16–3), which are also called otoconia or ear dust. tension in the tip link. This causes the channel to close and
The processes of the hair cells are embedded in the membrane. permits restoration of the resting state. Depolarization of hair
The nerve fibers from the hair cells join those from the cristae cells causes them to release a neurotransmitter that initiates
in the vestibular division of the eighth cranial nerve. depolarization of neighboring afferent neurons.
CHAPTER 16 Special Senses II: Hearing and Equilibrium 151

OM OM
OL

K
S

RC SC

A E

FIGURE 16–5 Structure of a hair cell in the saccule. Left: Hair cells in the membranous labyrinth of the ear have a common structure,
and each is within an epithelium of supporting cells (SC) surmounted by an otolithic membrane (OM) embedded with the otoliths (OL).
Projecting from the apical end are rod-shaped processes, or hair cells (RC), in contact with afferent (A) and efferent (E) nerve fibers. Except in the
cochlea, one of these, kinocilium (K), is a true but nonmotile cilium with nine pairs of microtubules around its circumference and a central pair
of microtubules. The other processes, stereocilia (S), are found in all hair cells; they have cores of actin filaments coated with isoforms of myosin.
There is an orderly structure within the clump of processes on each cell. Along an axis toward the kinocilium, the stereocilia increase
progressively in height; along the perpendicular axis, all stereocilia are the same height. (Reproduced with permission from Hillman DE: Morphology of
peripheral and central vestibular systems. In: Frog Neurobiology. Llinas R, Precht W (editors). Springer, 1976.) Right: Scanning electron photomicrograph of
processes on a hair cell in the saccule. The otolithic membrane has been removed. The small projections around the hair cell are microvilli on
supporting cells. (Courtesy of A.J. Hudspeth.)

Myosin
K+
Ca2+

Tip link

FIGURE 16–6 Schematic representation of the role of tip links in the responses of hair cells. When a stereocilium is pushed
toward a taller stereocilium, the tip link is stretched and opens an ion channel in its taller neighbor. The channel next is moved down the taller
stereocilium by a molecular motor, so the tension on the tip link is released. When hairs return to their resting position, the motor moves back
up the stereocilium. (Modified with permission from Kandel ER, Schwartz JH, Jessel TM [editors]: Principles of Neuroscience, 4th ed. McGraw-Hill, 2000.)
152 SECTION IV CNS/Neural Physiology

chain saw, lawn mower) is extremely high, 60–80 dB (e.g., alarm


clock, busy traffic, dishwasher, conversation) is very loud,
Scala vestibuli SL and
SV 40–50 dB (e.g., moderate rainfall, normal room noise) is mod-
Na+ 150
K+ 5 erate, and 30 dB (e.g., whisper, library) is faint.
Cl− 125 The sound frequencies audible to humans range from about
20 to 20,000 cycles per second (cps, Hz). The range decreases
with age, especially difficulty detecting higher frequency
Na+ 1 sounds. The threshold of the human ear varies with the pitch
K+ 150

Cl 130
of the sound; the greatest sensitivity is in the 1,000–4,000-Hz
range. The pitch of the average male and female voice in con-
versation is 120 and 250 Hz, respectively. The number of
pitches that can be distinguished by an average individual is
Organ of Corti Na+ 150
K+ 3 about 2,000, but trained musicians can improve on this figure
Cl− 125 considerably.
Scala tympani
FIGURE 16–7 Ionic composition (mmol/L) of perilymph in SOUND TRANSMISSION
the scala vestibuli, endolymph in the scala media, and perilymph
in the scala tympani. SL, spiral ligament; SV, stria vascularis. The The ear converts sound waves in the environment into action
dashed arrow indicates the path by which K+ recycles from the hair potentials in the auditory nerves. The waves are transformed
cells to the supporting cells to the spiral ligament and is then by the eardrum and auditory ossicles into movements of the
secreted back into the endolymph by cells in the stria vascularis. foot plate of the stapes. These movements set up waves in the
(Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H:
fluid of the inner ear. The action of the waves on the organ of
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
Corti generates action potentials in the nerve.
The tympanic membrane moves in and out in response to
the pressure changes produced by sound waves on its external
The K+ that enters hair cells via the mechanosensitive cation surface. Thus, the membrane functions as a resonator that
channels is recycled (Figure 16–7). It enters supporting cells reproduces the vibrations of the sound source. It stops vibrat-
and then passes on to other supporting cells via tight junc- ing almost immediately when the sound wave stops. The
tions. In the cochlea, it eventually reaches the stria vascularis motions of the tympanic membrane are imparted to the man-
and is secreted back into the endolymph, completing the cycle. ubrium. The malleus rocks on an axis through the junction of
The processes of the hair cells project into the endolymph and its long and short processes, so that the short process transmits
the bases are bathed in perilymph. The perilymph is formed the vibrations of the manubrium to the incus. The incus moves
mainly from plasma; endolymph is formed in the scala media in such a way that the vibrations are transmitted to the head of
by the stria vascularis and has a high concentration of K+ and the stapes. Movements of the head of the stapes swing its foot
a low concentration of Na+. Cells in the stria vascularis have a plate to and fro like a door hinged at the posterior edge of the
high concentration of Na+,K+-ATPase. oval window. The auditory ossicles function as a lever system
that converts the resonant vibrations of the tympanic mem-
brane into movements of the stapes against the perilymph-
HEARING filled scala vestibuli of the cochlea (Figure 16–8). This system
increases the sound pressure that arrives at the oval window,
SOUND WAVES because the lever action of the malleus and incus multiplies
the force 1.3 times and the area of the tympanic membrane is
Sound is the sensation produced when vibrations of molecules in
much greater than the area of the foot plate of the stapes.
the external environment strike the tympanic membrane. The
When the middle ear muscles (tensor tympani and stape-
loudness of a sound is typically correlated with the amplitude of
dius) contract, the manubrium of the malleus pulls inward
a sound wave and its pitch with its frequency (number of waves
and the foot plate of the stapes pushes outward (Figure 16–2),
per unit of time). The amplitude of a sound wave is expressed on
decreasing sound transmission. Loud sounds initiate the tym-
a relative scale, called a decibel scale. The intensity of a sound in
panic reflex, which contracts the middle ear muscles to pre-
bels is the logarithm of the ratio of the intensity of that sound to a
vent strong sound waves from causing excessive stimulation of
standard sound. A value of 0 dB does not mean the absence of
the auditory receptors.
sound; rather, it is a sound level whose intensity is equal to that of
a standard. The 0–160-dB range from threshold pressure to a
pressure that is potentially damaging to the organ of Corti actu- BONE AND AIR CONDUCTION
ally represents a 107-fold variation in sound pressure.
A range of 120–160 dB (e.g., firearms, jackhammer, jet plane Ossicular conduction is the normal conduction of sound
on takeoff) is painful, 90–110 dB (e.g., subway, bass drum, waves to the fluid of the inner ear via the tympanic
CHAPTER 16 Special Senses II: Hearing and Equilibrium 153

Malleus ACTION POTENTIALS IN


Pivot
Incus
AUDITORY NERVE FIBERS
Stapes

Oval Reissner's Inner hair cells are the primary sensory cells that generate
window membrane action potentials in auditory nerves and are stimulated by the
fluid movements noted above. Outer hair cells respond to
sound, but depolarization makes them short and hyperpolar-
Organ
ization makes them lengthy. They do this over a very flexible
of Corti part of the basal membrane, and this action increases the
amplitude and clarity of sounds. The frequency of the action
Round potentials in auditory nerve fibers is proportional to the loud-
window ness of the sound stimuli.
Basilar The major determinant of the pitch perceived when a sound
Auditory tube membrane wave strikes the ear is the place in the organ of Corti that is
maximally stimulated. The traveling wave set up by a tone pro-
FIGURE 16–8 Schematic representation of the auditory duces peak depression of the basilar membrane, and conse-
ossicles and the way their movement translates movements of quently maximal receptor stimulation, at one point. The
the tympanic membrane into a wave in the fluid of the inner ear.
distance between this point and the stapes is inversely related
The wave is dissipated at the round window. The movements of
the ossicles, the membranous labyrinth, and the round window are
to the pitch of the sound, with low tones producing maximal
indicated by dashed lines. (Reproduced with permission from Barrett KE, stimulation at the apex of the cochlea and high tones produc-
Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. ing maximal stimulation at the base.
McGraw-Hill Medical, 2009.)

CENTRAL PATHWAY
membrane and the auditory ossicles. Sound waves also ini-
tiate vibrations of the secondary tympanic membrane that The afferent fibers in the auditory division of the eighth cranial
closes the round window; this process, unimportant in nor- nerve end in dorsal and ventral cochlear nuclei (Figure 16–9).
mal hearing, is called air conduction. Bone conduction is From there, auditory impulses pass by various routes to the
the transmission of vibrations of the bones of the skull to inferior colliculi, the centers for auditory reflexes, and via the
the fluid of the inner ear; this plays a role in transmission of medial geniculate body in the thalamus to the auditory cor-
extremely loud sounds. Considerable bone conduction also tex. Other impulses enter the reticular formation. Information
occurs when a vibrating tuning fork is applied directly to from both ears converges on each superior olive, and beyond
the skull. this, most of the neurons respond to inputs from both sides.
The primary auditory cortex is Brodmann’s area 41. Low
tones are represented anterolaterally and high tones postero-
TRAVELING WAVES medially in the auditory cortex.
In the primary auditory cortex, most neurons respond to
The movements of the foot plate of the stapes set up a series inputs from both ears, but strips of cells are stimulated
of traveling waves in the perilymph of the scala vestibuli. by input from the contralateral ear and inhibited by input
The bony walls of the scala vestibuli are rigid, but Reissner’s from the ipsilateral ear. There are several additional auditory
membrane is flexible. The basilar membrane is not under receiving areas, just as there are several receiving areas for
tension, and it also is readily depressed into the scala tym- cutaneous sensation. The auditory association areas adjacent
pani by the peaks of waves in the scala vestibuli. Displace- to the primary auditory receiving areas are widespread.
ments of the fluid in the scala tympani are dissipated into air The olivocochlear bundle is a prominent bundle of efferent
at the round window. Sound distorts the basilar membrane, fibers in each auditory nerve that arises from both ipsilateral
and the site at which this distortion is maximal is deter- and contralateral superior olivary complexes and ends primar-
mined by the frequency of the sound wave. The tops of the ily around the bases of the outer hair cells of the organ of
hair cells in the organ of Corti are held rigid by the reticular Corti.
lamina, and the processes of the outer hair cells are embed-
ded in the tectorial membrane (Figure 16–4). When the sta-
pes moves, both membranes move in the same direction, but SOUND LOCALIZATION
they are hinged on different axes, so a shearing motion
bends the hairs. The processes of the inner hair cells are not Determination of the direction from which a sound ema-
attached to the tectorial membrane, but they are bent by nates in the horizontal plane depends on detecting the dif-
fluid moving between the membrane and the underlying ference in time between the arrival of the stimulus in the
hair cells. two ears and the consequent difference in phase of the sound
154 SECTION IV CNS/Neural Physiology

waves on the two sides; it also depends on the fact that the eighth cranial nerve or within central auditory pathways. It
sound is louder on the side closest to the source. The detect- can impair the ability to hear certain pitches while others
able time difference, which can be as little as 20 microsec- are unaffected. Aminoglycoside antibiotics such as strepto-
onds, is the most important factor at frequencies below mycin and gentamicin obstruct the mechanosensitive chan-
3,000 Hz and the loudness difference the most important nels in the stereocilia of hair cells and can cause the cells to
factor at frequencies above 3,000 Hz. Neurons in the audi- degenerate, producing sensorineural hearing loss and
tory cortex that receive input from both ears respond maxi- abnormal vestibular function. Damage to the outer hair cells
mally or minimally when the time of arrival of a stimulus at by prolonged exposure to noise is associated with hearing
one ear is delayed by a fixed period relative to the time of loss. Other causes include tumors of the eighth cranial nerve
arrival at the other ear. This fixed period varies from neuron and cerebellopontine angle and vascular damage in the
to neuron. medulla.
Sounds coming from directly in front of the individual dif- Conduction and sensorineural deafness can be differenti-
fer in quality from those coming from behind because each ated by simple tests with a tuning fork. Three of these tests,
pinna (the visible portion of the exterior ear) is turned slightly named for the individuals who developed them, are outlined
forward. Also, reflections of sound waves from the pinnal sur- in Table 16–1. The Weber and Schwabach tests demonstrate
face change as sounds move up or down; the change in the the important masking effect of environmental noise on the
sound waves is the primary factor in locating sounds in the auditory threshold.
vertical plane. Lesions of the auditory cortex disrupt sound
localization.
VESTIBULAR SYSTEM
DEAFNESS The vestibular system is divided into the vestibular apparatus
and central vestibular nuclei. The vestibular apparatus within
Hearing loss is the most common sensory defect in humans. the inner ear detects head motion and position and transduces
Presbycusis, the gradual hearing loss associated with aging, this information into a neural signal. The vestibular nuclei are
affects more than one third of those over 75 and is probably concerned with maintaining the position of the head in space;
due to gradual cumulative loss of hair cells and neurons. In the tracts that descend from these nuclei mediate head-on-
most cases, hearing loss is a multifactorial disorder caused by neck and head-on-body adjustments.
both genetic and environmental factors. The vestibular ganglia contain the cell bodies of the neurons
Conductive deafness refers to impaired sound transmission supplying the cristae and maculae. Each vestibular nerve ter-
in the external or middle ear and impacts all sound frequen- minates in the ipsilateral vestibular nucleus and in the floccu-
cies. Causes of conduction deafness include plugging of the lonodular lobe of the cerebellum (Figure 16–9). Fibers from
external auditory canals with wax (cerumen) or foreign bod- the semicircular canals end in the superior and medial divi-
ies, fluid accumulation due to otitis externa (inflammation of sions of the vestibular nucleus and project mainly to nuclei
the outer ear, “swimmer’s ear”) or otitis media (inflammation controlling eye movement. Fibers from the utricle and saccule
of the middle ear), perforation of the eardrum, and osteoscler- end in Deiters’ nucleus, which projects to the spinal cord.
osis in which bone is resorbed and replaced with sclerotic The vestibular nuclei also project to the thalamus and from
bone that grows over the oval window. there to the primary somatosensory cortex. The ascending
Sensorineural deafness is usually due to the loss of connections to cranial nerve nuclei are concerned with eye
cochlear hair cells but can also be due to problems with the movements.

TABLE 16–1 Common tests with a tuning fork to distinguish between sensorineural and conduction deafness.
Weber Rinne Schwabach

Method Base of vibrating tuning fork placed on Base of vibrating tuning fork placed on Bone conduction of patient
vertex of skull mastoid process until subject no longer compared with that of healthy
hears it, and then held in air next to ear subject

Normal Hears equally on both sides Hears vibration in air after bone
conduction is over

Conduction deafness Sound louder in diseased ear because Vibrations in air not heard after bone Bone conduction better than
(one ear) masking effect of environmental noise conduction is over normal (conduction defect
is absent on diseased side excludes masking noise)

Sensorineural deafness Sound louder in normal ear Vibration heard in air after bone Bone conduction worse than
(one ear) conduction is over, as long as nerve normal
deafness is partial
CHAPTER 16 Special Senses II: Hearing and Equilibrium 155

To somatosensory
cortex
To cortex (superior
temporal gyrus)

Thalamus

Thalamus
Medial
geniculate III
body
IV
Medial
Pineal To longitudinal
cerebellum fasciculus
Inferior colliculus VI
Reticular
formation
IV ventricle

Dorsal and ventral


cochlear nuclei Vestibular
ganglion
Vestibular nuclei:
superior, lateral
(Deiters’), medial,
Spiral Medulla Lateral spinal
ganglion vestibulo-
Superior spinal tract Anterior vestibulo-
olives spinal tracts
From utricle,
From semicircular
cochlea canals

AUDITORY VESTIBULAR
FIGURE 16–9 Simplified diagram of main auditory (left) and vestibular (right) pathways superimposed on a dorsal view of the
brain stem. Cerebellum and cerebral cortex have been removed. III, IV, and VI are the 3rd, 4th, and 6th cranial nerves. (Reproduced with permission
from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

RESPONSES TO ROTATIONAL Nystagmus is the characteristic jerky movement of the eye


ACCELERATION observed at the start and end of a period of rotation. It is a reflex
that maintains visual fixation on stationary points while the
Rotational acceleration in the plane of a given semicircular body rotates. When rotation starts, the eyes move slowly in a
canal stimulates its crista. The endolymph is displaced in a direction opposite to the direction of rotation, maintaining
direction opposite to the rotation, and the fluid pushes on the visual fixation (vestibulo-ocular reflex). At the end of this
cupula, deforming it. This bends the processes of the hair cells movement, the eyes quickly snap back to a new fixation point
(Figure 16–3). When a constant speed of rotation is reached, and then again move slowly in the other direction. The slow
the fluid spins at the same rate as the body and the cupula component is initiated by impulses from the vestibular laby-
swings back into the upright position. When rotation is rinths; the quick component is triggered by a center in the brain
stopped, deceleration produces displacement of the endo- stem. By convention, the direction of eye movement in nystag-
lymph in the direction of the rotation, and the cupula is mus is identified by the direction of the quick component. The
deformed in a direction opposite to that during acceleration. It direction of the quick component is the same as that of the rota-
returns to mid position in 25–30 seconds. Movement of the tion; postrotatory nystagmus, which is due to displacement of
cupula in one direction increases the activity of nerves from the cupula, is in the opposite direction. Nystagmus can be
the crista, and movement in the opposite direction inhibits observed at rest in patients with lesions of the brain stem. Caloric
neural activity. stimulation can be used to test the function of the vestibular
Rotation causes maximal stimulation of the semicircular labyrinth. The semicircular canals are stimulated by instilling
canals in the plane of rotation. Because the canals on one side warm (40°C) or cold (30°C) water into the ear. The temperature
of the head are a mirror image of those on the other side, the difference sets up convection currents in the endolymph, with
endolymph is displaced toward the ampulla on one side and consequent motion of the cupula. In normal subjects, warm
away from it on the other. The pattern of stimulation reaching water causes nystagmus that bears toward the stimulus, and cold
the brain varies with the direction and the plane of rotation. water induces nystagmus that bears toward the opposite ear.
156 SECTION IV CNS/Neural Physiology

RESPONSES TO LINEAR ACCELERATION


ear causing vertigo or severe dizziness, tinnitus, fluctuat-
The utricular and saccular maculae respond to horizontal and ing hearing loss, and the sensation of pressure or pain in
vertical acceleration, respectively. The otoliths are denser than the affected ear lasting for several hours. Symptoms can
the endolymph, and acceleration in any direction causes them occur suddenly and recur daily or very rarely. The hearing
to be displaced in the opposite direction, distorting the hair loss is initially transient but can become permanent. The
cell processes and generating neural activity. The maculae also pathophysiology may involve an immune reaction. An
discharge tonically in the absence of head movement, because inflammatory response can increase fluid volume within
of the pull of gravity on the otoliths. the membranous labyrinth, causing it to rupture and
The labyrinth righting reflexes are a series of responses allowing the endolymph and perilymph to mix together.
integrated in midbrain nuclei in response to head tilt. The There is no cure for Ménière disease but the symptoms
response is a compensatory contraction of neck muscles to can be controlled by reducing the fluid retention through
keep the head level. These reflexes stabilize the head and keep dietary changes (low-salt or salt-free diet, no caffeine, no
the eyes fixed on visual targets despite movements of the alcohol) or medication.
body.
Vestibular impulses that reach the cerebral cortex are likely
responsible for conscious perception of motion and supply CHAPTER SUMMARY
part of the information necessary for orientation in space.
■ The external ear funnels sound waves to the external auditory
Vertigo is the sensation of rotation in the absence of actual
meatus and tympanic membrane. From there, sound waves
rotation and is a prominent symptom when one labyrinth is
pass through three auditory ossicles (malleus, incus, and
inflamed. stapes) in the middle ear. The inner ear, or labyrinth, contains
the cochlea and organ of Corti.
SPATIAL ORIENTATION ■ The hair cells in the organ of Corti signal hearing. The
stereocilia provide a mechanism for generating changes in
Orientation in space depends on input from the vestibular membrane potential proportional to the direction and distance
the hair moves. Sound is the sensation produced when
receptors as well as visual cues, information from propriocep-
longitudinal vibrations of air molecules strike the tympanic
tors in joint capsules, and cutaneous touch and pressure recep- membrane.
tors. These four inputs are synthesized at a cortical level into a
■ The activity within the auditory pathway passes from the eighth
continuous picture of the individual’s orientation in the envi- cranial nerve afferent fibers to the dorsal and ventral cochlear
ronment. nuclei to the inferior colliculi to the thalamic medial geniculate
body and then to the auditory cortex.
■ Loudness is correlated with the amplitude of a sound wave and
pitch with the frequency.
■ Conductive deafness is due to impaired sound transmission in
CLINICAL CORRELATION the external or middle ear and impacts all sound frequencies.
Sensorineural deafness is due to loss of cochlear hair cells or to
A 26-year-old woman came to her primary care physi- damage to the eighth cranial nerve or central auditory pathways.
cian’s office because of a recent series of episodes in which ■ Rotational acceleration stimulates the crista in the semicircular
she has experienced severe dizziness, tinnitus (ringing in canal, displacing the endolymph in a direction opposite to the
the ears), and nausea and vomiting. During her latest epi- direction of rotation, deforming the cupula and bending the
sode, she realized that she could barely hear her daughter hair cell. The utricle responds to horizontal acceleration and
calling for her in the next room. When queried by the the saccule to vertical acceleration. Acceleration in any
direction displaces the otoliths, distorting the hair cell
physician’s assistant, she recalled that similar, although
processes and generating neural activity.
less severe, incidences of these symptoms had occurred
■ Spatial orientation is dependent on input from vestibular
while she was in college. In particular, she experienced
receptors, visual cues, proprioceptors in joint capsules, and
tinnitus, vertigo, and nausea on various occasions. Because cutaneous touch and pressure receptors.
the symptoms occurred only sporadically and lasted for
only hours or a day, she did not seek medical advice. The
latest episodes have her fearful that something serious is STUDY QUESTIONS
wrong with her.
1. A 40-year-old male, employed as a road construction worker for
A hearing test detected reduced hearing in one ear.
nearly 20 years, went to his physician to report that he has been
Based on the symptoms, her physician suspected that she
having difficulty hearing during normal conversations. A Weber
had Ménière disease. She was later seen by an otolaryn- test showed that sound from a vibrating tuning fork was localized
gologist and a neurologist to rule out other causes of her to the right ear. A Schwabach test showed that bone conduction
symptoms. Ménière disease is an abnormality of the inner was below normal. A Rinne test showed that both air and bone
CHAPTER 16 Special Senses II: Hearing and Equilibrium 157

conduction were abnormal, but air conduction lasted longer than 4. Some diseases damage the hair cells in the ear. When the damage to
bone conduction. The diagnosis was the outer hair cells is greater than the damage to the inner hair cells
A) sensorial deafness in both ears. A) perception of vertical acceleration is disrupted.
B) conduction deafness in the right ear. B) K+ concentration in endolymph is decreased.
C) sensorial deafness in the right ear. C) K+ concentration in perilymph is decreased.
D) conduction deafness in the left ear. D) there is severe hearing loss.
E) sensorineural deafness in the left ear. E) affected hair cells fail to shorten when exposed to sound.
2. What would the diagnosis be if a patient had the following test 5. Which of the following are incorrectly paired?
results? Weber test showed that sound from a vibrating tuning
A) tympanic membrane:manubrium of malleus
fork was louder than normal; Schwabach test showed that bone
B) helicotrema:apex of cochlea
conduction was better than normal; and Rinne test showed that
C) foot plate of stapes:oval window
air conduction did not outlast bone conduction.
D) otoliths:semicircular canals
A) sensorial deafness in both ears E) basilar membrane:organ of Corti
B) conduction deafness in both ears
6. The direction of nystagmus is vertical when a subject is rotated
C) normal hearing
D) both sensorial and conduction deafness A) after warm water is put in one ear.
E) a possible tumor on the eighth cranial nerve B) with the head tipped backward.
C) after cold water is put in both ears.
3. Postrotational nystagmus is caused by continued movement of
D) with the head tipped sideways.
A) aqueous humor over the ciliary body in the eye. E) after section of one vestibular nerve.
B) cerebrospinal fluid over the vestibular nuclei.
7. In the utricle, tip links in hair cells are involved in
C) endolymph in the semicircular canals, with consequent
bending of the cupula and stimulation of hair cells. A) formation of perilymph.
D) endolymph toward the helicotrema. B) depolarization of the stria vascularis.
E) perilymph over hair cells with processes embedded in the C) movements of the basement membrane.
tectorial membrane. D) perception of sound.
E) regulation of mechanosensitive ion channels.
This page intentionally left blank
17
C H A P T E R

Special Senses III:


Smell and Taste
Susan M. Barman

O B J E C T I V E S

■ Describe the basic features of the olfactory epithelium and olfactory bulb.
■ Explain signal transduction in odorant receptors.
■ Outline the pathway by which impulses generated in the olfactory epithelium
reach the olfactory cortex.
■ Describe the location and cellular composition of taste buds.
■ Name the five major taste receptors and their signal transduction
mechanisms.
■ Outline the pathways by which impulses generated in taste receptors reach
the insular cortex.

INTRODUCTION tors. The axons of the olfactory sensory neurons pass through
the cribriform plate of the ethmoid bone and enter the olfac-
Smell and taste are classified as visceral senses because of their tory bulbs (Figure 17–1). New olfactory sensory neurons are
close association with gastrointestinal function. Physiologi- generated by basal stem cells as needed to replace those dam-
cally, they are related to each other; the flavors of various foods aged by exposure to the environment.
are in large part a combination of their taste and smell. This In the olfactory bulbs, the axons of the olfactory sensory
explains why food may taste “different” if one has a cold that neurons (first cranial nerve) contact the primary dendrites
depresses the sense of smell. Both smell and taste receptors are of the mitral cells and tufted cells (Figure 17–3) to form
chemoreceptors that are stimulated by molecules in solution anatomically discrete synaptic units called olfactory glom-
in mucus in the nose and saliva in the mouth. eruli. Both types of neurons send axons into the olfactory
cortex. The olfactory bulbs also contain periglomerular
cells, which are inhibitory neurons connecting one glomeru-
lus to another, and granule cells, which have no axons and
SMELL make reciprocal synapses with the lateral dendrites of the
mitral and tufted cells. At these synapses, the mitral or tufted
OLFACTORY EPITHELIUM cell excites the granule cell by releasing glutamate, and the
AND OLFACTORY BULBS granule cell in turn inhibits the mitral or tufted cell by releas-
ing γ-Aminobutyric acid (GABA).
A specialized portion of the nasal mucosa, the yellowish, pig-
mented olfactory epithelium (Figure 17–1), contains 10–20
million bipolar olfactory sensory neurons interspersed with OLFACTORY CORTEX
glia-like supporting (sustentacular) cells and basal stem cells.
The olfactory epithelium is the place in the body where the ner- The axons of the mitral and tufted cells pass posteriorly
vous system is closest to the external world. Each neuron has a through the lateral olfactory stria to terminate on apical
short, thick dendrite that projects into the nasal cavity where it dendrites of pyramidal cells in five regions of the olfactory
terminates in a knob containing 10–20 cilia (Figure 17–2). The cortex: anterior olfactory nucleus, olfactory tubercle, piri-
cilia are unmyelinated processes that contain odorant recep- form cortex, amygdala, and entorhinal cortex (Figure 17–4).

159

Ch17_159-166.indd 159 11/26/10 4:40:38 PM


160 SECTION IV CNS/Neural Physiology

Olfactory
bulb Olfactory
bulb

Cribriform
plate

Olfactory
Olfactory
epithelium
sensory
neurons

FIGURE 17–1 Olfactory sensory neurons embedded within the olfactory epithelium in the dorsal posterior recess of the nasal
cavity. These neurons project axons to the olfactory bulb of the brain, a small ovoid structure that rests on the cribriform plate of the ethmoid
bone. (Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

From these regions, information travels directly to the frontal is involved with the emotional responses to olfactory stimuli,
cortex or via the thalamus to the orbitofrontal cortex. Con- and the pathway to the entorhinal cortex is concerned with
scious discrimination of odors relies on the pathway to the olfactory memories.
orbitofrontal cortex. The orbitofrontal activation is generally
greater on the right side than the left; thus, cortical representa-
tion of olfaction is asymmetric. The pathway to the amygdala

To olfactory bulb

Basal Axon
cells

Olfactory
sensory
neuron

Dendrite

Supporting
cell

Mucus Cilia

FIGURE 17–2 Structure of the olfactory epithelium. There


are three cell types: olfactory sensory neurons, supporting cells, and
basal stem cells at the base of the epithelium. Each sensory neuron FIGURE 17–3 Basic neural circuits in the olfactory bulb. Note
has a dendrite that projects to the epithelial surface. Numerous cilia that olfactory receptor cells with one type of odorant receptor project to
protrude into the mucosal layer lining the nasal lumen. A single axon one olfactory glomerulus (OG) and olfactory receptor cells with another
projects from each neuron to the olfactory bulb. Odorants bind to type of receptor project to a different olfactory glomerulus. CP, cribriform
specific odorant receptors on the cilia and initiate a cascade of events plate; PG, periglomerular cell; M, mitral cell; T, tufted cell; Gr, granule cell.
leading to generation of action potentials in the sensory axon. White arrows, excitatory synapses; black arrows, inhibitory synapses.
(Modified with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: (Adapted with permission from Mori K, Nagao H, Yoshihara Y. The olfactory bulb: Coding
Principles of Neural Science, 4th ed. McGraw-Hill, 2000.) and processing of odor molecular information. Science. 1999;286(5440):711–715.)
CHAPTER 17 Special Senses III: Smell and Taste 161

Accessory
Lateral
olfactory
olfactory tract
bulb
Mitral
cell
Mitral
cell
Tufted
cell

Olfactory Anterior Olfactory Piriform Amygdala Entorhinal


bulb olfactory tubercle cortex cortex
nucleus
Vomeronasal Olfactory
organ epithelium
Hypothalamus
Contralateral
olfactory
bulb
Hippocampus

Thalamus

Orbitofrontal Frontal cortex


cortex

FIGURE 17–4 Diagram of the olfactory pathway. Information is transmitted from the olfactory bulb by axons of mitral and tufted relay
neurons in the lateral olfactory tract. Mitral cells project to five regions of the olfactory cortex: anterior olfactory nucleus, olfactory tubercle,
piriform cortex, and parts of the amygdala and entorhinal cortex. Tufted cells project to anterior olfactory nucleus and olfactory tubercle; mitral
cells in the accessory olfactory bulb project only to the amygdala. Conscious discrimination of odor depends on the neocortex (orbitofrontal
and frontal cortices). Emotive aspects of olfaction derive from limbic projections (amygdala and hypothalamus). In rodents and some mammals,
a well-developed vomeronasal organ is concerned with perception of odors that act as pheromones; its receptors project to the accessory
olfactory bulb. (Reproduced with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

OLFACTORY DISCRIMINATION catalyze the production of cAMP, which acts as a second mes-
AND SIGNAL TRANSDUCTION senger to open cation channels, causing an inward-directed
Ca2+ current. This produces the graded receptor potential,
The olfactory epithelium is covered by a thin layer of mucus which then leads to an action potential in the olfactory
secreted by the supporting cells and Bowman glands that lie nerve.
beneath the epithelium. The mucus bathes the odorant recep- Although there are millions of olfactory sensory neurons,
tors on the cilia and provides the appropriate molecular and each expresses only 1 of the 1,000 different odorant recep-
ionic environment for odor detection. Odor-producing mole- tors. Each neuron projects to one or two glomeruli
cules are usually small, containing from 3 to 20 carbon atoms, (Figure 17–3). This provides a distinct two-dimensional map
and molecules with the same number of carbon atoms but dif- in the olfactory bulb that is unique to the odorant. The mitral
ferent structural configurations have different odors. Rela- cells with their glomeruli project to different parts of the
tively high water and lipid solubility are characteristic of olfactory cortex.
substances with strong odors. The olfactory glomeruli demonstrate lateral inhibition
Anosmia (inability to smell) and hyposmia or hypesthesia mediated by periglomerular and granule cells. This sharpens
(diminished olfactory sensitivity) can result from simple nasal and focuses olfactory signals. In addition, the extracellular
congestion or from damage to the olfactory nerves due to field potential in each glomerulus oscillates, and the granule
fractures of the cribriform plate, neuroblastomas or menin- cells can regulate the frequency of the oscillation. The exact
giomas, or infections (such as abscesses). Aging is also associ- function of the oscillation is unknown, but it may also help to
ated with abnormalities in smell sensation; more than 75% of focus the olfactory signals reaching the cortex.
humans over the age of 80 have an impaired ability to identify
odors.
The genes that code for about 1,000 different types of odor- TASTE
ant receptors make up the largest gene family so far described
in mammals. The amino acid sequences of odorant receptors TASTE BUDS
are diverse, but all the odorant receptors are coupled to het-
erotrimeric G proteins. When an odorant molecule binds to The specialized sense organ for taste (gustation) consists of
its receptor, the G protein subunits (α, β, γ) dissociate approximately 10,000 taste buds. There are four morphologi-
(Figure 17–5). The α-subunit activates adenylate cyclase to cally distinct types of cells within each taste bud: basal cells,
162 SECTION IV CNS/Neural Physiology

Odorant
Adenylate Na+/Ca2+
Odorant cyclase channel
receptor

G-proteins

(a)

Ca2+ Na+
Odorant Odorant Adenylate
receptor cyclase Na+/Ca2+
channel

cAMP
FIGURE 17–5 Signal transduction in an odorant receptor.
Olfactory receptors are G protein–coupled receptors that ATP
dissociate on binding to the odorant. The α-subunit of G proteins
activates adenylate cyclase to catalyze production of cAMP. cAMP cAMP
acts as a second messenger to open cation channels. Inward
diffusion of Na+ and Ca2+ produces depolarization. (Reproduced with (b)
permission from Fox SI: Human Physiology. McGraw-Hill, 2008.)

dark cells, light cells, and intermediate cells (Figure 17–6). tongue reach the brain stem via the glossopharyngeal nerve
The latter three cell types are referred to as Type I, II, and III (Figure 17–7). The fibers from areas other than the tongue
taste cells. They are the sensory neurons that respond to taste (e.g., pharynx) reach the brain stem via the vagus nerve. On
stimuli. The apical ends of taste cells have microvilli that proj- each side, the myelinated but relatively slowly conducting taste
ect into the taste pore, a small opening on the dorsal surface of fibers in these three nerves unite in the gustatory portion of
the tongue where taste cells are exposed to the oral contents. the nucleus of the tractus solitarius (NTS) in the medulla
Each taste bud is innervated by about 50 nerve fibers, and con- oblongata (Figure 17–7). From there, axons of second-order
versely, each nerve fiber receives input from an average of five neurons ascend in the ipsilateral medial lemniscus to pass
taste buds. The basal cells arise from the epithelial cells sur- directly to the ventral posteromedial nucleus of the thala-
rounding the taste bud. They differentiate into new taste cells, mus, from which fibers project to the anterior insula and
and the old cells are replaced with a half-time of about 10 days. frontal operculum in the ipsilateral cerebral cortex. This
If the sensory nerve is cut, the taste buds it innervates degener- region is rostral to the face area of the postcentral gyrus, which
ate and eventually disappear. may be the area that mediates conscious perception of taste
The taste buds are located in the mucosa of the epiglottis, and taste discrimination.
palate, and pharynx and in the walls of papillae of the tongue
(Figure 17–6). The fungiform papillae are rounded structures
most numerous near the tip of the tongue; the circumvallate TASTE MODALITIES, RECEPTORS,
papillae are prominent structures arranged in a V on the back AND TRANSDUCTION
of the tongue; the foliate papillae are on the posterior edge of
the tongue. Each fungiform papilla has up to 5 taste buds, A 30% change in the concentration of the substance being
mostly located at the top of the papilla; each vallate and foliate tasted is necessary before an intensity difference can be
papilla contains up to 100 taste buds, mostly located along the detected. A protein that binds taste-producing molecules is
sides of the papillae. produced by the Ebner gland that secretes mucus into the
cleft around vallate papillae. Ageusia (absence of the sense of
taste) and hypogeusia (diminished taste sensitivity) can be
TASTE PATHWAYS caused by damage to the lingual or glossopharyngeal nerve.
Neurological disorders such as vestibular schwannoma,
The sensory nerve fibers from the taste buds on the anterior Bell’s palsy, familial dysautonomia, multiple sclerosis, and
two thirds of the tongue travel in the chorda tympani branch certain infections (e.g., primary amoeboid meningoenceph-
of the facial nerve, and those from the posterior third of the alopathy) can also cause problems with taste sensitivity.
CHAPTER 17 Special Senses III: Smell and Taste 163

Chorda tympani Taste pore


nerve (VII)
Saliva
Glossopharyngeal
nerve (IX)

Epithelial cell

Circumvallate Taste cell

Basal cell

Gustatory afferent nerve


Serous
gland
A To sensory ganglion
Foliate
C

Taste
bud

Fungiform
B
FIGURE 17–6 Taste buds located in papillae of the human tongue. A) Taste buds on the anterior two thirds of the tongue are
innervated by the chorda tympani branch of the facial nerve; those on the posterior one third of the tongue are innervated by the lingual branch
of the glossopharyngeal nerve. B) The three major types of papillae (circumvallate, foliate, and fungiform) are located on specific parts of the
tongue. C) Taste buds are composed of basal stem cells and three types of taste cells (dark, light, and intermediate). Taste cells extend from the
base of the taste bud to the taste pore, where microvilli contact tastants dissolved in saliva and mucus. (Modified with permission from Kandel ER,
Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

Ageusia can also be an adverse side effect of various drugs or nucleotide–gated cation channel, and other mechanisms
vitamin B3 or zinc deficiencies. Aging and tobacco abuse also may contribute to sour transduction.
contribute to diminished taste. Dysgeusia or parageusia Substances that taste sweet also act via the G protein gust-
(unpleasant perception of taste) causes a metallic, salty, foul, ducin. The T1R3 family of G protein–coupled receptors is
or rancid taste. expressed by about 20% of taste cells, some of which also
Humans have five established basic tastes: sweet, sour, bit- express gustducin. Sugars taste sweet, but so do compounds
ter, salt, and umami. All tastants are sensed from all parts of such as saccharin that have an entirely different structure. Nat-
the tongue and adjacent structures. Afferent nerves to the NTS ural sugars such as sucrose and synthetic sweeteners may act
contain fibers from all types of taste receptors, without any via different receptors on gustducin. Like the bitter-responsive
clear localization of types. A fifth taste sense, umami, has been receptors, sweet-responsive receptors act via cyclic nucleotides
added to the four classic tastes. This taste is triggered by gluta- and inositol phosphate metabolism.
mate and particularly by the monosodium glutamate (MSG) Bitter taste is produced by a variety of unrelated com-
used very extensively in Asian cooking. The taste is pleasant pounds. Many of these are poisons, and bitter taste serves as
and sweet but differs from the standard sweet taste. a warning to avoid them. Some bitter compounds bind to
Figure 17–8 illustrates signal transduction in taste recep- and block K+-selective channels. Many G protein–linked
tors. The salty taste is triggered by NaCl. Salt-sensitive taste is receptors in the human genome are taste receptors (T2R
mediated by a Na+-selective channel known as ENaC, the family) and are stimulated by bitter substances such as
amiloride-sensitive epithelial sodium channel. The entry of strychnine. In some cases, these receptors couple to the het-
Na+ into the salt receptors depolarizes the membrane, gener- erotrimeric G protein, gustducin. Gustducin decreases
ating the receptor potential. In humans, the amiloride sensi- cAMP and increases the formation of inositol phosphates
tivity of salt taste is less pronounced than in some species, that could lead to depolarization. Some bitter compounds
suggesting that there are additional mechanisms to activate are membrane permeable and may not involve G proteins;
salt-sensitive receptors. quinine is an example.
The sour taste is triggered by protons (H+ ions). ENaCs per- Umami taste is due to activation of a truncated metabotro-
mit the entry of protons and may contribute to the sensation pic glutamate receptor, mGluR4, in the taste buds. The way
of sour taste. The H+ ions can also bind to and block a K+- activation of the receptor produces depolarization is unsettled.
sensitive channel. The decrease in K+ permeability can depo- Glutamate in food may also activate ionotropic glutamate
larize the membrane. A hyperpolarization-activated cyclic receptors to depolarize umami receptors.
164 SECTION IV CNS/Neural Physiology

Gustatory cortex
(anterior insula-
frontal operculum)

Ventral posterior
medial nucleus of
thalamus

Geniculate
Chorda ganglion
tympani

N. VII

Tongue Nucleus of
N. IX
Petrosal solitary tract
Glossopharyngeal ganglion
N. X Gustatory
area

Nodose
ganglion

Pharynx

FIGURE 17–7 Diagram of taste pathways. Signals from the taste buds travel via different nerves to gustatory areas of the nucleus of
the tractus solitarius that relays information to the thalamus; the thalamus projects to the gustatory cortex N. VII, N. IX, and N. X are the 7th, 9th,
and 10th cranial nerves, respectively. (Modified with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)

Salty Sour Predicted sweet Bitter Umami


receptor (L-glutamate)
α γ
N

N N N N
C C C C C C C
ENaC, others ENaC, HCN, others T1R3 (sac locus) T2R family, others Taste mGluR4

FIGURE 17–8 Signal transduction in taste receptors. Salt-sensitive taste is mediated by a Na -selective channel (ENaC); sour taste is
+

mediated by H+ ions permeable to ENaCs; umami taste is mediated by glutamate acting on a metabotropic receptor, mGluR4; bitter taste is
mediated by the T2R family of G protein–coupled receptors; sweet taste may be dependent on the T1R3 family of G protein–coupled receptors
that couple to the G protein gustducin. (Adapted with permission from Lindemann B. Receptors and transduction in taste. Nature. 2001;413:219.)
CHAPTER 17 Special Senses III: Smell and Taste 165

posteromedial nucleus of the thalamus, and on to the anterior


CLINICAL CORRELATION insula and frontal operculum in the ipsilateral cerebral cortex.

A 10-year-old boy was riding in the front passenger seat of STUDY QUESTIONS
an automobile being driven by his father. He dropped his
MP3 player and released his seat belt to retrieve it. At that 1. Odorant receptors are
moment, the car was hit from the rear by a speeding motor- A) located in the olfactory bulb.
ist. He received a sharp blow to his nose when he struck B) located on dendrites of mitral and tufted cells.
the dashboard. He was taken to the emergency room of a C) located on neurons that project directly to the olfactory cortex.
D) located on neurons in the olfactory epithelium that project to
nearby hospital. An x-ray showed that he had broken his
mitral cells and from there directly to the olfactory cortex.
ethmoid bone that separates the nasal cavity from the
E) located on sustentacular cells that project to the olfactory bulb.
brain. Following this accident, he lost the sense of smell
2. Taste receptors
(anosmia), and his sense of taste was also diminished.
A) for sweet, sour, bitter, salt, and umami tastes are spatially
The olfactory nerve runs through the ethmoid bone.
separated on the surface of the tongue.
The nerve can be damaged when the bone is broken,
B) are synonymous with taste buds.
resulting in the loss of the ability to smell. Because of the C) are a type of chemoreceptor.
close relationship between taste and smell, anosmia is D) are innervated by afferents in the facial, trigeminal,
associated with a reduction in taste sensitivity (hypogeusia). and glossopharyngeal nerves.
Major causes of anosmia include upper respiratory tract E) All of the above are correct.
infection, nasal polyps, head trauma, tumors of the frontal 3. Which of the following does not increase the ability to
lobe, toxins, and prolonged use of nasal decongestants. discriminate many different odors?
Anosmia is generally permanent in cases in which the A) many different receptors
olfactory nerve or other neural elements in the olfactory B) pattern of olfactory receptors activated by a given odorant
neural pathway are damaged. In addition to not being able C) projection of different mitral cell axons to different parts
to experience the enjoyment of pleasant aromas and full of the brain
spectrum of tastes, individuals with anosmia are at risk D) neural processing in the amygdala
because they are not being able to detect the odor from E) lateral inhibition
such dangers as gas leaks, fire, and spoiled food 4. Which of the following are incorrectly paired?
A) ENaC:sour
B) α-gustducin:bitter taste
C) nucleus tractus solitarius:taste
CHAPTER SUMMARY D) fungiform papillae:smell
E) Ebner glands:taste acuity
■ Olfactory sensory neurons, supporting (sustentacular) cells,
and basal stem cells are located in the olfactory epithelium 5. Which of the following is true about olfactory transmission?
within the upper portion of the nasal cavity. A) An olfactory sensory neuron expresses a wide range of
■ The cilia located on the dendritic knob of the olfactory sensory odorant receptors.
neuron contain odorant receptors that are coupled to heterotri- B) Lateral inhibition within the olfactory glomeruli reduces
meric G proteins. the ability to distinguish between different types of odorant
receptors.
■ Axons of olfactory sensory neurons contact the dendrites of
C) Conscious discrimination of odors is dependent on the
mitral and tufted cells in the olfactory bulbs to form olfactory
pathway to the orbitofrontal cortex.
glomeruli.
D) Olfaction is closely related to gustation because odorant
■ Information from the olfactory bulb travels via the lateral and gustatory receptors use the same central pathways.
olfactory stria directly to the olfactory cortex, including the E) All of the above are correct.
anterior olfactory nucleus, olfactory tubercle, piriform cortex,
6. Which of the following is not true about gustatory sensation?
amygdala, and entorhinal cortex.
A) The sensory nerve fibers from the taste buds on the anterior
■ Taste buds are the specialized sense organs for taste and are
two thirds of the tongue travel in the chorda tympani
composed of basal stem cells and Type I, II, and III taste cells
branch of the facial nerve.
that may represent various stages of differentiation of develop-
B) The sensory nerve fibers from the taste buds on the
ing taste cells. They are located in the mucosa of the epiglottis,
posterior third of the tongue travel in the petrosal branch
palate, and pharynx and in the walls of papillae of the tongue.
of the glossopharyngeal nerve.
■ There are taste receptors for sweet, sour, bitter, salt, and umami C) The pathway from taste buds on the left side of the tongue
tastes. Signal transduction mechanisms include passage is transmitted ipsilaterally to the cerebral cortex.
through ion channels, binding to and blocking ion channels, D) Sustentacular cells in the taste buds serve as stem cells to
and second messenger systems. permit growth of new taste buds.
■ The afferents from taste buds in the tongue travel via the 7th, E) The pathway from taste receptors includes synapses in the
9th, and 10th cranial nerves to synapse in the NTS. From there, nucleus of the tractus solitarius in the brain stem and
axons ascend via the ipsilateral medial lemniscus to the ventral ventral posterior medial nucleus in the thalamus.
This page intentionally left blank
18
C H A P T E R

Control of Posture
and Movement
Susan M. Barman

O B J E C T I V E S

■ Describe how skilled movements are planned and carried out.


■ Describe the posture-regulating parts of the central nervous system.
■ Describe decerebrate and decorticate rigidity.
■ Describe the function of the basal ganglia in control of movement.
■ Describe the symptoms of Parkinson’s disease.
■ Discuss the functions of the cerebellum and the neurological abnormalities
produced by diseases of this part of the brain.

INTRODUCTION CONTROL OF AXIAL


Somatic motor activity ultimately depends on the pattern and AND DISTAL MUSCLES
rate of discharge of the spinal motor neurons and homologous
neurons in the motor nuclei of the cranial nerves. These neu- Within the brain stem and spinal cord, pathways and neu-
rons, the final common pathway to skeletal muscle, receive in- rons that control skeletal muscles of the trunk and proximal
put from an array of descending pathways, other spinal neurons, portions of the limbs are located medially or ventrally. Path-
and peripheral afferents. The integration of these multiple in- ways and neurons that are concerned with the control of
puts regulates the posture of the body and makes coordinated skeletal muscles in the distal portions of the limbs are lo-
movement possible. The inputs bring about voluntary activity, cated laterally. The axial muscles are concerned with pos-
adjust body posture to provide a stable background for move- tural adjustments and gross movements; the distal limb
ment, and coordinate the action of the various muscles to make muscles mediate fine, skilled movements. For example, neu-
movements smooth and precise. As shown by the scheme in rons in the medial portion of the ventral horn innervate
Figure 18–1, voluntary movement is planned in the cortex, proximal limb muscles, particularly the flexors, and lateral
basal ganglia, and lateral cerebellum. The basal ganglia and ventral horn neurons innervate distal limb muscles. Simi-
cerebellum funnel information to the premotor and motor cor- larly, the ventral corticospinal tract and medial descending
tex via the thalamus. Posture is continually adjusted both before brain stem pathways (rubrospinal, reticulospinal, tectospi-
and during movement by descending brain stem pathways and nal, and vestibulospinal tracts) are concerned with adjust-
peripheral afferents. Movement is smoothed and coordinated ments of proximal muscles and posture, and the lateral
by the connections of medial and intermediate portions of the corticospinal and rubrospinal tracts are concerned with
cerebellum. The basal ganglia and lateral cerebellum are part of distal limb muscles and, particularly in the case of the corti-
a feedback circuit to the premotor and motor cortex that is con- cospinal tract, with skilled voluntary movements.
cerned with planning and organizing voluntary movement.

167

Ch18_167-176.indd 167 11/26/10 4:41:35 PM


168 SECTION IV CNS/Neural Physiology

Plan Execute

Basal ganglia

Cortical
Idea association Premotor and Movement
areas motor cortex

Lateral
cerebellum Intermediate
cerebellum

FIGURE 18–1 Control of voluntary movement. Commands for voluntary movement originate in cortical association areas. The cortex,
basal ganglia, and cerebellum work cooperatively to plan movements. Movement executed by the cortex is relayed via the corticospinal tracts
and corticobulbar tracts to motor neurons. The cerebellum provides feedback to adjust and smooth movement. (Reproduced with permission from
Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

CORTICOSPINAL AND Precentral


CORTICOBULBAR TRACTS gyrus
(area 4,
The axons of neurons from the motor cortex that project to etc)
spinal motor neurons form the corticospinal tracts, a large
bundle of about 1 million fibers. About 80% of these fibers
cross the midline in the medullary pyramids to form the lat-
eral corticospinal tract (Figure 18–2). The other 20% form the
ventral corticospinal tract, which does not cross the midline
Corticospinal tract
until it reaches the level of the spinal cord at which it termi-
nates. Lateral corticospinal tract neurons make monosynaptic
connections to motor neurons, especially those concerned Internal capsule
with skilled movements, and on spinal interneurons. The tra-
jectory from the cortex to the spinal cord passes through the Decussation of
corona radiata to the posterior limb of the internal capsule. the pyramids
Within the midbrain corticospinal tract fibers traverse the ce- Pyramids
rebral peduncle and the basilar pons until they reach the med-
ullary pyramids on their way to the spinal cord. Lateral cortico-
The corticobulbar tract is composed of the fibers that pass spinal tract
from the motor cortex to motor neurons in the trigeminal, Ventral cortico- (80% of fibers)
spinal tract
facial, and hypoglossal nuclei. Corticobulbar neurons end ei- (20% of fibers) Anterior
ther directly on the cranial nerve nuclei or on their antecedent horn cell
interneurons within the brain stem. Their axons traverse
through the genu of the internal capsule, the cerebral peduncle Interneuron
Spinal nerve
(medial to corticospinal tract neurons), to descend with corti-
cospinal tract fibers in the pons and medulla.
The motor system can be divided into lower and upper mo-
Distal
tor neurons. Lower motor neurons refer to the spinal and cra- muscle
nial motor neurons that directly innervate skeletal muscles.
Proximal
Upper motor neurons are those in the cortex and brain stem muscle
that activate the lower motor neurons. The pathophysiologic
responses to damage to lower and upper motor neurons are FIGURE 18–2 Corticospinal tracts. This tract originates in the
precentral gyrus and passes through the internal capsule. Most fibers
very distinctive.
decussate in the pyramids and descend in the lateral white matter of
Damage to lower motor neurons is associated with flaccid the spinal cord to form the lateral division of the tract that can make
paralysis, muscular atrophy, fasciculations (visible muscle monosynaptic connections with spinal motor neurons. The ventral
twitches that appear as flickers under the skin), hypotonia division of the tract remains uncrossed until reaching the spinal cord
(decreased muscle tone), and hyporeflexia or areflexia. where axons terminate on spinal interneurons antecedent to motor
Damage to upper motor neurons initially causes muscles to neurons. (Reproduced with permission from Barrett KE, Barman SM, Boitano S,
become weak and flaccid but eventually leads to spasticity, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
CHAPTER 18 Control of Posture and Movement 169

hypertonia (increased resistance to passive movement),


hyperactive stretch reflexes, and abnormal plantar extensor
reflex (Babinski sign). The Babinski sign is dorsiflexion of the

Shoulder
Elbow
Wrist
great toe and fanning of the other toes when the lateral aspect

Trunk
Hip
Kne Ank

d
Han
of the sole of the foot is scratched. In adults, the normal

e le

R ttle
M ing
To
response to this stimulation is plantar flexion in all the toes.

Li

e
e

dl
s x
de b

id
It is of value in the localization of disease processes, but its In um k
Th Nec w
o
physiologic significance is unknown. Br all
b
d eye
n e
li da Fac
Eye

VOCALIZATION
Lips

MOTOR CORTEX AND


Ja
VOLUNTARY MOVEMENT Ton w

N
Sw gue

TIO
allo
win

LIVA
N
g

IO
AT
Corticospinal and corticobulbar tract neurons are pyramidal

SA
IC
ST
MA
shaped and located in layer V of the cerebral cortex (see
Chapter 12). Figure 18–3 shows the major cortical regions in-
volved in motor control. About 31% of the corticospinal tract
neurons are from the primary motor cortex (M1; Brodmann’s
area 4) in the precentral gyrus of the frontal lobe, extending into FIGURE 18–4 Motor homunculus. The figure represents, on a
coronal section of the precentral gyrus, the location of the cortical
the central sulcus. The premotor cortex and supplementary
representation of the various parts. The size of the various parts is
motor cortex (Brodmann’s area 6) account for 29% of the cor-
proportionate to the cortical area devoted to them. Compare with
ticospinal tract neurons. The premotor area is anterior to the Figure 13–6. (Reproduced with permission from Penfield W, Rasmussen G:
precentral gyrus, on the lateral and medial cortical surface, and The Cerebral Cortex of Man. Macmillan, 1950.)
the supplementary motor area is on and above the superior bank
of the cingulate sulcus on the medial side of the hemisphere. The
other 40% of corticospinal tract neurons originate in the pari-
the musculature on the opposite side of the body. The size of the
etal lobe (Brodmann’s area 5, 7) and primary somatosensory
cortical representation of each body part is proportional to the
area (Brodmann’s area 3, 1, 2) in the postcentral gyrus.
number of corticospinal neurons supplying the musculature of
The various parts of the body are represented in M1, with the
that region of the body and its role in fine, voluntary movement.
feet at the top of the gyrus and the face at the bottom (Figure 18–4).
Thus, the areas involved in speech and hand movements are es-
The facial area is represented bilaterally, but the rest of the repre-
pecially large. A somatotopic organization continues through-
sentation is unilateral, with the cortical motor area controlling
out the corticospinal and corticobulbar pathways. The cells in
the cortical motor areas are arranged in columns. Neurons in
several cortical columns project to the same muscle; also, the
Motor cortex cells in each column receive extensive sensory input from
Supplementary
Primary somatic the peripheral area in which they produce movement, providing
motor area
Premotor sensory cortex the basis for feedback control of movement.
cortex Posterior The supplementary motor area (Figure 18–3), which proj-
parietal ects to M1, also contains a map of the body, but it is less precise
cortex
6 4
than in M1. It is involved in organizing or planning motor
3,1,2 5 sequences, while M1 executes the movements. When human
7 7 subjects count to themselves without speaking, the motor cor-
tex is quiescent, but when they speak the numbers aloud,
blood flow increases in M1 and the supplementary motor area.
Thus, both M1 and the supplementary motor area are involved
in voluntary movement when the movements being performed
are complex and involve planning.
Prefrontal The premotor cortex (Figure 18–3), which also contains a
cortex
somatotopic map, receives input from sensory regions of the
parietal cortex and projects to M1, the spinal cord, and the
FIGURE 18–3 A view of the human cerebral cortex, showing
the motor cortex (Brodmann’s area 4) and other areas concerned
brain stem reticular formation. This region is concerned with
with control of voluntary movement, along with the numbers setting posture at the start of a planned movement and with
assigned to the regions by Brodmann. (Reproduced with permission getting the individual prepared to move. It is most involved in
from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. control of proximal limb muscles needed to orient the body
McGraw-Hill, 2000.) for movement.
170 SECTION IV CNS/Neural Physiology

In addition to providing fibers that run in the corticospinal ullary reticulospinal, vestibulospinal, and tectospinal tracts.
and corticobulbar tracts, the somatic sensory area and por- These pathways descend in the ipsilateral ventral columns of
tions of the posterior parietal lobe project to the premotor the spinal cord and terminate predominantly on interneurons
area. Some of the neurons in area 5 (Figure 18–3) are con- in the ventromedial part of the ventral horn to control axial
cerned with aiming the hands toward an object and manipu- and proximal muscles. A few medial pathway neurons synapse
lating it, whereas some of the neurons in area 7 are concerned directly on motor neurons controlling axial muscles.
with hand–eye coordination. The medial and lateral vestibulospinal tracts were briefly
described in Chapter 16. The medial tract originates in the
medial and inferior vestibular nuclei and projects bilaterally to
MEDIAL AND LATERAL cervical spinal motor neurons that control neck musculature.
BRAIN STEM PATHWAYS: The lateral tract originates in the lateral vestibular nuclei and
projects ipsilaterally to neurons at all spinal levels. It activates
POSTURE AND VOLUNTARY motor neurons to antigravity muscles (e.g., proximal limb ex-
MOVEMENT tensors) to control posture and balance.
The pontine and medullary reticulospinal tracts project to
As mentioned above, spinal motor neurons are organized such all spinal levels. They are involved in the maintenance of pos-
that those innervating the most proximal muscles are located ture and in modulating muscle tone, especially via an input to
most medially and those innervating the more distal muscles γ-motor neurons. Pontine reticulospinal neurons are primar-
are located more laterally. This organization is also reflected in ily excitatory and medullary reticulospinal neurons are
descending brain stem pathways (Figure 18–5). primarily inhibitory. The tectospinal tract originates in the su-
The medial brain stem pathways, which work in concert perior colliculus of the midbrain. It projects to the contralat-
with the ventral corticospinal tract, are the pontine and med- eral cervical spinal cord to control head and eye movements.

A Medial brain stem pathways B Lateral brain stem pathways

Tectum Red nucleus


(magnocellular part)

Medial
reticular
formation

Tectospinal
tract
Lateral and medial
vestibular nuclei
Reticulospinal
tract

Vestibulospinal
tracts

Rubrospinal
tract

FIGURE 18–5 Medial and lateral descending brain stem pathways involved in motor control. A) Medial pathways (reticulospinal,
vestibulospinal, and tectospinal) terminate in ventromedial area of spinal gray matter and control axial and proximal muscles. B) Lateral pathway
(rubrospinal) terminates in dorsolateral area of spinal gray matter and controls distal muscles. (Reproduced with permission from Kandel ER, Schwartz JH,
Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
CHAPTER 18 Control of Posture and Movement 171

The main control of distal muscles is from the lateral corti-


cospinal tract, but neurons within the red nucleus of the mid-
BASAL GANGLIA
brain cross the midline and project to interneurons in the The basal ganglia are comprised of the caudate nucleus,
dorsolateral part of the spinal ventral horn to also influence putamen, globus pallidus, and the functionally related sub-
motor neurons that control distal limb muscles. This rubrospi- thalamic nucleus and substantia nigra (Figure 18–7). The
nal tract excites flexor motor neurons and inhibits extensor globus pallidus is divided into external and internal segments
motor neurons. (GPe and GPi). The substantia nigra is divided into pars com-
pacta and pars reticulata. The caudate nucleus and putamen
are collectively called the striatum; the putamen and globus
DECEREBRATION AND pallidus form the lenticular nucleus.
DECORTICATION The main inputs to the basal ganglia terminate in the stri-
atum (Figure 18–8). They include the excitatory corticostri-
A complete transection of the brain stem between the supe- ate pathway from M1 and premotor cortex. There is also a
rior and inferior colliculi permits the brain stem pathways projection from intralaminar nuclei of the thalamus to the
to function independent of their input from higher brain striatum (thalamostriatal pathway). The connections be-
structures. This is called a midcollicular decerebration and tween the parts of the basal ganglia include a dopaminergic
is diagramed in Figure 18–6 by the dashed line labeled A. nigrostriatal projection from the substantia nigra pars com-
This lesion interrupts all input from the cortex and red nu- pacta to the striatum and a corresponding GABAergic
cleus to distal muscles of the extremities. The excitatory and projection from the striatum to substantia nigra pars reticu-
inhibitory reticulospinal pathways (primarily to postural lata. The striatum projects to both GPe and GPi. GPe proj-
extensor muscles) remain intact. The dominance of drive ects to the subthalamic nucleus, which in turn projects to
from ascending sensory pathways to the excitatory reticu- both GPe and GPi.
lospinal pathway leads to decerebrate rigidity, which is The principal output from the basal ganglia is from GPi via
characterized by hyperactivity in extensor muscles in all the thalamic fasciculus to the ventral lateral, ventral anterior,
four extremities. This resembles what ensues after uncal and centromedian nuclei of the thalamus. From the thalamic
herniation resulting from a supratentorial lesion as seen in nuclei, fibers project to the prefrontal and premotor cortex.
patients with large tumors or a hemorrhage in the cerebral The substantia nigra also projects to the thalamus.
hemisphere. The main feature of the connections of the basal ganglia is
After decerebration, section of dorsal roots to a limb (dashed that the cerebral cortex projects to the striatum, the striatum
line labeled B in Figure 18–6) eliminates the hyperactivity of to GPi, GPi to the thalamus, and the thalamus back to the cor-
extensor muscles, suggesting that decerebrate rigidity is spas- tex, completing a loop. The output from GPi to the thalamus is
ticity due to facilitation of the myotatic stretch reflex. The ex- inhibitory, whereas the output from the thalamus to the cere-
citatory input from the reticulospinal pathway activates bral cortex is excitatory.
γ-motor neurons that indirectly activate α-motor neurons
(via Ia spindle afferent activity; see Chapter 14). This is called
the gamma loop. FUNCTION
Decerebrate rigidity may also lead to direct activation of
α-motor neurons. If the anterior lobe of the cerebellum is re- The basal ganglia are involved in the planning and program-
moved in a decerebrate animal (dashed line labeled C in ming of voluntary movement (Figure 18–1). They influence
Figure 18–6), extensor muscle hyperactivity is exaggerated the motor cortex via the thalamus. Also, GPi projects to brain
(decerebellate rigidity). This cut eliminates cortical inhibition stem nuclei and from there to motor neurons in the brain stem
of the cerebellar fastigial nucleus and secondarily increases ex- and spinal cord.
citation to vestibular nuclei. This rigidity is not reversed by Three biochemical pathways in the basal ganglia normally
cutting the dorsal roots. operate in a balanced fashion (Figure 18–8): (1) the nigrostri-
Removal of the cerebral cortex (decortication; dashed line atal dopaminergic system, (2) the intrastriatal cholinergic
labeled D in Figure 18–6) produces decorticate rigidity that is system, and (3) the GABAergic system, which projects from
characterized by flexion of the upper extremities at the elbow the striatum to the globus pallidus and substantia nigra. When
and extensor hyperactivity in the lower extremities. The flex- one or more of these pathways become dysfunctional, charac-
ion can be explained by rubrospinal excitation of flexor mus- teristic motor abnormalities occur. Diseases of the basal gan-
cles in the upper extremities; the hyperextension of lower glia lead to two general types of disorders: hyperkinetic and
extremities is due to the same changes that occur after midcol- hypokinetic. The hyperkinetic conditions are those in which
licular decerebration. Decorticate rigidity is seen on the hemi- movement is excessive and abnormal, including tremor,
plegic side in humans after hemorrhages or thromboses in the chorea, athetosis, and ballism. Hypokinetic abnormalities
internal capsule. Sixty percent of intracerebral hemorrhages include akinesia and bradykinesia.
occur in the internal capsule, and 10% each in the cerebral Chorea is characterized by rapid, involuntary “dancing”
cortex, pons, thalamus, and cerebellum. movements. Athetosis is characterized by continuous, slow
172 SECTION IV CNS/Neural Physiology

FIGURE 18–6 A circuit drawing representing lesions produced in experimental animals to replicate decerebrate and decorticate
deficits seen in humans. Bilateral transections are indicated by dashed lines A–D. Decerebration is at a midcollicular level (A), decortication is
rostral to the superior colliculus, dorsal roots sectioned for one extremity (B), and removal of anterior lobe of cerebellum (C). The objective was
to identify anatomic substrates responsible for decerebrate or decorticate rigidity/posturing seen in humans with lesions that either isolate the
forebrain from the brain stem or separate rostral from caudal brain stem and spinal cord. (Reproduced with permission from Haines DE [editor]: Fundamental
Neuroscience for Basic and Clinical Applications, 3rd ed. Elsevier, 2006.)

writhing movements. Choreiform and athetotic movements PARKINSON’S DISEASE


have been likened to the start of voluntary movements occur-
ring in an involuntary, disorganized way. In ballism, involun- Parkinson’s disease has both hypokinetic and hyperkinetic
tary flailing, intense, and violent movements occur. Akinesia features. It was the first disease identified as being due to a de-
is difficulty in initiating movement and decreased spontane- ficiency in a specific neurotransmitter; it is due to the degen-
ous movement. Bradykinesia is slowness of movement. eration of dopaminergic neurons in the substantia nigra pars
CHAPTER 18 Control of Posture and Movement 173

Caudate nucleus
Thalamus
Thalamus
Internal
capsule

Lateral ventricle

Amygdaloid
nucleus Caudate nucleus
Putamen and
globus pallidus Putamen

Lateral view Globus pallidus:


External segment
Caudate nucleus Thalamus Internal segment

Inte Subthalamic
rna Substantia nucleus
l ca
psu nigra
le
Amygdala
Globus
pallidus Tail of Frontal section
Putamen caudate nucleus

Horizontal section
FIGURE 18–7 Basal ganglia. The basal ganglia are composed of the caudate nucleus, putamen, and globus pallidus and the functionally
related subthalamic nucleus and substantia nigra. The frontal (coronal) section shows the location of the basal ganglia in relation to surrounding
structures. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

compacta. It is one of the most common neurodegenerative cogwheel rigidity and tremor at rest. The absence of motor
diseases; it is estimated to occur in 1–2% of individuals over activity and the difficulty in initiating voluntary movements
age 65. Symptoms appear when 60–80% of the nigrostriatal are striking. Normal, unconscious movements such as swing-
dopaminergic neurons degenerate. ing of the arms during walking, facial expressions, and fidg-
The hypokinetic features of Parkinson’s disease are akine- eting are absent in Parkinson’s disease. The rigidity is different
sia and bradykinesia, and the hyperkinetic features are from spasticity because motor neuron discharge increases to
both the agonist and antagonist muscles. Passive motion of
an extremity meets with a plastic, dead-feeling resistance
Cerebral that has been likened to bending a lead pipe and is therefore
cortex
called lead pipe rigidity. Sometimes a series of “catches”
Glu takes place during passive motion (cogwheel rigidity), but
Globus GABA Striatum the sudden loss of resistance seen in a spastic extremity is
pallidus, ES (acetylcholine) absent. The tremor, which is present at rest and disappears
with activity, is due to regular, alternating contractions of an-
GABA Glu GABA GABA DA
tagonistic muscles.
Subthalamic Glu Globus A common treatment for Parkinson’s disease is the adminis-
SNPR SNPC
nucleus pallidus, IS
tration of l-DOPA (levodopa). Unlike dopamine, this dop-
Brain stem GABA GABA GABA amine precursor crosses the blood–brain barrier and helps
and PPN Thalamus repair the dopamine deficiency. However, the degeneration of
spinal cord
these neurons continues and in 5–7 years, the beneficial effects
FIGURE 18–8 Diagrammatic representation of the principal of l-DOPA usually disappear.
connections of the basal ganglia. Solid lines indicate excitatory
pathways, dashed lines inhibitory pathways. The transmitters are
indicated in the pathways, where they are known. Glu, glutamate; DA,
dopamine. Acetylcholine is the transmitter produced by interneurons CEREBELLUM
in the striatum. SNPR, substantia nigra pars reticulata; SNPC,
substantia nigra pars compacta; ES, external segment; IS, internal
The cerebellum sits astride the main sensory and motor sys-
segment; PPN, pedunculopontine nuclei. The subthalamic nucleus tems in the brain stem and is connected to the brain stem by
also projects to the pars compacta of the substantia nigra; this the superior, middle, and inferior peduncles. From a func-
pathway has been omitted for clarity. (Reproduced with permission from tional point of view, the cerebellum is divided into three
Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, parts (Figure 18–9). The vestibulocerebellum has vestibular
23rd ed. McGraw-Hill Medical, 2009.) connections and is concerned with equilibrium and eye
174 SECTION IV CNS/Neural Physiology

Spinocerebellum ning and programming movements. The medial portion of


To medial
descending the cerebellum is called the vermis and it projects to the
systems Motor brain stem area concerned with control of axial and proximal
To lateral execution limb muscles (medial brain stem pathways). The area just
descending lateral to the vermis projects to the brain stem areas con-
systems
cerned with control of distal limb muscles (lateral brain stem
pathways).
To motor
and Motor
premotor planning
CELLULAR ORGANIZATION
cortices OF THE CEREBELLUM
Cerebrocerebellum To Balance
vestibular and eye The cerebellar cortex contains five types of neurons: Purkinje,
nuclei movements
granule, basket, stellate, and Golgi cells (Figure 18–10). The
Vestibulocerebellum Purkinje cells are among the biggest neurons in the CNS, with
FIGURE 18–9 Functional divisions of the cerebellum. extensive dendritic arbors. They are the only output neurons
(Modified with permission from Kandel ER, Schwartz JH, Jessell TM [editors]: of the cerebellar cortex, and their synaptic actions are inhibi-
Principles of Neural Science, 4th ed. McGraw-Hill, 2000.) tory via GABA release. The granule cells innervate the
Purkinje cells; their axons bifurcate to form a T. The branches
of the T are straight and run long distances; thus, they are
movements. The spinocerebellum receives proprioceptive called parallel fibers. The parallel fibers make synaptic con-
input from the body as well as a copy of the “motor plan” tact with the dendrites of many Purkinje cells and release glu-
from the motor cortex. It functions to smooth out and coor- tamate (an excitatory neurotransmitter).
dinate ongoing movements. The cerebrocerebellum in the The other three types of neurons in the cerebellar cortex are
lateral hemisphere interacts with the motor cortex in plan- inhibitory interneurons that release GABA. Basket cells

5 Stellate
cell Parallel fibers:
Axons of granule
cells
Axon
1 Purkinje
cell

Molecular
layer 2
1
Purkinje
layer
5
Axon Granular 4
layer 3 2 Golgi
cell
4 Granule
3 Basket
cell
cell
Axons

FIGURE 18–10 Location and structure of five neuronal types in the cerebellar cortex. Drawings are based on Golgi-stained
preparations. Purkinje cells (1) have processes aligned in one plane; their axons are the only output from the cerebellum. Axons of granule cells
(4) traverse and make connections with Purkinje cell processes in molecular layer. Golgi (2), basket (3), and stellate (5) cells have characteristic
positions, shapes, branching patterns, and synaptic connections. (Reproduced with permission from Kuffler SW, Nicholls JG, Martin AR: From Neuron to Brain,
2nd ed. Sinauer, 1984.)
CHAPTER 18 Control of Posture and Movement 175

Parallel fiber modulating or timing the excitatory output of the deep cere-
+ + + bellar nuclei to the brain stem and thalamus.
Cerebellar
+
cortex
CEREBELLAR DISEASE
BC

PC
GC Damage to the cerebellum leads to several characteristic
abnormalities, including hypotonia, ataxia, and intention trem-
− GR +
or. Most abnormalities are apparent during movement. The

Climbing + marked ataxia is characterized as incoordination due to errors in
fiber the rate, range, force, and direction of movement. Ataxia is man-
ifest not only in the wide-based, unsteady, “drunken” gait of pa-
− tients, but also in defects of the skilled movements involved in
Deep the production of speech. The individual pauses between words
nuclei NC + Mossy fiber
and syllables, a phenomenon referred to as scanning speech.
+ +
Voluntary movements are also highly abnormal. As an
Other
inputs
example, if one attempts to touch an object with a finger, there is
+ an overshoot to one side or the other. This dysmetria promptly
initiates a gross corrective action, but the correction overshoots
FIGURE 18–11 Diagram of neural connections in the
to the other side. Consequently, the finger oscillates back and
cerebellum. Plus (+) and minus (–) signs indicate whether endings
are excitatory or inhibitory. BC, basket cell; GC, Golgi cell; GR, granule forth. This oscillation is the intention tremor of cerebellar dis-
cell; NC, cell in deep nucleus; PC, Purkinje cell. Note that PCs and BCs ease. Another characteristic of cerebellar disease is the inability
are inhibitory. The connections of the stellate cells, which are not to stop movement promptly. Normally, for example, flexion of
shown, are similar to those of the basket cells, except that they end the forearm against resistance is quickly checked when the re-
for the most part on Purkinje cell dendrites. (Reproduced with permission sistance force is suddenly broken off. The patient with cerebel-
from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical lar disease cannot stop the movement of the limb, and the
Physiology, 23rd ed. McGraw-Hill Medical, 2009.) forearm flies backward in a wide arc (rebound phenomenon).
This is one of the reasons these patients show dysdiadochoki-
nesia, the inability to perform rapidly alternating opposite
receive input from the parallel fibers, and each projects to movements such as repeated pronation and supination of the
many Purkinje cells (Figure 18–10). Their axons form a basket hands. Finally, patients with cerebellar disease have difficulty
around the cell body and axon hillock of each Purkinje cell performing actions that involve simultaneous motion at more
they innervate. Stellate cells are similar to the basket cells but than one joint. They dissect such movements and carry them
more superficial in location. The dendrites of Golgi cells re- out one joint at a time (decomposition of movement).
ceive input from the parallel fibers. Their cell bodies receive Motor abnormalities associated with cerebellar damage vary
input from the mossy fibers, and their axons project to the depending on the region involved. The major dysfunctions
dendrites of the granule cells. seen after damage to the vestibulocerebellum are ataxia, dyse-
The primary afferent inputs to the cerebellum are the mossy quilibrium, and nystagmus. Damage to the vermis and fastigial
and climbing fibers, both of which are excitatory nucleus (part of the spinocerebellum) leads to disturbances in
(Figure 18–11). The climbing fibers relay proprioceptive in- control of axial and trunk muscles during attempted antigravity
put from a single source, the inferior olivary nuclei. The postures and scanning speech. Degeneration of this portion of
mossy fibers provide proprioceptive input as well as input the cerebellum can result from thiamine deficiency in alcohol-
from the cerebral cortex via the pontine nuclei. ics or malnourished individuals. The major dysfunctions seen
The fundamental circuits of the cerebellar cortex are rela- after damage to the cerebrocerebellum are delays in initiating
tively simple (Figure 18–11). Climbing fiber inputs exert a movements and decomposition of movement.
strong excitatory effect on a single Purkinje cell, and mossy
fiber inputs exert a weak excitatory effect on many Purkinje
cells via the granule cells. The basket and stellate cells are also
excited by granule cells via the parallel fibers, and their output
inhibits Purkinje cell discharge (feed-forward inhibition).
CLINICAL CORRELATION
Golgi cells are excited by the mossy fiber collaterals, Purkinje About 2 years ago at the age of 34, a man developed pro-
cell collaterals, and parallel fibers, and they inhibit transmis- gressive weakness in his right leg and eventually his
sion from mossy fibers to granule cells. whole right side weakened. He was unable to continue
The output of Purkinje cells is inhibitory to the deep cere- his work as an electrician. He experienced cramps in his
bellar nuclei. These nuclei also receive excitatory inputs via right calf muscle and muscle twitches in his arm and leg.
collaterals from the mossy and climbing fibers. Thus, almost A neurological examination revealed muscular atrophy,
all the cerebellar circuitry seems to be concerned solely with
176 SECTION IV CNS/Neural Physiology

to the striatum and a GABAergic projection from the striatum


fasciculations (muscle twitches that appear as flickers to substantia nigra.
under the skin), and hypotonia of muscles in the arm ■ Parkinson’s disease is due to degeneration of the nigrostriatal
and leg. He also had marked hyporeflexia. Sensory and dopaminergic neurons and is characterized by akinesia,
cognitive function tests were normal. All signs were in- bradykinesia, cogwheel rigidity, and tremor at rest.
dicative of a lower motor neuron disease affecting mul- ■ The cerebellar cortex contains five types of neurons: Purkinje,
tiple spinal cord levels. He was eventually diagnosed with granule, basket, stellate, and Golgi cells. The two main inputs
amyotrophic lateral sclerosis (ALS). Over the course of to the cerebellar cortex are climbing fibers and mossy fibers.
the next year, the disease progressed to the point where Purkinje cells are the only output from the cerebellar cortex
he had difficulty swallowing (dysphagia), so he had to be and they generally project to the deep nuclei.
fed via a gastric tube. About 6 months ago, he developed ■ Damage to the cerebellum leads to several characteristic
difficulty breathing and was placed on a ventilator. One abnormalities, including hypotonia, ataxia, and intention tremor.
week ago, he died of pneumonia.
ALS is a selective, progressive degeneration of α-motor
neurons. “Amyotrophic” means “no muscle nourishment” STUDY QUESTIONS
and describes the atrophy that muscles undergo because 1. A primary function of the basal ganglia is
of disuse. “Sclerosis” refers to the hardness felt when a pa- A) sensory integration.
thologist examines the spinal cord on autopsy; the hard- B) short-term memory.
ness is due to proliferation of astrocytes and scarring of C) planning voluntary movement.
the lateral columns of the spinal cord. This fatal disease is D) neuroendocrine control.
also known as Lou Gehrig’s disease in recognition of a fa- E) slow-wave sleep.
mous American baseball player who died of it. The world- 2. The therapeutic effect of l-DOPA in patients with Parkinson’s
wide annual incidence of ALS is estimated to be 0.5–3 disease eventually wears off because
cases per 100,000 people. Most cases are sporadic, but A) antibodies to dopamine receptors develop.
5–10% of the cases are familial. Forty percent of the famil- B) inhibitory pathways grow into the basal ganglia from
ial cases have a mutation in the gene for Cu/Zn superox- the frontal lobe.
ide dismutase (SOD-1) on chromosome 21. SOD is a free C) cholinergic neurons in the striatum degenerate.
D) the normal action of nerve growth factor (NGF) is
radical scavenger that reduces oxidative stress. A defec-
disrupted.
tive SOD-1 gene permits free radicals to accumulate and
E) the dopaminergic neurons in the substantia nigra continue
kill neurons. The disease has no racial, socioeconomic, or to degenerate.
ethnic boundaries. The life expectancy of ALS patients is
3. Increased neural activity before a skilled voluntary movement
usually 3–5 years after diagnosis. ALS is most commonly is first seen in the
diagnosed in middle age and affects men more often than
A) spinal motor neurons.
women. The causes of ALS are unclear, but may include B) precentral motor cortex.
viruses, neurotoxins, heavy metals, DNA defects (espe- C) midbrain.
cially in familial ALS), immune system abnormalities, and D) cerebellum.
enzyme abnormalities. There is no cure for ALS; treat- E) cortical association areas.
ments (e.g., physical and occupational therapy) focus on 4. After falling down a flight of stairs, a young woman is found to
relieving symptoms and maintaining the quality of life. have partial loss of voluntary movement on the right side of her
body and loss of pain and temperature sensation on the left side
below the midthoracic region. It is probable that she has a lesion
A) transecting the left half of the spinal cord in the
CHAPTER SUMMARY lumbar region.
■ The ventral corticospinal tract and medial descending brain B) transecting the left half of the spinal cord in the upper
stem pathways (tectospinal, reticulospinal, and vestibulospinal thoracic region.
tracts) regulate proximal muscles and posture. The lateral C) transecting sensory and motor pathways on the right
corticospinal and rubrospinal tracts control distal limb muscles side of the pons.
and skilled voluntary movements. D) transecting the right half of the spinal cord in the
■ Decerebrate rigidity leads to hyperactivity in extensor upper thoracic region.
muscles in all four extremities; it is actually spasticity due E) transecting the dorsal half of the spinal cord in the
to facilitation of the myotatic stretch reflex. Decorticate upper thoracic region.
posturing or decorticate rigidity is flexion of the upper 5. Which of the following are not correctly paired?
extremities at the elbow and extensor hyperactivity in the A) medial brain stem pathways:axial and proximal
lower extremities. muscle control
■ The basal ganglia include the caudate nucleus, putamen, globus B) lateral brain stem pathway:rubrospinal tract
pallidus, subthalamic nucleus, and substantia nigra. The C) upper motor neuron disease:hypotonia
connections between the parts of the basal ganglia include a D) cerebellar disease:intention tremor
dopaminergic nigrostriatal projection from the substantia nigra E) decerebrate rigidity:hyperactivity of extensor muscles
19
C H A P T E R

Autonomic Nervous
System
Susan M. Barman

O B J E C T I V E S

■ Describe the location of the cell bodies and axonal trajectories


of preganglionic sympathetic and parasympathetic neurons.
■ Describe the location and trajectories of postganglionic sympathetic
and parasympathetic neurons.
■ Name the neurotransmitters that are released by preganglionic and
postganglionic autonomic neurons.
■ List the major functions of the autonomic nervous system.
■ Identify some of the neural inputs to sympathetic and parasympathetic neurons.

INTRODUCTION α-motor neurons serve as the final common pathway linking


the central nervous system (CNS) to skeletal muscles. Simi-
The autonomic nervous system (ANS) is one of the control sys- larly, sympathetic and parasympathetic neurons serve as the
tems responsible for homeostasis and is the source of innerva- final common pathway from the CNS to visceral targets. How-
tion of organs other than skeletal muscle. Nerve terminals are ever, the peripheral portion of the ANS is composed of two
located in smooth muscle (e.g., blood vessels, gut wall, urinary neurons: preganglionic and postganglionic neurons. The cell
bladder), cardiac muscle, and glands (e.g., sweat glands, salivary bodies of the preganglionic neurons are located in the inter-
glands). Although survival is possible without an ANS, the abil- mediolateral column (IML) of the spinal cord and in motor
ity to adapt to environmental stressors and other challenges is nuclei of some cranial nerves. In contrast to the large-diameter
severely compromised in diseases that affect this component of and rapidly conducting α-motor neurons, preganglionic axons
the nervous system. The ANS has two major divisions: sympa- are small-diameter and relatively slowly conducting B fibers.
thetic and parasympathetic nervous systems. It is classically A preganglionic axon diverges to an average of about nine
defined by the preganglionic and postganglionic neurons within postganglionic neurons, making autonomic output diffuse.
the sympathetic and parasympathetic divisions. A more mod- The axons of the postganglionic neurons are mostly unmyeli-
ern definition of the ANS takes into account the descending nated C fibers and terminate on the visceral effectors.
pathways from several forebrain and brain stem regions as well
as visceral afferent pathways that set the level of activity in sym-
pathetic and parasympathetic nerves. SYMPATHETIC DIVISION
In contrast to α-motor neurons that are located at all spinal
segments, sympathetic preganglionic neurons are located in
ANATOMIC ORGANIZATION the IML of only the first thoracic to the third or fourth lum-
OF AUTONOMIC OUTFLOW bar segments. This is why the sympathetic nervous system is
sometimes called the thoracolumbar division of the ANS.
Figure 19–1 compares some fundamental characteristics of the The axons of sympathetic preganglionic neurons leave the
innervation of skeletal muscle and innervation of smooth mus- spinal cord at the level at which their cell bodies are located
cle, cardiac muscle, and glands. As described in Chapter 14, and exit via the ventral root along with axons of α- and

177

Ch19_177-184.indd 177 11/26/10 9:58:22 AM


178 SECTION IV CNS/Neural Physiology

CNS Somatic nervous system

Effector
organ
ACh

Autonomic nervous system:


CNS Parasympathetic division

Effector
organ

Ganglion ACh

Autonomic nervous system:


CNS Sympathetic division
Effector
organ

ACh Ganglion ACh NE

Effector
(via bloodstream) organ

Adrenal Epi (also NE, DA, peptides)


medulla
FIGURE 19–1 Comparison of peripheral organization and transmitters released by somatomotor and autonomic nervous systems
(NS). ACh, acetylcholine; DA, dopamine; NE, norepinephrine; Epi, epinephrine. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s
Human Physiology. McGraw-Hill, 2008.)

γ-motor neurons (Figure 19–2). They then separate from PARASYMPATHETIC DIVISION
the ventral root via the white rami communicans and proj-
ect to the adjacent sympathetic paravertebral ganglion, The parasympathetic nervous system is sometimes called the
where some of them end on the cell bodies of postganglionic craniosacral division of the ANS because of the location of its
neurons. Paravertebral ganglia are located adjacent to each preganglionic neurons (Figure 19–3). The parasympathetic
thoracic and upper lumbar segment; in addition, there are a nerves supply the visceral structures in the head via the oculo-
few ganglia adjacent to the cervical and sacral spinal seg- motor, facial, and glossopharyngeal nerves, and those in the
ments. Paravertebral ganglia form the sympathetic chain thorax and upper abdomen via the vagus nerves. The sacral
(or sympathetic trunk) bilaterally. The ganglia are connected outflow supplies the pelvic viscera via branches of the second
to each other via the axons of preganglionic neurons that to fourth sacral spinal nerves. Parasympathetic preganglionic
travel rostrally or caudally to terminate on postganglionic fibers synapse on ganglia cells clustered within the walls of vis-
neurons located at some distance. This arrangement is ceral organs; thus, these parasympathetic postganglionic fibers
shown in Figures 19–2 and 19–3. are very short.
Some preganglionic neurons pass through the sympathetic
chain and end on postganglionic neurons located in preverte-
bral (or collateral) ganglia close to the viscera, including the CHEMICAL TRANSMISSION AT
celiac, superior mesenteric, and inferior mesenteric ganglia
(Figure 19–3). There are also preganglionic neurons whose AUTONOMIC JUNCTIONS
axons terminate directly on an effector organ, the medulla of
the adrenal gland. ACETYLCHOLINE AND
The axons of some of the postganglionic neurons leave the NOREPINEPHRINE
chain ganglia and reenter the spinal nerves via the gray rami
communicans and are distributed to autonomic effectors in The principal transmitter agents released by autonomic
the areas supplied by these spinal nerves (Figure 19–2). These nerves are acetylcholine and norepinephrine (Figure 19–1).
postganglionic sympathetic nerves terminate on smooth mus- The neurons that are cholinergic (i.e., release acetylcholine)
cle of blood vessels and hair follicles and on sweat glands in the include (1) all preganglionic neurons, (2) all parasympa-
limbs. Other postganglionic fibers leave the chain ganglia to thetic postganglionic neurons, (3) sympathetic postgangli-
enter the thoracic cavity to terminate in visceral organs. Post- onic neurons that innervate sweat glands, and (4) sympathetic
ganglionic fibers from prevertebral ganglia also terminate in postganglionic neurons that end on blood vessels in some
visceral targets. skeletal muscles and produce vasodilation when stimulated
CHAPTER 19 Autonomic Nervous System 179

FIGURE 19–2 Projection of sympathetic preganglionic and postganglionic fibers. The drawing shows the thoracic spinal cord,
paravertebral, and prevertebral ganglia. Preganglionic neurons are shown in red, postganglionic neurons in dark blue, afferent sensory pathways
in blue, and interneurons in black. (Reproduced with permission from Boron WF, Boulpaep EL: Medical Physiology. Elsevier, 2005.)

(sympathetic vasodilator nerves). The remaining sympa- contain neuropeptide Y. Low-frequency stimulation may pro-
thetic postganglionic neurons are noradrenergic (i.e., release mote release of ATP, whereas high-frequency stimulation may
norepinephrine). The adrenal medulla is essentially a sym- cause release of neuropeptide Y. The viscera contain puriner-
pathetic ganglion in which the postganglionic cells have lost gic receptors, and ATP may be a mediator in the ANS along
their axons and secrete norepinephrine and epinephrine with norepinephrine.
directly into the bloodstream.
Transmission in autonomic ganglia is mediated primarily
by N2 nicotinic cholinergic receptors that are blocked by RESPONSES OF EFFECTOR
hexamethonium, whereas transmission at the neuromuscu-
lar junction is via N1 nicotinic cholinergic receptors that are ORGANS TO AUTONOMIC NERVE
blocked by D-tubocurare. The release of acetylcholine from IMPULSES
postganglionic fibers acts on muscarinic receptors, which
are blocked by atropine. The release of norepinephrine from The effects of stimulation of the noradrenergic and cholin-
sympathetic postganglionic fibers acts on α1-, α2-, β1-, or ergic postganglionic nerve fibers are indicated in Figure
β2-adrenoreceptors, depending on the target organ. 19–3 and Table 19–1. Release of acetylcholine onto smooth
Table 19–1 shows the types of receptors at various junctions muscle of some organs leads to contraction (e.g., walls of the
within the ANS. gastrointestinal tract), and release onto other organs leads
In addition to these “classical” neurotransmitters, some to relaxation (e.g., sphincters in the gastrointestinal tract).
autonomic fibers also release neuropeptides. The small granu- For some targets innervated by the ANS, one can shift from
lated vesicles in postganglionic noradrenergic neurons contain contraction to relaxation by switching from activation of
ATP and norepinephrine, and the large granulated vesicles the parasympathetic nervous system to activation of the
180 SECTION IV CNS/Neural Physiology

FIGURE 19–3 Organization of sympathetic (left) and parasympathetic (right) nervous systems. Preganglionic sympathetic and
parasympathetic neurons are shown in red and orange, respectively; postganglionic sympathetic and parasympathetic neurons in blue and
green, respectively. (Reproduced with permission from Boron WF, Boulpaep EL: Medical Physiology. Elsevier, 2005.)
CHAPTER 19 Autonomic Nervous System 181

TABLE 19–1 Responses of some effector organs to autonomic nerve activity.


Sympathetic Nervous System

Effector Organs Parasympathetic Nervous System Receptor Type Response

Eyes

Radial muscle of iris — a


α1 Contraction (mydriasis)

Sphincter muscle of iris Contraction (miosis) —

Ciliary muscle Contraction for near vision —

Heart

SA node Decreases heart rate β1 Increases heart rate

Atria and ventricle Decreases contractility β 1 , β2 Increases contractility

AV node and Purkinje Decreases conduction velocity β 1 , β2 Increases conduction velocity

Arterioles

Coronary — α1, α2 Constriction

β2 Dilation

Skin — α1, α2 Constriction

Skeletal muscle — α1 Constriction

β2, Muscarinic Dilation

Abdominal viscera — α1 Constriction

Salivary glands Dilation α1, α2 Constriction

Renal — α1 Constriction

Systemic veins — α1, α2 Constriction

β2 Dilation

Lungs

Bronchial muscle Contraction β2 Relaxation

Bronchial glands Stimulation α1 Inhibition

β2 Stimulation

Stomach

Motility and tone Increases α1, α2, β2 Decreases

Sphincters Relaxation α1 Contraction

Secretion Stimulation Unknown Inhibition

Intestine

Motility and tone Increases α1, α2, β1, β2 Decreases

Sphincters Relaxation α1 Contraction (usually)

Secretion Stimulation α2 Inhibition

Gall bladder Contraction β2 Relaxation

Urinary bladder

Detrusor Contraction β2 Relaxation

Sphincter Relaxation α1 Contraction

(Continued)
182 SECTION IV CNS/Neural Physiology

TABLE 19–1 Responses of some effector organs to autonomic nerve activity. (Continued)
Sympathetic Nervous System

Effector Organs Parasympathetic Nervous System Receptor Type Response

Uterus Variable α1 Contraction (pregnant)

β2 Relaxation

Male sex organs Erection α1 Ejaculation

Skin

Pilomotor muscles — α1 Contraction

Sweat glands α1 Slight, localized secretionb

Generalized abundant, dilute


Muscarinic secretion

Liver — α1, β2 Glycogenolysis

Pancreas

Exocrine glands Increases secretion α Decreases secretion

Endocrine glands — α2 Inhibits secretion

Salivary glands Profuse, watery secretion α1 Thick, viscous secretion

β Amylase secretion

Lacrimal glands Secretion —

Adipose tissue — α2, β3 Lipolysis


a
A dash means these cells are not innervated by this division of the autonomic nervous system.
b
On palms of hands and in some other locations (“adrenergic sweating”).
Modified with permission from Hardman JG, Limbird LE, Gilman AG [editors]: Goodman and Gilman’s The Pharmacological Basis of Therapeutics, 10th ed. McGraw-Hill, 2001.

sympathetic nervous system. This is the case for the many There are also several organs that are innervated by only one
organs that receive dual innervation with antagonistic division of the ANS. The adrenal medulla, most blood vessels,
effects, including the digestive tract, airways, and urinary the pilomotor muscles in the skin (hair follicles), and sweat
bladder. The heart is another example of an organ with glands are innervated exclusively by sympathetic nerves. The
dual antagonistic control. Stimulation of sympathetic lacrimal muscle (tear gland), ciliary muscle (for accommoda-
nerves increases heart rate, and stimulation of parasympa- tion for near vision), and the sublingual salivary gland are
thetic nerves decreases heart rate. innervated exclusively by parasympathetic nerves.
In other cases, the effects of sympathetic and parasympa- The parasympathetic nervous system is largely concerned
thetic activation are complementary. An example is the inner- with the vegetative aspects of day-to-day living and is some-
vation of salivary glands. Parasympathetic activation causes times called the anabolic nervous system. For example,
release of watery saliva, while sympathetic activation causes parasympathetic action favors digestion and absorption of
the production of thick, viscous saliva. food by increasing the activity of the intestinal muscula-
The two divisions of the ANS act in a synergistic or coop- ture, increasing gastric secretion, and relaxing the pyloric
erative manner in the control of some functions. One exam- sphincter.
ple is the control of pupil diameter in the eye. Both The sympathetic nervous system can prepare an individual
sympathetic and parasympathetic innervations are excit- to cope with an emergency and can be called the catabolic ner-
atory, but the former contracts the radial muscle to dilate the vous system. Sympathetic activity dilates the pupils (letting
pupil (mydriasis) and the latter contracts the sphincter mus- more light into the eyes), accelerates the heartbeat and raises
cle to constrict the pupil (miosis). Another example is the blood pressure (providing better perfusion of the vital organs
synergistic actions of these nerves on sexual function. and muscles), and constricts blood vessels in the skin (which
Activation of parasympathetic nerves to the penis increases limits bleeding from wounds). Sympathetic discharge also
blood flow and leads to erection while activation of sympa- leads to elevated plasma glucose and free fatty acid levels (sup-
thetic nerves to the male genitalia causes ejaculation. plying more energy).
CHAPTER 19 Autonomic Nervous System 183

REFLEX AND CENTRAL CONTROL absence of an ANS, it is difficult to regulate body tem-
OF AUTONOMIC ACTIVITY perature, fluid and electrolyte balance, and blood pres-
sure. MSA can also present with cerebellar, basal ganglia,
As is the case for α-motor neurons, the activity of autonomic locus ceruleus, inferior olivary nucleus, and pyramidal
nerves depends on reflexes (e.g., baroreceptor and chemore- tract deficits. It is defined as a sporadic, progressive, adult-
ceptor reflexes) and on descending excitatory and inhibitory onset disorder characterized by autonomic dysfunction,
inputs from several brain regions. For example, pregangli- parkinsonism, and cerebellar ataxia in any combination.
onic sympathetic neurons in the IML receive excitatory input The pathological hallmark of MSA is cytoplasmic and
from the rostral ventrolateral medulla and the paraventric- nuclear inclusions in oligodendrocytes and neurons in
ular nucleus of the hypothalamus and inhibitory input from central motor and autonomic areas. There is also depletion
medullary raphe neurons. In addition to these direct path- of monoaminergic, cholinergic, and peptidergic markers in
ways to the IML, there are many brain regions that feed into several brain regions and in the cerebrospinal fluid. Basal
these pathways to regulate autonomic nerve activity. These levels of sympathetic activity and plasma norepinephrine
include the caudal ventrolateral medulla, nucleus of the trac- levels are normal in MSA patients, but they fail to increase
tus solitarius, and locus ceruleus. This is analogous to the in response to standing up or other stimuli and lead to
control of somatomotor function by areas such as the basal severe orthostatic hypotension (low blood pressure upon
ganglia and cerebellum. Chapters 29 and 30 describe reflex standing). In addition to the decrease in blood pressure,
and central mechanisms that regulate autonomic nerves as orthostatic hypotension can lead to dizziness, dimness of
related to the control of the cardiovascular system in health vision, and fainting because of insufficient perfusion of
and disease. the brain. MSA is also accompanied by parasympathetic
The hypothalamus is often regarded as a major central dysfunction, including urinary and sexual dysfunction.
autonomic control area. Indeed, many of the complex auto- MSA is most often diagnosed in individuals between 50
nomic mechanisms that maintain homeostasis are integrated and 70 years of age; it affects more men than women.
in the hypothalamus. The hypothalamus also functions with Erectile dysfunction is often the first symptom of the dis-
the limbic system as a unit that regulates emotional and ease. There are also abnormalities in baroreceptor reflex
instinctual behavior. It interconnects with nuclei in the mid- and respiratory control mechanisms. Although auto-
brain, pons, and medulla to regulate autonomic activity. The nomic abnormalities are often the first symptoms, 75% of
autonomic responses triggered by activation of the hypothala- patients with MSA also experience motor disturbances.
mus are part of complex phenomena such as eating, emotions There is no cure for MSA. Treatment is directed toward
such as rage, and responses to stress. assuring patient comfort and maintaining bodily func-
tions as long as possible.

CLINICAL CORRELATION CHAPTER SUMMARY


A 67-year-old retired biology professor suddenly col- ■ Preganglionic sympathetic neurons are located in the IML of
lapsed at his home when he stood up from a reclining the thoracolumbar spinal cord and project to postganglionic
position. His wife called for an ambulance and reported neurons in the paravertebral or prevertebral ganglia or the
he was pale but never lost consciousness. He was taken to adrenal medulla. Preganglionic parasympathetic neurons are
located in motor nuclei of cranial nerves III, VII, IX, and X and
a local hospital. His blood pressure was normal in a reclin-
the sacral IML.
ing position but decreased to hypotensive levels when he
■ Nerve terminals of postganglionic neurons are located in
stood. On questioning, he reported that he began to expe-
smooth muscle (e.g., blood vessels, gut wall, urinary
rience erectile dysfunction about 6 months ago. He also bladder), cardiac muscle, and glands (e.g., sweat gland,
has noticed difficulty adapting to altered environmental salivary glands).
temperatures. Episodes of urinary incontinence have also ■ Acetylcholine is released at nerve terminals of preganglionic
been experienced. The physician detected a mild degree neurons, postganglionic parasympathetic neurons, and a few
of ataxia (staggering gait) and tremor. postganglionic sympathetic neurons (sweat glands, sympathetic
After a series of neurological exams, he was diagnosed vasodilator fibers). The remaining sympathetic postganglionic
with Shy–Drager syndrome that is a subtype of multiple neurons release norepinephrine.
system atrophy (MSA) in which autonomic failure domi- ■ Sympathetic activity can prepare the individual to cope with an
nates. MSA is a neurodegenerative disorder associated emergency by accelerating the heartbeat, raising blood pressure
with autonomic failure due to loss of preganglionic auto- (perfusion of the vital organs), and constricting the blood
nomic neurons in the spinal cord and brain stem. In the vessels of the skin (limits bleeding from wounds). Parasympa-
thetic activity is concerned with the vegetative aspects of
184 SECTION IV CNS/Neural Physiology

day-to-day living and favors digestion and absorption of food 3. Parasympathetic nerve activity
by increasing the activity of the intestinal musculature, A) relaxes most vascular smooth muscle.
increasing gastric secretion, and relaxing the pyloric sphincter. B) affects only smooth muscles and glands.
■ The activity of autonomic nerves is dependent on both reflexes C) causes contraction of the radial muscle of the eye to allow
(e.g., baroreceptor and chemoreceptor reflexes) and descending accommodation for near vision.
excitatory and inhibitory inputs from several brain regions D) contracts smooth muscle of the gastrointestinal wall and
including the hypothalamus and brain stem. relaxes the gastrointestinal sphincter.
E) all of the above
4. Which of the following are correctly paired?
STUDY QUESTIONS A) sinoatrial (SA) node:nicotinic cholinergic receptors
B) autonomic ganglia:muscarinic cholinergic receptors
1. Administration of a β-adrenergic receptor antagonist would
C) pilomotor smooth muscle:β2-adrenergic receptors
not be expected to
D) vasculature of some skeletal muscles:muscarinic cholinergic
A) decrease the conduction velocity in the Purkinje fibers receptors
of the heart. E) sweat glands:α2-adrenergic receptors
B) decrease the heart rate.
C) decrease the force of cardiac contraction.
D) relax the detrusor muscle of the bladder.
E) contract bronchial smooth muscle.
2. Sympathetic nerve activity
A) is essential for survival.
B) causes contraction of some smooth muscles and relaxation
of others.
C) causes relaxation of the radial muscle of the eye to dilate
the pupil.
D) relaxes smooth muscle of the gastrointestinal wall and
gastrointestinal sphincter.
E) all of the above
20
C H A P T E R

Electrical Activity of the


Brain, Sleep–Wake States,
and Circadian Rhythms
Susan M. Barman

O B J E C T I V E S

■ List the main clinical uses of the electroencephalogram.


■ Describe the primary types of brain rhythms.
■ Summarize the behavioral and electrophysiological characteristics of each
of the stages of nonrapid eye movement and rapid eye movement sleep.
■ Describe the pattern of normal nighttime sleep in adults and the variations
in this pattern from birth to old age.
■ Discuss the circadian rhythm and the role of the suprachiasmatic nuclei
in its regulation.
■ Describe the diurnal regulation of synthesis of melatonin from serotonin
in the pineal gland and its secretion into the bloodstream.

INTRODUCTION malities are often difficult to demonstrate. Seizures are divided


into partial (focal) seizures and generalized seizures.
The spectrum of behavioral states ranges from deep sleep to Partial seizures originate in a small group of neurons and
light sleep, rapid eye movement (REM) sleep, and the two can result from head injury, brain infection, stroke, or tumor.
awake states: relaxed awareness and awareness with concen- Symptoms depend on the seizure focus and are subdivided
trated attention. Specific patterns of brain electrical activity into simple (without loss of consciousness) and complex par-
correlate with each of these states. Some disease processes lead tial seizures (with altered consciousness). An example of a
to changes in these activity patterns. Arousal can be produced partial seizure is localized jerking movements in one hand
by sensory stimulation and by impulses ascending in the retic- progressing to clonic movements of the entire arm. Auras typ-
ular core of the midbrain. Many of these activities have rhyth- ically precede the onset of a partial seizure and include abnor-
mic fluctuations that are approximately 24 hours in length mal sensations. The time after the seizure until normal
(circadian rhythm). neurological function returns is called the postictal period.
Generalized seizures are associated with widespread electri-
cal activity and involve both hemispheres simultaneously and
ELECTROENCEPHALOGRAM are subdivided into convulsive and nonconvulsive categories
depending on whether tonic or clonic movements occur.
EPILEPSY Absence seizures (formerly called petit mal seizures) are one of
the forms of nonconvulsive generalized seizures characterized
The electroencephalogram (EEG) recorded from the scalp is by a momentary loss of consciousness.
a measure of the summation of dendritic postsynaptic poten- The most common convulsive generalized seizure is tonic–
tials in underlying cortical neurons. The EEG is of value in clonic seizure (formerly called grand mal seizure). This is
localizing pathologic processes in the brain. Epilepsy is a syn- associated with sudden onset of contraction of limb muscles
drome with multiple causes and is characterized by both (tonic phase) lasting about 30 seconds, followed by a phase
behavioral and EEG changes. In some forms, characteristic with symmetric jerking of the limbs as a result of alternating
EEG patterns occur during seizures; between attacks, abnor- contraction and relaxation (clonic phase) lasting 1–2 minutes.

185

Ch20_185-190.indd 185 11/26/10 10:05:03 AM


186 SECTION IV CNS/Neural Physiology

beta rhythm (Figure 20–1). This phenomenon is called alpha


(a) Alpha rhythm (relaxed with eyes closed)
block (or arousal or alerting response) and can be pro-
duced by any form of sensory stimulation or mental concen-
tration (e.g., solving arithmetic problems). It has also been
called desynchronization, because it represents breaking up
of the highly synchronized neural activity. However, the rapid
EEG activity seen in the alert state is also synchronized, but at
(b) Beta rhythm (alert) a higher rate. Therefore, the term desynchronization is mis-
leading. Gamma oscillations at 30–80 Hz are often observed
when an individual is aroused and focuses attention on some-
thing. This can be replaced by irregular fast activity when the
Time
individual initiates motor activity in response to the stimulus.

FIGURE 20–1 EEG records showing the alpha and beta


rhythms. a and b) When attention is focused on something, the SLEEP STAGES
8–13-Hz alpha rhythm is replaced by an irregular 13–30-Hz low-voltage
activity, the beta rhythm. (Reproduced with permission from Widmaier EP, Raff H, There are two kinds of sleep: REM sleep and non-REM (NREM)
Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.) or slow-wave sleep. REM sleep is so named because of the char-
acteristic eye movements that occur during this stage of sleep.
NREM sleep is divided into four stages (Figure 20–2). A person
There is fast EEG activity during the tonic phase. Slow waves, falling asleep enters stage 1 sleep, and the EEG shows a low-
each preceded by a spike, occur at the time of each clonic jerk. voltage, mixed frequency pattern. A theta rhythm (4–7 Hz) can
For a while after the attack, slow waves are present. be seen at this early stage of slow-wave sleep. Throughout
NREM sleep, there is some activity of skeletal muscle but no
eye movements. Stage 2 is marked by the appearance of sleep
spindles (12–14 Hz) and occasional high-voltage biphasic
BRAIN RHYTHMS IN K complexes. In stage 3, a high-amplitude delta rhythm
WAKEFULNESS AND SLEEP (0.5–4 Hz) dominates the EEG waves. Maximum slowing with
large waves is seen in stage 4. Thus, the characteristic of deep
In adult humans who are awake but at rest with the mind wan- sleep is a pattern of rhythmic slow waves, indicating marked
dering and the eyes closed, the most prominent component of synchronization, and it is referred to as slow-wave sleep.
the EEG is a fairly regular pattern of waves at a frequency of Whereas theta and delta rhythms are normal during sleep, their
8–13 Hz and amplitude of 50–100 μV when recorded from the appearance during wakefulness is a sign of brain dysfunction.
scalp. This pattern is the alpha rhythm (Figure 20–1). It is The high-amplitude slow waves seen in the EEG during
most marked in the parietal and occipital lobes and is associ- sleep are periodically replaced by rapid, low-voltage EEG
ated with decreased levels of attention. activity, which resembles that seen in the awake, aroused state
When attention is focused on something, the alpha rhythm (Figure 20–2). However, sleep is not interrupted; indeed, the
is replaced by an irregular 13–30-Hz low-voltage activity, the threshold for arousal by sensory stimuli is elevated. Rapid,

Awake Sleep stage 1 2 3 4 REM

EOG

EMG

EEG
50 V
1s

FIGURE 20–2 EEG and muscle activity during various stages of the sleep–wake cycle. NREM sleep has four stages. Stage 1 is
characterized by a slight slowing of the EEG. Stage 2 has high-amplitude K complexes and spindles. Stages 3 and 4 have slow, high-amplitude
delta waves. REM sleep is characterized by eye movements, loss of muscle tone, and a low-amplitude, high-frequency activity pattern. The higher
voltage activity in the EOG tracings during stages 2 and 3 reflects high-amplitude EEG activity in the prefrontal areas rather than eye movements.
EOG, electrooculogram registering eye movements; EMG, electromyogram registering skeletal muscle activity. (Reproduced with permission from
Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
CHAPTER 20 Electrical Activity of the Brain, Sleep–Wake States, and Circadian Rhythms 187

roving movements of the eyes occur during this sleep stage, Children
Awake
accounting for its name. Another characteristic of REM sleep
is the occurrence of large phasic potentials that originate in the REM

Sleep stages
cholinergic neurons in the pons and pass rapidly to the lateral 1
geniculate body and from there to the occipital cortex. They 2
are called pontogeniculooccipital (PGO) spikes. The tone of 3
the skeletal muscles in the neck is markedly reduced during 4
REM sleep.
Humans aroused during REM sleep often report that they 1 2 3 4 5 6 7
were dreaming, whereas individuals awakened from slow-
wave sleep do not. Thus, REM sleep and dreaming are closely Awake Young Adults
associated. Positron emission tomography (PET) scans
REM
of humans in REM sleep show increased activity in the pon-

Sleep stages
1
tine area, amygdala, and anterior cingulate gyrus, but decreased
2
activity in the prefrontal and parietal cortex. Activity in visual
association areas is increased, but there is a decrease in the 3
primary visual cortex. This is consistent with increased emo- 4
tion and operation of a closed neural system cut off from the
areas that relate brain activity to the external world. 1 2 3 4 5 6 7

Elderly
DISTRIBUTION OF SLEEP STAGES Awake
REM
Sleep stages

In a typical night of sleep, a young adult first enters NREM sleep, 1


passes through stages 1 and 2, and spends 70–100 minutes in 2
stages 3 and 4. Sleep then lightens, and a REM period follows. 3
This cycle is repeated at intervals of about 90 minutes through- 4
out the night (Figure 20–3). The cycles are similar, though there
is less stage 3 and 4 sleep and more REM sleep toward morning. 1 2 3 4 5 6 7
Four to six REM periods occur per night. Hours of sleep
If humans are awakened every time they show REM sleep
and then permitted to sleep without interruption, they show a FIGURE 20–3 Normal sleep cycles at various ages. REM sleep
great deal more than the normal amount of REM sleep for a is indicated by the darker-colored areas. (Reproduced with permission from
few subsequent nights. Various studies imply that sleep is Kales AM, Kales JD. Sleep disorders. Recent findings in the diagnosis and treatment
needed to maintain metabolic–caloric balance, thermal equi- of disturbed sleep. N Engl J Med. 1974; 290:487.)

librium, and immune competence.


Sleepwalking (somnambulism), bed-wetting (nocturnal
SLEEP DISORDERS enuresis), and night terrors are referred to as parasomnias,
which are sleep disorders associated with arousal from NREM
Narcolepsy is a chronic neurological disorder caused by the and REM sleep. Episodes of sleepwalking are more common
brain’s inability to regulate sleep–wake cycles normally. There in children than in adults, occur predominantly in males, and
is a sudden loss of voluntary muscle tone (cataplexy), an may last several minutes. Somnambulists walk with their eyes
eventual irresistible urge to sleep during daytime, and some- open and avoid obstacles, but when awakened, they cannot
times brief episodes of total paralysis at the beginning or end recall the episodes.
of sleep. Narcolepsy is characterized by a sudden onset of
REM sleep, unlike normal sleep that begins with NREM,
slow-wave sleep. Brains from humans with narcolepsy often CIRCADIAN RHYTHMS AND
contain fewer hypocretin (orexin)-producing neurons in the
hypothalamus. An immune attack on these neurons can lead
THE SLEEP–WAKE CYCLE
to their degeneration. Obstructive sleep apnea (OSA) is a Most, if not all, living cells in plants and animals have rhyth-
common sleep disorder that involves periodic cessation of air mic fluctuations in their function on a circadian cycle.
flow during sleep and causes abnormal sleep architecture and Normally, they become entrained (synchronized) to the day–
daytime hypersomnolence. Central sleep apnea is a rare night light cycle in the environment. If they are not entrained,
sleep disorder that occurs when the brain temporarily stops the cycles become progressively more out of phase with the
sending signals to the phrenic motor neurons controlling the light–dark cycle because they are longer or shorter than
diaphragm. 24 hours. The entrainment process in most cases is dependent
188 SECTION IV CNS/Neural Physiology

Melatonin
H H H O
CH3O C C N C CH3
N H H
H

Pineal
gland

Day

Inhibition

Sympathetic
neurons
Night

Retinohypothalamic
Stimulation tract
Superior cervical
Suprachiasmatic nucleus
ganglion
(the "biological clock")

FIGURE 20–4 Secretion of melatonin. Retinohypothalamic fibers synapse in the suprachiasmatic nuclei (SCN), and there are connections
from the SCN to sympathetic preganglionic neurons in the spinal cord that project to the superior cervical ganglion. Postganglionic neurons
project from this ganglion to the pineal gland that secretes melatonin. The cyclic activity of SCN sets up a circadian rhythm for melatonin release.
This rhythm is entrained to light/dark cycles by neurons in the retina. (Reproduced with permission from Fox SI: Human Physiology. McGraw-Hill, 2008.)

on the suprachiasmatic nuclei (SCN) that are located above system (RAS) is composed of several groups of neurons that
the optic chiasm (Figure 20–4). The SCN receives information release norepinephrine, serotonin, or acetylcholine. In the
about the light–dark cycle via a special neural pathway, the case of the forebrain neurons involved in control of the
retinohypothalamic tract. Efferents from the SCN initiate sleep–wake cycles, preoptic neurons in the hypothalamus
neural and humoral signals that entrain a wide variety of cir- release GABA and posterior hypothalamic neurons release
cadian rhythms including the sleep–wake cycle and the secre- histamine.
tion of the pineal hormone melatonin. One theory regarding the basis for transitions from sleep to
The exposure to bright light can advance, delay, or have no wakefulness involves alternating reciprocal activity of differ-
effect on the sleep–wake cycle depending on the time of day ent groups of RAS neurons. In this model (Figure 20–5), wake-
when it is experienced. During the usual daytime it has no fulness and REM sleep are at opposite extremes. When the
effect, but just after dark it delays the onset of the sleep period, activity of norepinephrine- and serotonin-containing neurons
and just before dawn it accelerates the onset of the next sleep (locus ceruleus and raphe nuclei) is dominant, there is a
period. Injections of melatonin have similar effects. reduced level of activity in acetylcholine-containing neurons
in the pontine reticular formation. This pattern of activity
contributes to the appearance of the awake state. The reverse
NEUROCHEMICAL MECHANISMS of this pattern leads to REM sleep. When there is a more even
PROMOTING SLEEP AND AROUSAL balance in the activity of the aminergic and cholinergic neu-
rons, NREM sleep occurs.
Transitions between sleep and wakefulness manifest a circa- In addition, an increased release of GABA and reduced
dian rhythm consisting of an average of 8 hours of sleep and release of histamine increase the likelihood of NREM sleep via
16 hours of wakefulness. Nuclei in both the brain stem and deactivation of the thalamus and cortex. Wakefulness occurs
hypothalamus are critical for the transitions between these when GABA release is reduced and histamine release is
states of consciousness. The brain stem reticular activating increased.
CHAPTER 20 Electrical Activity of the Brain, Sleep–Wake States, and Circadian Rhythms 189

Brainstem nuclei that


melatonin secretion may function as a timing signal to coordi-
are part of the reticular nate events with the light–dark cycle in the environment.
activating system Melatonin synthesis and secretion are increased during the
dark period of the day and maintained at a low level dur-
ing daylight hours. This diurnal variation in secretion is
Norepinephrine Norepinephrine brought about by norepinephrine secreted by the postgangli-
and and
serotonin
onic sympathetic nerves that innervate the pineal gland
serotonin
(Figure 20–4). Norepinephrine acts via β-adrenergic receptors
to increase intracellular cAMP, and the cAMP in turn pro-
Acetylcholine Acetylcholine duces a marked increase in N-acetyltransferase activity. This
results in increased melatonin synthesis and secretion.
Circulating melatonin is rapidly metabolized in the liver by
6-hydroxylation followed by conjugation, and over 90% of the
Waking NREM sleep REM sleep
melatonin that appears in the urine is in the form of 6-hydroxy
conjugates and 6-sulfatoxymelatonin. The mechanism by
which the brain metabolizes melatonin may involve cleavage
Activation Activation of the indole nucleus.
of the thalamus of the thalamus The discharge of the sympathetic nerves to the pineal is
and cortex and cortex
entrained to the light–dark cycle in the environment via the
retinohypothalamic nerve fibers to the SCN. From the hypo-
Histamine Histamine thalamus, descending pathways converge onto preganglionic
sympathetic neurons that in turn innervate the superior cervi-
cal ganglion, the site of origin of the postganglionic neurons to
GABA GABA the pineal gland.

Hypothalamus with
circadian and
CLINICAL CORRELATION
homeostatic centers
A 47-year-old male research scientist at a biotechnology
FIGURE 20–5 A model of how alternating activity of brain institute has always been liked for his positive attitude and
stem and hypothalamic neurons may influence the different high energy level. Recently, his colleagues have noticed
states of consciousness. (Reproduced with permission from Widmaier EP, Raff that he becomes agitated if things do not go well in the
H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.) laboratory, and he appears to be tired all of the time. His
wife has also been concerned about his being more
grumpy than normal. She accompanies him to his physi-
cian’s office for his annual physical. When the doctor
MELATONIN AND THE questions him about his sleep pattern, he insists there has
SLEEP–WAKE STATE been no change. But his wife mentions that he has been
waking up suddenly at night and making very loud noises,
Melatonin release from the richly vascularized pineal gland sounding somewhat like gasping. He is referred to a sleep
also plays a role in sleep mechanisms (Figure 20–4). The clinic where he undergoes a polysomnogram (PSG) that
pineal arises from the roof of the third ventricle in the dien- records brain activity, eye movements, body movements,
cephalon and is encapsulated by the meninges. The pineal breathing and heart rate, and blood oxygen saturation
stroma contains glia and pinealocytes with features suggest- over the course of a sleep cycle.
ing that they have a secretory function. Like other endocrine The results of PSG confirmed the doctor’s suspicion
glands, it has highly permeable fenestrated capillaries. In that the scientist has obstructive sleep apnea OSA. OSA is
infants, the pineal is large and the cells tend to be arranged in the most common cause of daytime sleepiness (hyper-
alveoli. It begins to involute before puberty and small concre- somnolence) due to fragmented sleep at night and affects
tions of calcium phosphate and carbonate (pineal sand) about 24% of middle-aged men and 9% of women in the
appear in the tissue. Because the concretions are radiopaque, United States. Breathing ceases for more than 10 seconds
the pineal is often visible on x-ray films of the skull in adults. during frequent episodes of obstruction of the upper air-
Displacement of a calcified pineal from its normal position way (especially the pharynx) due to reduction in upper
indicates the presence of a space-occupying lesion such as a airway muscle tone despite continued activity of the
tumor in the brain. inspiratory muscles. The apnea causes brief arousals from
Melatonin is synthesized by pinealocytes and secreted into sleep in order to reestablish upper airway tone. Snoring is
the blood and the cerebrospinal fluid. The diurnal change in
190 SECTION IV CNS/Neural Physiology

2. Which of the following pattern of changes in central


a common patient complaint, and OSA is often associated neurotransmitters or neuromodulators is associated
with obesity. There is actually not a reduction in total sleep with the transition from NREM to wakefulness?
time, but individuals with OSA experience a much greater A) decrease in norepinephrine, increase in serotonin,
time in stage 1 NREM sleep (from an average of 10% of increase in acetylcholine, decrease in histamine, and
total sleep to 30–50%) and a marked reduction in slow- decrease in GABA
wave sleep (stage 3 and 4 NREM sleep). The pathophysiol- B) decrease in norepinephrine, increase in serotonin,
ogy of OSA includes both a reduction in neuromuscular increase in acetylcholine, decrease in histamine, and
tone at the onset of sleep and a change in the central respi- increase in GABA
C) decrease in norepinephrine, decrease in serotonin,
ratory drive. A common treatment of OSA is the use of a
increase in acetylcholine, increase in histamine, and
continuous positive airway pressure (CPAP) mask during increase in GABA
sleep to prevent airway occlusions. D) increase in norepinephrine, increase in serotonin,
decrease in acetylcholine, increase in histamine, and
decrease in GABA
E) increase in norepinephrine, decrease in serotonin,
CHAPTER SUMMARY decrease in acetylcholine, increase in histamine, and
decrease in GABA
■ The EEG is of some value in localizing pathologic processes,
and it is useful in characterizing different types of epilepsy. 3. A gamma rhythm (30–80 Hz)
■ The major rhythms in the EEG are alpha (8–13 Hz), beta A) is characteristic of seizure activity.
(13–30 Hz), theta (4–7 Hz), delta (0.5–4 Hz), and gamma B) is seen in an individual who is awake but not focused.
(30–80 Hz) oscillations. C) may be a mechanism to bind together sensory information
into a single percept and action.
■ Throughout NREM sleep, there is some activity of skeletal
D) is also called an alerting response.
muscle. A theta rhythm can be seen during stage 1 of sleep.
E) is characteristic of obstructive sleep apnea.
Stage 2 is marked by the appearance of sleep spindles and
occasional K complexes. In stage 3, a delta rhythm is dominant. 4. Melatonin secretion would probably not be increased by
Maximum slowing with large slow waves is seen in stage 4. A) stimulation of the superior cervical ganglia.
■ REM sleep is characterized by low-voltage, high-frequency B) a reduction in sunlight.
EEG activity and rapid, roving movements of the eyes. C) intravenous infusion of norepinephrine.
D) stimulation of the optic nerve.
■ A young adult typically passes through stages 1 and 2, and
E) an increase in N-acetyltransferase activity.
spends 70–100 minutes in stages 3 and 4. Sleep then lightens,
and a REM period follows. This cycle repeats at 90-minute 5. Absence seizures are a form of
intervals throughout the night. A) nonconvulsive generalized seizures accompanied by
■ Transitions from sleep to wakefulness may involve alternating momentary loss of consciousness.
reciprocal activity of different groups of RAS neurons. When B) complex partial seizures accompanied by momentary
the activity of norepinephrine- and serotonin-containing loss of consciousness.
neurons is dominant, the activity in acetylcholine-containing C) nonconvulsive generalized seizures without loss of
neurons is reduced, leading to the appearance of wakefulness. consciousness.
The reverse of this pattern leads to REM sleep. Also, wakeful- D) simple partial seizures without loss of consciousness.
ness occurs when GABA release is reduced and histamine E) convulsive generalized seizures accompanied by momentary
release is increased. loss of consciousness.
■ The entrainment of biological processes to the light–dark cycle 6. Narcolepsy is triggered by abnormalities in the
is regulated by the SCN. A) skeletal muscles.
■ The diurnal change in melatonin secretion from serotonin in B) medulla oblongata.
the pineal gland may function as a timing signal to coordinate C) hypothalamus.
events with the light–dark cycle, including the sleep–wake cycle. D) olfactory bulb.
E) neocortex.

STUDY QUESTIONS
1. In a healthy, alert adult sitting with the eyes closed, the dominant
EEG rhythm observed with electrodes over the occipital lobes is
A) delta (0.5–4 Hz).
B) theta (4–7 Hz).
C) alpha (8–13 Hz).
D) beta (18–30 Hz).
E) fast, irregular low-voltage activity.
21
C H A P T E R

Learning, Memory,
Language, and Speech
Susan M. Barman

O B J E C T I V E S

■ Describe the various types of long-term memory.


■ Define synaptic plasticity, long-term potentiation, long-term depression,
habituation, and sensitization, and their roles in learning and memory.
■ List the parts of the brain that are involved in memory and their role in
memory processing and storage.
■ Describe the abnormalities of brain structure and function found in
Alzheimer disease.
■ Define the terms categorical hemisphere and representational hemisphere.
■ Explain the differences between fluent and nonfluent aphasia.

INTRODUCTION From a physiologic point of view, memory is divided into


explicit and implicit forms (Figure 21–2). Explicit or declara-
A revolution in our understanding of brain function has been tive memory is associated with consciousness (or at least
brought about by the development and widespread availabil- awareness) and is dependent on the hippocampus and other
ity of positron emission tomography (PET), functional parts of the medial temporal lobes of the brain for its reten-
magnetic resonance imaging (fMRI), and related tech- tion. Implicit or nondeclarative memory does not involve
niques. PET is often used to measure local glucose metabo- awareness, and its retention does not usually involve process-
lism, which is proportionate to neural activity, and fMRI is ing in the hippocampus.
used to measure local amounts of oxygenated blood. These
techniques make it possible to determine the activity in vari-
ous parts of the brain in healthy subjects and in those with
various diseases. They have been used to study not only sim-
ple responses, but also complex aspects of learning, memory,
and perception. An example of the use of PET scans to study
the functions of the cerebral cortex in processing words is
shown in Figure 21–1. Different portions of the cortex are
activated when a person is hearing, seeing, speaking, or gen-
erating words.

LEARNING AND MEMORY


Learning is acquisition of information that makes it possi- FIGURE 21–1 Images of active areas of the brain in a male
ble to alter behavior on the basis of experience, and memory (left) and female (right) during a language task. Note that males use
is the retention and storage of that information. The two only one side of the brain whereas females use both sides of the
are obviously closely related and should be considered brain when language is being processed. (Reproduced with permission
together. from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology. McGraw-Hill, 2008.)

191

Ch21_191-198.indd 191 11/26/10 4:42:30 PM


192 SECTION IV CNS/Neural Physiology

Two forms of
long term memory

Explicit Implicit
(declarative) (nondeclarative)

Facts Events Priming Procedural Associative learning: Nonassociative learning:


(Semantic) (Episodic) (skills and classical and habituation and
habits) operant conditioning sensitization

Emotional Skeletal
FIGURE 21–2 Forms of long-term responses musculature
memory. (Modified with permission from Kandel
ER, Schwartz JH, Jessell TM [editors]: Principles of Medial temporal lobe Neocortex Striatum Amygdala Cerebellum Reflex
Neural Science, 4th ed. McGraw-Hill, 2000.) Hippocampus pathways

Explicit memory is divided into episodic memory for events synaptic conduction can be strengthened or weakened on the
and semantic memory for facts (e.g., words, rules, and lan- basis of past experience. These changes, which can be presyn-
guage). Explicit memories initially required for activities such aptic or postsynaptic, are of great interest because they repre-
as riding a bicycle can become implicit once the task is thor- sent forms of learning and memory.
oughly learned. One form of plastic change is post-tetanic potentiation, the
Implicit memory is subdivided into four types. Procedural production of enhanced postsynaptic potentials in response to
memory includes skills and habits, which, once acquired, stimulation. This enhancement lasts up to 60 seconds and
become unconscious and automatic. Priming is facilitation of occurs after a brief (tetanizing) train of stimuli in a presynap-
recognition of words or objects by prior exposure to them. An tic neuron. The stimulation causes Ca2+ to accumulate in
example is improved recall of a word when presented with the the presynaptic neuron to such a degree that the intracellu-
first few letters of it. In nonassociative learning, one learns lar binding sites that keep cytoplasmic Ca2+ low are over-
about a single stimulus. In associative learning, one learns whelmed.
about the relation of one stimulus to another. Habituation is a simple form of learning in which a neutral
Explicit memory and many forms of implicit memory stimulus is repeated many times. The first time it is applied it
involve (1) short-term memory, which lasts seconds to hours, is novel and evokes a reaction (the “what is it?” response);
during which processing in the hippocampus and elsewhere however, it evokes less and less electrical response as it is
leads to long-term changes in synaptic strength; and (2) long- repeated. Eventually, the subject becomes habituated to the
term memory, which stores memories for years. During short- stimulus and ignores it. This is associated with decreased
term memory, the memory traces are subject to disruption by release of neurotransmitter from the presynaptic terminal
trauma and various drugs, whereas long-term memory traces because of decreased intracellular Ca2+. The decrease in intra-
are remarkably resistant to disruption. Working memory is a cellular Ca2+ is due to a gradual inactivation of Ca2+ channels.
form of short-term memory that keeps information available, It can be short-term, or it can be prolonged if exposure to the
usually for very short periods, while the individual plans action benign stimulus is repeated many times. Habituation is a clas-
based on it. sic example of nonassociative learning.
Sensitization is the prolonged occurrence of augmented
SYNAPTIC PLASTICITY AND LEARNING postsynaptic responses after a stimulus to which one has
become habituated is paired once or several times with a nox-
The key to memory is alteration in the strength of selected ious stimulus. It is due to presynaptic facilitation and may
synaptic connections. In all but the simplest of cases, the alter- occur as a transient response. If it is reinforced by additional
ation involves activation of genes and protein synthesis. This pairings of the noxious stimulus and the initial stimulus, it can
occurs during the change from short-term working memory exhibit features of short- or long-term memory. The short-
to long-term memory. If an intervention occurs too soon after term prolongation of sensitization is due to a Ca2+-mediated
a training session, acquisition of long-term memory is change in adenylyl cyclase that increases production of cAMP.
impaired. This is exemplified by the loss of memory for the The long-term potentiation (LTP) involves protein synthesis
events immediately preceding brain concussion or electro- and growth of the presynaptic and postsynaptic neurons and
shock therapy (retrograde amnesia). their connections.
Short- and long-term changes in synaptic function can LTP is a rapidly developing persistent enhancement of the
occur as a result of the history of discharge at a synapse, that is, postsynaptic potential response to presynaptic stimulation
CHAPTER 21 Learning, Memory, Language, and Speech 193

after a brief period of rapidly repeated stimulation of the pre- Prefrontal


synaptic neuron. It resembles post-tetanic potentiation but is cortex
much more prolonged and can last for days. Unlike post-tetanic
potentiation, it is initiated by an increase in intracellular Ca2+
in the postsynaptic rather than the presynaptic neuron. It
occurs in many parts of the CNS but has been studied in great-
est detail in the hippocampus where there are two forms: mossy
fiber LTP, which is presynaptic and independent of N-methyl-
d-aspartate (NMDA) receptors, and Schaffer collateral LTP,
which is postsynaptic and NMDA receptor-dependent. The
hypothetical basis of the latter form is summarized in Figure
21–3. The basis of mossy fiber LTP appears to include cAMP
and Ih, a hyperpolarization-activated cation channel.
Long-term depression (LTD) is found throughout the brain Parahippocampal cortex Hippocampus
in the same fibers as LTP. It is characterized by a decrease in FIGURE 21–4 Areas concerned with encoding explicit
synaptic strength. It is produced by slower stimulation of pre- memories. The prefrontal cortex and the parahippocampal cortex
synaptic neurons and is associated with a smaller rise in intra- of the brain are active during the encoding of memories. (Adapted
cellular Ca2+ than occurs in LTP. with permission from Rugg Russ MD. Memories are made of this. Science.
1998;281(5380):1151–1152.)

WORKING MEMORY
Working memory areas are connected to the hippocampus
and the adjacent parahippocampal portions of the medial
temporal cortex (Figure 21–4). Bilateral destruction of
the ventral hippocampus, or Alzheimer disease (described
below) and similar disease processes that destroy its CA1
neurons, causes striking defects in short-term memory.
Individuals with such destruction have intact working mem-
Ca2+ Glu Na+ ory and remote memory. Their implicit memory processes
Mg2+ are generally intact. They perform adequately in terms of
PS conscious memory as long as they concentrate on what they
NMDA AMPA are doing. However, if they are distracted for even a very
P short period, all memory of what they were doing and what
they proposed to do is lost. They are thus capable of new
Ca2+ AMPA learning and retain old prelesion memories, but they cannot
form new long-term memories.
CaM CaM kII
The hippocampus is closely associated with the overlying
parahippocampal cortex in the medial frontal lobe (Figure
21–4). When subjects recall words, activity in their left frontal
lobe and their left parahippocampal cortex increases, but when
they recall pictures or scenes, activity takes place in their right
FIGURE 21–3 Production of long-term potentiation (LTP) frontal lobe and the parahippocampal cortex on both sides.
in Schaffer collaterals in the hippocampus. Glutamate (Glu) The connections of the hippocampus to the diencephalon
released from the presynaptic neuron binds to α-amino-3-hydroxyl-5- are also involved in memory. Some people with alcoholism-
methyl-4-isoxazole-propionate (AMPA) and N-methyl-d-aspartate related brain damage develop impairment of recent memory,
(NMDA) receptors in the membrane of the postsynaptic neuron. The and the memory loss correlates well with the presence of
depolarization triggered by activation of the AMPA receptors relieves pathologic changes in the mamillary bodies, which have
the Mg2+ block in the NMDA receptor channel, and Ca2+ enters the
extensive efferent connections to the hippocampus via the
neuron with Na+. The increase in cytoplasmic Ca2+ activates calmodulin
fornix. The mamillary bodies project to the anterior thala-
(CaM), which in turn activates Ca2+/calmodulin kinase II (CaM kII).
The kinase phosphorylates the AMPA receptors (P), increasing their
mus via the mamillothalamic tract. From the thalamus, the
conductance, and moves more AMPA receptors into the synaptic cell fibers concerned with memory project to the prefrontal cortex
membrane from cytoplasmic storage sites. In addition, a chemical and from there to the basal forebrain. From the basal fore-
signal (PS) may pass to the presynaptic neuron, producing a long-term brain, a diffuse cholinergic projection goes to all of the neo-
increase in the quantal release of glutamate. (Courtesy of R. Nicoll) cortex, the amygdala, and the hippocampus from the nucleus
194 SECTION IV CNS/Neural Physiology

basalis of Meynert. Severe loss of these fibers occurs in


Alzheimer disease.
ALZHEIMER DISEASE AND
The amygdala is closely associated with the hippocampus SENILE DEMENTIA
and is concerned with encoding and recalling emotionally
charged memories. During retrieval of fearful memories, the Alzheimer disease is the most common age-related neurode-
theta rhythms of the amygdala and the hippocampus become generative disorder. Memory decline initially manifests as a
synchronized. In healthy subjects, events associated with loss of episodic memory, which impedes recollection of recent
strong emotions are remembered better than events without events. Loss of short-term memory is followed by general loss
an emotional charge, but in patients with bilateral lesions of of cognitive and other brain functions, the need for constant
the amygdala, this difference is absent. care, and, eventually, death. The cytopathologic hallmarks of
the disease are intracellular neurofibrillary tangles, made up
in part of hyperphosphorylated forms of the tau protein that
LONG-TERM MEMORY normally binds to microtubules, and extracellular senile
plaques, which have a core of β-amyloid peptides (Aβ)
While the encoding process for short-term explicit mem- surrounded by altered nerve fibers and reactive glial cells.
ory involves the hippocampus, long-term memories are Figure 21–5 compares a normal nerve cell to one showing
stored in various parts of the neocortex. Various parts of the abnormalities associated with Alzheimer disease.
memories—visual, olfactory, auditory, etc.—are located in The Aβ peptides are products of a normal protein, amyloid
the cortical regions concerned with these functions, and the precursor protein (APP), a transmembrane protein that proj-
pieces are tied together by long-term changes in the strength ects into the extracellular fluid (ECF) from all nerve cells. This
of transmission at relevant synaptic junctions so that all the protein is hydrolyzed at three different sites by α-, β-, and
components are brought to consciousness when the memory γ-secretase, respectively. When APP is hydrolyzed by α-secretase,
is recalled. nontoxic peptide products are produced. However, when it is
Once long-term memories have been established, they can hydrolyzed by β- and γ-secretase, polypeptides with 40–42
be recalled or accessed by many different associations. For amino acids are produced; the actual length varies because of
example, the memory of a vivid scene can be evoked not variation in the site at which γ-secretase cuts the protein chain.
only by a similar scene, but also by a sound or smell associ- These polypeptides are toxic, the most toxic being Aβσ1–42. The
ated with the scene. Thus, each stored memory must have polypeptides form extracellular aggregates, which can stick to
multiple routes, and many memories have an emotional α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate
component. (AMPA) receptors and Ca2+ ion channels, increasing Ca2+ influx.

A Normal Nerve
terminals

B Alzheimer disease Aβ Paired helical


(fibrillar) filaments
Neuropil Abnormal
threads membranous
Neurofibrillary organelles
tangles

Neurites

Senile plaque

FIGURE 21–5 Comparison of a normal neuron (A) and one with abnormalities associated with Alzheimer disease (B). (Reproduced with
permission from Kandel ER, Schwartz JH, Jessell TM [editors]: Principles of Neural Science, 4th ed. McGraw-Hill, 2000.)
CHAPTER 21 Learning, Memory, Language, and Speech 195

They also initiate an inflammatory response, with production of normally thicker than the left, and the left occipital lobe is
intracellular tangles. The damaged cells eventually die. wider and protrudes across the midline.
An interesting finding that may have broad physiologic In patients with schizophrenia, a disorder characterized by
implications is that frequent effortful mental activities, such as a distorted sense of reality, MRI studies show reduced volumes
doing difficult crossword puzzles and playing board games, of gray matter on the left side in the anterior hippocampus,
slow the onset of cognitive dementia due to Alzheimer disease amygdala, parahippocampal gyrus, and posterior superior
and vascular disease. The explanation for this “use it or lose it” temporal gyrus. The degree of reduction in the left superior
phenomenon is as yet unknown, but it certainly suggests that temporal gyrus correlates with the degree of disordered think-
the hippocampus and its connections have plasticity like other ing in the disease. There are also apparent abnormalities of
parts of the brain. dopaminergic systems and cerebral blood flow in this disease.

LANGUAGE AND SPEECH PHYSIOLOGY OF LANGUAGE


Memory and learning are functions of large parts of the brain, The primary brain areas concerned with language are located
but the centers controlling some of the other “higher functions along and near the Sylvian fissure (lateral cerebral sulcus) of
of the nervous system,” particularly the mechanisms related to the categorical hemisphere. A region at the posterior end of the
language, are more or less localized to the neocortex. superior temporal gyrus called Wernicke’s area (Figure 21–6) is
Human language functions depend more on one cerebral concerned with comprehension of auditory and visual informa-
hemisphere than on the other. This is called the dominant tion. It projects via the arcuate fasciculus to Broca’s area (area 44)
hemisphere and is concerned with categorization and sym- in the frontal lobe immediately in front of the inferior end of the
bolization. The other hemisphere is not less developed or motor cortex. Broca’s area processes the information received
“nondominant”; instead, it is specialized in the area of spa- from Wernicke’s area into a detailed and coordinated pattern for
tiotemporal relations. It is this hemisphere that is concerned, vocalization and then projects the pattern via a speech articula-
for example, with the identification of objects by their form tion area in the insula to the motor cortex, which initiates the
and plays a primary role in the recognition of faces. This con- appropriate movements of the lips, tongue, and larynx to pro-
tributes to the concept of complementary specialization of the duce speech. The probable sequence of events that occurs when
hemispheres, one for sequential-analytic processes (the cate- a subject names a visual object is shown in Figure 21–7. The
gorical hemisphere) and one for visuospatial relations (the angular gyrus behind Wernicke’s area processes information
representational hemisphere). The categorical hemisphere is from words that are read in such a way that they can be con-
concerned with language functions. Lesions in the categorical verted into the auditory forms of the words in Wernicke’s area.
hemisphere produce language disorders. In contrast, lesions in In individuals who learn a second language in adulthood,
the representational hemisphere lead to astereognosis, the fMRI reveals that the portion of Broca’s area concerned with it
inability to identify objects by feeling them. is adjacent to but separate from the area concerned with the
Hemispheric specialization is related to handedness. In 96% native language. However, in children who learn two languages
of right-handed individuals, who constitute 91% of the human early in life, only a single area is involved with both. It is well
population, the left hemisphere is the dominant or categorical known, of course, that children acquire fluency in a second
hemisphere, and in the other 4%, the right hemisphere is dom- language more easily than adults.
inant. In 70% of left-handers, the left hemisphere is also the
categorical hemisphere; in 15%, the right hemisphere is the
categorical hemisphere; and in 15%, there is no clear lateral-
Arcuate
ization. Learning disabilities such as dyslexia (an impaired fasciculus
ability to learn to read) are 12 times as common in left-handers
as they are in right-handers, possibly because some funda-
mental abnormality in the left hemisphere led to a switch in
handedness early in development. The spatial talents of left-
handers may be well above average as a disproportionately
large number of artists, musicians, and mathematicians are
left-handed.
Some anatomic differences between the two hemispheres
Broca’s
may correlate with the functional differences. The planum area Angular gyrus
temporale, an area of the superior temporal gyrus that is Wernicke’s area
involved in language-related auditory processing, is regularly FIGURE 21–6 Location of some of the areas in the categorical
larger on the left side than the right. Imaging studies show that hemisphere that are concerned with language functions.
other portions of the upper surface of the left temporal lobe (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H:
are larger in right-handed individuals, the right frontal lobe is Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
196 SECTION IV CNS/Neural Physiology

Left Right RECOGNITION OF FACES


An important part of the visual input goes to the inferior tem-
Broca’s area poral lobe, where representations of objects, particularly faces,
are stored. Faces are particularly important in distinguishing
Facial area of friends from foes and the emotional state of those seen. Storage
motor cortex
(area 4) 6 Arcuate and recognition of faces is more strongly represented in the
fasciculus right inferior temporal lobe in right-handed individuals.
Lesions in this area cause prosopagnosia, the inability to recog-
nize faces. Patients with this abnormality can recognize forms
Wernicke’s 5 and reproduce them. They can recognize people by their voices,
area From lateral
(area 22) geniculate nucleus and many of them show autonomic responses when they see
4
familiar as opposed to unfamiliar faces. However, they cannot
1 identify the familiar faces they see.
Angular gyrus
3
(area 39) 2

Higher order visual


cortical areas
(area 18) Primary visual cortex CLINICAL CORRELATION
(area 17)
FIGURE 21–7 Path taken by impulses when a subject names
A 9-year-old boy began to have epileptic seizures follow-
a visual object projected on a horizontal section of the human ing a bicycle accident. An electroencephalogram (EEG)
brain. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks showed that the seizures originated in the temporal lobes
H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) bilaterally. He had numerous partial seizures and several
tonic–clonic seizures by the age of 16. The frequent occur-
rence of seizures and lapses in consciousness made com-
LANGUAGE DISORDERS pleting high school very difficult, despite his intellectual
abilities. He was also unable to hold down a job as an
Aphasias are abnormalities of language functions that are not assembly worker. At the age of 27, he underwent experi-
due to defects of vision or hearing or to motor paralysis. They mental brain surgery to remove the amygdala, large por-
are caused by lesions in the categorical hemisphere. The most tions of the hippocampal formation, and portions of the
common cause is embolism or thrombosis of a cerebral blood association area of the temporal cortex. The seizures were
vessel. A convenient classification divides them into fluent, better controlled after surgery, reduced to only about one
nonfluent, and anomic aphasias. major seizure a year. However, the surgery led to devastat-
In one form of fluent aphasia, the lesion is in Wernicke’s area. ing memory deficits. He has maintained long-term mem-
Speech itself is normal, and sometimes the patients talk exces- ory for events that occurred before surgery, but he suffers
sively; however, what they say is full of jargon and neologisms that from anterograde amnesia. His short-term memory is
make little sense. The patient also fails to comprehend the mean- intact, but he is unable to commit new events to long-
ing of spoken or written words, so other aspects of the use of lan- term memory. He has normal procedural memory, and he
guage are compromised. Another form of fluent aphasia is a can learn new puzzles and motor tasks.
condition in which patients can speak relatively well and have This is an actual patient who underwent the experi-
good auditory comprehension but cannot put parts of words mental brain surgery in 1953. This case has been studied
together or conjure up words. It appears to be due to lesions in and by many scientists and has led to a greater understanding
around the auditory cortex (Brodmann’s areas 40, 41, and 42). of the link between the temporal lobe and declarative
In nonfluent aphasia, the lesion is in Broca’s area. Speech is memory. His case is the first to bring attention to the crit-
slow, and words are hard to come by. Patients with severe dam- ical role of temporal lobes in formation of long-term
age to this area are limited to two or three words with which to memories and to implicate this region in the conversion of
express the whole range of meaning and emotion. Sometimes short- to long-term memories. Later work showed that the
the words retained are those that were being spoken at the time hippocampus is the primary structure within the tempo-
of the injury or vascular accident that caused the aphasia. ral lobe involved in this conversion. Because he retained
When a lesion damages the angular gyrus in the categorical memories from before surgery, his case also shows that
hemisphere without affecting Wernicke’s or Broca’s area, there the hippocampus is not involved in the storage of declara-
is no difficulty with speech or the understanding of auditory tive memory. A fascinating audio recording by National
information; instead, there is trouble understanding written Public Radio from the 1990s of the patient talking to
language or pictures, because visual information is not pro- scientists was released in 2007 and is available at http://
cessed and transmitted to Wernicke’s area. The result is a con- www.npr.org/templates/story/story.php?storyId=7584970.
dition called anomic aphasia.
CHAPTER 21 Learning, Memory, Language, and Speech 197

CHAPTER SUMMARY 2. The effects of bilateral loss of hippocampal function include


A) disappearance of remote memories.
■ Long-term memory is divided into explicit (declarative) and
B) loss of working memory.
implicit (nondeclarative). Explicit is further subdivided into
C) loss of the ability to encode events of the recent past
semantic and episodic. Implicit is further subdivided into
in long-term memory.
priming, procedural, associative learning, and nonassociative
D) loss of the ability to recall faces and forms but not the ability
learning.
to recall printed or spoken words.
■ Synaptic plasticity is the ability of neural tissue to change as E) production of inappropriate emotional responses when
reflected by LTP (an increased effectiveness of synaptic recalling events of the recent past.
activity) or LTD (a reduced effectiveness of synaptic activity)
3. Which of the following are incorrectly paired?
after continued use.
A) lesion of the parietal lobe of the representational
■ Hippocampal and other temporal lobe structures and associa-
hemisphere:unilateral inattention and neglect
tion cortex are involved in declarative memory.
B) loss of cholinergic neurons in the nucleus basalis of
■ Alzheimer disease is characterized by progressive loss of Meynert and related areas of the forebrain:loss of
short-term memory followed by general loss of cognitive recent memory
function. The cytopathologic hallmarks of Alzheimer disease C) lesions of mamillary bodies:loss of recent memory
are intracellular neurofibrillary tangles and extracellular senile D) lesion of the angular gyrus in the categorical
plaques. hemisphere:nonfluent aphasia
■ Categorical and representational hemispheres are for sequen- E) lesion of Broca’s area in the categorical hemisphere:slow
tial-analytic processes and visuospatial relations, respectively. speech
Lesions in the categorical hemisphere produce language 4. The representational hemisphere is better than the categorical
disorders, whereas lesions in the representational hemisphere hemisphere at
produce astereognosis.
A) language functions.
■ Aphasias are abnormalities of language functions and are B) recognition of objects by their form.
caused by lesions in the categorical hemisphere. They are C) understanding printed words.
classified as fluent (Wernicke’s area; auditory cortex), nonfluent D) understanding spoken words.
(Broca’s area), and anomic (angular gyrus) based on the E) mathematical calculations.
location of brain lesions.
5. A lesion of Wernicke’s area (the posterior end of the superior
temporal gyrus) in the categorical hemisphere causes patients to
A) lose short-term memory.
STUDY QUESTIONS B) speak in a slow, halting voice.
1. The representational hemisphere C) experience déjà vu.
D) talk rapidly but make little sense.
A) is the right cerebral hemisphere in most right-handed
E) lose the ability to recognize faces.
individuals.
B) is the left cerebral hemisphere in most left-handed 6. Which of the following is most likely not to be involved in
individuals. production of LTP?
C) includes the part of the brain concerned with language A) cAMP
functions. B) Ca2+
D) is the site of lesions in most patients with aphasia. C) NMDA receptors
E) is morphologically identical to the opposite D) membrane hyperpolarization
nonrepresentational hemisphere. E) membrane depolarization
This page intentionally left blank
SECTION V CARDIOVASCULAR PHYSIOLOGY

22
C H A P T E R

Overview of the
Cardiovascular System
Lois Jane Heller and David E. Mohrman

O B J E C T I V E S

■ Define the role of the cardiovascular system in overall body homeostasis.


■ Identify the major body fluid compartments and state the approximate
volume of each.
■ Diagram the blood flow pathways between the heart and other major body
organs.
■ State the relationship among blood flow, blood pressure, and vascular
resistance.
■ Predict the relative changes in flow through a tube caused by changes in
tube length, tube radius, fluid viscosity, and pressure difference.
■ Identify the chambers and valves of the heart and describe the pathway of
blood flow through the heart.
■ Define cardiac output.
■ Describe the pathway of action potential propagation in the heart.
■ List five factors essential to proper ventricular pumping action.
■ State the relationship between ventricular filling and cardiac output
(Starling’s law of the heart) and describe its importance in the control of
cardiac output.
■ Identify the distribution of sympathetic and parasympathetic nerves in the
heart and list the basic effects of these nerves on the cardiac function.
■ List the major different types of vessels in a vascular bed and describe the
morphological differences among them.
■ Describe the basic anatomical features and functions of the different vessel
types.
■ Identify the major mechanisms in vascular control and blood flow
distribution.
■ Describe the basic composition of the fluid and cellular portions of blood and
list the events associated with blood clotting.

199

Ch22_199-210.indd 199 11/26/10 9:58:03 AM


200 SECTION V Cardiovascular Physiology

HOMEOSTATIC ROLE OF THE The well-being of individual cells therefore depends heavily
on the homeostatic mechanisms that regulate the composition
CARDIOVASCULAR SYSTEM of the interstitial fluid. This task is accomplished by continu-
ously exposing the interstitial fluid to “fresh” circulating
Three compartments of watery fluids, known collectively as plasma fluid.
the total body water, account for about 60% of body weight. As blood passes through capillaries, solutes exchange
This water is distributed among the intracellular, interstitial, between plasma and interstitial fluid by the process of diffu-
and plasma compartments, as indicated in Figure 22-1. Note sion. The net result of transcapillary diffusion is always that
that about two thirds of our body water is contained within interstitial fluid tends to take on the composition of the incom-
cells and communicates with the interstitial fluid across the ing blood. If, for example, potassium ion concentration in the
plasma membranes of cells. Of the fluid that is outside cells interstitium of a particular skeletal muscle were higher than
(i.e., extracellular fluid), only a small amount, the plasma vol- that in the plasma entering the muscle, potassium would dif-
ume, circulates within the cardiovascular system. Blood is fuse into the blood as it passed through the muscle’s capillar-
composed of plasma and roughly an equal volume of formed ies. Since this removes potassium from the interstitial fluid,
elements (primarily red cells). The circulating plasma fluid interstitial potassium ion concentration would decrease. It
communicates with the interstitial fluid across the walls of would stop decreasing when net movement of potassium into
small capillary vessels within organs. capillaries no longer occurred, that is, when interstitial con-
The interstitial fluid is the immediate environment of indi- centration reached that of incoming plasma.
vidual cells. The cells must draw their nutrients from and Three conditions are essential for this circulatory mecha-
release their products into the interstitial fluid. The interstitial nism to effectively control the composition of interstitial
fluid cannot, however, be considered a large reservoir for fluid: (1) there must be adequate blood flow through the tis-
nutrients or a large sink for metabolic products since its vol- sue capillaries, (2) the chemical composition of the incom-
ume is less than half that of the cells that it serves. ing (or arterial) blood must be controlled to be that which is

LUNGS

RIGHT
LEFT
HEART
HEART
BODY ORGANS

CAPILLARIES

CELLS

Circulating plasma compartment


≈3 L

Interstitial compartment
(internal environment) Intracellular compartment
≈12 L ≈30 L

FIGURE 22–1 Major body fluid compartments with average volumes indicated for a 70-kg human. Total body water is about 60% of
body weight. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 22 Overview of the Cardiovascular System 201

100%
LUNGS

RIGHT HEART PUMP LEFT HEART PUMP


100% 100%

3%
HEART MUSCLE

14%
BRAIN

15%
SKELETAL MUSCLE

5%
BONE
VEINS ARTERIES
21%
GASTROINTESTINAL SYSTEM, SPLEEN

6%
LIVER

22%
KIDNEY

6%
SKIN

8%
OTHER

FIGURE 22–2 Cardiovascular circuitry indicating the percentage distribution of cardiac output to various organ systems in a
resting individual. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)

optimal in the interstitial fluid, and (3) diffusion distances The overall functional arrangement of the cardiovascular
must be short. Figure 22–1 shows how the cardiovascular system is illustrated in Figure 22–2. Since a functional rather
transport system operates to accomplish these tasks. As than an anatomical viewpoint is expressed in this figure, the
discussed above, substances are transported between cells heart appears in three places: as the right heart pump, as the
and plasma in capillary vessels within organs by the process left heart pump, and as the heart muscle tissue. It is common
of diffusion. This transport occurs over extremely small dis- practice to view the cardiovascular system as (1) the pulmo-
tances because almost no cell in the body is located farther nary circulation, composed of the right heart pump and the
than about 10 μm from a capillary. Over such microscopic lungs, and (2) the systemic circulation, in which the left heart
distances, diffusion is a very rapid process; however, it is a pump supplies blood to the systemic organs (all structures
very poor mechanism by which to move substances from the except the gas exchange portion of the lungs). The pulmonary
capillaries of one organ, such as the lungs, to the capillaries and systemic circulations are arranged in series, that is, one
of another organ that may be 1 m or more distant. Conse- after the other. Consequently, the right and left hearts must
quently, substances are transported between organs by the each pump the same volume of blood each minute. This
process of convection, by which the substances move along amount is called the cardiac output. A cardiac output of
with blood flow simply because they are dissolved or other- 5–6 L/min is normal for a resting individual.
wise contained within blood. The relative distances involved As indicated in Figure 22–2, the systemic organs are gen-
in cardiovascular transport are not well illustrated in erally arranged in parallel (i.e., side by side) within the car-
Figure 22–1. If the figure were drawn to scale with 1 inch diovascular system. There are two important consequences
representing the distance from capillaries to cells within a of this parallel arrangement. First, nearly all systemic organs
calf muscle, the capillaries in the lungs would have to be receive blood of identical composition—that which has just
located about 1.5 miles away! left the lungs and is known as arterial blood. Second, the
202 SECTION V Cardiovascular Physiology

flow through any one of the systemic organs can be con- only slightly greater than that required for their metabolism,
trolled independently of the flow through the other organs. and they do not tolerate blood flow interruptions well. Uncon-
Thus, for example, the cardiovascular response to whole sciousness can occur within a few seconds after stoppage of
body exercise can involve increased blood flow through cerebral flow, and permanent brain damage can occur in as
some organs, but decreased or unchanged blood flow little as 4 minutes without flow. Similarly, the heart muscle
through others. (myocardium) normally consumes about 75% of the oxygen
Many of the organs in the body help perform the task of supplied to it, and the heart’s pumping ability begins to dete-
continuously reconditioning the blood circulating in the car- riorate within beats of a coronary flow interruption. As we
diovascular system. Organs that communicate with the exter- shall see later, the task of providing adequate blood flow to the
nal environment, such as the lungs, play key roles. As is evident brain and the heart muscle receives a high priority in the over-
from the arrangement shown in Figure 22–2, blood that has all operation of the cardiovascular system.
just passed through a systemic organ returns to the right heart
and is pumped through the lungs, where oxygen and carbon
dioxide are exchanged. Thus, the blood’s gas composition is
always reconditioned immediately after leaving a systemic THE BASIC PHYSICS
organ. OF BLOOD FLOW
Like the lungs, many of the systemic organs also serve to
recondition the composition of blood, although the flow cir- As outlined above, the task of maintaining interstitial
cuitry precludes their doing so each time the blood completes homeostasis requires that an adequate quantity of blood
one circuit. The kidneys, for example, adjust the electrolyte flow continuously through the millions of capillaries in the
composition of the blood passing through them. Because the body. In a resting individual, this adds up to a cardiac output
blood conditioned by the kidneys mixes freely with all the cir- of about 5 L/min (about 80 gal/h). As people go about their
culating blood and because electrolytes and water freely pass daily lives, the metabolic rates and therefore the blood flow
through most capillary walls, the kidneys control the electro- requirements in different organs and regions throughout the
lyte balance of the entire internal environment. To achieve body change from moment to moment. Thus, the cardiovas-
this, it is necessary that a given unit of blood pass often through cular system must continuously adjust both the magnitude
the kidneys. In fact, the kidneys (under resting conditions) of cardiac output and how that cardiac output is distributed
normally receive about one fifth of the cardiac output. This to different parts of the body. One of the most important
greatly exceeds the amount of flow that is necessary to supply keys to comprehending how the cardiovascular system oper-
the nutrient needs of the renal tissue. This situation is com- ates is a thorough understanding of the relationship among
mon to organs that have a blood-conditioning function. the physical factors that determine the rate of fluid flow
Blood-conditioning organs can also withstand, at least tem- through a tube.
porarily, severe reductions of blood flow. Skin, for example, The tube depicted in Figure 22–3 might represent a segment
can easily tolerate a large reduction in blood flow when it is of any blood vessel in the body. It has a certain length (L) and
necessary to conserve body heat (see Chapter 70). Most of the a certain internal radius (r) through which blood flows. Fluid
large abdominal organs also fall into this category. The reason flows through the tube only when the pressures in the fluid at
is simply that because of their blood-conditioning functions, the inlet and outlet ends (Pi and Po) are unequal, that is, when
their normal blood flow is far in excess of that necessary to there is a pressure difference (ΔP) between the ends. Pressure
maintain their basal metabolic needs. differences supply the driving force for flow. Because friction
The brain, heart muscle, and skeletal muscles typify organs develops between moving fluid and the stationary walls of a
in which blood flows primarily to supply the metabolic needs tube, vessels tend to resist fluid movement through them. This
of the tissue. They do not recondition blood for the benefit of vascular resistance is a measure of how difficult it is to make
any other organ. Flow to brain and heart muscle is normally fluid flow through the tube, that is, how much of a pressure

Length (L)

Radius (r )
Pi P0
·
Flow (Q )

Inlet ΔP = Pi − P0 Outlet
pressure pressure

FIGURE 22–3 Factors influencing fluid flow through a tube. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology,
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 22 Overview of the Cardiovascular System 203

difference it takes to cause a certain flow. The all-important through the vessels within an organ only because a pressure
relation among flow, pressure difference, and resistance is difference exists between the blood in the arteries supplying
described by the basic flow equation as follows: the organ and the veins draining it. The primary job of the
heart pump is to keep the pressure within arteries higher than
Flow = Pressure difference
______________
Resistance
(1) that within veins. Normally, the average pressure in systemic
arteries is approximately 100 mm Hg, and the average pressure
Q· = ___
ΔP
R in systemic veins approaches 0 mm Hg.
where Q· is the flow (volume/time), ΔP the pressure difference Therefore, because the pressure difference (ΔP) is identical
(mm Hg), and R the resistance to flow (mm Hg × time/vol- across all systemic organs, cardiac output is distributed among
ume). the various systemic organs solely on the basis of their indi-
The basic flow equation may be applied not only to a single vidual resistances to flow. Because blood flows along the path
tube, but also to complex networks of tubes, for example, to of least resistance, organs with relatively low resistance receive
the vascular bed of an organ or to the entire systemic circula- relatively high flow.
tion. The blood flow through the brain, for example, is deter-
mined by the difference in pressure between cerebral arteries
and veins divided by the overall resistance to flow through the
vessels in the cerebral vascular bed. It should be evident from THE HEART
the basic flow equation that there are only two ways in which
blood flow through any organ can be changed: (1) by changing PUMPING ACTION
the pressure difference across its vascular bed or (2) by chang-
The heart lies in the center of the thoracic cavity suspended by
ing its vascular resistance. Most often, it is the change in an
its attachments to the great vessels within a thin fibrous sac
organ’s vascular resistance that causes the flow through the
called the pericardium. A small amount of fluid in the sac
organ to change.
lubricates the surface of the heart and allows it to move freely
The resistance to flow through a cylindrical tube depends
during contraction and relaxation. Blood flow through all
on several factors, including the radius and length of the tube
organs is passive and occurs only because arterial pressure is
and the viscosity of the fluid flowing through it. These factors
kept higher than venous pressure by the pumping action of the
influence resistance to flow as follows:
heart. The right heart pump provides the energy necessary to
8 Lη move blood through the pulmonary vessels, and the left heart
R = ____
πr4
(2)
pump provides the energy to move blood through the systemic
where r is the inside radius of the tube, L the tube length, and organs.
η the fluid viscosity. The amount of blood from each ventricle pumped per min-
Note that resistance is inversely proportional to the internal ute (the cardiac output, CO) depends on the volume of blood
radius of the tube to the fourth power. Thus, even small ejected per beat (the stroke volume, SV) and the number of
changes in the internal radius of a tube have a very large influ- heart beats per minute (the heart rate, HR) as follows:
ence on its resistance to flow. For example, halving the inside
radius of a tube will increase its resistance to flow by 16-fold. CO[volume/minute] = SV[volume/beat] × HR[beats/min] (4)
Equations (1) and (2) may be combined into one expression
It should be evident from the above relationship that all
known as the Poiseuille equation (equation (3)), which
influences on cardiac output must act by changing either the
includes all the terms that influence flow through a cylindrical
heart rate or the stroke volume. These influences will be
vessel:
described in detail in subsequent chapters.
Q· = ΔP ____
π r4
8 Lη
(3) The pathway of blood flow through the chambers of the
heart is indicated in Figure 22–4. Venous blood returns from
Again note that flow occurs only when a pressure difference the systemic organs to the right atrium via the superior and
exists. It is not surprising then that arterial blood pressure is an inferior venae cavae. It passes through the tricuspid valve into
extremely important and carefully regulated cardiovascular the right ventricle and from there is pumped through the pul-
variable. Also note once again that, for any given pressure dif- monic valve into the pulmonary circulation via the pulmo-
ference, tube radius has a very large influence on the flow nary arteries. Oxygenated pulmonary venous blood flows in
through a tube. It is logical, therefore, that organ blood flows pulmonary veins to the left atrium and passes through the
are regulated primarily through changes in the radius of blood mitral valve into the left ventricle. From there it is pumped
vessels within organs. Whereas vessel length and blood viscos- through the aortic valve into the aorta to be distributed to the
ity are factors that influence vascular resistance, they are not systemic organs.
variables that can be easily manipulated for the purpose of Although the gross anatomy of the right heart pump is
moment-to-moment control of blood flow. somewhat different than the left heart pump, the pumping
In regard to the overall cardiovascular system as depicted in principles are identical. Each pump consists of a ventricle,
Figures 22–1 and 22–2, one can conclude that blood flows which is a closed chamber surrounded by a muscular wall, as
204 SECTION V Cardiovascular Physiology

by rhythmic and synchronized contraction and relaxation of


the individual cardiac muscle cells that lie in a circumferential
Superior Aorta
vena cava orientation within the ventricular wall.
When the ventricular muscle cells are contracting, they gen-
Pulmonary erate a circumferential tension in the ventricular walls that
artery
causes the pressure within the chamber to increase. As soon as
Right
Pulmonary the ventricular pressure exceeds the pressure in the pulmo-
atrium nary artery (right pump) or aorta (left pump), blood is forced
veins
Pulmonic out of the chamber through the outlet valve (pulmonic or aor-
valve Left tic) as shown in Figure 22–5. This phase of the cardiac cycle
atrium during which the ventricular muscle cells are contracting is
called systole. Because the pressure is higher in the ventricle
Aortic
valve
than in the atrium during systole, the inlet or atrioventricular
(AV) valve (tricuspid or mitral) is closed. When the ventricu-
Mitral
valve
lar muscle cells relax, the pressure in the ventricle decreases
Tricuspid
valve below that in the atrium, the AV valve opens, and the ventricle
Left refills with blood as shown on the right of Figure 22–5. This
ventricle portion of the cardiac cycle is called diastole. The outlet valve
Inferior
vena cava is closed during diastole because arterial pressure is greater
than intraventricular pressure. After the period of diastolic
Right filling, the systolic phase of a new cardiac cycle is initiated.
ventricle

FIGURE 22–4 Pathway of blood flow through the heart.


(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, EXCITATION
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
Efficient pumping action of the heart requires a precise coor-
dination of the contraction of millions of individual cardiac
illustrated in Figure 22–5. The valves are structurally designed muscle cells. Contraction of each cell is triggered when an
to allow flow in only one direction and passively open and electrical excitatory impulse (action potential) sweeps over its
close in response to the direction of the pressure differences membrane. Proper coordination of the contractile activity of
across them. Ventricular pumping action occurs because the the individual cardiac muscle cells is achieved primarily by the
volume of the intraventricular chamber is cyclically changed conduction of action potentials from one cell to the next via

VENTRICULAR SYSTOLE VENTRICULAR DIASTOLE

Atrium Outlet
valve

Inlet
valve

Ventricular wall Intraventricular chamber

FIGURE 22–5 Ventricular pumping action. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange
Medical Books/McGraw-Hill, 2006.)
CHAPTER 22 Overview of the Cardiovascular System 205

3. The valves must not leak (not insufficient or regurgitant).


4. The muscle contractions must be forceful (not failing).
5. The ventricles must fill adequately during diastole.
Sinoatrial In the subsequent chapters, we will study in detail how the
node above-mentioned requirements are met in the normal heart.
Atrioventricular
node
Atrial CONTROL OF THE HEART AND
muscle CARDIAC OUTPUT
Cartilage
Diastolic Filling
Bundle As cardiac filling increases during diastole, the volume ejected
of His during systole also increases. As a consequence, and as illus-
trated in Figure 22–7, with other factors equal, stroke volume
Left bundle
branch increases as cardiac end-diastolic volume increases. This phe-
nomenon (Starling’s law of the heart) is an intrinsic property
Right bundle of the cardiac muscle and is one of the primary regulators of
branch
cardiac output. The mechanisms responsible for this phenom-
Ventricular enon depend largely on the cardiac muscle cell’s length–tension
muscle relationship and will be described in detail in subsequent
chapters.
FIGURE 22–6 Electrical conduction system of the heart.
(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology,
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) Autonomic Neural Influences
While the heart can inherently beat on its own, cardiac func-
gap junctions that connect all cells of the heart into a func- tion can be influenced profoundly by neural inputs from both
tional syncytium (i.e., acting as one synchronous unit). In the sympathetic and parasympathetic divisions of the auto-
addition, muscle cells in certain areas of the heart are specifi- nomic nervous system. These inputs allow us to modify car-
cally adapted to control the frequency of cardiac excitation, diac pumping as appropriate to meet changing homeostatic
the pathway of conduction, and the rate of the impulse propa- needs of the body. All portions of the heart are richly inner-
gation through various regions of the heart. The major com- vated by adrenergic sympathetic fibers. When active, these
ponents of this specialized excitation and conduction system sympathetic nerves release norepinephrine (noradrenalin) on
are shown in Figure 22–6. They include the sinoatrial node cardiac cells. Norepinephrine interacts with β1-adrenergic
(SA node), the atrioventricular (AV) node, the bundle of receptors on cardiac muscle cells to increase heart rate, increase
His, and the right and left bundle branches made up of spe- action potential conduction velocity, and increase force of con-
cialized cells called Purkinje fibers.
The SA node contains specialized cells that normally func-
tion as the heart’s pacemaker and initiate the action potential
that is conducted through the heart. The AV node contains
slowly conducting cells that normally function to create a
slight delay between atrial contraction and ventricular con-
traction. The Purkinje fibers are specialized for rapid conduc-
tion and assure that all ventricular cells contract at nearly the
Stroke volume

same instant.

REQUIREMENTS FOR EFFECTIVE


OPERATION
For effective efficient ventricular pumping action, the heart
must be functioning properly in five basic respects:
1. The contractions of individual cardiac muscle cells must Ventricular end-diastolic volume
occur at regular intervals and be synchronized (not FIGURE 22–7 Starling’s law of the heart. (Reproduced with
arrhythmic). permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York:
2. The valves must open fully (not stenotic). Lange Medical Books/McGraw-Hill, 2006.)
206 SECTION V Cardiovascular Physiology

traction and rates of contraction and relaxation. Overall, sym- distinguished from one another by differences in physical
pathetic activation acts to increase cardiac pumping. dimensions, morphological characteristics, and function. One
Cholinergic parasympathetic nerve fibers travel to the thing that blood vessels have in common is that they are lined
heart via the vagus nerve and innervate the SA node, the AV with a contiguous single layer of endothelial cells. In fact, this
node, and atrial muscle. When active, these parasympathetic is true for the entire circulatory system including the heart
nerves release acetylcholine on cardiac muscle cells. Acetyl- chambers and even the valve leaflets.
choline interacts with muscarinic receptors on cardiac muscle Some representative physical characteristics are shown in
cells to decrease heart rate (SA node) and decrease action Figure 22–8 for each of the major vessel types. It should be
potential conduction velocity (AV node). Parasympathetic realized, however, that the vascular bed is a continuum and
nerves may also act to decrease the force of contraction of that the transition from one type of vascular segment to
atrial (not ventricular) muscle cells. Overall, parasympathetic another does not occur abruptly. The total cross-sectional area
activation acts to decrease cardiac pumping. Usually an through which blood flows at any particular level in the vascu-
increase in parasympathetic nerve activity is accompanied by lar system is equal to the sum of the cross-sectional areas of all
a decrease in sympathetic nerve activity, and vice versa. the individual vessels arranged in parallel at that level. The
number and total cross-sectional area values presented in
Figure 22–8 are estimates for the entire systemic circulation.
THE VASCULATURE Arteries are thick-walled vessels that contain, in addition to
some smooth muscle, a large component of elastin and colla-
Blood that is ejected into the aorta by the left heart passes con- gen fibers. Primarily because of the elastin fibers, which can
secutively through many different types of vessels before it stretch to twice their unloaded length, arteries can expand to
returns to the right heart. As diagrammed in Figure 22–8, the accept and temporarily store some of the blood ejected by the
major vessel classifications are arteries, arterioles, capillaries, heart during systole and then, by passive recoil, supply this
venules, and veins. These consecutive vascular segments are blood to the organs downstream during diastole. The aorta is

ARTERIES ARTERIOLES CAPILLARIES VENULES VEINS

One-way
valves

Venae
Aorta cavae
Internal
2.5 cm 0.4 cm 30 mm 5 mm 70 mm 0.5 cm 3 cm
diameter
Wall
2 mm 1 mm 20 mm 1 mm 7 mm 0.5 mm 1.5 mm
thickness
Number 1 160 5 = 107 1010 108 200 2
Total
cross-
4.5 cm2 20 cm2 400 cm2 4500 cm2 4000 cm2 40 cm2 18 cm2
sectional
area

FIGURE 22–8 Structural characteristics of the peripheral vascular system. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular
Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 22 Overview of the Cardiovascular System 207

the largest artery and has an inside (luminal) diameter of about greatly influence cardiac filling and therefore cardiac pump-
25 mm. Arterial diameter decreases with each consecutive ing. Thus, peripheral veins actually play an extremely impor-
branching, and the smallest arteries have diameters of approx- tant role in controlling cardiac output.
imately 0.1 mm. The consecutive arterial branching pattern
causes an exponential increase in arterial numbers. Thus,
while individual vessels get progressively smaller, the total CONTROL OF BLOOD VESSELS
cross-sectional area available for blood flow within the arterial
system increases to several-fold that in the aorta. Arteries are Blood flow through individual vascular beds is profoundly
often referred to as conduit vessels because they have rela- influenced by changes in activity of sympathetic nerves inner-
tively low and unchanging resistance to flow. vating arterioles. These nerves release norepinephrine from
Arterioles are smaller and structured differently than arter- their endings that interacts with α-adrenergic receptors on
ies. In proportion to lumen size, arterioles have much thicker the smooth muscle cells to cause contraction and thus arterio-
walls with more smooth muscle and less elastic material than lar constriction. The reduction in arteriolar diameter increases
arteries. Because arterioles are so muscular, their diameters vascular resistance and decreases blood flow. These neural
can be actively changed to regulate the blood flow through fibers provide the most important means of reflex control of
peripheral organs. Despite their minute size, arterioles are so vascular resistance and organ blood flow.
numerous that in parallel their collective cross-sectional area Arteriolar smooth muscle is also very responsive to changes
is much larger than that at any level in arteries. Arterioles are in the local chemical conditions within an organ that accom-
often referred to as resistance vessels because of their high pany changes in the metabolic rate of the organ. For reasons to
and changeable resistance, which regulates peripheral blood be discussed later, increased tissue metabolic rate leads to arte-
flow through individual organs. riolar dilation and increased tissue blood flow.
Capillaries are the smallest vessels in the vasculature. In Venules and veins are also richly innervated by sympathetic
fact, red blood cells with diameters of about 7 μm must nerves and constrict when these nerves are activated. The
deform to pass through them. The capillary wall consists of a mechanism is the same as that involved with arterioles. Thus,
single layer of endothelial cells, which separate the blood increased sympathetic nerve activity is accompanied by
from the interstitial fluid by only about 1 μm. Capillaries con- decreased venous volume. The importance of this phenome-
tain no smooth muscle and thus lack the ability to change non is that venous constriction tends to increase cardiac filling
their diameters actively. They are so numerous that the total and therefore cardiac output via Starling’s law of the heart.
collective cross-sectional area of all the capillaries in systemic There is no important neural or local metabolic control of
organs is more than 1,000 times that of the root of the aorta. either the arterial or capillary vessels.
Given that capillaries are about 0.5 mm in length, the total
surface area available for exchange of material between blood
and interstitial fluid can be calculated; it exceeds 100 m2. For BLOOD
obvious reasons, capillaries are viewed as the exchange ves-
sels of the cardiovascular system. In addition to the transcap- Blood is a complex fluid that serves as the medium for
illary diffusion of solutes that occurs across these vessel walls, transporting substances between the tissues of the body and
there can sometimes be net movements of fluid (volume) into it performs a host of other functions as well. Normally about
and/or out of capillaries. For example, tissue swelling (edema) 40% of the volume of whole blood is occupied by blood cells
is a result of net fluid movement from plasma into the inter- that are suspended in the plasma, which accounts for the
stitial space. rest of the volume. The fraction of blood volume occupied
After leaving capillaries, blood is collected in venules and by cells is a clinically important parameter termed the
veins and returned to the heart. Venous vessels have very thin hematocrit:
walls in proportion to their diameters. Their walls contain cell volume
Hematocrit = ______________ (5)
smooth muscle and the diameters of venous vessels can actively total blood volume
change. Because of their thin walls, venous vessels are quite
distensible. Therefore, their diameters change passively in
response to small changes in transmural distending pressure BLOOD CELLS
(i.e., the difference between the internal and external pressures
across the vessel wall). Venous vessels, especially the larger Blood contains three general types of “formed elements”:
ones, also have one-way valves that prevent reverse flow. As red cells, white cells, and platelets (Table 22–1). All are
will be discussed later, these valves are especially important in formed in bone marrow from a common stem cell. Red cells
the cardiovascular system’s operation during standing and (erythrocytes) are by far the most abundant. They are spe-
during exercise. It turns out that peripheral venules and veins cialized to carry oxygen from the lungs to other tissues by
normally contain more than 50% of the total blood volume. binding oxygen to hemoglobin, an iron-containing heme
Consequently, they are commonly thought of as the capaci- protein concentrated within red blood cells. Because of the
tance vessels. More importantly, changes in venous volume presence of hemoglobin, blood can transport 50–60 times the
208 SECTION V Cardiovascular Physiology

TABLE 22–1 Normal values of erythrocytes, leukocytes, and platelets in adult human blood.a
Erythrocytes 4.0–5.5 million/μL of blood
Platelets 130,000–400,000/μL of blood
Leukocytes 4,000–10,000/μL of blood

Type of Leukocyte Percent Total Leukocytes Primary Role

Polymorphonuclear granulocytes

Neutrophils 50–70 Phagocytosis

Eosinophils 1–4 Allergic hypersensitivity reactions

Basophils 0–0.75 Allergic hypersensitivity reaction

Monocytes 2–8 Phagocytosis and antibody


production

Lymphocytes 15–40 Antibody production and cell-


mediated immunity
a
Normal reference ranges vary somewhat with age, gender, and race. They also may vary from laboratory to laboratory. To confuse the issue further, various unit measurements
are used to report blood values, so caution must be used in interpreting data.

amount of oxygen that plasma alone could carry. In addition, Proteins do not readily cross capillary walls and, in gen-
the hydrogen ion buffering capacity of hemoglobin is vitally eral, their plasma concentrations are much greater than their
important to the blood’s capacity to transport carbon dioxide. concentrations in the interstitial fluid. As will be dis-
A small but important fraction of the cells in blood is white cussed, plasma proteins play an important osmotic role in
cells or leukocytes. Leukocytes are involved in immune pro- transcapillary fluid movement and thus the distribution of
cesses and have specific roles as indicated in Table 22–1. extracellular volume between the plasma and interstitial
Platelets are small cell fragments that are important in the compartments. Albumin plays an especially important role
blood clotting process. in this regard simply because it is by far the most abundant of
the plasma proteins.
Plasma also serves as the vehicle for transporting nutrients
PLASMA and waste products. Thus, a plasma sample contains many
small organic molecules such as glucose, amino acids, urea,
Plasma is the liquid component of blood and, as indicated in creatinine, and uric acid whose measured values are useful in
Table 22–2, is a complex solution of electrolytes and proteins. clinical diagnosis.
Serum is the fluid obtained from a blood sample after it has
been allowed to clot. For all practical purposes, the composi-
tion of serum is very similar to that of plasma except that it
contains none of the clotting proteins. HEMOSTASIS
Inorganic electrolytes (ions such as sodium, potassium,
Whenever damage occurs to a blood vessel, a variety of pro-
chloride, and bicarbonate) are the most concentrated solutes
cesses are initiated that are aimed at preventing or stopping
in plasma. Of these, sodium and chloride are by far the most
blood from exiting the vascular space. The three primary pro-
abundant and, therefore, are primarily responsible for plasma’s
cesses are summarized in the following outlines:
normal osmolarity of about 300 mOsm/L. To a first approxi-
mation, plasma is a 150-mM solution of sodium chloride. 1. Platelet aggregation and plug formation: occur as a re-
Such a solution is called isotonic saline and has many clinical sult of the following steps:
uses as a fluid that is compatible with cells. A. vessel injury with endothelial damage and collagen
Plasma normally contains many different proteins. Most exposure;
plasma proteins can be classified as albumins, globulins, or B. platelet adherence to collagen (mediated by von
fibrinogen on the basis of different physical and chemical Willebrand factor);
characteristics used to separate them. More than 100 distinct C. platelet shape change (from disks to spiny spheres);
plasma proteins have been identified and each presumably D. platelet degranulation with release of the following:
serves some specific function. Many plasma proteins are (i) adenosine diphosphate, which causes platelet ag-
involved in blood clotting or immune/defense reactions. Many gregation to “plug” the hole,
others are important carrier proteins for a variety of substances (ii) thromboxane, causing vasoconstriction, and
including fatty acids, iron, copper, and certain hormones. platelet adhesion and aggregation.
CHAPTER 22 Overview of the Cardiovascular System 209

TABLE 22–2 Normal constituents of adult human plasma.


Class Constituent Normal Concentration Range
+
Cations Sodium (Na ) 136–145 mEq/L
+
Potassium (K ) 3.5–5.0 mEq/L
2+
Calcium (Ca ) 4.3–5.2 mEq/L
2+
Magnesium (Mg ) 1.2–1.8 mEq/L
3+
Iron (Fe ) 60–160 μg/dL
+
Hydrogen (H ) 35–45 nmol/L (pH 7.35–7.45)

Anions Chloride (Cl ) 98–106 mEq/L

Bicarbonate (HCO3 ) 23–28 mEq/L

Lactate 0.67–1.8 mEq/L


2–
Phosphate (HPO4 mostly) 3.0–4.5 mg/dL

Proteins Total (7% of plasma weight) 6–8 g/dL

Albumin 3.4–5.0 g/dL

Globulins 2.2–4.0 g/dL

Fibrinogen 0.3 g/dL

(Note: Aspirin and other cyclooxygenase inhibi- (b) converts fibrinogen to fibrin;
tors are anticoagulants because they prevent the (c) recruits the “intrinsic pathway,” which amplifies fur-
formation of thromboxane.) ther formation of factor Xa and facilitates the conver-
sion of prothrombin to thrombin by promoting the
2. Local vasoconstriction: mediated largely by thrombox-
following reactions:
ane but also may be induced by local release of other
(i) conversion of factor XI to its activated form, XIa,
chemical signals that constrict local vessels and reduce
which then converts factor IX to IXa, which then
blood flow.
attaches to activated platelets and converts factor
3. Blood clotting: the formation of a solid gel made up of
X to Xa,
the protein, fibrin, platelets, and trapped blood cells.
(ii) conversion of factor VIII (missing in people with
The critical step in blood clotting is the formation of hemophilia) to its activated form, VIIIa, which
thrombin from prothrombin, which then catalyzes the attaches to activated platelets and accelerates con-
conversion of fibrinogen to fibrin. The final clot is stabi- version of factor X to Xa,
lized by covalent cross-linkages between fibrin strands cata- (iii) conversion of factor V to its activated form, Va,
lyzed by factor XIIIa (the formation of which is catalyzed which attaches to activated platelets and acceler-
by thrombin). ates conversion of prothrombin to thrombin.
The cascade of reactions that leads from vessel injury to the
Several agents clinically used as anticoagulants interfere
formation of thrombin is as follows:
with various steps in this clotting process. Dicoumarol and
(1) Vessel injury or tissue damage with blood exposure to coumadin block the activity of vitamin K, which is neces-
subendothelial cells that release thromboplastin (“tissue sary for synthesis of many of the clotting factors by the liver.
factor”). Heparin activates a plasma protein called antithrombin
(2) The plasma protein factor VII binds to the tissue factor, III, which, in turn, inactivates thrombin and several of the
which converts it to an activated form, factor VIIa. other clotting factors. Because calcium is an important clot-
(3) VIIa catalyzes conversion of both factors IX and X to ac- ting cofactor, calcium chelators such as EDTA, oxalate,
tivated forms, IXa and Xa, respectively. and citrate are used to prevent stored blood from clotting.
(4) IXa also helps convert factor X to Xa (Stuart factor). Various thrombolytic agents modeled after the endogenous
(5) Xa converts prothrombin to thrombin. tissue plasminogen activator (tPA) are also available that
(6) Thrombin: promote dissolution of the fibrin clot after it is formed.
(a) activates platelets (makes them sticky, induces de- These agents promote the formation of plasmin from plas-
granulation, promotes attachment of various factors minogen that enzymatically attacks the clot, turning it into
that participate in clotting); soluble peptides.
210 SECTION V Cardiovascular Physiology

■ Blood flow through individual organs is regulated by changes


CLINICAL CORRELATION in the diameters of their arterioles.
■ Changes in arteriolar diameter can be accomplished by
A 45-year-old investment banker passed out at the mall. alterations in sympathetic nerve activity and by variations in
He was conscious by the time the paramedics arrived a local conditions.
few minutes later but was brought to the emergency room ■ Blood is a complex suspension of red cells, white cells, and
for evaluation. He reported that he had been very busy platelets in plasma that is ideally suited to carry gases, salts,
and somewhat stressed lately but otherwise was healthy. nutrients, and waste molecules throughout the system.
He looked pale and felt somewhat nauseated and dizzy ■ Hemostasis involves platelet aggregation, local vasoconstric-
just before the episode, but did not remember falling or tion, and blood clotting.
anything else until the paramedic was leaning over him
on the floor. Physical findings included: weight = 90 kg,
height = 5΄10˝, arterial blood pressure = 130/85 mm Hg, STUDY QUESTIONS
heart rate = 85 b/min, normal heart and breath sounds, 1. You need to determine the correct dose of an i.v. drug that distributes
normal reflexes, and normal cognition. An electrocardio- only within the extracellular space. Which of the following values
gram was obtained and found to be normal. Blood sam- would be the closest estimate of the extracellular fluid volume of a
ples were obtained for markers of myocardial infarction healthy young adult male weighing 100 kg (220 lb)?
and heart failure, and also found to be normal. He was A) 3 L
admitted to the hospital and ECG was monitored over- B) 5 L
night with no significant findings. C) 8 L
Unexplained syncopal (fainting) episodes may be a D) 10 L
result of either a neural problem (seizure) or a significant E) 20 L
decrease in blood flow to the brain. The latter may be 2. You have transfused a liter of blood into your dehydrated patient.
caused by either a failure of the heart to maintain suffi- At any instant, most of this blood will be found in which segment
cient cardiac output (due to an arrhythmia, valve failure, of the systemic vascular bed?
or infarction affecting significant myocardial mass) or a A) arteries
failure within the systemic vasculature either to maintain B) arterioles
C) capillaries
sufficient contractile activity (due to a withdrawal of sym-
D) veins
pathetic neural influences as in vasovagal syncope) or to
E) right atrium
provide sufficient arterial blood supply to the brain (due
3. Which of the following will produce the greatest increase in
to severe, often transient, carotid artery occlusions pro-
blood flow through exercising muscles?
voking a transient ischemic attack [TIA]). Diagnostic
A) halve the length of the capillaries
testing was performed to evaluate these possibilities. The
B) halve the viscosity of the blood
man was outfitted with a Holter monitor that recorded his C) double the radius of the venules
ECG for a 24-hour period, and found to be normal. By D) double the blood pressure difference across the bed
process of elimination, he was given the diagnosis of vaso- E) double the radius of the arterioles
vagal syncope that is usually due to an increase in efferent 4. An individual has had the “flu” for 3 days with severe vomiting
vagal (parasympathetic) input to the heart that will be dis- and diarrhea without increasing his fluid intake. How is this
cussed in Chapters 23 and 29. In most cases of vasovagal likely to influence his hematocrit?
syncope, no treatment is necessary. A) no effect
B) higher than normal
C) lower than normal
D) cannot predict
CHAPTER SUMMARY E) eliminate it
5. Calculate the cardiac output from the following data: pulmonary
■ The primary role of the cardiovascular system is to maintain
arterial pressure = 20 mm Hg; left atrial pressure = 5 mm Hg;
homeostasis of the interstitial fluid.
pulmonary vascular resistance = 3 mm Hg per L/min.
■ The physical law that governs cardiovascular operation is that
A) 3 L/min
flow through any segment is equal to pressure difference across
· B) 5 L/min
that segment divided by its resistance to flow, that is, Q = ΔP/R.
C) 4 L/min
■ The heart pumps blood by rhythmically filling with and D) 15 L/min
ejecting blood from the ventricular chambers that are served by E) 60 mm Hg
passive one-way inlet and outlet valves.
■ Changes in heart rate and stroke volume (and therefore cardiac
output) can be accomplished by alterations in ventricular filling
and by alterations in autonomic nerve activity to the heart.
23
C H A P T E R

Cardiac Muscle Cells


Lois Jane Heller and David E. Mohrman

O B J E C T I V E S

■ Describe the characteristics of cardiac resting potentials and “fast” and “slow”
response cardiac action potentials.
■ Identify the refractory periods of the cardiac cell electrical cycle.
■ Define threshold potential and describe the interaction between ion channel
conditions and membrane potential during the depolarization phase of the
action potential.
■ Define pacemaker potential and describe the basis for rhythmic electrical
activity of cardiac cells.
■ List the phases of the cardiac cell electrical cycle and state the membrane
permeability alterations responsible for each phase.
■ Describe gap junctions and their role in cardiac excitation.
■ Describe the normal pathway of action potential conduction through the heart.
■ Indicate the timing of electrical excitation of various areas of the heart and
identify the characteristic action potential shapes and conduction velocities
in each major part of the conduction system.
■ State the relationship between electrical events of cardiac excitation and the P,
QRS, and T waves, the PR interval, and the ST segment of the electrocardiogram.
■ State how diastolic potentials of pacemaker cells can be altered to produce
changes in heart rate.
■ Describe how cardiac sympathetic and parasympathetic nerves alter heart
rate and conduction of cardiac action potentials.
■ Define the terms chronotropic and dromotropic.
■ Define and describe the excitation–contraction process in cardiac muscle.
■ Define isometric, isotonic, and afterloaded contractions of cardiac muscle.
■ Identify the influence of altered preload on the tension-producing and
shortening capabilities of cardiac muscle.
■ Describe the influence of altered afterload on the shortening capabilities
of cardiac muscle.
■ Define the terms contractility and inotropic state and describe the influence
of altered contractility on the tension-producing and shortening capabilities
of cardiac muscle.
■ Describe the effect of altered sympathetic neural activity on cardiac
inotropic state.
■ State the relationships between ventricular volume and muscle length, and
between intraventricular pressure and muscle tension; explain the law of Laplace.

211

Ch23_211-222.indd 211 11/26/10 10:00:01 AM


212 SECTION V Cardiovascular Physiology

CHARACTERISTICS OF potentials, respectively. As shown in panel A of this figure, fast


response action potentials are characterized by a rapid
CARDIAC MUSCLE CELLS depolarization (phase 0) with a substantial overshoot (positive
inside voltage), a rapid reversal of the overshoot potential
Coordination of the activity of cardiac muscle cells (myocytes) (phase 1), a long plateau (phase 2), and a repolarization
depends on an electrical stimulus that is regularly initiated at (phase 3) to a stable, high (i.e., large negative) resting mem-
an appropriate rate and reliably conducted through the entire brane potential (phase 4). In comparison, the slow response
heart. Mechanical pumping action depends on a robust con- action potentials are characterized by a slower initial depolar-
traction of the muscle cells that results in repeating cycles of ization phase (phase 0), a lower amplitude overshoot, a shorter
tension development, shortening, and relaxation. In addition, and less stable plateau phase (phase 2), and a repolarization
mechanisms to adjust the excitation and contraction charac- (phase 3) to an unstable, slowly depolarizing “resting” poten-
teristics must be available to meet changing demands on the tial (phase 4) (Figure 23–1B). The unstable resting potential
circulatory system. seen in pacemaker cells with slow response action potentials is
variously referred to as the phase 4 depolarization, diastolic
depolarization, or pacemaker potential. Such cells are usu-
ELECTRICAL PROPERTIES ally found in the sinoatrial (SA) and atrioventricular (AV)
Contraction of cardiac myocytes is triggered by action poten- nodes.
tials that occur on the cell membrane. Cardiac action poten- As indicated at the bottom of Figure 23–1A, cells are in an
tials differ sharply from those of skeletal muscle in three absolute refractory state during most of the action potential
important ways that promote synchronous rhythmic excita- (i.e., they cannot be stimulated to fire another action poten-
tion of the heart: (1) they can be self-generating; (2) they can tial). Near the end of the action potential, the membrane is
be conducted directly from cell to cell; and (3) they have long relatively refractory and can be re-excited only by a larger-
durations, which preclude fusion of individual twitch contrac- than-normal stimulus. Immediately after the action potential,
tions. To understand these special electrical properties of car- the membrane is transiently hyperexcitable and is said to be in
diac muscle and how cardiac function depends on them, the a “vulnerable” or “supranormal” period. Similar alterations in
basic electrical properties of excitable cell membranes membrane excitability occur during slow action potentials but
described in Chapters 3 and 6 should be carefully reviewed. at present are not well characterized.
Recall that the membrane potential of any cell at any given
instant depends on the relative permeability of the cell mem-
SUMMARY OF CARDIAC CELL brane to specific ions. As in all excitable cells, cardiac cell
MEMBRANE POTENTIALS action potentials are the result of transient changes in the ionic
permeability of the cell membrane that are triggered by an ini-
At rest, cardiac cell membranes are more permeable to potas- tial depolarization. Panels C and D of Figure 23–1 indicate the
sium than to any other ion. Because the potassium concentra- changes in membrane permeabilities to K+, Na+, and Ca2+ that
tion inside the cells is significantly higher than that of the produce the various phases of the fast and slow response action
interstitial fluid (150 mM vs. 4 mM, respectively), the outward potentials.
diffusion of potassium down its concentration gradient is bal- Note that during the resting phase, the membranes of both
anced by the generation of a membrane potential (i.e., the types of cells are dominated by a higher permeability to K+
potassium equilibrium potential). Both the electrical and the than to Na+ or Ca2+. Therefore, the membrane potentials are
concentration gradients favor Na+ and Ca2+ entry into the rest- closer to the potassium equilibrium potential during this
ing cells. However, the very low permeability of the resting period than to the equilibrium potential of any other ion. In
membrane to Na+ and Ca2+, in combination with an energy- the pacemaker-type cells, at least three mechanisms are
requiring sodium pump that extrudes Na+ from the cell, pre- thought to contribute to the slow depolarization of the mem-
vents Na+ and Ca2+ from gradually accumulating inside the brane observed during the diastolic interval: (1) there is a pro-
resting cell. The steep sodium gradient promotes Ca2+ removal gressive slow decrease in the membrane permeability to K+,
from the cytoplasm via the sodium/calcium exchanger. (2) there is a slow increase in permeability to Na+, and (3) there
Action potentials of cells from different regions of the heart is a slight increase in the permeability of the membrane to
are not identical but have varying characteristics that depend calcium ions.
on differences in the patterns of changes in their ion permea- When the membrane potential depolarizes to the threshold
bilities. Some cardiac cells have the ability to act as pacemakers potential in either type of cell, major rapid alterations in the
and to spontaneously initiate action potentials, whereas ordi- permeability of the membrane to specific ions are triggered.
nary cardiac muscle cells do not (except under unusual Once initiated, these permeability changes cannot be stopped
conditions). Basic membrane electrical features of an ordinary and they proceed to completion.
cardiac muscle cell and a cardiac pacemaker-type cell are The characteristic rapid rising phase of the fast response
shown in Figure 23–1. Action potentials from these cell types action potential is a result of a sudden increase in Na+
are referred to as “fast response” and “slow response” action permeability. As indicated in panel C of Figure 23–1, this
CHAPTER 23 Cardiac Muscle Cells 213

A Fast-response action B Slow-response action


potentials potentials

Transmembrane potential (mV)


Phase
0 2

Phase 0

Phas
–50

e3
se 4
Pha
Phase 4

–100
Absolute refractory period
Relative refractory period
Supranormal period

C D

10.0
Na+
Relative membrane permeability

Ca2+
Ca2+
K+ K+
1.0

Na+

0.1

0 0.15 0.30 0 0.15 0.30


Time (s) Time (s)
FIGURE 23–1 Time course of membrane potential and ion permeability changes that occur during “fast response” (A and C) and
“slow response” (B and D) action potentials. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical
Books/McGraw-Hill, 2006.)

period of very high sodium permeability (phase 0) is short- membrane of a single cell as discussed in Chapter 6. Table 23–1
lived and is followed by a very brief increase in potassium per- summarizes some of the major currents and channel types
meability (not shown in Figure 23–1C). This brief current (both voltage- and ligand-gated) involved in cardiac cell elec-
accounts for the very early repolarization that immediately trical activity.
follows the initial rising phase of the action potential (phase 1). Some of the voltage-gated channels respond to a sudden-
Development and maintenance of a depolarized plateau state onset, sustained change in membrane potential with only a
(phase 2) depends on: (1) a sustained reduction in K+ perme- brief period of activation. However, changes in membrane
ability, (2) a slowly developed and sustained increase in the potential of slower onset but the same magnitude may fail to
membrane permeability to Ca2+, and (3) electrogenic action of activate these channels at all. To explain such behavior, it is
an Na+/Ca2+ exchanger in which three Na+ ions move into the postulated that these channels have two independently
cell in exchange for a single Ca2+ moving out of the cell. In cells operating “gates”—an activation gate and an inactivation
with the slow response action potential, the initial fast inward gate—both of which must be open for the channel as a whole
current is small (or even absent). The slow rising phase of to be open. Both of these gates respond to changes in mem-
these action potentials is therefore primarily a result of an brane potential but do so with different voltage sensitivities
inward movement of Ca2+ ions. In both types of cells, the and time courses. With an abrupt depolarization to threshold,
membrane is repolarized (phase 3) to its original resting sodium channel activation gates open and within a few milli-
potential as the K+ permeability increases and the Ca2+ and seconds their inactivation gates close. In pacemaker cells with
Na+ permeabilities return to their low resting values. slow diastolic depolarization, the inactivation gates of the
Recall that the smoothly graded permeability changes that sodium channels are closed before the activation gates have a
produce action potentials are the net result of alterations in chance to open. When threshold is reached, only the calcium
each of the many individual ion channels within the plasma channel is available to open, thus accounting for the initial
214 SECTION V Cardiovascular Physiology

TABLE 23–1 Characteristics of important cardiac ion channels in order of their participation in an action potential.
Gating
Current Channel Mechanism Functional Role

iK1 Kir+ channel (inward rectifier) Voltage Maintains high K+ permeability during phase 4
Its decay contributes to diastolic depolarization
Its suppression during phases 0–2 contributes to plateau
+
iNa Na channel (fast) Voltage Accounts for phase 0 of action potential
Inactivation may contribute to phase 1 of action potential
+
Ito K channel (transient outward) Voltage Contributes to phase 1 of action potential
2+
iCa Ca channel (slow inward, Both Contributes to phase 2 of action potential
L channels) Inactivation may contribute to phase 3 of action potential
Is enhanced by sympathetic stimulation and β-adrenergic agents
+
iK K channel (delayed rectifier) Voltage Causes phase 3 of action potential
Is enhanced by increased intracellular Ca2+

iKATP K+ channel (ATP-sensitive) Ligand Increases K+ permeability when [ATP] is low

iKACh K+ channel Ligand Responsible for effects of vagal stimulation


(acetylcholine-activated) Decreases diastolic depolarization (and heart rate)
Hyperpolarizes resting membrane potential
Shortens phase 2 of the action potential
+
if (“funny”) Nạ (pacemaker current) Both Is activated by hyperpolarization and contributes to the diastolic depolarization
Is enhanced by sympathetic stimulation and β-adrenergic agents
Is suppressed by vagal stimulation

slow rising phase of the action potentials in pacemaker cells. however, can inactivate both the fast and slow channels and
Calcium channel inactivation gate closing is delayed for more thus make the cardiac muscle cells completely unexcitable.
than 100 milliseconds until near the end of the plateau phase.
Inactivation gates on the sodium and calcium channels remain
closed until the membrane repolarizes. This accounts for the CONDUCTION OF CARDIAC
long cardiac muscle cell refractory period. ACTION POTENTIALS
Multiple factors influence the operation of K+ channels,
some of which are summarized in Table 23–1. For example, Action potentials are conducted over the surface of individual
high intracellular Ca2+ concentration contributes to activation cells because active depolarization in any one area of the mem-
of some K+ channels during repolarization. brane produces local currents in the intracellular and extracel-
Although cells in certain areas of the heart typically have lular fluids that passively depolarize immediately adjacent
fast-type action potentials and cells in other areas normally areas of the membrane to their voltage threshold for active
have slow-type action potentials, it is important to recognize depolarization.
that all cardiac cells are potentially capable of having either In the heart, cardiac muscle cells are connected end-to-end
type of action potential depending on their resting membrane by structures called intercalated disks. These disks contain
potentials and how fast they depolarize to the threshold poten- the following: (1) firm mechanical attachments between
tial. Rapid depolarization to the threshold potential is usually adjacent cell membranes by proteins called adherins in struc-
an event forced on a cell by the occurrence of an action poten- tures called desmosomes and (2) low-resistance electrical
tial in an adjacent cell. Slow depolarization to threshold occurs connections between adjacent cells through channels formed
when a cell itself spontaneously and gradually loses its resting by a protein called connexin in structures called gap junc-
polarization, which normally happens only in the SA node. A tions. Figure 23–2 shows schematically how these gap junc-
chronic moderate depolarization of the resting membrane tions allow action potential propagation from cell to cell.
(caused, e.g., by moderately high extracellular K+ concentra- Cells B, C, and D are shown in the resting phase with more
tion) can inactivate the fast sodium channels (i.e., prevent negative charges on the inside than the outside. Cell A is shown
them from opening) without inactivating the slow calcium in the plateau phase of an action potential and has more positive
channels. Under these conditions, all cardiac cell action poten- charges inside than out. Because of the gap junctions, electro-
tials will be of the slow type. Large sustained depolarizations, static attraction can cause a local current flow (ion movement)
CHAPTER 23 Cardiac Muscle Cells 215

+ + + + + + + + +
+ – – – – – – – – –
+ + –
+ + + – – Resting cell C
– – – – – – – – + – –
– – – – – – – – –
+ + + + + + + + – –+ + + + + + + +
+
Cell A with action potential Resting cell B ++ + + + + –


+ + + +
+ + + + + + + + – – – – – – – – – –
– – – – – – – – – – – –
+ + + – – – – Resting cell D – –+ +
+ – – –
+ + + – – – – –
+ +
+ + + – – – –
+ + +
+
Gap junction (nexus)

FIGURE 23–2 Local currents and cell-to-cell conduction of cardiac muscle cell action potentials. (Modified with permission from Mohrman
DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)

between the depolarized membrane of active cell A and the small-diameter cells in the AV node is significantly slower
polarized membrane of resting cell B, as indicated by the arrows than conduction over large-diameter cells in the ventricu-
in the figure. This ion movement depolarizes the membrane of lar Purkinje system.
cell B. Once the local currents from active cell A depolarize the 2. Conduction velocity is also directly dependent on the
membrane of cell B near the gap junction to the threshold level, intensity of the local depolarizing currents, which are in
an action potential will be triggered at that site and will be con- turn directly determined by the rate of rise of the action
ducted over cell B. Because cell B branches (a common morpho- potential. Rapid depolarization favors rapid conduction.
logical characteristic of cardiac muscle fibers), its action 3. Conduction velocity is dependent on the capacitive and/
potential will evoke action potentials on cells C and D. This pro- or resistive properties of the cell membranes, gap junc-
cess is continued through the entire myocardium. Thus, an tions, and cytoplasm. Electrical characteristics of gap
action potential initiated at any site in the myocardium will be junctions can be influenced by external conditions that
conducted from cell to cell throughout the entire myocardium. promote phosphorylation/dephosphorylation of the con-
The speed at which an action potential propagates through nexin proteins.
a region of cardiac tissue is called the conduction velocity.
Details of the overall consequences of the cardiac conduc-
The conduction velocity varies considerably in different areas
tion system are shown in Figure 23–3. As noted earlier, the
in the heart and is determined by three variables:
specific electrical adaptations of various cells in the heart are
1. Conduction velocity is directly dependent on the diame- reflected in the characteristic shape of their action potentials
ter of the muscle fiber involved. Thus, conduction over that are shown in the right half of Figure 23–3. Note that the

SA node A
mV

–100
Atrial muscle B

Atrial muscle C

AV node D

Purkinje fiber E
Ventricular muscle
F
Ventricular muscle
G

P wave T wave
ECG
Q S
PR Interval ST segment

QT Interval
1.0 second
FIGURE 23–3 Electrical activity of the heart: single-cell voltage recordings (traces A–G) and lead II electrocardiogram. (Modified with
permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
216 SECTION V Cardiovascular Physiology

action potentials shown in Figure 23–3 have been positioned tent pathway, the temporal pattern of voltage change recorded
to indicate the time at which the electrical impulse that origi- between two points on the body surface is also consistent and
nates in the SA node reaches other areas of the heart. Cells of will repeat itself with each heart cycle.
the SA node act as the heart’s normal pacemaker and deter- The lower trace of Figure 23–3 represents a typical record-
mine the heart rate. This is because the spontaneous diastolic ing of the voltage changes normally measured between the
depolarization of the resting membrane is most rapid in SA right arm and the left leg as the heart goes through two cycles
nodal cells, and they reach their threshold potential before of electrical excitation; this record is called a lead II electrocar-
cells elsewhere in the heart. diogram. The major features of an electrocardiogram are the
The action potential initiated by an SA nodal cell first spreads P wave, the QRS complex, and the T wave. The P wave
progressively through the atrial wall. Action potentials from corresponds to atrial depolarization, the QRS complex to
cells in two different regions of the atria are shown in Figure 23–3: ventricular depolarization, and the T wave to ventricular
one close to the SA node and one more distant from the SA repolarization.
node. Both cells have similarly shaped action potentials, but
their temporal displacement reflects the fact that it takes some
time for the impulse to spread over the atria. Action potential CONTROL OF HEART RATE
conduction is greatly slowed as it passes through the AV node.
This is because of the small size of the AV nodal cells and Normal rhythmic contractions of the heart occur because of
the slow rate of rise of their action potentials. Since the AV node spontaneous electrical pacemaker activity (automaticity) of
delays the transfer of the cardiac excitation from the atria to the cells in the SA node. The interval between heartbeats (and
ventricles, atrial contraction can contribute to ventricular filling thus the heart rate) is determined by how long it takes the
just before the ventricles contract. Note also that AV nodal cells membranes of these pacemaker cells to spontaneously depo-
have a faster spontaneous depolarization during the resting larize to the threshold level. The SA nodal cells fire at a spon-
period than other cells of the heart except those of the SA node. taneous or intrinsic rate (≈100 beats/min) in the absence of
The AV node is sometimes referred to as a latent pacemaker, any outside influences.
and in many pathological situations it (rather than the SA node) The two most important outside influences on automaticity
controls the heart rhythm. of SA nodal cells come from the autonomic nervous system
Because of sharply rising action potentials and other factors, (see Chapter 19). Fibers from both the sympathetic and para-
such as large cell diameters, electrical conduction is extremely sympathetic divisions of the autonomic system terminate on
rapid in Purkinje fibers. This allows the Purkinje system to cells in the SA node and these fibers can modify the intrinsic
transfer the cardiac impulse to cells in many areas of the ven- heart rate. Activation of the cardiac sympathetic nerves
tricle nearly in unison. Action potentials from muscle cells (increasing cardiac sympathetic tone) increases the heart rate.
in two areas of the ventricle are shown in Figure 23–3. Because Increasing cardiac parasympathetic tone decreases the heart
of the high conduction velocity in ventricular tissue, there is rate. As shown in Figure 23–4, the parasympathetic and sym-
only a small discrepancy in their time of onset. Note that the pathetic nerves both influence heart rate by altering the course
ventricular cells that are the last to depolarize have of spontaneous depolarization of the resting potential in SA
shorter-duration action potentials and thus are the first to pacemaker cells.
repolarize. The physiological importance of this unexpected Cardiac parasympathetic fibers, which travel to the heart
behavior is not clear but it does have an influence on the elec- through the vagus nerves, release the transmitter substance
trocardiograms that will be discussed in Chapter 25. acetylcholine on SA nodal cells. Acetylcholine increases the
permeability of the resting membrane to K+ and decreases the
diastolic permeability to Na+. The signaling process involves
ELECTROCARDIOGRAMS acetylcholine interaction with muscarinic receptors on the SA
nodal cell membrane that in turn are linked to inhibitory G
Fields of electrical potential caused by the electrical activity of proteins, Gi. The activation of Gi has two effects: (1) an
the heart extend through the extracellular fluid of the body increase in K+ permeability resulting from an increased
and can be measured with electrodes placed on the body sur- opening of the KAch channels and (2) a suppression of adeny-
face. Electrocardiography provides a record of how the volt- late cyclase leading to a decrease in intracellular cyclic ade-
age difference between two points on the body surface changes nosine monophosphate (cAMP) concentration that reduces
with time as a result of the electrical events of the cardiac cycle. the inward-going pacemaker current carried by Na+ (if). As
At any instant of the cardiac cycle, the electrocardiogram indi- indicated in Figure 23–4, these permeability changes have
cates the net electrical field that is the summation of many two effects on the resting potential of cardiac pacemaker cells:
weak electrical fields being produced by voltage changes (1) they cause an initial hyperpolarization of the resting mem-
occurring on individual cardiac cells. When a large number of brane potential by bringing it closer to the K+ equilibrium
cells are simultaneously depolarizing or repolarizing, large potential and (2) they slow the rate of spontaneous depolar-
voltages are observed on the electrocardiogram. Since the ization of the resting membrane. Both of these effects increase
electrical impulse spreads through the heart tissue in a consis- the time between beats by prolonging the time required for
CHAPTER 23 Cardiac Muscle Cells 217

decreases conduction velocity (has a negative dromotropic


effect). These effects are most notable at the AV node and
can influence the time between the P and R waves (the PR
interval).

Sympathetic tone
–10

tone
Membrane potential (mV)

pathetic
MECHANICAL PROPERTIES
Intrinsic
–30

Parasym
Contraction of the cardiac muscle cell is initiated by the action
potential acting on intracellular organelles to evoke tension
–50 generation and/or shortening of the cell. The reader is encour-
aged to carefully review the materials presented in Chapters 9
and 10 for specific cellular details describing contraction of
–70 skeletal and cardiac muscle.

Threshold potential

–90
EXCITATION–CONTRACTION COUPLING
Time The major event in excitation–contraction coupling in car-
FIGURE 23–4 Effect of sympathetic and parasympathetic diac muscle is a dramatic rise in the intracellular free Ca2+ con-
tones on pacemaker potential. (Modified with permission from Mohrman centration from less than 0.1 μM to as much as 100 μM. When
DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/ the wave of depolarization passes over the muscle cell mem-
McGraw-Hill, 2006.) brane, Ca2+ is released from the sarcoplasmic reticulum (SR)
into the intracellular fluid. The specific trigger is a small local-
ized increase in calcium concentration triggering a massive
release of calcium from the SR. Although the amount of Ca2+
the resting membrane to depolarize to the threshold level. that enters the cell during a single action potential is quite
Since there is normally some continuous tonic activity of car- small compared with that released from the SR, it is not only
diac parasympathetic nerves, the normal resting heart rate is essential for triggering the SR calcium release, but also essen-
approximately 70 beats/min. tial for maintaining adequate levels of Ca2+ in the intracellular
Sympathetic nerves release the transmitter substance nor- stores over the long run.
epinephrine on cardiac cells. In addition to other effects dis- The contractile process initiated by the increase in intracel-
cussed later, norepinephrine increases the inward currents lular calcium concentration has been described in Chapters 9
carried by Na+ (if ) and by Ca2+ during the diastolic interval. and 10. Recall that excitation–contraction coupling in cardiac
The signaling process involves norepinephrine interaction muscle differs from that in skeletal muscle in that it may be
with β1-adrenergic receptors on the SA nodal cell membrane modulated; different intensities of actin–myosin interaction
that in turn are linked to stimulatory G proteins, Gs. The acti- (contraction) can result from a single action potential trigger
vation of Gs increases adenylate cyclase, leading to an increase in cardiac muscle. The mechanism for this is largely depen-
in intracellular cyclic AMP that increases the open-state prob- dent on variations in the amount of Ca2+ reaching the myofila-
ability of the pacemaker Na+ current channel (if ). These ments and therefore the number of cross-bridges activated
changes increase heart rate by increasing the rate of diastolic during the twitch. This ability of cardiac muscle to vary its
depolarization as shown in Figure 23–4. contractile strength—that is, change its contractility—is
In addition to sympathetic and parasympathetic nerves, extremely important to cardiac function, as will be discussed
there are many (usually less important) factors that can alter later in this chapter.
heart rate. These include a number of ions and circulating hor-
mones, as well as physical influences such as temperature and
atrial wall stretch. All act by altering the time required for the RELAXATION
resting membrane to depolarize to the threshold potential. An
abnormally high concentration of Ca2+ in the extracellular The processes that participate in the reduction of intracellular
fluid, for example, tends to decrease heart rate by shifting the Ca2+ that terminates the contraction include (1) active uptake
threshold potential. Factors that increase heart rate are said to of ~80% of the calcium back into the SR by the action of
have a positive chronotropic effect. Those that decrease heart Ca2+-ATPase pumps, (2) active extrusion of ~5% of the cal-
rate have a negative chronotropic effect. cium from the cell via sarcolemmal Ca2+-ATPase pumps, and
An increase in sympathetic activity also increases action (3) passive exchange of ~15% of the calcium with extracellular
potential conduction velocity (has a positive dromotropic sodium via the Na+–Ca2+ exchanger located in the sarcolemma.
effect), whereas an increase in parasympathetic activity The Na+–Ca2+ exchanger is powered by the sodium gradient
218 SECTION V Cardiovascular Physiology

across the sarcolemma that in turn is maintained by the Na+/ CARDIAC MUSCLE CELL MECHANICS
K+-ATPase. This exchanger is electrogenic in that three Na+
ions move into the cell in exchange for each Ca2+ ion that As has been described in Chapters 9 and 10, the cross-bridge
moves out. This net inward movement of positive charge may interaction that occurs after a muscle is activated to contract
contribute to the maintenance of the plateau phase of the gives the muscle the potential to develop force and/or
action potential. The cardiac glycoside, digitalis, slows down shorten. Whether it does one, the other, or some combina-
the Na+/K+ pump and thus reduces the sodium gradient, tion of the two depends primarily on what is allowed to hap-
resulting in an increase in intracellular Ca2+ that gets seques- pen by the external constraints placed on the muscle during
tered into the SR. This mechanism contributes importantly to the contraction. Muscle cells in the ventricular wall operate
the positive effect of cardiac glycosides on the contractile force under different constraints during different phases of each
of the failing heart. cardiac cycle and undergo both isometric and isotonic con-
The duration of the cardiac muscle cell contraction is tractions.
approximately the same as that of its action potential. There-
fore, the electrical refractory period of a cardiac muscle cell is
not over until the mechanical response is completed. Mechan- ISOMETRIC CONTRACTIONS:
ical relaxation accompanies electrical repolarization. As a con- LENGTH–TENSION RELATIONSHIPS
sequence, heart muscle cells cannot be activated rapidly
enough to cause a fused (tetanic) state of prolonged contrac- Recall that the cardiac muscle maximum isometric contractile
tion. This is fortunate because intermittent contraction and force is strongly influenced by the initial length of the mus-
relaxation are essential for the heart’s pumping action. cle as is indicated in Figure 23–5. The top panel shows the

sarcomere

1 2
3 4
Resting Contracting 5 6
Resting Contracting
Resting Contracting

4 6
Muscle tension

2
Active tension
5 5

3 3
1 1
Stimulus Resting tension

Time

4
Peak isometric tension
Muscle tension

2 Active tension
FIGURE 23–5 Isometric contractions and
the effect of muscle length on resting tension Resting tension
and active tension development. (Modified with 3
permission from Mohrman DE, Heller LJ: Cardiovascular 1 Lmax
Physiology, 6th ed. New York: Lange Medical Books/
McGraw-Hill, 2006.) Muscle length
CHAPTER 23 Cardiac Muscle Cells 219

experimental arrangement for measuring muscle force at rest ISOTONIC & AFTERLOADED
and during contraction at three different lengths. The middle
panel shows time records of muscle tensions recorded at each
CONTRACTIONS
of the three lengths in response to an external stimulus, and During an isotonic (“fixed load”) contraction, muscle short-
the bottom panel shows a graph of the resting and peak ten- ens against a constant load as shown in Figure 23–6. When a
sion results plotted against muscle length. 1-g weight is suspended from a resting muscle, it will result in
The length-dependent influence on the resting tension of some specific resting muscle length, which is determined by
cardiac muscle is represented by the lower curve in the graph the muscle’s resting length–tension curve. If the muscle were
in Figure 23–5. When a muscle is stimulated to contract while to contract isometrically at this length, it would be capable of
its length is held constant (i.e., isometric contraction), it devel- generating a certain amount of tension, for example, 4.5 g as
ops active tension. The total tension exerted by a muscle dur- indicated by the dashed line in the graph of Figure 23–6. How-
ing contraction is the sum of the active and resting tensions ever, a contractile tension of 4.5 g will not be generated when
and is represented by the upper curve in Figure 23–5. Active lifting a 1-g weight. When a muscle has contractile potential in
tension development is shown to be maximal at some interme- excess of the tension it is actually developing, it shortens. Thus,
diate length referred to as Lmax. Normally, cardiac muscle oper- in an isotonic contraction, muscle length decreases at constant
ates at lengths well below Lmax, so that increasing muscle length tension, as illustrated by the horizontal arrow from point 1 to
increases the tension developed during an isometric contrac- point 3 in Figure 23–6. As the muscle shortens, however, its
tion. The mechanisms involved in the relationship between contractile potential inherently decreases, as indicated by the
cardiac muscle length and developed tension are discussed in downward slope of the peak isometric tension curve in Figure
Chapter 10. The important point is that the dependence of 23–6. There exists some short length at which the muscle is
active tension development and shortening on muscle length capable of generating only 1 g of tension, and when this length
is a fundamental property of cardiac muscle that has extremely is reached, shortening must cease. Thus, the curve on the car-
powerful effects on heart function. diac muscle length–tension diagram that indicates how much

Isotonic contraction Afterloaded contraction

Isometric Isotonic

3
5
2 1g 3-g total
1 4
1 1g 1g load
2-g 1g
1g 1g
afterload 2g
Resting Contracting 2g 2g

Resting
Contracting

5 Peak isometric tension


Muscle tension (g)

5
3 4

2 Resting tension

3 2
1
1

Muscle length

FIGURE 23–6 Relationship of isotonic and afterloaded contractions to the cardiac muscle length–tension diagram. (Modified with
permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
220 SECTION V Cardiovascular Physiology

isometric tension a muscle can develop at various lengths also A Isometric contraction
establishes the limit on how far muscle shortening can pro-
ceed with different loads. NE Peak isometric tension
ith
Figure 23–6 also shows an afterloaded isotonic contrac- W
5
tion, in which the load on the muscle at rest (the preload) and

Muscle tension (g)


the load on the muscle during contraction (the total load) are 4

NE
different. In the example of Figure 23–6, the preload is equal to

t
ou
3

it h
1 g, and because an additional 2-g weight (the afterload) is

E
engaged during contraction, the total load equals 3 g.

N
2
ut Resting tension
Since preload determines the resting muscle length, both i t ho
1 or w
isotonic contractions shown in Figure 23–6 begin from the With
same length. Because of the different loading arrangement,
however, the afterloaded muscle must increase its total ten- Muscle length
sion to 3 g before it can shorten. This initial tension will be
developed isometrically and can be represented as going
B Afterloaded contraction
from point 1 to point 4 on the length–tension diagram. Once
the muscle generates enough tension to equal the total load, Peak isometric tension
its tension output is fixed at 3 g and it will now shorten iso- NE
5 ith E
tonically because its contractile potential still exceeds its ten- W tN
ou
Muscle tension (g)
sion output. This isotonic shortening is represented as a 4 ith
W
horizontal movement on the length–tension diagram along
the line from point 4 to point 5. As in any isotonic contrac- 3
More shortening with NE Resting tension
tion, shortening must cease when the muscle’s tension-pro-

E
2 N
ducing potential is decreased sufficiently by the length ut
it ho
change to be equal to the load on the muscle. Note that the 1 or w
With
afterloaded muscle shortens less than the non-afterloaded
muscle even though both muscles began contracting at the
Muscle length
same initial length. Increases in afterload will further
decrease the shortening of the muscle. The factors that affect FIGURE 23–7 Effect of norepinephrine (NE) on isometric
the extent of cardiac muscle shortening during an afterloaded (A) and afterloaded (B) contractions of cardiac muscle. (Modified
with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New
contraction are of special interest because stroke volume is
York: Lange Medical Books/McGraw-Hill, 2006.)
determined by how far cardiac muscle shortens under these
conditions.

it enhances the forcefulness of muscle contraction even when


CARDIAC MUSCLE CONTRACTILITY length is constant. Changes in contractility and initial length
can occur simultaneously, but by definition, a change in con-
A number of factors in addition to initial muscle length can tractility must involve a shift from one peak isometric length–
affect the tension-generating potential of cardiac muscle. Any tension curve to another.
intervention that increases the peak isometric tension that a Figure 23–7B shows how raising the peak isometric length–
muscle can develop at a fixed length is said to increase cardiac tension curve with norepinephrine increases the amount of
muscle contractility. Such an agent is said to have a positive shortening in afterloaded contractions of cardiac muscle. With
inotropic effect on the heart. preload and total load constant, more shortening occurs in the
The most important physiological regulator of cardiac mus- presence of norepinephrine than in its absence. This is because
cle contractility is norepinephrine. When norepinephrine is when contractility is increased, the tension-generating poten-
released on cardiac muscle cells from sympathetic nerves, it tial is equal to the total load at a shorter muscle length. Note
has not only the chronotropic effect on heart rate discussed that norepinephrine has no effect on the resting length–tension
earlier, but also a pronounced positive inotropic effect that relationship of cardiac muscle. Thus, norepinephrine causes
causes cardiac muscle cells to contract more rapidly and increased shortening by changing the final but not the initial
forcefully. muscle length associated with afterloaded contractions.
The positive effect of norepinephrine on the isometric ten- The cellular mechanism of the norepinephrine effect on
sion-generating potential is illustrated in Figure 23–7A. When contractility is mediated by its interaction with a β1-adrenergic
norepinephrine is present, cardiac muscle will, at every length, receptor. The signaling pathway involves an activation of the
develop more isometric tension, thus raising the peak isomet- Gs protein–cAMP–protein kinase A, which then phosphory-
ric tension curve on the cardiac muscle length–tension graph. lates the Ca2+ channel, increasing the inward calcium current
Norepinephrine increases cardiac muscle contractility because during the plateau of the action potential. This increase in
CHAPTER 23 Cardiac Muscle Cells 221

calcium influx not only contributes to the magnitude of the the length of the individual cardiac muscle cells. Thus, the
increase in intracellular Ca2+ for a given beat, but also loads the extent of diastolic filling of the ventricle determines the
internal calcium stores, which allows more to be released “preload.”
during subsequent depolarizations. This increase in free Ca2+ 2. At any given ventricular volume, an increase in the ten-
during activation allows more cross-bridges to be formed, sion of individual cardiac muscle cells in the wall causes
increases the speed of cross-bridge turnover, and allows greater an increase in intraventricular pressure.
tension to be developed at a faster rate. 3. As ventricular volume decreases (i.e., as the ventricular
Because norepinephrine also causes phosphorylation of the radius decreases), a lesser total (collective) force is re-
regulatory protein, phospholamban, on the sarcoplasmic quired by the muscle cells in the ventricular walls to pro-
reticular Ca2+-ATPase pump, the rate of calcium retrapping duce any given intraventricular pressure.
into the SR is enhanced and the rate of relaxation is also
The last point is a reflection of the law of Laplace that states
increased. This is called a positive lusitropic effect. In addi-
the physical relationship that must exist between total wall
tion to more rapid calcium retrapping by the SR, there is also a
tension and internal pressure in any hollow vessel with circu-
norepinephrine-induced decrease in the action potential dura-
lar containing walls. Regardless of whether the ventricle is
tion. This effect is achieved by a potassium channel alteration,
envisioned as a hollow cylinder or a hollow sphere, the law of
occurring in response to the elevated intracellular [Ca2+] that
Laplace says that the total wall tension (T) depends on both
increases potassium permeability, terminates the plateau phase
intraventricular pressure (P) and its internal radius (r) as fol-
of the action potential, and contributes to the early relaxation.
lows: T = P × r.
(Such shortening of the systolic interval is helpful in the pres-
One implication of the law of Laplace is that the muscle
ence of elevated heart rates that might otherwise significantly
cells in the ventricular wall have a somewhat easier job of
compromise diastolic filling time.)
producing internal pressure at the end of systole (when the
Enhanced parasympathetic activity has been shown to have
radius is small) than at the beginning of systole (when the
a small negative inotropic effect on the heart. In the atria,
radius is large). More importantly, the law of Laplace has
where this effect is most pronounced, the negative inotropic
significant clinical relevance in some pathological
effect is thought to be due to a decrease in the duration of the
situations.
action potential and a decrease in the amount of Ca2+ that
enters the cell during the action potential.
Changes in heart rate also influence cardiac contractility.
Recall that a small amount of extracellular Ca2+ enters the cell
during the plateau phase of each action potential. As the heart CLINICAL CORRELATION
rate increases, more Ca2+ enters the cells per minute. There is a
buildup of intracellular Ca2+ and a greater amount of Ca2+ is An elderly man is brought to the emergency room by his
released into the sarcoplasm with each action potential. Thus, daughter. She reports that he has recently complained of
a sudden increase in beating rate is followed by a progressive severe weakness, fatigue, some dizziness, and seems to be
increase in contractile force to a higher plateau (the force– a bit confused. This condition appeared only a few days
frequency relationship). ago and does not seem to be getting better or worse. He
has been healthy otherwise and usually likes to square
dance. The patient is alert and responsive but very weak
and pale. His blood pressure on admission is 100/60 mm Hg
RELATING CARDIAC and his heart rate is 41 beats/min. An electrocardiogram
taken at the time of admission verifies the bradycardia
MUSCLE CELL MECHANICS (slow heart rate) with a ventricular rate of 41 beats/min
TO VENTRICULAR FUNCTION and atrial rate at 95 beats/min. There are no ECG signs of
cardiac ischemia or infarction but the occurrence of
Certain geometric factors dictate how the length–tension rela- P waves is very rapid and not correlated with slow occur-
tionships of cardiac muscle fibers in the ventricular wall deter- rence of QRS waves.
mine the volume and pressure relationships of the ventricular This patient has a third degree (total) AV nodal heart
chamber. The actual relationships are complex because the block in which the normal conduction of action potentials
shape of the ventricle is complex. The ventricle is often mod- originating in the SA node is not conducted through the
eled as either a cylinder or a sphere, although its actual shape AV node to the ventricles. The ventricles are being driven
lies somewhere between the two. Because cardiac muscle cells at an "escape" rhythm established by a pacemaker located
are oriented circumferentially in the ventricular wall, either below the AV node firing at a rate that is significantly
model can be used to illustrate three important functional slower than that of SA nodal pacemaker cells. The atria
points: are beating much faster due to sympathetic activation
1. An increase in ventricular volume causes an increase in (triggered by the low blood pressure) but the signals are
ventricular circumference and therefore an increase in
222 SECTION V Cardiovascular Physiology

not conducted through the AV nodal block. His symp-


STUDY QUESTIONS
toms of weakness, fatigue, dizziness, and confusion are a 1. A drug that promotes early activation of the “delayed rectifier”
result of his low blood pressure that, in turn, is a result of K+ channel (Ik) in cardiac muscle will do which of the following?
his low ventricular rate and reduced cardiac output. A) The resting potential will be increased (hyperpolarized).
Causes include drug toxicities (i.e., β-adrenergic blockers, B) The action potential’s duration will be decreased.
calcium channel blockers), metabolic disturbances (i.e., C) The action potential’s peak amplitude will be decreased.
hyperkalemia), anterior wall myocardial infarctions D) The action potential conduction velocity will be increased.
E) The absolute refractory period will be prolonged.
(i.e., septal ischemia), and cardiomyopathy (i.e., from a
viral infection). Diagnostic tests will be conducted to 2. Action potential conduction velocity in cardiac muscle tissue
is influenced by all of the following except
determine what might be responsible for his condition.
His treatment will involve implantation of a cardiac A) cell diameter.
B) resting membrane potential.
pacemaker. This is a subcutaneously implanted battery
C) extracellular potassium concentration.
pack that sends repetitive stimuli through wire electrodes D) rate of rise (phase 0) of the action potential.
positioned in both the right atrium and right ventricle via E) duration of the plateau phase (phase 2) of the action
the vena cava. This dual-chamber pacemaker will syn- potential.
chronize his atrial and ventricular beating rate at a more 3. The primary route of removal of [Ca2+] from the sarcoplasm
normal rate and rhythm and perhaps restore circulatory during relaxation of a cardiac muscle cell is by
status and his activity levels back to normal. A) active transport out of the cell.
B) passive exchange with extracellular sodium.
C) active transport into the sarcoplasmic reticulum.
D) trapping of calcium by troponin in the myofilaments.
CHAPTER SUMMARY E) passive movement out of the cell via L-type calcium
channels.
■ Cardiac myocyte membrane potentials are a result of the
relative permeability of the membrane to various ions and their 4. A therapeutic strategy to improve the amount of active
concentration differences across the membrane. shortening of cardiac muscle might include which one of
the following?
■ Action potentials of cardiac myocytes are a result of changes in
the membrane permeability to various ions. A) decrease preload
B) decrease afterload
■ Action potentials of cardiac myocytes have long plateau phases
C) decrease contractility
that generate long refractory periods and preclude summated
D) give a negative chronotropic agent
or tetanic contractions.
E) give a negative inotropic agent
■ Action potentials are spontaneously generated by pacemaker
5. Your patient has accidentally been given a large bolus injection
cells in the SA node and are conducted from cell to cell via gap
of potassium chloride and dies. Why?
junctions throughout the entire heart.
A) Cardiac muscle is depolarized and his heart has stopped
■ The rate of spontaneous diastolic depolarization of the SA
in diastole.
nodal cells (and thus the heart rate) is modulated by the
B) Cardiac muscle is depolarized and his heart has stopped
autonomic nervous system.
in systole.
■ Excitation of the cardiac myocyte initiates contraction by C) Cardiac muscle is hyperpolarized and his heart has stopped
increasing the cytostolic calcium concentration that activates in diastole.
the contractile apparatus. D) Action potential conduction is sped up and heart
■ Mechanical response of the myocyte depends on preload is fibrillating.
(determined by the initial resting length), afterload (deter- E) Gap junctions between cardiac muscle cells are disrupted.
mined by the tension that must be developed), and contractility
(the degree of activation of the contractile apparatus dependent
on the amount of calcium released on activation).
■ The cardiac myocyte length–tension relationships are
correlated with changes in volume and pressure in the
intact ventricle.
24
C H A P T E R

The Heart Pump


Lois Jane Heller and David E. Mohrman

O B J E C T I V E S

■ Correlate the electrocardiographic events with the mechanical events during


the cardiac cycle.
■ List the major distinct phases of the cardiac cycle as delineated by valve
opening and closure.
■ Describe the pressure and volume changes in the atria, the ventricles, and the
aorta during each phase of the cardiac cycle.
■ Define and state normal values for (1) ventricular end-diastolic volume,
end-systolic volume, stroke volume, diastolic pressure, and peak systolic
pressure, and (2) aortic diastolic pressure, systolic pressure, and pulse
pressure.
■ State similarities and differences between mechanical events in the left and
right heart pumps.
■ State the origin of the heart sounds.
■ Diagram the relationship between left ventricular pressure and volume
during the cardiac cycle.
■ Define cardiac output and cardiac index.
■ State the relationship between cardiac output, heart rate, and stroke volume.
■ Identify the major determinants of stroke volume.
■ State Starling’s law of the heart.
■ Predict the effect of altered ventricular preload on stroke volume and the
ventricular pressure–volume relationship.
■ Predict the effect of altered ventricular afterload on stroke volume and the
ventricular pressure–volume relationship.
■ Predict the effect of altered ventricular contractility (inotropic state) on stroke
volume and the ventricular pressure–volume relationship.
■ Draw a family of cardiac function curves describing the relationship
between filling pressure and cardiac output under various levels of
sympathetic tone.
■ Summarize the effect of sympathetic neural stimulation on cardiac
function.
■ List the determinants of myocardial oxygen consumption.

223

Ch24_223-234.indd 223 11/26/10 10:00:10 AM


224 SECTION V Cardiovascular Physiology

This chapter describes (1) the basic mechanical features of VENTRICULAR DIASTOLE
the cardiac pump, (2) the factors that influence and/or regu-
late cardiac output, and (3) the sources of energy and the The diastolic phase of the cardiac cycle begins with the open-
energy costs required for myocardial activity. ing of the atrioventricular (AV) valves. (Unless otherwise
noted, systole and diastole denote phases of ventricular
operation.) As shown in Figure 24–1, the mitral valve opens
CARDIAC CYCLE when left ventricular pressure decreases below left atrial pres-
sure and the period of ventricle filling begins. Blood that had
LEFT PUMP previously accumulated in the atrium behind the closed mitral
valve empties rapidly into the ventricle and this causes an ini-
The mechanical function of the heart can be described by the tial decrease in atrial pressure. Later, the pressures in both
pressure, volume, and flow changes that occur within it during chambers slowly increase together as the atrium and ventricle
one cardiac cycle. A cardiac cycle is defined as one complete continue passively filling with blood returning to the heart
sequence of contraction and relaxation. The normal mechani- through the veins.
cal events of a cycle of the left heart pump are correlated in Atrial contraction is initiated near the end of ventricular
Figure 24–1. This important figure summarizes a great deal of diastole by the depolarization of the atrial muscle cells, which
information and should be studied carefully. causes the P wave of the electrocardiogram. As the atrial

A B
Cardiac cycle C D E
phase

R
Lead II P T
electrocardiogram

Atrial
Muscle contraction
Ventricular
Incisura
Systolic pressure
120 Aortic
pressure
Pulse pressure

80 Diastolic pressure
Pressure (mm Hg)
Left ventricular pressure
40 Left atrial
pressure

Closed Closed
Aortic valve Open
Closed Open
Mitral valve Open
S3 S4 S1 S2 S3
Heart sounds

120
End-diastolic volume Stroke
Left ventricular volume volume
(mL)
End-systolic volume
60
FIGURE 24–1 Cardiac cycle—left heart.
Cardiac cycle phases: A) diastole; B) systole that is
divided into three periods; C) isovolumetric Outflow
contraction; D) ejection, and E) isovolumetric Aortic flow 0 Inflow
relaxation. (Modified with permission from Mohrman DE,
Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange 0 0.2 0.4 0.6 0.8
Medical Books/McGraw-Hill, 2006.) Time (s)
CHAPTER 24 The Heart Pump 225

muscle cells develop tension and shorten, atrial pressure rises pressure decreases below atrial pressure, the AV valve opens,
and an additional amount of blood is forced into the ventricle. and a new cardiac cycle begins.
At normal heart rates, atrial contraction is not essential for Note that atrial pressure progressively increases during ven-
adequate ventricular filling. This is evident in Figure 24–1 tricular systole because blood continues to return to the heart
showing that the ventricle has nearly reached its maximum and fill the atrium. The increased atrial pressure at the end of
(end-diastolic volume) before atrial contraction begins. Atrial systole promotes rapid ventricular filling once the AV valve
contraction plays an increasingly significant role in ventricu- opens to begin the next heart cycle.
lar filling as heart rate increases because the time interval The ventricle has reached its minimum (end-systolic
between beats for passive filling becomes progressively shorter. volume) at the time of aortic valve closure. The amount of
Note that throughout diastole, atrial and ventricular pressures blood ejected from the ventricle during a single beat, the
are almost the same. This is because a normal open mitral stroke volume, is equal to ventricular end-diastolic volume
valve has very little resistance to flow and thus only a very minus ventricular end-systolic volume.
small atrial–ventricular pressure difference is necessary to During the early, most rapid phase of systolic ejection, the
produce ventricular filling. aorta distends because more blood is being put into it from the
left heart than is leaving it to the systemic organs. That disten-
sion is caused by the increasing pressure within the aorta.
VENTRICULAR SYSTOLE During the later, weakening phase of cardiac ejection,
the opposite is true. The overall result is that the aortic pres-
Ventricular systole begins when the action potential breaks sure reaches a maximum value (systolic pressure) near the
through the AV node and sweeps over the ventricular muscle middle of ventricular systole. During diastole, the arterial
resulting in the QRS complex of the electrocardiogram. Con- pressure is maintained by the elastic recoil of walls of the aorta
traction of the ventricular muscle cells causes intraventricular and other large arteries. Nonetheless, aortic pressure gradually
pressure to increase above that in the atrium, which causes decreases during diastole as the aorta supplies blood to the
abrupt closure of the AV valve. systemic vascular beds. The lowest aortic pressure, reached at
Pressure in the left ventricle continues to increase sharply as the end of diastole, is called diastolic pressure. The difference
the ventricular contraction intensifies. When the left ventricu- between diastolic and peak systolic pressure in the aorta is
lar pressure exceeds that in the aorta, the aortic valve opens. called the arterial pulse pressure. Typical values for systolic
The period of time between mitral valve closure and aortic and diastolic pressures in the aorta are 120 and 80 mm Hg,
valve opening is referred to as the isovolumetric contraction respectively, with a pulse pressure of 40 mm Hg.
phase because, during this interval, the ventricle is a closed At a normal resting heart rate of about 70 beats/min, the
chamber with a fixed volume. Ventricular ejection begins with heart spends approximately two thirds of the cardiac cycle in
the opening of the aortic valve. In early ejection, blood enters diastole and one third in systole. When increases in heart rate
the aorta rapidly and causes the pressure there to rise. Pressure occur, both diastolic and systolic intervals become shorter.
builds simultaneously in both the ventricle and the aorta as Action potential durations are shortened and conduction
the ventricular muscle cells continue to contract in early sys- velocity is increased. Contraction and relaxation rates are also
tole. This interval is often called the rapid ejection period. enhanced. This shortening of the systolic interval tends to
Left ventricular and aortic pressures ultimately reach a max- blunt the potential adverse effects of increases in heart rate on
imum called peak systolic pressure. At this point, the strength diastolic filling time.
of ventricular muscle contraction begins to wane. Muscle
shortening and ejection continue, but at a reduced rate. Aortic
pressure begins to decrease because blood is leaving the aorta RIGHT PUMP
and large arteries faster than blood is entering from the left
ventricle. Throughout ejection, there are very small pressure Because the entire heart is served by a single electrical excita-
differences between the left ventricle and the aorta because the tion system, similar mechanical events occur almost simulta-
aortic valve orifice is so large that it presents very little resis- neously in both the left heart and the right heart. Both ventricles
tance to flow. have synchronous systolic and diastolic periods and the valves
Eventually, the strength of the ventricular contraction of the right and left heart normally open and close nearly in
diminishes to the point where intraventricular pressure unison. Because the two sides of the heart are arranged in
decreases below aortic pressure. This causes abrupt closure of series in the circulation, they must pump the same amount of
the aortic valve. A dip, called the incisura or dicrotic notch, blood and therefore must have the same cardiac output.
appears in the aortic pressure trace because a small volume of The major difference between the right and left pumps is in
aortic blood must flow backward to fill the aortic valve leaf- the magnitude of the peak systolic pressure. The pressures
lets as they close. After aortic valve closure, intraventricular developed by the right heart as shown in Figure 24–2 are
pressure decreases rapidly as the ventricular muscle relaxes. considerably lower than those for the left heart (Figure 24–1).
For a brief interval, called the isovolumetric relaxation phase, This is because the pulmonary vessels provide considerably
the mitral valve is also closed. Ultimately, intraventricular less resistance to blood flow than that offered collectively by
226 SECTION V Cardiovascular Physiology

vibrations of the cardiac structures and the blood in the


ventricular chambers. S1 can be heard most clearly by placing
the stethoscope over the apex of the heart. Note that this
Lead II electrocardiogram
sound occurs immediately after the QRS complex of the
electrocardiogram.
25 Pulmonary
artery The second heart sound, S2, arises from the closure of the
aortic and pulmonic valves at the beginning of the period of
Pressure (mm Hg)

20 pressure
isovolumetric relaxation. This sound is heard at about the time
15 of the T wave in the electrocardiogram. The pulmonic valve
Right
10 ventricular usually closes slightly after the aortic valve. Because this dis-
Right atrial a pressure crepancy is enhanced during the inspiratory phase of the
5 pressure c v
respiratory cycle, inspiration causes what is referred to as the
0 physiological splitting of the second heart sound. The dis-
crepancy in valve closure during inspiration may range from
0.0 0.2 0.4 0.6 0.8 1.0
30 to 60 milliseconds. One of the factors that leads to pro-
Time (s)
longed ejection of the right ventricle during inspiration is that
FIGURE 24–2 Cardiac cycle—right heart. (Modified with the decreased intrathoracic pressure that accompanies
permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York:
inspiration transiently enhances venous return and diastolic
Lange Medical Books/McGraw-Hill, 2006.)
filling of the right heart. For reasons that will be detailed later
in this chapter, this extra filling volume will be ejected but a
little extra time is required.
those of the systemic organs. Therefore, less arterial pressure is The third and fourth heart sounds, shown in Figure 24–1,
required to drive the cardiac output through the lungs than are not normally present. When they are present, however,
through the systemic organs. Typical pulmonary artery sys- they, along with S1 and S2, produce what are called gallop
tolic and diastolic pressures are 25 and 8 mm Hg, respectively. rhythms (resembling the sound of a galloping horse). When
The pressure pulsations that occur in the right atrium are present, the third heart sound occurs shortly after S2 during
transmitted in retrograde fashion to the large veins near the the period of rapid passive ventricular filling and, in
heart. These pulsations, shown on the atrial pressure trace of combination with heart sounds S1 and S2, produces what is
Figure 24–2, can be visualized in the neck over the jugular called ventricular gallop rhythm. Although S3 may sometimes
veins in a recumbent individual. They are collectively referred be detected in normal children, it is heard more commonly in
to as the jugular venous pulse, and can provide clinically use- patients with left ventricular failure. The fourth heart sound,
ful information about the heart. Atrial contraction produces which occasionally is heard shortly before S1, is associated
the first pressure peak, called the a wave. The c wave, which with atrial contraction and rapid active filling of the ventricle.
follows shortly thereafter, coincides with the onset of ventricu- Thus, the combination of S1, S2, and S4 sounds produces what
lar systole and is caused by an initial bulging of the tricuspid is called an atrial gallop rhythm. The presence of S4 often
valve into the right atrium. Right atrial pressure decreases after indicates an increased ventricular diastolic stiffness, which
the c wave because of atrial relaxation and a downward dis- can occur with several cardiac disease states.
placement of the tricuspid valve during ventricular emptying.
Right atrial pressure then begins to increase toward a third
peak, the v wave, as the central veins and right atrium fill CARDIAC CYCLE PRESSURE–VOLUME
behind a closed tricuspid valve with blood returning to the
heart from the peripheral organs. With the opening of the tri-
& LENGTH–TENSION RELATIONSHIPS
cuspid valve at the conclusion of ventricular systole, right Intraventricular pressure and volume are intimately linked to
atrial pressure again decreases as blood moves into the relaxed the tension and length of the cardiac muscle cells in the ven-
right ventricle. Shortly afterward, right atrial pressure begins tricular wall. Figure 24–3A and B shows the correspondence
to increase once more toward the next a wave as returning between a ventricular pressure–volume loop and a cardiac
blood fills the central veins, the right atrium, and right ven- muscle length–tension loop during one cardiac cycle. It is clear
tricle together during diastole. that cardiac muscle length–tension behavior is the underlying
basis for ventricular function. Each major phase of the ventric-
HEART SOUNDS ular cardiac cycle has a corresponding phase of cardiac muscle
length and tension change. During diastolic ventricular filling,
A record of the heart sounds, which occur in the cardiac cycle, for example, the progressive increase in ventricular pressure
is included in Figure 24–1. These sounds are normally heard stretches the resting cardiac muscle to greater lengths along its
by auscultation with a stethoscope placed on the chest. The resting length-tension curve and causes a corresponding
first heart sound, S1, occurs at the beginning of systole because increase in muscle tension. End-diastolic ventricular pressure
of the abrupt closure of the AV valves, which produces is referred to as ventricular preload because it sets the end-
CHAPTER 24 The Heart Pump 227

A
120 Ejection
Intraventricular pressure (mm Hg)

Reaches end-
systolic volume

80 Aortic valve opens

Systole

Isovolumetric
relaxation Isovolumetric contraction

Mitral valve
opens Diastolic filling
Reaches end-diastolic volume

60 130
Stroke volume
Intraventricular volume (mL)

Shortening
Muscle tension

Reaches end-
systolic length
Active Isometric tension development

Isometric relaxation
Passive stretch Reaches end-diastolic length FIGURE 24–3 Ventricular pressure–volume
cycle (A), and corresponding cardiac muscle
length–tension cycle (B). (Modified with permission
from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th
Muscle length ed. New York: Lange Medical Books/McGraw-Hill, 2006.)

diastolic ventricular volume and therefore the resting length of against which they are shortening. Once shortening ceases
the cardiac muscle fibers at the end of diastole. and the output valve closes, the cardiac muscle cells relax
At the onset of systole, the ventricular muscle cells develop isometrically. Ventricular wall tension and intraventricular
tension isometrically (during the isovolumetric contraction pressure decrease in unison during isovolumetric relaxation.
phase) and intraventricular pressure increases accordingly.
After the intraventricular pressure increases sufficiently to
open the outlet valve, ventricular ejection begins as a conse- DETERMINANTS OF
quence of ventricular muscle shortening. Systemic arterial CARDIAC OUTPUT
pressure is often referred to as the ventricular afterload
because it determines the tension that must be developed by The cardiac output (liters of blood pumped by each of the
cardiac muscle fibers before they can shorten. It should be ventricles per minute) is an extremely important cardiovascu-
noted that other factors that influence the actual wall tension lar variable that is continuously adjusted so that the cardiovas-
required to eject blood from the ventricle (such as end-diastolic cular system operates to meet the body’s moment-to-moment
volume, velocity of contraction, viscosity of the blood) do con- circulatory needs. In going from rest to strenuous exercise, for
tribute to ventricular afterload but we choose to ignore them example, the cardiac output of an average person will increase
at this point. from approximately 5.5 to perhaps 15 L/min. The extra car-
During cardiac ejection, cardiac muscle is simultaneously diac output provides the exercising skeletal muscles with the
generating active tension and shortening (i.e., an afterloaded additional nutritional supply needed to sustain an increased
isotonic contraction). The stroke volume is determined by metabolic rate. To understand the cardiovascular system’s
how far ventricular muscle cells are able to shorten during response not only to exercise, but also to all other physiologi-
contraction. This, as already discussed, depends on the length– cal or pathological demands placed on it, we must understand
tension relationship of the cardiac muscle cells and the load what determines, and therefore controls, cardiac output.
228 SECTION V Cardiovascular Physiology

As stated in Chapter 22, cardiac output is the product of A


heart rate and stroke volume (CO = HR × SV). Therefore, all
5
changes in cardiac output must be produced by changes in

Muscle tension (g)


Peak isometric
heart rate and/or stroke volume. 4 tension
Factors influencing heart rate do so by altering the charac-
3
teristics of the diastolic depolarization of the pacemaker cells More shortening
as discussed in Chapter 23 (see Figure 23–4). Recall that varia- 2
tions in activity of the sympathetic and parasympathetic nerves Resting
tension
leading to cells of the sinoatrial (SA) node constitute the most 1
Larger preload
important regulators of heart rate. Increases in sympathetic
activity increase heart rate, whereas increases in parasympa- Muscle length
thetic activity decrease heart rate. These neural inputs have
immediate effects (within one beat) and therefore can cause B
very rapid adjustments in cardiac output. 120

LV pressure (mm Hg)


INFLUENCES ON STROKE VOLUME 80
More
EFFECT OF CHANGES IN 40
stroke volume

VENTRICULAR PRELOAD: Larger


STARLING’S LAW OF THE HEART ventricular
preload

The volume of blood that the heart ejects with each beat can 60 120
vary significantly. One of the most important factors respon- LV volume (mL)
sible for these variations in stroke volume is the extent of car- FIGURE 24–4 Effect of changes in preload on cardiac
diac filling during diastole. This concept was introduced in muscle shortening during afterloaded contractions (A) and
Chapter 22 (Figure 22–7) and is known as Starling’s law of the on ventricular stroke volume (B). (Modified with permission from
heart. To review (and to reemphasize its importance), this law Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange
states that, with other factors equal, stroke volume increases as Medical Books/McGraw-Hill, 2006.)

cardiac filling increases. As shown below, this phenomenon is


based on the intrinsic mechanical properties of myocardial
muscle. One form of heart failure is called diastolic failure and is
Figure 24–4A illustrates how increasing muscle preload will characterized by an abnormally stiff ventricle and a distur-
increase the extent of shortening during a subsequent contrac- bance in the relationship between cardiac filling pressure and
tion with a fixed total load. Recall from the nature of the rest- end-diastolic volume. In this situation, diastolic filling is lim-
ing length–tension relationship that an increased preload is ited, stroke volume is reduced, cardiac output is inadequate,
necessarily accompanied by increased initial muscle fiber and function of the cardiovascular system is compromised.
length. As was described in Chapter 23, when a muscle starts It should be noted in Figure 24–4A that increasing preload
from a greater length, it has more distance to shorten before it increases initial muscle length without significantly changing
reaches the length at which its tension-generating capability is the final length to which the muscle shortens against a con-
no longer greater than the load upon it. Cardiac muscle cells stant total load. Thus, increasing ventricular filling pressure
exhibit the same behavior when they are operating in the ven- increases stroke volume primarily by increasing end-diastolic
tricular wall. Increases in ventricular preload increase both volume. As shown in Figure 24–4B, this is not accompanied by
end-diastolic volume and stroke volume almost equally, as a significant alteration in end-systolic volume.
illustrated in Figure 24–4B.
The precise relationship between cardiac preload (cardiac
filling pressure) and end-diastolic volume has especially EFFECT OF CHANGES IN
important physiological and clinical consequences. Although VENTRICULAR AFTERLOAD
the relationship is somewhat curvilinear, especially at very
high filling pressures, it is nearly linear over the normal oper- Figure 24–5A shows how increased afterload, at constant pre-
ating range of the heart. The low slope of this relationship load, has a negative effect on cardiac muscle shortening
indicates the incredible distensibility of the normal ventricle because muscle cannot shorten beyond the length at which its
during diastole. (For example, a change in filling pressure of peak isometric tension-generating potential equals the total
only 1 mm Hg normally will change end-diastolic volume by load upon it. Thus, shortening must stop at a greater muscle
about 25 mL!) length when afterload is increased.
CHAPTER 24 The Heart Pump 229

A EFFECT OF CHANGES IN CARDIAC


Peak isometric
MUSCLE CONTRACTILITY
5
Muscle tension (g)

tension
4 Less shortening Recall that activation of the sympathetic nervous system
results in release of norepinephrine from cardiac sympathetic
3 nerves, which increases contractility of the individual cardiac
Resting
tension
muscle cells. This results in an upward shift of the peak iso-
2
Larger total load metric length–tension curve. As shown in Figure 24–6A, such
1 a shift will result in an increase in the shortening of a muscle
contracting with constant preload and total load. Thus, as
shown in Figure 24–6B, the norepinephrine released by sym-
Muscle length
pathetic nerve stimulation will increase ventricular stroke vol-
ume by decreasing the end-systolic volume without directly
B influencing the end-diastolic volume.
120 In addition to these changes in the extent of myocyte short-
ening, an increase in contractility will also cause an increase in
LV pressure (mm Hg)

the rates of myocyte tension development and of shortening.


80 This will result in an increase in the rate of isovolumetric pres-
Lower Larger sure development (dP/dt) and the rate of ejection during
stroke volume ventricular systole.
40 afterload Systolic failure is characterized by a severely depressed abil-
ity of the cardiac muscle cells to produce tension and shorten.
In this situation, systolic contraction is limited, stroke volume

60 120
LV volume (mL) A Afterloaded contraction
FIGURE 24–5 Effect of changes in afterload on cardiac Peak isometric
muscle shortening during afterloaded contractions (A) and on NE tension
h
ventricular stroke volume (B). Dashed line shows how stroke 5 NE
it
t
Muscle tension (g)

ou
volume is decreased by an increase in afterload. Dotted line shows ith
4
W

the relationship between end-systolic pressure and end-systolic


volume obtained at a constant preload but different afterloads. 3
(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, More shortening N
E Resting tension
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) 2 ut
wi th o
1 or
With
Normally, mean ventricular afterload is nearly constant,
because mean arterial pressure is held within tight limits by Muscle length
the cardiovascular control mechanisms described in
B
Chapter 29. In many pathological situations such as hyperten-
Wi
sion (high blood pressure) and aortic valve obstruction, ven- th
120 NE
tricular function is adversely influenced by abnormally high
LV pressure (mm Hg)

With
o ut
ventricular afterload. When this occurs, stroke volume may be N E
decreased as shown by the changes in the pressure–volume
80
loop indicated by the dashed line in Figure 24–5B. Under these Increased
conditions, note that stroke volume is decreased because end- stroke volume
systolic volume is increased. 40
The relationship between end-systolic pressure and
end-systolic volume obtained at a constant preload but different
afterloads is indicated by the dotted line in Figure 24–5B. In a
normally functioning heart, the effect of changes in afterload 60 120
on end-systolic volume (and therefore stroke volume) is quite LV volume (mL)
small (about 0.5 mL/mm Hg). However, in systolic cardiac FIGURE 24–6 Effect of norepinephrine (NE) on afterloaded
failure, the end-systolic pressure-volume line is shifted down- contractions on cardiac muscle (A) and on ventricular stroke
ward and is flattened so that the effect of afterload on end- volume (B). (Modified with permission from Mohrman DE, Heller LJ:
systolic volume is greatly exaggerated. Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
230 SECTION V Cardiovascular Physiology

Negative chronotropic
CARDIAC PARASYMPATHETIC NERVE ACTIVITY LEVEL –
HEART RATE
+
Positive chronotropic

CARDIAC SYMPATHETIC NERVE ACTIVITY LEVEL


+
CARDIAC OUTPUT
Contractility (positive inotropic) +

ARTERIAL PRESSURE

FIGURE 24–7 Influences on cardiac


Afterload
– STROKE VOLUME
FILLING PRESSURE
output. (Modified with permission from Mohrman DE,
Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Preload
+
Medical Books/McGraw-Hill, 2006.) (Starling’s law)

is reduced, cardiac output is inadequate, and function of the CARDIAC FUNCTION CURVES
cardiovascular system is compromised.
One very useful way to summarize the influences on cardiac
function and the interactions between them is by cardiac
SUMMARY OF DETERMINANTS function curves such as those shown in Figure 24–8. Cardiac
filling pressure (“cardiac preload”) is plotted as the independent
OF CARDIAC OUTPUT variable on the horizontal axis in this figure and cardiac out-
put as the dependent variable on the vertical axis. Each curve
The major influences on cardiac output are summarized in
in this figure shows the effect of changes in cardiac preload on
Figure 24–7. Heart rate is controlled by chronotropic influ-
ences on the spontaneous electrical activity of SA nodal cells.
Cardiac parasympathetic nerves have a negative chronotro-
pic effect, and sympathetic nerves have a positive chronotro-
Activity levels of
pic effect on the SA node. Stroke volume is controlled by cardiac sympathetic nerves
influences on the contractile performance of ventricular car- Greatly increased
diac muscle—in particular its degree of shortening in the
afterloaded situation. The three distinct influences on stroke
volume are contractility, preload, and afterload. Increased car- 10.0 Moderately increased
diac sympathetic nerve activity tends to increase stroke vol-
Cardiac output (L /min)

ume by increasing the contractility of cardiac muscle (a


8.0 Normal
positive inotropic effect). Increased arterial pressure tends to
decrease stroke volume by increasing the afterload on cardiac C B
muscle fibers. Increased ventricular filling pressure increases 6.0
end-diastolic volume, which tends to increase stroke volume Decreased
through Starling’s law of the heart. A
It is important to recognize at this point that both heart 4.0
rate and stroke volume are subject to more than one influ-
ence. Thus, the fact that increased contractility tends to
increase stroke volume should not be taken to mean that, in 2.0
the intact cardiovascular system, stroke volume is always high
when contractility is high. Following blood loss caused by
hemorrhage, for example, stroke volume may be low in spite 0 2.0 4.0 6.0
of a high level of sympathetic nerve activity and increased Cardiac filling pressure (mm Hg)
contractility. Because arterial pressure is normal or low fol- FIGURE 24–8 Influence of cardiac sympathetic nerves on
lowing hemorrhage, the low stroke volume associated with cardiac function curves. (Modified with permission from Mohrman DE,
severe blood loss must be (and is) the result of low cardiac Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/
filling pressure. McGraw-Hill, 2006.)
CHAPTER 24 The Heart Pump 231

cardiac output at one constant level of cardiac sympathetic 2. a decrease in cardiac action potential duration by early
nerve activity. Different curves are used for different levels of activation of the delayed iK current in cardiac myocytes,
cardiac sympathetic nerve activity. Thus, Figure 24–8 shows which minimizes the detrimental effect of high heart rates
how the cardiac filling pressure and the activity level of cardiac on diastolic filling time;
sympathetic nerves interact to determine cardiac output. 3. an increase in the rate of action potential conduction,
When cardiac filling pressure is 2 mm Hg and the activity of particularly evident in the AV node (positive dromotropic
cardiac sympathetic nerves is normal, the heart will operate at effect), by altering conductivity of gap junctions;
point A and will have a cardiac output of 5 L/min. Each single 4. an increase in cardiac contractility (positive inotropic ef-
curve in Figure 24–8 shows how cardiac output would be fect) by activating the iCa2+ current and increasing Ca2+
changed by changes in cardiac filling pressure if cardiac sym- release from the sarcoplasmic reticulum, which increas-
pathetic nerve activity were held at a fixed level. For example, es the contractile ability of cardiac muscle at any given
if cardiac sympathetic nerve activity remained normal, increas- preload; and
ing cardiac filling pressure from 2 to 4 mm Hg would cause the 5. an increase in rate of cardiac relaxation (positive lusitropic
heart to shift its operation from point A to point B on the car- effect) by increasing Ca2+ uptake by the sarcoplasmic
diac function diagram. In this case, cardiac output would reticulum, which also helps minimize the detrimental
increase from 5 to 7 L/min solely as a result of the increased effect of high heart rates on diastolic filling time.
filling pressure (Starling’s law). If, on the other hand, cardiac
filling pressure were fixed at 2 mm Hg while the activity of Most catecholamine influences on the heart are a result
cardiac sympathetic nerves was moderately increased from of increases in sympathetic neural activity. Although circulat-
normal, the heart would change from operating at point A to ing catecholamines of adrenal origin can potentially evoke
operating at point C. Cardiac output would again increase similar effects, their concentrations are normally so low that
from 5 to 7 L/min. In this instance, however, cardiac output their contributions are negligible. Specific drugs called
does not increase through the length-dependent mechanism β1-adrenergic receptor blockers (antagonists) can block all of
because cardiac filling pressure did not change. Cardiac out- the effects of catecholamines from whatever source on cardiac
put increases at constant filling pressure with an increase in muscle. These drugs may be useful in the treatment of coro-
cardiac sympathetic activity for two reasons. First, increased nary artery disease to thwart increased metabolic demands
cardiac sympathetic nerve activity increases heart rate. Second, placed on the heart by activity of sympathetic nerves.
but just as importantly, increased sympathetic nerve activity As we will see in subsequent chapters, increases in sympa-
increases stroke volume by increasing cardiac contractility. thetic activity can also have indirect influences on cardiac
Cardiac function graphs thus consolidate knowledge of function that are a consequence of sympathetic-induced alter-
many mechanisms of cardiac control, and are most helpful in ations in arteriolar and venous tone (i.e., alterations in after-
describing how the heart interacts with other elements in the load and preload, respectively).
cardiovascular system. Furthermore, these graphs reempha-
size the important point that a change in cardiac filling pres-
sure alone will have a very potent effect on cardiac output at DETERMINANTS OF MYOCARDIAL
any level of sympathetic activity. OXYGEN CONSUMPTION
In many pathological situations, such as severe coronary
SUMMARY OF SYMPATHETIC atherosclerosis, oxygen requirements of myocardial tissue
may exceed the capacity of coronary blood flow to deliver
NEURAL INFLUENCES ON oxygen to the heart muscle. This mismatch can result in
CARDIAC FUNCTION severe chest pain or discomfort called angina pectoris. It is
important to understand what factors determine energy
The effects of the sympathetic nervous system on the electrical costs, and therefore the myocardial oxygen consumption,
and mechanical properties of cardiac muscle, and thus on car- because reduction of oxygen demand may be of significant
diac pumping ability, are initiated by the interaction of norepi- clinical benefit to the patient.
nephrine with β1-adrenergic receptors on cardiac muscle Because the heart derives its energy almost entirely from
cells. This results in a cascade of events involving the Gs acti- aerobic metabolism, myocardial oxygen consumption is
vation of adenylate cyclase, formation of cAMP, and activa- directly related to myocardial ATP use. The basal metabo-
tion of protein kinase A with subsequent phosphorylation of lism of the heart tissue (the energy consumed in cellular pro-
many molecules that play key regulatory roles in intracellular cesses other than contraction such as energy-dependent ion
processes. These cellular events combine to evoke the follow- pumping) normally accounts for about 25% of myocardial
ing improvements in pumping capabilities of the heart: ATP use and therefore myocardial oxygen consumption in a
1. an increase in heart rate (positive chronotropic effect) by resting individual. The processes associated with muscle
activating the inward-going sodium if current in SA contraction account for about 75% of myocardial energy
nodal cells; use. Primarily, this reflects ATP splitting associated with
232 SECTION V Cardiovascular Physiology

cross-bridge cycling during the isovolumetric contraction energy cost of the isovolumetric pressure development phase
and ejection phases of the cardiac cycle. Some ATP is also of the cardiac cycle. The less pressure (wall tension) devel-
used for Ca2+ sequestration at the termination of each oped and the less often pressure development occurs, the less
contraction. energy used.
The energy expended during the isovolumetric contraction
phase of the cardiac cycle accounts for the largest portion
(~50%) of total myocardial oxygen consumption despite
the fact that the heart does no external work during this
period. The energy needed for isovolumetric contraction is
CLINICAL CORRELATION
mainly dependent on the intraventricular pressure that must A 40-year-old woman comes to the emergency room
develop during this time, that is, on the cardiac afterload. because of the sudden onset of weakness and dizziness
Cardiac afterload then is a major determinant of myocardial about an hour earlier. In addition, she reports a sense of
oxygen consumption. Reductions in cardiac afterload can pro- fluttering in her chest and throat. Examination reveals
duce clinically significant reductions in myocardial energy her heart rate to be rapid (165 beats/min; tachycardia)
requirements and therefore myocardial oxygen consumption. and regular and her blood pressure to be on the low side
Energy utilization during isovolumetric contraction is actu- at 80/60 mm Hg. Her ECG shows a tachycardia associ-
ally more directly related to isometric wall tension develop- ated with a narrow QRS complex. After attempts
ment than to intraventricular pressure development. Recall to restore normal rates by massaging the neck at the
that wall tension is related to intraventricular pressure and to location of the carotid sinuses, and by putting a cold
ventricular radius through the law of Laplace (T = P × r). compress on her face, she was given an intravenous injec-
Consequently, reductions in cardiac preload (i.e., end-diastolic tion of adenosine that converted her heart rate to
volume, radius) will also tend to reduce the energy required 80 beats/min and her blood pressure to 130/85 mm Hg.
for isovolumetric contraction. The primary complaint of weakness and dizziness
It is during the ejection phase of the cardiac cycle that the suggests a decrease in systemic blood flow including
heart actually performs external work and the energy the the cerebral circulation caused by the low arterial pres-
heart expends during ejection depends on how much exter- sure. This could be caused by a decrease either in car-
nal work it is doing. In a fluid system, work (force × dis- diac output or in total peripheral resistance. In our
tance) is equal to pressure (force/distance2) × volume patient, the very fast heart rate severely shortened the
(distance3). The external physical work done by the left ven- duration of diastole and, as a result, cardiac filling was
tricle in one beat, that is, the stroke work, is equal to the area significantly compromised. Because of Starling’s law of
enclosed by the left ventricular pressure–volume loop the heart relating end-diastolic volume with stroke vol-
(see Figure 24–3A). Stroke work is increased either by an ume, the decrease in filling results in a decrease in
increase in stroke volume (increased “volume” work) or by stroke volume. In spite of the rapid heart rate, the low
an increase in afterload (increased “pressure” work). In terms stroke volume produces a significant decrease in car-
of ATP utilization and oxygen consumption, increases in the diac output. The therapeutic strategy involves attempts
pressure work of the heart are more costly than increases in to decrease heart rate.
volume work. Thus, reductions in afterload are especially The ECG suggests a supraventricular tachycardia
helpful in reducing the myocardial oxygen requirements for with either an atrial ectopic focus firing rapidly or a
doing external work. reentrant AV nodal circuit rapidly exciting the normal
Changes in myocardial contractility can have important ventricular conduction system. (See Chapter 25 for more
consequences on the oxygen requirement for basal metabo- details.) Mechanical ways to interrupt these processes
lism, isovolumic wall tension generation, and external work. include strategies to increase vagal firing to the atrial tis-
Heart muscle cells use more energy in rapidly developing a sue by (1) massage of neck at the region of the carotid
given tension and shortening by a given amount than in doing sinus (carotid massage), stretching the arterial barore-
the same thing more slowly. Also, with increased contractility, ceptors, and tricking the medullary cardiovascular cen-
more energy is expended in active Ca2+ transport. The net ters to think arterial pressure is elevated (see Chapter
result of these influences is often referred to as the “energy- 29) or (2) instituting the diving reflex with a cold com-
wasting” effect of increased contractility. press on the face in which bradycardia is induced by
Heart rate is one of the most important determinants activation of the trigeminal nerve afferents to the brain
of myocardial oxygen consumption because the energy (see Chapter 71). The primary pharmacological strategy
costs per minute must equal the energy cost per beat used in the emergency room involved intravenous injec-
times the number of beats per minute. In general, it has been tion of a bolus of adenosine. This can have a transient
found that it is more efficient (i.e., less oxygen is required) to influence on supraventricular conduction within the
achieve a given cardiac output with low heart rate and high heart and can often interrupt the abnormal reentrant
stroke volume than with high heart rate and low stroke vol- pathway. Adenosine was effective in our patient. If it had
ume. This again appears to be related to the relatively high
CHAPTER 24 The Heart Pump 233

2. Your patient has coronary artery disease with significant


not been, drugs such as β-adrenergic receptor blockers occlusion of the left anterior descending coronary artery.
or calcium channel blockers can be effective. She was Although all of the following will increase cardiac work,
referred to a cardiologist who will perform extensive which of the following will be most likely to result in an attack
testing of her cardiac function to try to identify the ana- of angina pectoris?
tomical source of her abnormal cardiac rhythm. A) high heart rates
B) elevated arterial blood pressures
C) increased end-diastolic volume
D) low arterial blood pressure
E) increased cardiac contractility
CHAPTER SUMMARY 3. A drug is given that blocks cardiac β1-adrenergic receptors.
■ Effective cardiac pumping of blood requires coordinated filling What will be the direct consequences of this drug on
of the chambers, excitation and contraction of the cardiac the heart?
muscle cells, pressure generation within the chambers, opening A) Heart rate will increase.
and closing of cardiac valves, and one-way movement of blood B) The PR interval of the ECG will shorten.
through the chambers into the aorta or pulmonary artery. C) Metabolic demands will be reduced.
■ Except for lower ejection pressures, events of the right side of D) Coronary flow rate will increase.
the heart are identical to those of the left side. E) Cardiac contractility will increase.
■ Heart sounds associated with valve movements and detected 4. A cannula is placed in the pulmonary artery (PA) of a normal
on auscultation can be used to identify the beginnings of healthy individual and pressure is reported to be 25/8 mm Hg.
diastolic and systolic phases of the cardiac cycle. Which of the following can be surmised from these values?
■ The events of a single ventricular cardiac cycle can be displayed A) RV systolic pressure is 25 mm Hg; PA diastolic pressure
as records of electrical, mechanical, pressure, sound, or flow is 8 mm Hg.
changes against time or as a record of volume against pressure. B) RV systolic pressure is 25 mm Hg; left atrial pressure
■ Cardiac output is defined as the amount of blood pumped by is 8 mm Hg.
either of the ventricles per minute and is determined by the C) RV systolic pressure must be significantly higher
product of heart rate and stroke volume. than 25 mm Hg.
■ Stroke volume can be altered by changes in ventricular preload D) RV diastolic pressure must be significantly higher
(filling), ventricular afterload (arterial pressure), and/or cardiac than 8 mm Hg.
muscle contractility. E) Pulmonary capillary pressure is 8 mm Hg.
■ A cardiac function curve describes the relationship between 5. An individual has an autonomic nervous system dysfunction
ventricular filling and cardiac output and can be shifted up in which cardiac parasympathetic nerve activity abruptly
(left) or down (right) by changes in sympathetic activity to the increases while cardiac sympathetic activity decreases.
heart or by changes in cardiac muscle contractility. Which of the following situations is most likely
■ Energy for cardiac muscle contraction is derived primarily to occur?
from aerobic metabolic pathways such that cardiac work is A) LV end-diastolic volume will increase.
tightly related to myocardial oxygen consumption. B) Cardiac output will increase.
C) Heart rate will increase.
D) LV end-systolic volume will decrease.
E) Coronary blood flow will increase.
STUDY QUESTIONS
1. You are listening to your patient’s heart sounds and have
identified the systolic and diastolic intervals. Four of the
conditions listed below exist during the same phase of
the cardiac cycle and one does not. Which one is the odd one?
A) The mitral valve is open.
B) The ST segment of the ECG is occurring.
C) The “v” wave of the jugular venous pulse has just occurred.
D) Ventricular volume is rapidly increasing.
E) Aortic pressure is decreasing.
This page intentionally left blank
25
C H A P T E R

Cardiac Function
Assessments
Lois Jane Heller and David E. Mohrman

O B J E C T I V E S

■ State the relationship between electrical events of cardiac excitation and


the P, QRS, and T waves, the PR and QT intervals, and the ST segment of the
electrocardiogram.
■ State Einthoven’s basic electrocardiographic conventions and, given data,
determine the mean electrical axis of the heart.
■ Describe the standard 12-lead electrocardiogram.
■ Detect common cardiac arrhythmias from the electrocardiogram, identify
their physiological bases, and describe their physiological consequences.
■ Calculate cardiac output using the Fick principle.
■ Recognize echocardiographic images obtained during the cardiac cycle.
■ Define ejection fraction and identify visualization methods used to determine it.
■ Describe the end-systolic pressure–volume relationship.
■ List stenotic and regurgitant valve abnormalities for the left heart and
describe their consequences in terms of intracardiac and arterial pressures,
flow patterns, and heart sounds that accompany them.

TECHNIQUES FOR ASSESSING MEASUREMENT OF CARDIAC


CARDIAC FUNCTION EXCITATION—THE
There are a variety of methods available to assess cardiac ELECTROCARDIOGRAM
function. Some of these are noninvasive (e.g., electrocardiog-
raphy to evaluate electrical characteristics, auscultation of the The electrocardiogram is a powerful clinical tool that is used to
chest to evaluate valve function, and echocardiography to evaluate cardiac beating rate, rhythm, and the conduction char-
visualize mechanical pumping action) and others require vari- acteristics of cardiac tissue. As briefly described in Chapter 23,
ous types of invasive instrumentation. This chapter will pro- the electrocardiogram is the result of currents propagated
vide a brief overview of some of these commonly used clinical through the extracellular fluid that are generated by the spread
tools and an introduction to some of the common cardiac of the wave of excitation throughout the heart. Electrodes
functional abnormalities. placed on the surface of the body record the small potential dif-
ferences between various recording sites that vary over the time
course of the cardiac cycle.

235

Ch25_235-250.indd 235 11/26/10 10:00:28 AM


236 SECTION V Cardiovascular Physiology

R cells have already returned to the resting phase, whereas ven-


1 mV
tricular muscle cells are in the plateau phase of their action
potentials. (Myocardial injury or inadequate blood flow, how-
ever, can produce elevations or depressions in the ST segment.)
0.5
When ventricular cells begin to repolarize, a voltage once
Voltage

T
P again appears on the body surface and is measured as the
0 T wave of the electrocardiogram. The T wave is wider and not
Q S
as large as the R wave because ventricular repolarization is less
synchronous than depolarization. At the conclusion of the
−0.5 PR ST T wave, all the cells in the heart are in the resting state. The QT
QRS
segment segment interval roughly approximates the duration of the ventricular
interval
PR
myocyte depolarization and thus the period of ventricular sys-
interval QT interval
tole. At a normal heart rate of 60 beats/min, the QT interval is
normally less than 380 milliseconds. No body surface poten-
tial is measured until the next impulse is generated by the
Time sinoatrial (SA) node.
FIGURE 25–1 Typical electrocardiogram. (Modified with The operation of the specialized conduction system is a pri-
permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: mary factor in determining the normal electrocardiographic
Lange Medical Books/McGraw-Hill, 2006.) pattern. For example, the AV nodal transmission time deter-
mines the PR interval. Also, the effectiveness of the ventricular
Purkinje system in synchronizing ventricular depolarization
A typical electrocardiographic record of voltages recorded is reflected in the large magnitude and short duration of the
on the surface of the body between the left leg and the right QRS complex. Remember that nearly every heart muscle cell is
arm (called “lead II”) is indicated in Figure 25–1. The major inherently capable of rhythmicity and that all cardiac cells are
features of the electrocardiogram are the P, QRS, and T waves electrically interconnected through gap junctions. Thus, a
that are caused by atrial depolarization, ventricular depolar- functional heart rhythm can and often does occur without the
ization, and ventricular repolarization, respectively. The involvement of part or all of the specialized conduction sys-
period of time from the initiation of the P wave to the begin- tem. Such a situation is, however, abnormal, and the existence
ning of QRS complex is designated as the PR interval and of abnormal conduction pathways produces an abnormal elec-
indicates the time it takes for an action potential to spread trocardiogram.
through the atria and the atrioventricular (AV) node. During
the latter portion of the PR interval (PR segment), no voltages
are detected on the body surface. This is because atrial muscle BASIC ELECTROCARDIOGRAPHIC
cells are depolarized (in the plateau phase of their action CONVENTIONS
potentials), ventricular cells are still resting, and the electrical
field set up by the action potential progressing through the Recording electrocardiograms is a routine diagnostic proce-
small AV node is not intense enough to be detected. The dura- dure standardized by universal application of certain conven-
tion of the normal PR interval ranges from 120 to 200 millisec- tions. The conventions for recording and analysis of
onds. Shortly after the cardiac impulse breaks out of the AV electrocardiograms from the three standard bipolar limb leads
node and into the rapidly conducting Purkinje system, all the are briefly described here and summarized in Figure 25–2.
ventricular muscle cells depolarize rapidly and cause the QRS Recording electrodes are placed on both arms and the left leg—
complex. The R wave is the largest event in the electrocardio- usually at the wrists and ankle. The arms and legs act as con-
gram because ventricular muscle cells are so numerous and ductive extensions from the body, and voltage measurements
because they depolarize nearly in unison. The normal QRS are actually between points that form an equilateral triangle
complex lasts between 60 and 100 milliseconds. (The repolar- over the thorax. This is called Einthoven’s triangle in honor of
ization of atrial cells is also occurring during the time period the Dutch physiologist who devised it at the turn of the 19th
in which ventricular depolarization generates the QRS com- century. Any single electrocardiographic trace is a recording of
plex on the electrocardiogram [see Figure 23–3]. Atrial repo- the voltage difference measured between any two vertices of
larization is not evident on the electrocardiogram because it is Einthoven’s triangle. An example of the lead II electrocardio-
a poorly synchronized event in a relatively small mass of heart gram measured between the right arm and the left leg
tissue and is completely overshadowed by the major electrical has already been shown in Figure 25–2. Similarly, lead I and
events occurring in the ventricles at this time.) lead III electrocardiograms represent voltage measurements
The QRS complex is followed by the ST segment. Normally, taken along the other two sides of Einthoven’s triangle, as
no electrical potentials are measured on the body surface dur- indicated in Figure 25–2. The + and – symbols in Figure 25–2
ing the ST segment because no rapid changes in membrane indicate polarity conventions that have been universally adopted.
potential are occurring in any of the cells of the heart; atrial For example, an upward deflection in a lead II electrocardiogram
CHAPTER 25 Cardiac Function Assessments 237

Right arm − Lead I + Left arm


III
Le

ad
ad

Le
II
Lead selector

+
Chart recorder and amplifier

+
Left leg

FIGURE 25–2 Einthoven’s


Voltage scale Paper speed electrocardiographic conventions. (Modified
10 mm 25 mm = 1 s
= +1 mV with permission from Mohrman DE, Heller LJ: Cardiovascular
upward Physiology, 6th ed. New York: Lange Medical Books/
McGraw-Hill, 2006.)

(as normally occurs during the P, R, and T waves) indicates that voltages appear on the body surface as a result of the cardiac
an electrical polarity exists at that instant between the left leg electrical activity must be examined.
and the right shoulder electrodes, with the left leg electrode
being positive. Conversely, a downward deflection in a lead
II record indicates that an electrical polarity exists between this CARDIAC DIPOLES AND
pair of electrodes but this time the electrode on the left leg is
recording negative charges instead of positive charges. Similar ELECTROCARDIOGRAPHIC
polarity conventions have been established for lead I and lead RECORDS
III recordings and are indicated by the + and – symbols in
Figure 25–2. In addition, electrocardiographic recording equip- Einthoven’s conceptualization of how cardiac electrical activ-
ment is usually standardized so that a 1-cm deflection on the ity causes potential differences on the surface of the body is
vertical axis always represents a potential difference of 1 mV, illustrated in Figure 25–3. In this example, the heart is shown
and that a 25-mm segment of the horizontal tracing of any elec- at one instant in the atrial depolarization phase. The cardiac
trocardiographic record represents 1 second. Most electrocar- impulse, after having arisen in the SA node, is spreading as a
diographic records contain calibration signals so that abnormal wavefront of depolarization through the atrial tissue. At each
rates and wave amplitudes can be easily detected. point along this wavefront of electrical activity, a small charge
As shown in the subsequent section, many cardiac electrical separation exists in the extracellular fluid between polarized
abnormalities can be detected in recordings from a single elec- membranes (positive outside) and depolarized membranes
trocardiographic lead. However, certain clinically useful infor- (negative outside). Thus, the wavefront may be thought of as
mation can only be derived by combining the information a series of individual electrical dipoles (regions of charge
obtained from two electrocardiographic leads. To understand separation). Each individual dipole is oriented in the direction
these more complex electrocardiographic analyses, the way of local wavefront movement. The large black arrow in

Depolarized − I
RA LA
cells − −
SA Wavefront of
node −
− electrical activity


− −− Net dipole
II III
Atria
FIGURE 25–3 Net cardiac dipole during atrial
depolarization and its components on the limb
Ventricles leads. (Modified with permission from Mohrman DE, Heller LJ:
Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/
LL
McGraw-Hill, 2006.)
238 SECTION V Cardiovascular Physiology

Figure 25–3 represents the total net dipole created by the orientation during the course of atrial depolarization. The
summed contributions of all the individual dipoles distributed nature of these changes will determine the shape of the P wave
along the wavefront of atrial depolarization. The salty extra- on each of the electrocardiogram leads.
cellular fluid acts as an excellent conductor, allowing these The P wave terminates when the wave of depolarization
instantaneous net dipoles generated on the surface of the heart reaches the nonmuscular border between the atria and the
muscle to be recorded by electrodes on the surface of the ventricles and the number of individual dipoles becomes very
body. small. At this time, the cardiac impulse is still being slowly
The net dipole that exists at any instant is oriented (i.e., transmitted toward the ventricles through the AV node. How-
points) in the general direction of wavefront movement at that ever, the electrical activity in the AV node involves so few cells
instant. The magnitude or strength of the dipole (represented that it generates no detectable net cardiac dipole. Thus, no
here by the arrow length) is determined by: (1) how extensive voltages are measured on the surface of the body for a brief
the wavefront is (i.e., how many cells are simultaneously depo- period following the P wave. A net cardiac dipole reappears
larizing at the instant in question) and (2) the consistency of only when the depolarization completes its passage through
orientation between individual dipoles at different points in the AV node, enters the Purkinje system, and begins its rapid
the wavefront (dipoles with the same orientation reinforce passage over the ventricular muscle cells. Because the Purkinje
each other; dipoles with opposite orientation cancel each fibers terminate in the intraventricular septum and in the
other). endocardial layers at the apex of the ventricles, ventricular
The net dipole in the example of Figure 25–3 causes the depolarization occurs first in these areas and then proceeds
lower left portion of the body to be generally positive with outward and upward through the ventricular myocardium.
respect to the upper right portion. This particular dipole will
cause positive voltages to exist on all three of the electrocar-
diogram limb leads. As shown in the right half of Figure 25–3, VENTRICULAR DEPOLARIZATION
this can be deduced from Einthoven’s triangle by observing AND THE QRS COMPLEX
that the net dipole has some component that points in the
positive direction of leads I, II, and III. As illustrated in It is the rapid and large changes in the magnitude and direction
Figure 25–3, the component that a cardiac dipole has on a of the net cardiac dipoles that exist during ventricular depolar-
given electrocardiogram lead is found by drawing perpendic- ization that are responsible for the QRS complex of the electro-
ular lines from the appropriate side of Einthoven’s triangle to cardiogram. The normal process is illustrated in Figure 25–4.
the tip and tail of the dipole. (It may be helpful to think of the The initial ventricular depolarization usually occurs on the left
component on each lead as the “shadow” cast by the dipole on side of the intraventricular septum as diagrammed in the upper
that lead as a result of a “sun” located far beyond the corner of panel of the figure. Analysis of the cardiac dipole formed by
Einthoven’s triangle that is opposite the lead.) Note that the this initial ventricular depolarization with the aid of Einthoven’s
dipole in this example is most parallel to lead II and therefore triangle shows that this dipole has a negative component on
has a large component in the lead II direction. Thus, it will lead I, a small negative component on lead II, and a positive
create a larger voltage on lead II than on leads I or III. This component on lead III. The upper right panel shows the actual
dipole has a rather small component on lead III because it is deflections on each of the electrocardiographic limb leads that
oriented nearly perpendicular to lead III. will be produced by this dipole. Note that it is possible for a
The limb lead configuration may be thought of as a way to given cardiac dipole to produce opposite deflections on differ-
view the heart’s electrical activity from three different perspec- ent leads. For example, in Figure 25–4, Q waves appear on leads
tives (or axes). The vector representing the heart’s instanta- I and II but not on lead III.
neous dipole strength and orientation is the object under The second row of panels in Figure 25–4 shows the ventri-
observation, and its appearance depends on the position from cles during the instant in ventricular depolarization when the
which it is viewed. The instantaneous voltage measured on the number of individual dipoles is greatest and/or their orienta-
axis of lead I, for example, indicates how the dipole being gen- tion is most similar. This phase generates the large net cardiac
erated by the heart’s electrical activity at that instant appears dipole, which is responsible for the R wave of the electrocar-
when viewed from directly above. A cardiac dipole that is ori- diogram. In Figure 25–4, this net cardiac dipole is nearly par-
ented horizontally appears large on lead I, whereas a vertically allel to lead II. As indicated, such a dipole produces large
oriented cardiac dipole, however large, produces no voltage on positive R waves on all three limbs leads.
lead I. Thus, it is necessary to have views from two directions The third row of diagrams in Figure 25–4 shows the situa-
to establish the magnitude and orientation of the heart’s dipole. tion near the end of the spread of depolarization through the
A vertically oriented dipole would be invisible on lead I but ventricles and indicates how the small net cardiac dipole pres-
would be readily apparent if viewed from the perspective of ent at this time produces the S wave. Note that an S wave does
lead II or lead III. not necessarily appear on all electrocardiogram leads (as in
It is important to recognize that the example of Figure 25–3 lead I of this example).
pertains only to one instant during atrial depolarization. The The bottom row of diagrams in Figure 25–4 shows that
net cardiac dipole continuously changes in magnitude and during the ST segment, all ventricular muscle cells are in a
CHAPTER 25 Cardiac Function Assessments 239

− I I
− −

II
Q II III
wave
III

− I
I
− −

II III II
R
wave

III

− I I
− −

II
II III
S
wave
III

− I
I
− −

II FIGURE 25–4 Ventricular depolarization


ST II III and the generation of the QRS complex.
segment (Modified with permission from Mohrman DE, Heller LJ:
III Cardiovascular Physiology, 6th ed. New York: Lange Medical
Books/McGraw-Hill, 2006.)

depolarized state. There are no waves of electrical activity ventricular repolarization results in a positive T wave recorded,
moving through the heart tissue. Consequently, no net cardiac for example, on lead II. The T wave is broader and smaller
dipole exists at this time and no voltage differences exist than the R wave because the repolarization of ventricular mus-
between points on the body surface. All electrocardiographic cle cells is less well synchronized than is their depolarization.
traces will be flat at the isoelectric (zero voltage) level.

VENTRICULAR REPOLARIZATION MEAN ELECTRICAL AXIS


AND THE T WAVE AND AXIS DEVIATIONS
As illustrated in Figure 25–1, the T wave is normally positive The orientation of the cardiac dipole during the most intense
on lead II as is the R wave. This indicates that the net cardiac phase of ventricular depolarization (i.e., at the instant the R wave
dipole generated during ventricular repolarization is oriented reaches its peak) is called the mean electrical axis of the heart. It
in the same general direction as that which exists during ven- is used clinically as an indicator of whether or not ventricular
tricular depolarization. This may be somewhat surprising. depolarization is proceeding over normal pathways. The mean
However, recall from Figure 24–3 that the last ventricular cells electrical axis is reported in degrees according to the convention
to depolarize are the first to repolarize. The reasons for this are indicated in Figure 25–5. Note that the downward direction is
not well understood but the result is that the wavefront of elec- designated as plus 90° in this polar coordinate system describing
trical activity during ventricular repolarization tends to axis orientation in the frontal plane (a vertical plane that divides
retrace, in reverse direction, the course followed during ven- the body into anterior and posterior sections.) As indicated, a
tricular depolarization. Therefore, the dipole formed during mean electrical axis that lies anywhere in the patient’s lower left-
repolarization has the same polarity as that during depolariza- hand quadrant is considered normal (between 0° and 90°). A
tion. This reversed wavefront propagation pathway during left axis deviation exists when the mean electrical axis falls in
240 SECTION V Cardiovascular Physiology

RA LA The standard limb leads (I, II, and III) and the augmented uni-
−90 degrees polar limb leads (aVR, aVL, and aVF) record the electrical activity
Left axis
of the heart as it appears from six different “perspectives.” As
deviation
180 0 < 0 degrees shown in Figure 25–6A, the axes for leads I, II, and III are those
degrees degrees
of the sides of Einthoven’s triangle, while those for aVR, aVL, and
Right axis Range of normal aVF are specified by lines drawn from the center of Einthoven’s
deviation 0 to +90 degrees triangle to each of its vertices. As indicated in Figure 25–6B,
> +90 degrees +90 degrees
these six limb leads can be thought of as a hexaxial reference
system for observing the cardiac vectors in the frontal plane.
The other six leads of the standard 12-lead electrocardio-
gram are also unipolar leads that “look” at the electrical vector
LL projections in the transverse plane (a horizontal plane that
FIGURE 25–5 Mean electrical axis and axis deviations.
divides the body into superior and inferior segments). These
(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology,
potentials are obtained by placing an additional (exploring)
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) electrode in six specified positions on the chest wall as shown
in Figure 25–6C. The indifferent electrode in this case is
formed by electrically connecting the limb electrodes. These
the patient’s upper left-hand quadrant and may indicate any of leads are identified as precordial or chest leads and are desig-
the several conditions such as a physical displacement of the nated as V1–V6. As shown in this figure, the wave of ventricu-
heart to the left, left ventricular hypertrophy, or loss of electrical lar excitation sweeps away from V1, resulting in a downward
activity in part of the right ventricle (e.g., after an infarct). A deflection. The wave of ventricular excitation sweeps toward
right axis deviation exists when the mean electrical axis falls in V6, resulting in an upward deflection.
the patient’s lower right-hand quadrant and may indicate, among In summary, the electrocardiogram is a powerful tool for
several conditions, a physical displacement of the heart to the evaluating cardiac excitation characteristics. It must be recog-
right, right ventricular hypertrophy, or loss of electrical activity nized, however, that the ECG does not provide direct evidence
in part of the left ventricle. of mechanical pumping effectiveness. For example, a leaky
The mean electrical axis of the heart can be determined from heart valve will usually have no direct electrocardiographic
the electrocardiogram. The process involves determining what consequences but may adversely influence pumping ability of
single net dipole orientation will produce the R-wave ampli- the heart.
tudes recorded on any two leads. For example, if the R waves on
leads II and III are both positive (upright) and of equal magni-
tude, the mean electrical axis must be +90°. As should be obvi- ABNORMAL CARDIAC
ous, in this case, the amplitude of the R wave on lead I will be EXCITATION AND RHYTHMICITY
zero. Alternatively, one can scan the electrocardiographic
records for the lead tracing with the largest R waves and then The material presented here is an introduction to the more
deduce that the mean electrical axis must be nearly parallel to common abnormalities in cardiac rate and rhythm with an
that lead. In Figure 25–4, for example, the largest R wave occurs emphasis on the primary physiological consequences of these
on lead II. Lead II has an orientation of +60°, which is very abnormal situations. Many cardiac excitation problems can be
close to the actual mean electrical axis in this example. diagnosed from the information in a single lead of an electro-
cardiogram. The lead II electrocardiogram trace at the top of
Figure 25–7 is identified as normal sinus rhythm based on the
THE STANDARD 12-LEAD following characteristics: (1) the frequency of QRS complexes is
ELECTROCARDIOGRAM ~1/s, indicating a normal beating rate of 60 beats/min; (2) the
shape of the QRS complex is normal for lead II and its duration
The standard clinical electrocardiogram involves voltage mea- is less than 120 milliseconds, indicating rapid depolarization of
surements recorded from 12 different leads. Three of these are the ventricles via normal conduction pathways; (3) each QRS
the bipolar limb leads I, II, and III, which have already been complex is preceded by a P wave of proper configuration, indi-
discussed. The other nine leads are unipolar leads. Three of cating SA nodal origin of the excitation; (4) the PR interval is
these leads are generated by using the limb electrodes. Two of less than 200 milliseconds, indicating proper conduction delay
the electrodes are electrically connected to form an indifferent of the impulse propagation through the AV node; (5) the QT
electrode while the third limb electrode is made the positive interval is less than half of the R-to-R interval, indicating nor-
pole of the pair. Recordings made from these electrodes are mal ventricular repolarization; and (6) there are no extra P
called augmented unipolar limb leads. The voltage record waves, indicating that no AV nodal conduction block is present.
obtained between the electrode at the right arm and the indif- The subsequent electrocardiographic tracings in Figures 25–7
ferent electrode is called a lead aVR electrocardiogram. Simi- and 25–9 represent irregularities commonly found in clinical
larly, lead aVL is recorded from the electrode on the left arm practice. Examination of each of these traces with the above
and lead aVF is recorded from the electrode on the left leg. characteristics in mind will aid in the differential diagnosis.
CHAPTER 25 Cardiac Function Assessments 241

A B − C
RA − I + LA − −
aVR aVL
− aV − + +
+ R aV L +

− − − + I
aVF

II III
− −
+ V6
V1
+ + V2
+ + III + II
LL aVF
V5
V3 V4

FIGURE 25–6. The standard 12-lead electrocardiogram. A and B) Leads in the frontal plane. C) Electrode positions for precordial leads in the
transverse plane. D) A 12 lead ECG. The bottom line is a rhythm strip taken from lead V1 (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular
Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006. Fig. 25–6D courtesy of Dr. David Gutterman)

The physiological consequences of abnormal excitation and arrhythmia because the extremely high heart rate does not
conduction in the heart depend on whether the electrical allow sufficient diastolic time for ventricular filling.
abnormality evokes a tachycardia, which will limit the time There are two mechanisms that may account for supraven-
for cardiac filling between beats; evokes a bradycardia, which tricular tachycardia. First, an atrial region, usually outside
is inadequate to support sufficient cardiac output; or decreases the SA node, may become irritable (perhaps because of local
the coordination of myocyte contraction, which will reduce interruption in blood flow) and begin to fire rapidly to take
stroke volume (SV). over the pacemaker function for the entire heart. Such an
abnormal pacemaker region is called an ectopic focus. Alter-
SUPRAVENTRICULAR ABNORMALITIES natively, atrial conduction may become altered so that a
single wave of excitation does not die out but continually
Traces 2–6 below the normal trace in Figure 25–7 represent travels around some abnormal atrial conduction loop. In
typical supraventricular arrhythmias (i.e., originating in the this case, the continual activity in the conduction loop may
atria or AV node). Supraventricular tachycardia (shown in drive the atria and AV node at a very high frequency. This
trace 2 of Figure 25–7 and sometimes called paroxysmal atrial self-sustaining process is called a reentry phenomenon and
tachycardia) occurs when the atria are abnormally excited is diagrammed in Figure 25–8. This situation may develop
and drive the ventricles at a very rapid rate. These paroxysms as a result of abnormal repolarization and altered refractory
may begin abruptly, last for a few minutes to a few hours, and periods in local areas of the myocardium. Atrial flutter is a
then, just as abruptly, disappear and heart rate reverts to special form of tachycardia of atrial origin in which a large
normal. QRS complexes appear normal (albeit frequent) with reentrant pathway drives the atria at very fast rates (250–300
simple paroxysmal atrial tachycardia because the ventricular beats/min) and normal refractory periods of AV nodal tis-
conduction pathways operate normally. The P and T waves sue are overwhelmed. Thus, ventricular rate is often some
may be superimposed because of the high heart rate. Low fixed ratio of the atrial rate (2:1, 4:1) with frequencies often
blood pressure and dizziness may accompany bouts of this 150–220 beats/min.
242 SECTION V Cardiovascular Physiology

1. Normal sinus rhythm

2. Supraventricular
tachycardia

3. First-degree block

2:1 4:1
4. Second-degree block

5. Third-degree block
FIGURE 25–7 Supraventricular
arrhythmias. (Reproduced with permission from 6. Atrial fibrillation
Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. 1 mV
New York: Lange Medical Books/McGraw-Hill, 2006.) 1s

Conduction blocks occur at the AV node and generally rep- node from the atria at unpredictable times. Fibrillation is a self-
resent impaired conduction through this tissue. In first-degree sustaining process. The mechanisms behind it are not well
heart block (trace 3 of Figure 25–7), the only electrical understood, but impulses are thought to progress repeatedly
abnormality is unusually slow conduction through the AV around irregular conduction pathways (sometimes called circus
node. This condition is detected by an abnormally long PR pathways, which imply a reentry phenomenon as described ear-
interval (>0.2 second). Otherwise, the electrocardiogram may lier and in Figure 25–8). However, because atrial contraction
be normal. At normal heart rates, the physiological effects of a usually plays a negligible role in ventricular filling, atrial fibrilla-
first-degree block are usually inconsequential. The danger, tion may be well tolerated by most patients as long as ventricular
however, is that the slow conduction may deteriorate to an rate is sufficient to maintain the cardiac output. The real danger
actual interruption of conduction. with atrial fibrillation lies in the tendency for blood to form clots
A second-degree heart block (trace 4 of Figure 25–7) is said in the atria in the absence of the normal vigorous, coordinated
to exist when some but not all atrial impulses are transmitted atrial contraction. These clots can fragment and move out of the
through the AV node to the ventricle. Impulses are blocked in heart to lodge in small arteries throughout the systemic or pul-
the AV node if the cells of the region are still in a refractory monary circulation. These emboli can have devastating effects
period from a previous excitation. The situation is aggravated on function of critical organs. Consequently, anticoagulant ther-
by high atrial rates and slower-than-normal conduction apy is used as prophylaxis for patients in atrial fibrillation.
through the AV nodal region. In second-degree block, some
but not all P waves are accompanied by corresponding QRS
complexes and T waves. Atrial rate is often faster than ven- VENTRICULAR ABNORMALITIES
tricular rate by a certain ratio (e.g., 2:1, 3:1, 4:1). This condi-
Traces 2–6 below the normal trace in Figure 25–9 show typical
tion may not represent a serious clinical problem as long as the
ventricular electrical abnormalities. Conduction blocks called
ventricular rate is adequate to meet the pumping needs.
bundle branch blocks or hemiblocks (trace 2 of Figure 25–9)
In third-degree heart block (trace 5 of Figure 25–7), no
impulses are transmitted through the AV node. In this event,
some area in the ventricles—often in the common bundle or
bundle branches near the exit of the AV node—assumes the
pacemaker role for the ventricular tissue. Atrial rate and ven-
tricular rate are completely independent, and P waves and QRS
complexes are totally dissociated in the electrocardiogram.
Ventricular rate is likely to be slower than normal (bradycardia)
and sometimes is slow enough to impair cardiac output.
Atrial fibrillation (trace 6 of Figure 25–8) is characterized by
a complete loss of the normally close synchrony of the excitation
and resting phases between individual atrial cells. Cells in differ-
ent areas of the atria depolarize, repolarize, and are excited again Normal Reentrant
randomly. Consequently, no P waves appear in the electrocar- pathway pathway
diogram although there may be rapid irregular small waves FIGURE 25–8 Normal and reentrant (circus) cardiac excitation
apparent throughout diastole. The ventricular rate is often very pathways. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular
irregular in atrial fibrillation because impulses enter the AV Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 25 Cardiac Function Assessments 243

1. Normal sinus rhythm

2. Bundle branch block

3. Premature ventricular
contraction

4. Ventricular tachycardia

5. Long QT syndrome with


torsades des pointes

FIGURE 25–9 Ventricular arrhythmias.


6. Ventricular fibrillation (Modified with permission from Mohrman DE, Heller LJ:
1 mV Cardiovascular Physiology, 6th ed. New York: Lange Medical
1s Books/McGraw-Hill, 2006.)

can occur in either of the branches of the Purkinje system of filling time limited by the rapid rate, but also the abnormal
the intraventricular septum often as a result of a myocardial excitation pathways make ventricular contraction less syn-
infarction. Ventricular depolarization is less synchronous than chronous and therefore less effective than normal. In addition,
normal in the half of the heart with the nonfunctional Purkinje ventricular tachycardia often precedes ventricular fibrillation.
system. This results in a widening of the QRS complex (>0.12 Prolonged QT intervals (left side of trace 5 in Figure 25–9)
second) because a longer time is required for cell-to-cell ven- are a result of delayed ventricular myocyte repolarization,
tricular depolarization of the blocked side to be completed. which may be due to inappropriate opening of sodium chan-
The direct physiological effects of bundle branch blocks are nels or prolonged closure of potassium channels during the
usually inconsequential. action potential plateau phase. Although the normal QT inter-
Premature ventricular contractions (PVCs; trace 3 of Figure val varies with heart rate, it is normally less than 40% of the
25–9) are caused by action potentials initiated by and propa- cardiac cycle length (except at very high heart rates). Long QT
gated away from an ectopic focus in the ventricle. As a result, syndrome is identified when the QT interval is greater than
the ventricle depolarizes and contracts before it normally 50% of the cycle duration. It may be genetic in origin (muta-
would. A PVC is often followed by a missed beat (called a com- tions influencing various ion channels involved with cardiac
pensatory pause) because the ventricular cells are still refrac- excitability), may be acquired from several electrolyte distur-
tory when the next normal impulse emerges from the SA node. bances (low blood levels of Ca2+, Mg2+, or K+), or may be
The highly abnormal ventricular depolarization pattern of a induced by several pharmacological agents (including some
PVC produces the large-amplitude, long-duration deflections antiarrhythmic drugs). The prolongation of the myocyte
on the electrocardiogram. The shapes of the electrocardio- refractory period, which accompanies the long QT syndrome,
graphic records of these extra beats are highly variable and extends the vulnerable period during which extra stimuli can
depend on the ectopic site of their origin and the depolariza- evoke tachycardia or fibrillation. Patients with long QT syn-
tion pathways involved. The volume of blood ejected by the drome are predisposed to a particularly dangerous type of
premature beat itself is smaller than normal (if a volume is ventricular tachycardia called torsades de pointes (“twisting of
ejected at all), whereas the stroke volume of the beat following points” as shown on the right side of trace 5 in Figure 25–9).
the compensatory pause is larger than normal. This is partly This differs from the ordinary ventricular tachycardia in
due to the differences in filling times and partly to an inherent that the ventricular electrical complexes cyclically vary in
phenomenon of cardiac muscle called postextrasystolic poten- amplitude around the baseline and can deteriorate rapidly into
tiation. Single PVCs occur occasionally in most individuals ventricular fibrillation.
and, although sometimes alarming to the individual experi- In ventricular fibrillation (trace 6 of Figure 25–9), various
encing them, are not dangerous. Frequent occurrence of PVCs, areas of the ventricle are excited and contract asynchronously.
however, may be a signal of possible myocardial damage or The mechanisms are similar to those in atrial fibrillation. The
perfusion problems and can lead to ventricular tachycardia ventricle is especially susceptible to fibrillation whenever a
and even ventricular fibrillation (discussed below). premature excitation occurs at the end of the T wave of the
Ventricular tachycardia (trace 4 of Figure 25–9) occurs previous excitation, that is, when most ventricular cells are in
when the ventricles are driven at high rates, usually by impulses the “hyperexcitable” or “vulnerable” period of their electrical
originating from a ventricular ectopic focus. Ventricular cycle. In addition, because some cells are repolarized and some
tachycardia is a very serious condition. Not only is diastolic are still refractory, circus pathways can be triggered easily at
244 SECTION V Cardiovascular Physiology

A niques. One of the most accurate methods of measuring car-


Increased Normal diac output by invasive means makes use of the Fick principle,
contractility contractility which is discussed in more detail in Chapter 26. Briefly, this
principle states that the amount of a substance consumed by
120 the tissues, Xtc, is equal to what goes in minus what goes out
LV pressure (mm Hg)

Increased (which is the arterial–venous concentration difference in the


afterload substance ([X]a – [X]v) times the blood flow rate, Q̇). This
80
relationship can be algebraically arranged to solve for blood
flow as follows:
Ẋtc
Q̇ = ________
[X] _ [X] (1)
Increased a v
40 preload
A common method of determining cardiac output is to use
the Fick principle to calculate the collective flow through the
systemic organs from (1) the whole body oxygen consumption
60 120 rate (Ẋ tc), (2) the oxygen content of the arterial blood ([X]a),
LV volume (mL) and (3) the concentration of oxygen in mixed venous blood
([X]v). Of the values required for this calculation, the oxygen
B Normal Cardiac failure content of mixed venous blood is the most difficult to obtain.
individual patient
Generally, the sample for venous blood oxygen measurement
d
se must be taken from a venous catheter positioned in the pul-
LV pressure (mm Hg)

120 r es tility
p c
y
ntr rmal

De ntra monary artery to ensure that it is a mixed sample of venous


tilit

co
ac

blood from all systemic organs.


No

Untreated
80 The calculation of cardiac output from the Fick principle
co

is best illustrated by an example. Suppose a patient is con-


Treated with
afterload suming 250 mL of O2/min when systemic arterial blood
40
reducer contains 200 mL of O2/L and the right ventricular blood
contains 150 mL of O2/L. This means that, on the average,
each liter of blood loses 50 mL of O2 as it passes through the
60 120 180
systemic organs. In order for 250 mL of O2 to be consumed
LV volume (mL)
per minute, 5 L of blood must pass through the systemic cir-
FIGURE 25-10 Left ventricular end systolic pressure- culation each minute:
volume relationships. A) The effect of increased contractility shifts 250[mL O2/min]
the line upward and to the left. B) The effect of systolic cardiac failure Q̇ = __________________
200–150[mL O /L blood]
= 5[L blood/min] (2)
2
shifts the line downward and to the right. (Modified with permission from
Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Although use of the Fick Principle as described above pro-
Books/McGraw-Hill, 2006.) vides the gold standard for cardiac output determination,
there are several other techniques that give good estimates
of cardiac output. Indicator dilution techniques involve
this time. Since no pumping action occurs with ventricular
injection of a known quantity of indicator (dye or a thermal
fibrillation, the situation is fatal unless quickly corrected by
bolus) into blood entering the right heart and appropriate
cardiac conversion (commonly referred to as external electri-
detectors are arranged to continuously record the concen-
cal defibrillation or cardioversion). During this procedure,
tration of the indicator in blood as it leaves the left heart.
the artificial application of large currents to the entire heart
The dilution of the indicator is proportional to the cardiac
(via paddle electrodes applied across the chest) may be effec-
output. Other techniques for imaging the heart (echocar-
tive in depolarizing all heart cells simultaneously and thus
diography, ventricular angiography, radionuclide ventricu-
allowing a normal excitation pathway to be reestablished.
lography) can be used to estimate stroke volume, cardiac
Cardiopulmonary resuscitation (CPR) must be administered
output and other indices of ventricular function as are
until a defibrillation is achieved.
described below.
Cardiac index is the cardiac output corrected for the indi-
vidual’s size. For example, the cardiac output of a 50-kg woman
MEASUREMENT OF will be significantly lower than that of a 90-kg man. It has been
CARDIAC OUTPUT found, however, that cardiac output correlates better with body
surface area than with body weight. Therefore, it is common to
Fick Principle: Measurement of cardiac output is not a simple express the cardiac output per square meter of surface area.
task and usually involves either some invasive maneuver or Under resting conditions, the cardiac index is normally
some significant assumptions based on noninvasive tech- approximately 3 (L/min)/m2.
CHAPTER 25 Cardiac Function Assessments 245

CARDIAC CONTRACTILITY tions in contractility will cause shifts in the end-systolic


pressure–volume relationship.)
ESTIMATIONS Note in Figure 25–10A that both the “normal” and “increased
contractility” end-systolic pressure–volume lines nearly proj-
It is often important to assess an individual’s cardiac function ect to the origin at zero pressure, zero volume. Thus, it is pos-
without using major invasive procedures. Advances in several sible to get a reasonable clinical estimate of the slope of the
techniques have made it possible to obtain two- and three- end-systolic pressure–volume relationship (read “myocardial
dimensional images of the heart throughout the cardiac cycle. contractility”) from a single measurement of end-systolic pres-
Visual or computer-aided analysis of such images provides sure and volume. This avoids the need to do multiple expen-
information useful in clinically evaluating cardiac function. sive tests with vasodilator or vasoconstrictor infusions.
Echocardiography is the most widely used of the several A decrease in contractility (as may be caused by heart dis-
imaging techniques currently available. This noninvasive ease) is associated with a downward shift of the end-systolic
technique is based on the fact that sound waves reflect back pressure–volume relationship and is known as systolic heart
toward the source when encountering abrupt changes in the failure. In this situation, increases in sympathetic drive have
density of the medium through which they travel. A trans- limited influence on cardiac output. Part of the compensatory
ducer, placed at specified locations on the chest, generates process involves a significant increase in body fluid retention
pulses of ultrasonic waves and detects waves reflected from that results in an increase in circulating blood volume and
the cardiac tissue interfaces. The longer the time between the ventricular EDV (see Chapter 29). A left ventricular pressure–
transmission of the wave and the arrival of the reflection, the volume loop describing the events of a cardiac cycle from a
deeper the structure is in the thorax. Such information can be failing heart (shown in Figure 25–10B) is displaced far to the
reconstructed by computer in various ways to produce a con- right of that of a normal heart. The untreated patient described
tinuous image of the heart and its chambers throughout the in this figure is in serious trouble with a reduced SV and EF
cardiac cycle. and high filling pressure with possible pulmonary vascular
Echocardiography is especially well suited for detecting congestion. Furthermore, the slope of the line describing the
abnormal operation of cardiac valves or contractile function end-systolic pressure–volume relationship is shifted down-
in portions of the heart walls. It also can provide estimates of ward and is less steep, indicating the reduced contractility of
heart chamber volumes at different times in the cardiac cycle the cardiac muscle. However, because of this flatter relation-
that are used in a number of ways to assess cardiac function. ship, small reductions in cardiac afterload (i.e., arterial blood
Ejection fraction (EF) is an extremely useful clinical mea- pressure) will produce substantial increases in EF and SV that
surement that can be calculated from an echocardiogram. It is will significantly help this patient.
defined as the ratio of stroke volume (SV) to end-diastolic
volume (EDV):
SV
EF = ____ (3)
ABNORMAL CARDIAC
EDV
VALVE FUNCTION
Estimates of end-diastolic and -systolic volumes can be
made from the images and SV calculated. EF is commonly Pumping action of the heart is impaired when the valves do
expressed as a percentage and normally ranges from 55% to not function properly. A number of techniques, ranging from
80% (mean 67%) under resting conditions. EFs of less than simple auscultation (listening to the heart sounds) to echocar-
55% indicate depressed myocardial contractility. diography or cardiac catheterization, are used to obtain infor-
The end-systolic pressure–volume relationship is another mation about the nature and extent of these valve malfunctions.
useful clinical technique to assess cardiac contractility. End- Often, abnormal heart sounds called murmurs accompany
systolic volume for a given cardiac cycle is estimated by one of cardiac valve defects. These sounds are caused by abnormal
the imaging techniques described above while end-systolic pressure gradients and turbulent blood flow patterns that
pressure for that cardiac cycle can obtained from the arterial occur during the cardiac cycle.
pressure recorded at the point of the closure of the aortic valve In general, when a valve does not open fully (i.e., is stenotic),
(the incisura). Values for several different cardiac cycles may the chamber upstream of the valve has to develop more pres-
be obtained during infusion of a vasoconstrictor (which sure during its systolic phase to achieve a given flow through
increases afterload), and the data plotted as in Figure 25–10 in the valve. This increase in “pressure” work will induce hypertro-
the context of overall ventricular pressure–volume loops. As phy of cardiac muscle cells and thickening of the walls of that
shown, increases in myocardial contractility are associated chamber. (This is analogous to the hypertrophied skeletal mus-
with a leftward rotation in the end-systolic pressure–volume cles of the weightlifter doing isometric or high-tension work.)
relationship. This method of assessing cardiac function is par- When a valve does not close completely (i.e., is insufficient,
ticularly important because it provides an estimate of contrac- regurgitant, or incompetent), the regurgitant blood flow repre-
tility that is independent of the EDV (preload). (Recall from sents an additional volume that must be ejected in order to get
Figure 24–4 that increases in preload cause increases in SV sufficient forward flow out of the ventricle into the tissues. This
without changing the end-systolic volume. Thus, only altera- increase in “volume” work often leads to chamber dilation but
246 SECTION V Cardiovascular Physiology

not to an increase in wall thickness. (This is analogous to the malities can occur in right ventricular valves with similar con-
nonhypertrophied but well-toned skeletal muscles of the long- sequences on right ventricular function.
distance runner doing isotonic or shortening work.)
A second generality about valve abnormalities is that when-
ever there is an increase in the atrial pressure as a result of AV AORTIC STENOSIS
valve stenosis or regurgitation, this will result in higher pres-
sures in the upstream capillary beds. If capillary hydrostatic Some characteristics of aortic stenosis are shown in
pressures are increased, tissue edema will ensue with substan- Figure 25–11A. Normally, the aortic valve opens widely and
tial negative consequences on the function of those upstream offers a pathway of very low resistance through which blood
organs. leaves the left ventricle. If this opening is narrowed
A brief overview of four of the common valve defects influ- (stenotic), resistance to flow through the valve increases. A
encing left ventricular function is given in Figure 25–11. The significant pressure difference between the left ventricle
reader should note that similar stenotic and regurgitant abnor- and the aorta may be required to eject blood through a

A B

150

Aortic pressure
100

Left ventricular
pressure

Left atrial
50
pressure

ECG

Phonocardiogram

C D

Aortic pressure
100
Left ventricular
pressure

Left atrial
pressure
50

ECG

Phonocardiogram

FIGURE 25–11 Common valve abnormalities. A) Aortic stenosis. B) Mitral stenosis. C) Aortic regurgitation (insufficiency).
D) Mitral insufficiency. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 25 Cardiac Function Assessments 247

stenotic aortic valve. As shown in Figure 25–11A, intraven- tolic and a diastolic murmur are present. The primary physi-
tricular pressures may rise to very high levels during systole ological consequences of aortic insufficiency are a reduction
while aortic pressure rises more slowly than normal to a in forward flow out to the tissues (if the insufficiency is severe)
systolic value that is subnormal. Pulse pressure is usually and an increase in the volume workload of the left ventricle.
low with aortic stenosis. High intraventricular pressure
development is a strong stimulus for cardiac muscle cell
hypertrophy, and an increase in left ventricular muscle mass MITRAL REGURGITATION
invariably accompanies aortic stenosis. This tends to pro-
Typical characteristics of mitral regurgitation (insufficiency,
duce a leftward deviation of the electrical axis. (The mean
incompetence) are shown in Figure 25–11D. When the mitral
electrical axis will fall in the upper right-hand quadrant of
valve is insufficient, some blood regurgitates from the left ven-
Figure 25–5.) Blood being ejected through the narrowed
tricle into the left atrium during systole. A systolic murmur
orifice may reach very high velocities, and turbulent flow
may accompany this abnormal flow pattern. Left atrial pressure
may occur as blood enters the aorta. This abnormal turbu-
is increased to abnormally high levels, and left ventricular EDV
lent flow can be heard as a systolic (or ejection) murmur
and pressure increase. Mitral valve prolapse is a common form
with a properly placed stethoscope. The primary physiolog-
of mitral insufficiency in which the valve leaflets evert into the
ical consequence of aortic stenosis is a high ventricular
left atrium during systole. The primary physiological conse-
afterload that is caused by restriction of the outflow tract.
quences of mitral regurgitation are somewhat similar to aortic
This imposes an increased pressure workload on the left
insufficiency in that forward flow out of the left ventricle into
ventricle.
the aorta may be compromised (if the insufficiency is severe)
and there is an increase in the volume workload of the left ven-
MITRAL STENOSIS tricle. In addition, the elevated left atrial pressure can also lead
to pulmonary effects with shortness of breath.
Some characteristics of mitral stenosis are shown in
Figure 25–11B. A pressure difference of more than a few mil-
limeters of mercury across the mitral valve during diastole is
distinctly abnormal and indicates that this valve is stenotic. CLINICAL CORRELATION
The high resistance mandates an increased pressure difference
to achieve normal flow across the valve (Q̇ = ΔP/R). Conse- A 72-year-old man comes to the doctor’s office with com-
quently, as shown in Figure 25–11B, left atrial pressure is plaints of diminished exercise tolerance. He has had dys-
increased with mitral stenosis. The high left atrial workload pnea (shortness of breath) on exertion for several years
may induce hypertrophy of the left atrial muscle. Increased left but it has gotten worse lately. He now experiences chest
atrial pressure is reflected back into the pulmonary bed and, if pain and some dizziness with only mild exertion and, the
high enough, causes pulmonary edema and pulmonary vascu- day before his appointment, he fainted when getting out
lar congestion. A diastolic murmur associated with turbulent of bed. His heart rate is 75 beats/min and his blood pres-
flow through the stenotic mitral valve can often be heard. The sure is 113/90 mm Hg. A loud systolic murmur is heard
primary physiological consequences of mitral stenosis are using a stethoscope placed above the aorta, and a slowly
increases in left atrial pressure and pulmonary capillary pres- rising pulse is detected in his radial artery. An ECG reveals
sure. The latter can cause interference with normal gas exchange normal rate and rhythm but significant left ventricular
in the lungs, leading to dyspnea (shortness of breath). hypertrophy (positive R wave in lead I, negative R wave in
lead AVF , and large R-wave amplitudes in leads aVl, V5,
and V6). Echocardiography indicated significant left ven-
AORTIC INSUFFICIENCY tricular wall thickening and significant narrowing of the
aortic valve opening during systole.
Typical characteristics of aortic regurgitation (insufficiency, This man’s symptoms can all be a result of an increasing
incompetence) are shown in Figure 25–11C. When the leaflets severity of aortic valve stenosis. Because of the elevated
of the aortic valve do not provide an adequate seal, blood resistance to outflow (essentially an increased afterload),
regurgitates from the aorta back into the left ventricle during the left ventricular muscles must develop more force to
the diastolic period. Aortic pressure falls faster and further generate sufficient pressure to eject blood during systole.
than normal during diastole, which causes a low diastolic pres- Left ventricular pressure during systole will be much
sure and a large pulse pressure. In addition, ventricular EDV higher than aortic pressure during systole, thereby pro-
and pressure are higher than normal because of the extra blood ducing a significant pressure gradient. Over time, this
that reenters the chamber through the incompetent aortic increased workload induces hypertrophy of the left ven-
valve during diastole. Turbulent flow of the blood reentering tricular muscle that accounts for the leftward shift in the
the left ventricle during early diastole produces a characteris- mean electrical axis. The dyspnea on exertion is a result of
tic diastolic murmur. Often the aortic valve is altered so that it an exercise-induced imbalance in SV from the normal
is both stenotic and insufficient. In these instances, both a sys-
248 SECTION V Cardiovascular Physiology

decreases the coordination of myocyte contractions resulting in


right side and the abnormal left side of the heart and inadequate pressure development and ejection.
blood backing up into the pulmonary circulation. ■ Supraventricular arrhythmias are a result of abnormal action
Fainting may be a common symptom in patients with potential initiation at the SA node or altered propagation
aortic stenosis and, although it certainly reflects a decrease characteristics through the atrial tissue and the AV node.
in brain blood flow, the specific causes are not entirely ■ Tachycardias may originate either in the atria or ventricles and
clear. A popular theory is that because the left ventricular are a result of increased pacemaker automaticity or of continu-
SV (and therefore cardiac output) is nearly fixed and not ously circling pathways setting up a reentrant circuit.
able to adjust to the many cardiovascular challenges asso- ■ Abnormal conduction through the AV node results in
ciated with even mild exertion, arterial pressure decreases conduction blocks.
as a result of the unopposed decrease in peripheral vascu- ■ Abnormal conduction pathways in the Purkinje system or in
lar resistance. Other possibilities include a hypertrophy- the ventricular tissue result in significant QRS alterations.
induced predisposition to arrhythmias or a vasodilator ■ Ventricular tachycardia and ventricular fibrillation represent
reflex evoked by high left ventricular pressures. severe abnormalities that are incompatible with effective
The chest pain (angina pectoris) is a result of inade- cardiac pumping.
quate coronary blood flow to meet the myocardial meta- ■ A variety of methods are available for measuring various
bolic demands. Ischemia can be a result of either an aspects of cardiac mechanical function. These methods are
impediment to coronary flow (as might occur with coro- based on the Fick principle and various imaging techniques
nary artery disease or atherosclerosis) or an increase in including echocardiography.
metabolic demands. In this case, the increase in myocar- ■ The ejection fraction (which is the stroke volume divided by
dial work because of the aortic stenosis plus the accomp- the end-diastolic volume) and the ventricular end-systolic
pressure–volume relationship are very useful indices of cardiac
anying hypertrophy outstrips the ability of the coronary
contractility.
bed to provide sufficient flow. (Eventually, there will be
■ Failure of cardiac valves to open fully (stenosis) can result in
ischemia even at rest and ECG signs of ventricular strain
elevated upstream chamber pressure and abnormal pressure
and subendocardial ischemia will appear. These signs gradients, congestion in upstream vascular beds, chamber wall
include ST-segment depression and T-wave inversion.) hypertrophy, turbulent forward flow across the valve, and
The treatment for aortic stenosis is surgical replace- murmurs during systole or diastole.
ment of the aortic valve. ■ Failure of cardiac valves to close completely (insufficiency,
incompetence, regurgitation) can result in large stroke
volumes, abnormal pressure pulses, congestion in upstream
vascular beds, turbulent backward flow across the valve, and
CHAPTER SUMMARY murmurs during systole or diastole.

■ The electrocardiogram is a record of the voltage changes


that occur on the surface of the body as a result of the STUDY QUESTIONS
propagation of the action potential through the heart during
1. Your 75-year-old male patient is alert with complaints of general
a cardiac cycle.
fatigue. His heart rate = 90 beats/min and arterial pressure =
■ There are standardized conventions used for recording 140/50 mm Hg. A diastolic murmur is present. There are no ECG
electrocardiograms. abnormalities identified and mean electrical axis = 10°. Cardiac
■ The magnitude and direction of the net dipole formed by the catheterization indicate that LV pressure = 140/20 mm Hg and
wavefront of the action potential at any instant in time can be left atrial pressure = 10/3 mm Hg (as peak systolic/end diastolic).
deduced from the magnitude and orientation of the electrocar- Which of the following is most consistent with these findings?
diographic deflections. A) aortic stenosis
■ The mean electrical axis describes the orientation of the net B) aortic insufficiency
dipole at the instant of maximum wavefront propagation C) mitral stenosis
during ventricular depolarization and normally falls between D) mitral insufficiency
0° and +90° on a polar coordinate system. E) right ventricular hypertrophy
■ The standard 12-lead electrocardiogram is widely used to 2. Evaluation of your patient’s electrocardiogram shows that P
evaluate cardiac electrical activity and consists of a combina- waves occur at a regular rate of 90/min and QRS complexes occur
tion of bipolar and unipolar records from limb electrodes and at a regular rate of 37/min. Which of the following is the most
chest electrodes. likely diagnosis?
■ Cardiac arrhythmias can often be detected and diagnosed from A) supraventricular tachycardia
a single electrocardiographic lead. B) first-degree heart block
■ Physiological consequences of abnormal excitation and C) second-degree heart block
conduction in the heart depend on whether the electrical D) third-degree heart block
abnormality limits the time for adequate cardiac filling or E) bundle branch block
CHAPTER 25 Cardiac Function Assessments 249

3. Given the following information, calculate cardiac output and 5. Your patient’s ECG shows that R-wave amplitude on leads I
determine whether this would be a normal value for a healthy and aVF is upright and equally large. Which of the following
70 kg young adult: systemic arterial blood oxygen concentration, statements is true?
[O2]SA = 200 mL/L; pulmonary arterial blood oxygen A) This indicates a significant left electrical axis deviation.
concentration, [O2]PA = 140 mL/L; total body oxygen B) The mean electrical axis is +90°.
consumption, VO2 = 600 mL/min. C) The R-wave amplitude will be smallest on lead aVL.
A) 10 L/min that is normal for mild exercise D) The R-wave amplitude will be positive on lead aVR.
B) 10 L/min that is abnormally low at rest E) The left ventricle is hypertrophied.
C) 6 L/min that is close to a normal resting value
D) 0.6 L/min that is abnormally low at rest
E) 60 L/min that is impossible for normal individuals
4. Your patient takes a drug that decreases AV nodal action potential
conduction velocity. The direct effect of this drug will be seen on
the ECG as
A) a decrease in QRS-wave frequency.
B) an increase in P-wave amplitude.
C) an increase in the PR interval.
D) a widening of the QRS interval.
E) an increase in the ST-segment duration.
This page intentionally left blank
26
C H A P T E R

Peripheral Vascular
System
David E. Mohrman and Lois Jane Heller

O B J E C T I V E S

■ Define convective transport and diffusion and list the factors that determine
the rate of each.
■ Given data, use the Fick principle to calculate the rate of removal of a solute
from blood as it passes through an organ.
■ Describe how capillary wall permeability to a solute is related to the size and
lipid solubility of the solute.
■ List the factors that influence transcapillary fluid movement and, given data,
predict the direction of transcapillary fluid movement.
■ Describe the lymphatic vessel system and its role in preventing fluid
accumulation in the interstitial space.
■ Given data, calculate the vascular resistances of networks of vessels arranged
in parallel and in series.
■ Describe differences in the blood flow velocity in the various segments and
how these differences are related to their total cross-sectional area.
■ Describe laminar and turbulent flow patterns and the origin of flow sounds in
the cardiovascular system.
■ Identify the approximate percentage of the total blood volume that is
contained in the various vascular segments in the systemic circulation.
■ Define peripheral venous pool and central venous pool.
■ Describe the pressure changes that occur as blood flows through a vascular
bed and relate them to the vascular resistance of the various vascular
segments.
■ State how the resistance of each consecutive vascular segment contributes to
an organ’s overall vascular resistance and, given data, calculate the overall
resistance.
■ Define total peripheral resistance (systemic vascular resistance) and
state the relationship between it and the vascular resistance of each
systemic organ.
■ Define vascular compliance and state how the volume–pressure curves for
arteries and veins differ.
■ Predict what will happen to venous volume when venous smooth muscle
contracts or when venous transmural pressure increases.
■ Describe the role of arterial compliance in storing energy for blood
circulation.
(Continued)

251

Ch26_251-262.indd 251 11/26/10 11:50:20 AM


252 SECTION V Cardiovascular Physiology

O B J E C T I V E S (Continued)

■ Describe the auscultation technique of determining arterial systolic and


diastolic pressures.
■ Identify the physical bases of the Korotkoff sounds.
■ Indicate the relationship between arterial pressure, cardiac output, and
total peripheral resistance and predict how arterial pressure will be
altered when cardiac output and/or total peripheral resistance change.
■ Given arterial systolic and diastolic pressures, estimate mean arterial
pressure.
■ Indicate the relationship between pulse pressure, stroke volume, and arterial
compliance and predict how pulse pressure will be changed by changes in
stroke volume, or arterial compliance.
■ Describe how arterial compliance changes with age and how this affects
arterial pulse pressure.

OVERVIEW OF THE PERIPHERAL CARDIOVASCULAR TRANSPORT


VASCULAR SYSTEM THE FICK PRINCIPLE
Homeostasis implies that each and every cell in the body is
Substances are carried between organs within the cardiovas-
continuously bathed in a local environment of constant com-
cular system by the process of convective transport, the sim-
position that is optimal for cell function. In essence, the
ple process of being swept along with the flow of the blood in
peripheral vascular system is a sophisticated irrigation system.
which they are contained. The rate at which a substance (X) is
Blood flow is continuously delivering nutrients to and remov-
transported by this process depends solely on the concentra-
ing waste products from the local interstitial environment
tion of the substance in the blood and the blood flow:
throughout the body.
The heart supplies the pumping power required to create Transport rate = Flow × Concentration
flow through the system. Because of the heart’s action, pres-
sure at the inlet (the aorta) of the vascular network is higher or
than that at its outlets (the vena cavae). Everywhere within the . .
vascular system, blood always flows from higher pressure to X = Q [X] (1)
. .
lower pressure according to well-known physical rules. Like where X is the rate of transport of X (mass/time), Q is the
water flowing downhill, blood seeks to travel along the path of blood flow (volume/time), and [X] is the concentration of X in
least resistance. Consequently, the peripheral vascular system blood (mass/volume).
changes the resistance of its various pathways to direct blood It is evident from the preceding equation that only two
flow to where it is needed. methods are available for altering the rate at which a substance
This chapter begins with a description of the mechanisms is carried to an organ: (1) a change in the blood flow through
responsible for the transport of dissolved substances through the organ or (2) a change in the arterial blood concentration of
the vascular system and the movement of these substances the substance. The preceding equation might be used, for
and fluid from capillaries to and from the interstitial space. example, to calculate how much oxygen is carried to a certain
Next, the basic equation for flow though a single vessel (Q = skeletal muscle each minute. Note, however, that this calcula-
ΔP/R, presented in Chapter 22) is applied to the complex tion would not indicate whether the muscle actually used the
network of branching vessels that actually exists in the car- oxygen carried to it.
diovascular system. Then, the consequences of the elastic One can extend the convective transport principle to determine
properties of the large diameter arteries and veins on overall a tissue’s rate of utilization (or production) of a substance by
cardiovascular system operation are considered. Finally, the simultaneously considering the transport rate of the substance to
principles of the routine clinical measurement of arterial and from the tissue. The relationship that results is referred to as
blood pressure are presented along with the conclusions the Fick principle and may be formally stated as follows:
about overall cardiovascular function that can be made from . .
the information. Xtc= Q ([X]a − [X]v) (2)
CHAPTER 26 Peripheral Vascular System 253

. .
where X tc is the transcapillary efflux rate of X (mass/time), Q Capillary beds allow huge amounts of materials to enter
the blood flow (volume/time), and [X]a,v the arterial and and leave blood because they maximize the area across
venous concentrations of X. which exchange can occur while minimizing the distance
The Fick principle demonstrates that the amount of a. sub- over which the diffusing substances must travel. Capillaries
stance that goes into an organ in a given are extremely fine vessels with a lumen (inner) diameter of
. period of time (Q[X]a)
minus the amount that comes out (Q [X]v) must equal the tis- about 5 μm, a wall thickness of approximately 1 μm, and an
sue utilization rate of that substance. (If the tissue is producing average length of perhaps 0.5 mm. (For comparison, a
substance X, then the above equation will yield a negative uti- human hair is roughly 100 μm in diameter.) Capillaries are
lization rate.) distributed in incredible numbers in organs and communi-
Recall that one method for determining cardiac output cate intimately with all regions of the interstitial space. It is
(CO) described in Chapter 25 used the Fick principle to calcu- estimated that there are about 1010 capillaries in the systemic
late the blood flow through the systemic circulation. In that organs with a collective surface area of about 100 m2. That is
case, the known variables included the systemic tissue oxygen roughly the area of one player’s side of a singles tennis court.
consumption rate and the concentrations of oxygen in arterial Recall from Chapter 22 that most cells are no more than
blood and mixed venous blood and the. above equation was about 10 μm (less than one tenth the thickness of paper)
rearranged to solve for the blood flow (Q ). from a capillary. Diffusion is a tremendously powerful
mechanism for material exchange when operating over such
a short distance and through such a large area. We are far
TRANSCAPILLARY SOLUTE DIFFUSION from being able to duplicate—in an artificial lung or kidney,
for example—the favorable geometry for diffusional
Capillaries act as efficient exchange sites where most substances exchange that exists in our own tissues.
cross the capillary walls by passively diffusing from regions of As diagrammed in Figure 26–1, the capillary wall itself con-
high concentration to regions of low concentration (see Chap- sists of only a single thickness of endothelial cells joined to
form a tube. The ease with which a particular solute crosses
ter 1). There are four factors that determine the diffusion rate
the capillary wall is expressed in a parameter called its capil-
of a substance between the blood and the interstitial fluid: (1) lary permeability. Permeability takes into account all the fac-
the concentration difference, Δ[X], (2) the surface area for tors (diffusion coefficient, diffusion distance, and surface
exchange, A, (3) the diffusion distance, ΔL, and (4) the perme- area)—except concentration difference—that affect the rate at
ability of the capillary wall to the diffusing substance repre- which a solute crosses the capillary wall.
sented as the diffusion coefficient, D. These factors are Two fundamentally distinct pathways exist for transcapillary
exchange. Lipid-soluble substances, such as the gases oxygen
combined in an equation (Fick’s first law of diffusion) that
. and carbon dioxide, cross the capillary wall easily. Because the
describes the diffusion (X d) of a substance X across a barrier: lipid endothelial cell plasma membranes are not a significant
. Δ[X] diffusion barrier for lipid-soluble substances, transcapillary
Xd= DA ____
ΔL
(3)

Interstitium

Endothelial cell
Water-filled
channels Cytoplasm
1 μ m thick
Plasma
Plasma membranes

40 Å

O2, CO2,
Na+, K+
ethanol
Cl– , H2O,
glucose Lipid-soluble
Proteins substances

FIGURE 26–1 Pathways for transcapillary


Small solute diffusion. (Modified with permission from
water-soluble
Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed.
substances
New York: Lange Medical Books/McGraw-Hill, 2006.)
254 SECTION V Cardiovascular Physiology

movement of these substances can occur through the entire important for a host of physiological functions, including the
capillary surface area. maintenance of circulating blood volume, intestinal fluid
The capillary permeability to small polar particles such as absorption, tissue edema formation, and saliva, sweat, and
sodium and potassium ions is about 10,000-fold less than that urine production. Net fluid movement out of capillaries is
for oxygen. Nevertheless, the capillary permeability to small referred to as filtration, and fluid movement into capillaries is
ions is several orders of magnitude higher than the permeabil- called reabsorption.
ity that would be expected if the ions were forced to move Fluid flows through transcapillary channels in response to
through the lipid plasma membranes. It is therefore postulated pressure differences between the interstitial and intracapillary
that capillaries are somehow perforated at intervals with water- fluids according to the basic flow equation. However, both
filled channels or pores (which may actually be clefts between hydrostatic and osmotic pressures influence transcapillary
the endothelial cells). Calculations from diffusion data indi- fluid movement. How hydrostatic pressure provides the driv-
cate that the collective cross-sectional area of the pores relative ing force for causing blood flow along vessels has been dis-
to the total capillary surface area varies greatly between capil- cussed previously. The hydrostatic pressure inside capillaries,
laries in different organs. Brain capillaries appear to be very Pc, is about 25 mm Hg and is the driving force that causes blood
tight (have few pores), whereas capillaries in the kidney and to return to the right heart from the capillaries of systemic
fluid-producing glands are much more leaky. On the average, organs. In addition, however, the 25-mm Hg hydrostatic intra-
however, pores constitute only a very small fraction of total capillary pressure tends to cause fluid to flow through the tran-
capillary surface area—perhaps 0.01%. This area is, neverthe- scapillary pores into the interstitium where the hydrostatic
less, sufficient to allow very rapid equilibration of small water- pressure (Pi) is near 0 mm Hg. Thus, there is normally a large
soluble substances between the plasma and interstitial fluids of hydrostatic pressure difference favoring fluid filtration across
most organs. Thus, the concentrations of inorganic ions mea- the capillary wall. Our entire plasma volume would soon be in
sured in a plasma sample can be taken to indicate their con- the interstitium if there were not some counteracting force
centrations throughout the entire extracellular space. tending to draw fluid into the capillaries. The balancing force is
In general, albumin and other large plasma proteins cannot an osmotic pressure that arises from the fact that plasma has a
easily cross capillary walls. The precise mechanism for the low higher protein concentration than interstitial fluid.
capillary permeability to proteins is in dispute. One hypothesis Recall that water always tends to move from regions of low
is that capillary pores are just physically smaller than the diam- to regions of high total solute concentration in establishing
eter of plasma protein molecules. Whatever the mechanism(s), osmotic equilibrium. Also recall that the driving force for
the result is that much higher protein concentrations normally osmotic water movement between one solution and another
exist in blood plasma than in interstitial fluid. can be expressed as an osmotic pressure difference between the
two. The osmotic pressure difference is directly related to the
difference in total solute concentration in the two solutions in
ENDOTHELIAL CELLS question. Because plasma and interstitial fluid are essentially
identical except for their protein concentrations, plasma pro-
In addition to forming capillaries, a layer of endothelial cells
teins are primarily responsible for the net osmotic pressure
lines the entire cardiovascular system—including the heart
difference across capillary walls. The component of total
chambers and valves. Because of their ubiquitous and intimate
osmotic pressure due to proteins has been given the special
contact with blood, endothelial cells have evolved to serve
name, oncotic pressure (or colloid osmotic pressure).
many functions in addition to acting as a barrier to transcapil-
Because of plasma proteins, the oncotic pressure of plasma
lary solute and water exchange. For example, endothelial cell
(πc) is about 25 mm Hg. Due to the absence of proteins, the
membranes contain specific enzymes that convert some circu-
oncotic pressure of the interstitial fluid (πi) is near 0 mm Hg.
lating hormones from inactive to active forms. Endothelial
Thus, there is normally a large osmotic force for fluid reab-
cells are also intimately involved in producing substances that
sorption into capillaries. The forces that influence transcap-
lead to blood clot formation and the stemming of bleeding in
illary fluid movement are summarized on the left side of
the event of tissue injury. Moreover, and as will be discussed in
Figure 26–2.
the next chapter, the endothelial cells lining muscular vessels
The relationship among the factors that influence transcap-
such as arterioles can produce vasoactive substances that act
illary fluid movement, known as the Starling hypothesis, can
on the smooth muscle cells that surround them to influence
be expressed by the following equation:
arteriolar diameter.
Net filtration rate = k[(Pc − Pi) − (πc − πi)] (4)
TRANSCAPILLARY FLUID MOVEMENT where Pc is the hydrostatic pressure of intracapillary fluid,
πc the oncotic pressure of intracapillary fluid, Pi and πi the
In addition to providing a diffusion pathway for polar mole- hydrostatic and oncotic pressures for interstitial fluid,
cules, the water-filled channels that traverse capillary walls respectively, and k a constant expressing how readily fluid can
permit fluid flow through the capillary wall. Net shifts of fluid move across capillaries (essentially the reciprocal of the resis-
between the capillary and interstitial compartments are tance to fluid flow through the capillary wall).
CHAPTER 26 Peripheral Vascular System 255

Interstitium The lymphatic system begins in the tissues with blind-end


lymphatic capillaries that are roughly equivalent in size to but
less numerous than regular capillaries. These capillaries are
Endothelial cell very porous and easily collect large particles accompanied by
interstitial fluid. This fluid, called lymph, moves through the
Net fluid converging lymphatic vessels, is filtered through lymph nodes
Plasma filtration where bacteria and particulate matter are removed, and reen-
Pc – Pi > πc – πi ters the circulatory system through the thoracic duct near the
Hydrostatic
Pi Pc point where the blood enters the right heart.
Flow of lymph from the tissues toward the entry point into
No net
movement
the circulatory system is promoted by (1) increases in tissue
Osmotic Pc – Pi = πc – πi interstitial pressure (due to fluid accumulation or to move-
πi πc ment of surrounding tissue), (2) contractions of the lymphatic
vessels, and (3) valves located in these vessels to prevent back-
ward flow.
Net fluid Roughly 2.5 L of lymphatic fluid enters the cardiovascular
reabsorption system each day. In the steady state, this indicates a total body
Pc – Pi < πc – πi
net transcapillary fluid filtration rate of 2.5 L per day. When
FIGURE 26–2 Factors influencing transcapillary fluid compared with the total amount of blood that circulates each
movement. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular day (about 7,000 L), this may seem like an insignificant amount
Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
of net capillary fluid leakage. However, lymphatic blockage is a
very serious problem and is accompanied by severe swelling
(lymphedema). Thus, the lymphatics play a critical role in
Fluid balance within a tissue (the absence of net transcapil- keeping the interstitial protein concentration low and in
lary water movement) occurs when the bracketed term in this removing excess capillary filtrate from the tissues.
equation is zero. This equilibrium may be upset by alterations
in any of the four pressure terms. The pressure imbalances that
cause capillary filtration and reabsorption are indicated on the BASIC VASCULAR FUNCTION
right side of Figure 26–2.
In most tissues, rapid net filtration of fluid is abnormal and RESISTANCE AND FLOW IN
causes tissue swelling as a result of excess fluid in the intersti- NETWORKS OF VESSELS
tial space (edema). For example, a substance called histamine
is often released in damaged tissue. One of the actions of his- In. Chapter 22, it was asserted that the basic flow equation
tamine is to increase capillary permeability to the extent that (Q =ΔP/R) may be applied to networks of tubes as well as to
proteins leak into the interstitium. Net filtration and edema individual tubes. The reason is that any network of resis-
accompany histamine release, in part because the oncotic tances, however complex, can always be reduced to a single
pressure difference (πc – πi) is reduced below normal. “equivalent” resistor that relates the total flow through the
Transcapillary fluid filtration is not always detrimental. network to the pressure difference across the network. To do
Indeed, fluid-producing organs such as salivary glands and so, one must make use of the two equations below for series
kidneys utilize high intracapillary hydrostatic pressure to pro- (one after another) and parallel (side-by-side) networks of
duce continuous net filtration. Moreover, in certain abnormal individual vessels. When vessels with individual resistances
situations, such as severe loss of blood volume through hem- R1, R2, …, Rn are connected in series, the overall resistance of
orrhage, the net fluid reabsorption accompanying diminished the network is given by the following formula:
intracapillary hydrostatic pressure helps restore the volume of
circulating fluid. Rs = R1 + R2 + … + Rn (5)

Figure 26–3A shows an example of three vessels connected


LYMPHATIC SYSTEM in series between some region where the pressure is Pi and
another region with a lower pressure Po, so that the total pres-
Despite the extremely low capillary permeability to proteins, these sure difference across the network, ΔP, is equal to Pi – Po. By
molecules as well as other large particles such as long-chain fatty the series resistance equation, the total resistance across this
acids and bacteria find their way into the interstitial space. If such network (Rs) is equal to R1 + R2 + R3. By
. the basic flow equa-
particles are allowed to accumulate in the interstitial space, filtration tion, the flow through the network. (Q ) is equal to ΔP/Rs. It
forces will ultimately exceed reabsorption forces and edema will should be intuitively obvious that Q is also the flow (volume/
result. The lymphatic system represents a pathway by which large time) through each of the elements in the series as indicated
molecules reenter the circulating blood. in Figure 26–3B. (Fluid particles may move with different
256 SECTION V Cardiovascular Physiology

velocities [distance/time] in different elements of a series net- A


R1
R3
work, but the volume that passes through each element in a R2
minute must be identical.) Pi Q P0
As shown in Figure 26–3C, a portion of the total pressure a b
c d
decrease across the network occurs within each element of the
series. The pressure decrease across any element in the series Rs = R1 + R2 + R3
can be calculated by applying . the basic flow equation to that ele- ΔP• = Pi − P0
ment, for example, ΔP1 = Q R1. Note that the largest portion of Q = ΔP/Rs
the overall pressure decrease will occur across the element in the B
series with the largest resistance to flow (R2 in Figure 26–3). Q

As indicated in Figure 26–4, when several tubes with indi-

Flow
vidual resistances R1, R2, …, Rn are brought together to form a
parallel network of vessels, one can calculate a single overall
resistance for the parallel network Rp according to the follow- a b c d
ing formula: Position along network
70
1 __ C
__ = 1 + __
Rp R1 R2
1 1
+ … + __
Rn
(6) Pi

ΔP1 =Q ·R1
The total flow through a parallel network is determined by
ΔP/Rp. As the preceding equation implies, the overall resis- •
ΔP2 =Q ·R2 ΔP
Pressure
tance of any parallel network will always be less than that of

any of the elements in the network. (In the special case where ΔP3 =Q ·R3
the individual elements that form the network have identical
resistances Rx, the overall resistance of the network is equal P0
to the resistance of an individual element divided by the
number (n) of parallel elements in the network: Rp = Rx/n.) In a b c d
general, the more parallel elements that occur in the net- Position along network
work, the lower the overall resistance of the network. Thus, FIGURE 26–3 A–C) Series resistance network. (Modified with
for example, a capillary bed that consists of many individual permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York:
capillary vessels in parallel can have a very low overall resis- Lange Medical Books/McGraw-Hill, 2006.)
tance to flow even though the resistance of a single capillary
is relatively high.
As indicated in Figure 26–4, the basic flow equation may be PERIPHERAL BLOOD FLOW VELOCITIES
applied to any single element in the network or to the network
as a whole. For example,
. the flow through
. only the first element It is important to make the distinction between blood flow
of the network (Q 1) is given by Q 1 = ΔP/R1 , whereas
. the flow (volume/time) and blood flow velocity (distance/time) in the
through the entire parallel network is given by Qp = ΔP/Rp . peripheral vascular system. Linear velocity of flow at any point


R1 Q1 = ΔP/R1

Pi • P0
R2 Q2 = ΔP/R2


R3 Q3 = ΔP/R3

1 1 1 1
= + +
Rp R1 R2 R3

ΔP = Pi − P0
• • • •
Qtotal = Q1 + Q2 + Q3

Qtotal = ΔP/Rp
FIGURE 26–4 Parallel resistance network. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange
Medical Books/McGraw-Hill, 2006.)
CHAPTER 26 Peripheral Vascular System 257

is equal to the flow divided by the cross-sectional area. Con- across the tube as shown on the left side of Figure 26–6. Veloc-
sider the analogy of a stream whose water moves with greater ity is fastest along the central axis of the tube and falls to zero
velocity through shallow rapids than it does through an adja- at the wall. The concentric layers of fluid with different veloci-
cent deep pool. The volume of water passing through the pool ties slip smoothly over one another. Little mixing occurs
in a day (volume/time = flow), however, must equal that pass- between fluid layers so that individual particles move in
ing through the rapids in the same day. In such a series arrange- straight streamlines parallel to the axis of the flow. Laminar
ment, the flow is the same at all points along the channel but flow is very efficient because little energy is wasted on any-
the flow velocity varies inversely with the local cross-sectional thing but producing forward fluid motion.
area. The situation is the same in the peripheral vasculature, Because blood is a viscous fluid, its movement through a ves-
where blood flows most rapidly in the region with the smallest sel exerts a shear stress on the walls of the vessel. This is a force
total cross-sectional area (the aorta) and most slowly in the that wants to drag the inside surface (the endothelial cell layer)
region with the largest total cross-sectional area (the capillary of the vessel along with the flow. The endothelial cells that line
beds). Regardless of the differences in velocity, when the CO a vessel are able to sense (and possibly respond to) changes in
(flow into the aorta) is 5 L/min, the flow through the systemic the rate of blood flow through the vessel by detecting changes
capillaries (or arterioles, or venules) is also 5 L/min. The in the shear stress on them. Shear stress may also be an impor-
changes in flow velocity that occur as blood passes through tant factor in certain pathological situations. For example, ath-
the peripheral vascular system are shown in the top trace of erosclerotic plaques tend to form preferentially near branches
Figure 26–5. of large arteries where, for complex hemodynamic reasons
The important consequence of this slow flow through the cap- beyond the scope of this text, high shear stresses exist.
illaries is that it allows sufficient time for adequate solute and fluid When blood is forced to move with too high a velocity
exchange between the vascular and interstitial compartments. through a narrow opening, the normal laminar flow pattern
Blood normally flows through all vessels in the cardiovascu- may break down into the turbulent flow pattern shown on the
lar system in an orderly streamlined manner called laminar right side of Figure 26–6. With turbulent flow, there is much
flow. With laminar flow, there is a parabolic velocity profile internal mixing and friction. When the flow within a vessel is

Arteries Arterioles Capillaries Venules and veins


500 mm/s

Flow velocity
0.5 mm/s

Blood
volume
60%

2% 5%
12%

Systolic
Mean 100 mm Hg

25 mm Hg
Diastolic
blood pressure

Vascular
resistance

FIGURE 26–5 Flow velocities, blood volumes, blood pressures, and vascular resistances in the peripheral vasculature from aorta to
right atrium. Approximately 20% of the total volume is contained in the pulmonary system and the heart chambers and is not accounted for in
this figure. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
258 SECTION V Cardiovascular Physiology

Streamlines Velocity profile

FIGURE 26–6 Laminar and turbulent flow patterns.


(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Turbulent flow
Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) Laminar flow

turbulent, the resistance to flow is significantly higher than that waves (traveling back toward the heart) caused by discontinui-
predicted from the Poiseuille equation given in Chapter 22. ties such as branching points in the arterial system.
Turbulent flow also generates sound, which can be heard with A large pressure drop occurs in the arterioles, where the pul-
the aid of a stethoscope. Cardiac murmurs, for example, are satile nature of the pressure also nearly disappears. The aver-
manifestations of turbulent flow patterns generated by cardiac age capillary pressure is approximately 25 mm Hg. Pressure
valve abnormalities. Detection of sounds from peripheral continues to decrease in the venules and veins as blood returns
arteries (bruits) is abnormal and usually indicates a pathologi- to the right heart. The central venous pressure (which is the
cal reduction of a large vessel’s cross-sectional area or an abnor- filling pressure for the right heart) is normally very close to
mally high blood flow through an organ. 0 mm Hg.

PERIPHERAL BLOOD VOLUMES PERIPHERAL VASCULAR RESISTANCES


The second trace in Figure 26–5 shows the approximate per- The bottom trace in Figure 26–5 indicates the relative resistance
centage of the total circulating blood volume that is contained to flow that exists in each of the consecutive vascular regions.
in the different vascular regions of the systemic organs at any Because arterioles have such a large vascular resistance in com-
instant. parison to the other vascular segments, the overall vascular
Note that most of the circulating blood is contained within resistance of any organ is determined to a very large extent by
the veins of the systemic organs. This diffuse but large blood the resistance of its arterioles. Arteriolar resistance is strongly
reservoir is often referred to as the peripheral venous pool. A influenced by arteriolar radius (R is proportional to 1/r4). Thus,
second but smaller reservoir of venous blood, called the central the blood flow through an organ is primarily regulated by
venous pool, is contained in the great veins of the thorax and adjustments in the internal diameter of arterioles caused by
the right atrium. When peripheral veins constrict, blood is dis- contraction or relaxation of their muscular arteriolar walls.
placed from the peripheral venous pool and enters the central When the arterioles of an organ change diameter, not only
pool. An increase in the central venous volume, and thus pres- does the flow to the organ change, but the manner in which
sure, enhances cardiac filling, which in turn augments stroke the pressures decrease within the organ is also modified. The
volume (SV) according to the Starling law of the heart. This is effects of arteriolar dilation and constriction on the pressure
an extremely important mechanism of cardiovascular regula- profile within a vascular bed are illustrated in Figure 26–7.
tion and will be discussed in greater detail in Chapter 28. Arteriolar constriction causes a greater decrease in pressure
across the arterioles, and this tends to increase the arterial
pressure while it decreases the pressure in capillaries and veins.
PERIPHERAL BLOOD PRESSURES Conversely, increased organ blood flow caused by arteriolar
dilation is accompanied by decreased arterial pressure and
Blood pressure decreases in the consecutive segments with the increased capillary pressure. Because of the changes in capil-
pattern shown in the third trace of Figure 26–5. Recall from lary hydrostatic pressure, arteriolar constriction tends to cause
Figure 24–1 that aortic pressure fluctuates between a systolic transcapillary fluid reabsorption, whereas arteriolar dilation
and a diastolic value with each heartbeat, and the same is true tends to promote transcapillary fluid filtration.
throughout the arterial system. The average pressure in the
arch of the aorta, however, is about 100 mm Hg, and this mean
arterial pressure decreases by only a small amount within the TOTAL PERIPHERAL RESISTANCE
arterial system.
As indicated in Figure 26–5, arterial pulse pressure actually The overall resistance to flow through the entire systemic circu-
increases with distance from the heart, a phenomenon referred lation is called the total peripheral resistance (TPR; sometimes
to as peripheral peaking of pulse pressure. The hemodynamic called the systemic vascular resistance [SVR]). Because the
reasons for this are very complex but involve the positive addi- systemic organs are generally arranged in parallel (see Figure
tion of primary pressure waves produced by the heart (that 22–2), the vascular resistance of each organ contributes to the
travel much faster than blood flow does) and reflected pressure TPR according to the parallel resistance equation (6).
CHAPTER 26 Peripheral Vascular System 259

Arterial
compartment

Distending pressure (mm Hg)


100 ΔP
Blood pressure

ΔV

Arteriolar
dilation 50
Venous
Normal compartment
C B
D
Arteriolar d ΔP
tricte
constriction Cons A ΔV
l
Arteries Arterioles Capillaries Veins 0 Norma
Volume
FIGURE 26–7 Effect of changes in arteriolar resistance on FIGURE 26–8 Volume–pressure curves of arterial and venous
vascular pressures. (Modified with permission from Mohrman DE, Heller LJ:
compartments. (Modified with permission from Mohrman DE, Heller LJ:
Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)

ELASTIC PROPERTIES OF (pooling) in these vessels as might be represented by a shift from


ARTERIES AND VEINS point A to point B in Figure 26–8. Fortunately, this process can
be counteracted by active venous constriction. The dashed line
Arteries and veins contribute only a small portion to the overall in Figure 26–8 shows the venous volume–pressure relationship
resistance to flow through a vascular bed. Therefore, changes in that exists when veins are constricted by activation of venous
their diameters have no significant effect on the blood flow through smooth muscle. In constricted veins, volume may be normal
systemic organs. The elastic behavior of arteries and veins is, how- (point C) or even below normal (point D) despite greater-than-
ever, very important to overall cardiovascular function because normal venous pressure. Peripheral venous constriction tends
they can act as reservoirs and substantial amounts of blood can be to increase peripheral venous pressure and shift blood out of the
stored in them. Arteries or veins behave more like balloons with peripheral venous compartment.
one pressure throughout rather than as resistive pipes with a flow- The elasticity of arteries allows them to act as a reservoir on a
related pressure difference from end-to-end. Thus, think of an beat-to-beat basis. Arteries play an important role in converting
“arterial compartment” and a “venous compartment,” each with the pulsatile flow output of the heart into a steady flow through
an internal pressure that is related to the volume of blood within it the vascular beds of systemic organs. During the early rapid
at any instant and how easily its walls can be stretched. phase of cardiac ejection, the arterial volume increases because
This is characterized by a parameter called compliance (C; blood is entering the aorta more rapidly than it is passing into
see Chapter 1) given as follows that describes how much its systemic arterioles. Thus, part of the work the heart does in
volume changes (ΔV) in response to a given change in distend- ejecting blood goes to stretching the elastic walls of arteries.
ing pressure (ΔP), which is the difference between the internal Toward the end of systole and throughout diastole, arterial vol-
and external pressures on the vascular walls: ume decreases because the flow out of arteries exceeds flow into
the aorta. Previously stretched arterial walls recoil to shorter
ΔV
C = ___
ΔP
(7) lengths, and in the process give up their stored potential energy.
This reconverted energy is what actually does the work of pro-
Volume–pressure curves for the systemic arterial and venous pelling blood through the peripheral vascular beds during dias-
compartments are shown in Figure 26–8. It is apparent from the tole. If arteries were rigid tubes that could not store energy by
disparate slopes of the curves in this figure that the elastic proper- expanding elastically, arterial pressure would fall immediately
ties of arteries and veins are very different. For the arterial com- to zero with the termination of each cardiac ejection.
partment, the ΔV/ΔP measured near a normal operating pressure
of 100 mm Hg indicates a compliance of about 2 mL/mm Hg. By
contrast, the venous pool has a compliance of over 100 mL/mm
Hg near its normal operating pressure of 5–10 mm Hg. MEASUREMENT OF
Because veins are so compliant, even small changes in periph- ARTERIAL PRESSURE
eral venous pressure can cause a significant amount of the circu-
lating blood volume to shift into or out of the peripheral venous Recall that the systemic arterial pressure fluctuates with each
pool. Standing upright, for example, increases venous pressure heart cycle between a diastolic value (PD) and a higher systolic
in the lower extremities and promotes blood accumulation value (PS). Obtaining estimates of an individual’s systolic and
260 SECTION V Cardiovascular Physiology

diastolic pressures is one of the most routine diagnostic tech- blood through the systemic organs. One of the most fundamen-
niques available to the clinician. The basic principles of the tal equations of cardiovascular physiology is that which indi-

auscultation technique used to measure blood pressure are cates how mean arterial pressure (Pa) is related to CO and TPR:
described as follows with the aid of Figure 26–9: –
Pa = CO × TPR (8)
1. An inflatable cuff is wrapped around the upper arm, with a
The above equation
. is simply a rearrangement of the basic
recording device attached to monitor the pressure within the
flow equation Q = ΔP /R applied to the entire systemic circula-
cuff. The cuff is initially inflated with air to a pressure (usually
tion with the single assumption that central venous pressure is
175–200 mm Hg) that is well above normal systolic values. –
approximately zero so that ΔP = Pa . Note that mean arterial
This pressure collapses all blood vessels under the cuff.
pressure is influenced both by the heart (via CO) and by the
2. After initial inflation, air is gradually “bled” from the cuff
peripheral vasculature (via TPR). All changes in mean arterial
so that the pressure within it decreases slowly and steadily
pressure result from changes in either CO or TPR.
through the range of arterial pressure fluctuations.
Calculating the true value of mean arterial pressure requires
3. The moment cuff pressure decreases below peak systolic ar-
mathematically averaging the arterial pressure waveform over
terial pressure, some blood is able to pass beneath the cuff
one or more complete heart cycles. Most often, however, we
during the systolic phase of the cycle. Because the flow
know from auscultation only the systolic and diastolic pres-
through these partially compressed vessels is intermittent
sures, yet wish to make some estimate of the mean arterial
and turbulent, tapping sounds can be detected with a stetho-
pressure. Mean arterial pressure necessarily falls between the
scope placed over the radial artery at the elbow. As indicated
systolic and diastolic pressures. A useful rule of thumb is that
in Figure 26–9, sounds of varying character, known collec- –
mean arterial pressure (Pa) is approximately equal to diastolic
tively as Korotkoff sounds, are heard whenever the cuff
pressure (PD) plus one third of the difference between systolic and
pressure is between the systolic and diastolic arterial pres-
diastolic pressures (PS – PD).
sures. The highest cuff pressure at which tapping sounds are
heard is taken as the systolic arterial pressure.
4. When the cuff pressure decreases below the diastolic pres- ARTERIAL PULSE PRESSURE
sure, blood flows through the vessels beneath the cuff
The arterial pulse pressure (Pp) is defined simply as systolic
without periodic interruption and again no sound is de-
pressure minus diastolic pressure (PS – PD). To be able to use
tected over the radial artery. The cuff pressure at which the
pulse pressure to deduce something about how the cardiovascu-
sounds become muffled or disappear is taken as the dia-
lar system is operating, one must do more than just define it. It
stolic arterial pressure.
is important to understand what determines pulse pressure, that
is, what causes it to be what it is and what can cause it to change.
In a previous section of this chapter, there was a brief discussion
DETERMINANTS OF about how, as a consequence of the compliance of the arterial
ARTERIAL PRESSURE vessels, arterial pressure increases as arterial blood volume is
expanded during cardiac ejection. The magnitude of the pres-
MEAN ARTERIAL PRESSURE sure increase (ΔP) caused by an increase in arterial volume
depends on how large the volume change (ΔV) is and on how
Mean arterial pressure is a critically important cardiovascular compliant (Ca) the arterial compartment is: ΔP = ΔV/Ca. If, for
variable because it is the average effective pressure that drives the moment, the fact that some blood leaves the arterial com-

Cuff pressure
Arterial pressure
A
120
mm Hg

80

FIGURE 26–9 Blood pressure measurement by


auscultation. Point A indicates systolic pressure and Lou
der fter
point B indicates diastolic pressure. (Modified with So
permission from Mohrman DE, Heller LJ: Cardiovascular Physiology,
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) No sound Korotkoff sounds No sound
CHAPTER 26 Peripheral Vascular System 261

partment during cardiac ejection is neglected, then the increase in SV. Changes in TPR, however, have little or no effect on pulse
in arterial volume during each heartbeat is equal to the SV. pressure, because a change in TPR causes parallel changes in
Thus, pulse pressure is, to a first approximation, equal to SV both systolic and diastolic pressures.
divided by arterial compliance: A common misconception in cardiovascular physiology is
SV that the systolic pressure alone or the diastolic pressure alone
Pp ∼ ___
− C (9) indicates the status of a specific cardiovascular variable. The
Arterial pulse pressure is about 40 mm Hg in a normal resting reader should not attempt to interpret systolic and diastolic
young adult because SV is about 80 mL and arterial compliance pressure values independently. Interpretation is much more
is about 2 mL/mm Hg. Pulse pressure tends to increase with age straightforward when the focus is on mean arterial pressure

in adults because of a decrease in arterial compliance (and (Pa = CO × TPR) and arterial pulse pressure (Pp ∼ − SV/Ca).
increase in arterial stiffness also referred to as “hardening of the
arteries” or arteriosclerosis. This differs from atherosclerosis
which involves deposition of fat in the vessel wall). Arterial vol-
ume–pressure curves for a 20- and a 70-year-old are shown in
Figure 26–10. The increase in arterial stiffness with age is indi- CLINICAL CORRELATION
cated by the steeper curve for the 70-year-old (more ΔP for a A 27-year-old woman comes to the clinic because of the
given ΔV) than for the 20-year-old. Thus, a 70-year-old will nec- overnight onset of a pain in her left leg and swelling in her
essarily have a larger pulse pressure for a given SV than a left ankle and foot. She describes the pain as a cramping
20-year-old. As indicated in Figure 26–10, the increase in arte- sort of deep ache. She had returned to the United States
rial stiffness is sufficient to cause increased pulse pressure even yesterday on a 12-hour flight from Brazil where she had
though SV tends to decrease with age (Also see Figure 73–1). spent several weeks on an expedition to the rain forests.
Figure 26–10 also illustrates the fact that arterial blood vol- She takes no drugs except birth control pills containing
ume and mean arterial pressure tend to increase with age. The estrogen (see Chapter 68). She is 1.73-m tall and weighs
increase in mean arterial pressure is not caused by the decreased 93 kg. Vital signs are all within normal ranges. On exami-
arterial compliance, however, because compliance changes do nation, it is noted that her left lower leg is sensitive to
not directly influence either CO or TPR, which are the sole touch and her left foot feels warmer than her right.

determinants of Pa. Mean arterial pressure tends to increase Furthermore, there is edema with her left ankle and foot
with age because of an age-dependent increase in TPR that is significantly swollen compared to her right.
controlled primarily by arterioles, not arteries. The symptoms suggest that there is an imbalance between
Arterial compliance also decreases with increasing mean filtration and absorptive forces at work in the capillaries of
arterial pressure as evidenced by the curvature of the volume– the lower left leg. Because these symptoms are restricted to
pressure relationships shown in Figure 26–10. Otherwise, one leg and not both, overall circulatory abnormalities that
arterial compliance is a relatively stable parameter. Thus, most might cause ankle edema can be eliminated (e.g., decreased
acute changes in arterial pulse pressure are the result of changes

200

20-year-old 70-year-old
Arterial pressure (mm Hg)

ΔP
100 ΔP

ΔV
ΔV

0
Arterial volume
FIGURE 26–10 Effect of age on systemic arterial volumes and pressures and arterial stiffness. (Modified with permission from Mohrman DE,
Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
262 SECTION V Cardiovascular Physiology

■ Turbulent blood flow is usually abnormal and makes noise


plasma proteins which would decrease plasma oncotic (murmurs and bruits).
pressure; or right-sided heart failure, liver or kidney disease ■ Veins contain most of the total blood volume.
which would increase fluid retention and peripheral venous ■ Arterioles contribute most to the resistance to flow through
pressure). Local factors that might cause fluid to accumu- organs.
late in the interstitial space could include situations that ■ Arteriolar constriction tends to reduce flow through an
either prevent lymphatic drainage of this space (i.e., organ, reduce capillary hydrostatic pressure, and promote
lymphedema from tropical pathogenic filarial parasites) or transcapillary fluid reabsorption within the organ.
increase hydrostatic pressure in the veins draining the tis- ■ Venous constriction is important for cardiac filling and the
sue. Because of the rapid onset and symptoms, it is much ability to cope with blood loss.
more likely that a clot has formed in one of the large veins ■ Because arteries and arterioles are elastic, the intermittent
draining her left leg (deep vein thrombosis [DVT]) that has flow from the heart is converted to continuous flow through
increased the upstream capillary hydrostatic pressure, and capillaries.
caused the pain, filtration of fluid out of the vascular space, ■ Mean systemic arterial pressure is determined by the product
and edema of the tissue. of CO and TPR.
Being overweight and taking birth control pills contain- ■ Changes in arterial pulse pressure reflect changes in SV and/or
ing estrogens are risk factors that predispose this woman the compliance of the arterial space.
toward the development of such a clot. In addition, a long
period of time spent in a sitting position without moving
her legs (as might have occurred on her long airplane ride) STUDY QUESTIONS
allows blood to pool in these lower extremities and is an 1. Determine the rate of glucose uptake by an exercising skeletal
added risk factor for DVT or inflammation of the more muscle from the following data: arterial blood (glucose) = 50 mg
superficial veins (thrombophlebitis). In addition to the per 100 mL blood; muscle venous blood (glucose) = 30 mg per
localized discomfort, there is the real danger that these 100 mL blood; muscle blood flow = 60 mL/min.
clots can become dislodged from their anchorage in the A) 3,000 mg/min
leg vein, travel to the heart as an embolus, and become B) 1,200 mg/min
lodged in the lungs (pulmonary embolism). This can be a C) 30 mg/min
life-threatening event and requires immediate treatment. D) 20 mL/mg
E) 12 mg/min
Doppler ultrasonic examination of the patient’s leg
revealed the presence of DVT and she was treated with an 2. Which of the following conditions favor net absorption of
fluid out of the interstitial space and into the capillary bed
anticoagulant (heparin at first and then warfarin) as well
within an organ?
as drugs that can help dissolve clots. It is also possible that
A) increased interstitial protein concentration
this patient has an increased tendency to form clots (i.e.,
B) venous clot
she is hypercoagulable) in part due to being overweight.
C) decreased plasma protein concentration
There are some inherited forms of hypercoagulability that D) increased capillary pore size
she and her blood relatives can be tested for. It is possible E) arteriolar constriction
that she will require lifelong treatment with anticlotting 3. Which of the following is consistent with a normal mean arterial
drugs. Finally, she will be encouraged to switch to a differ- pressure but an abnormally high arterial pulse pressure?
ent birth control method, as estrogens can increase the A) low stroke volume
tendency to form clots. B) high heart rate
C) decreased total peripheral resistance
D) increased arterial stiffness
E) aortic valve stenosis
CHAPTER SUMMARY 4. Which of the following substances is likely to move most easily
■ Within the cardiovascular system, convection is used to across a skeletal muscle capillary wall?
transport substances between capillary beds and diffusion
A) potassium
is used to transport substances between blood and tissue.
B) glucose
■ Water may move out of (filtration) or into (reabsorption) C) oxygen
capillaries depending on the net balance of hydrostatic and D) water
osmotic forces across capillary walls. E) albumin
■ Plasma proteins are responsible for the major osmotic force 5. In which of the following vessels do red cells move with the
across capillary walls. fastest speed (distance/time)?
■ Lymphatic vessels serve to remove excess filtrate from tissues A) arteries
and keep interstitial protein concentration low. B) arterioles
■ The velocity of blood flow is indirectly proportional to the total C) capillaries
cross-sectional area of the vascular segment and is slowest in D) venules
capillaries. E) veins
27
C H A P T E R

Vascular Control
David E. Mohrman and Lois Jane Heller

O B J E C T I V E S

■ Define basal tone.


■ List several substances potentially involved in local metabolic control
of vascular tone.
■ State the local metabolic vasodilator hypothesis.
■ Describe how vascular tone is influenced by locally produced
endothelial factors and chemicals such as prostaglandins, histamine,
and bradykinin.
■ Describe the myogenic response of blood vessels.
■ Define active and reactive hyperemia and indicate a possible mechanism
for each.
■ Define autoregulation of blood flow and briefly describe the metabolic,
myogenic, and tissue pressure theories of autoregulation.
■ Define neurogenic tone and describe how sympathetic
(and parasympathetic) neural influences can alter it.
■ Describe how vascular tone is influenced by circulating catecholamines,
vasopressin, and angiotensin II.
■ List the major influences on the diameter of veins.
■ Describe in general how control of blood flow differs between organs with
strong local metabolic control of arteriolar tone and organs with strong
neurogenic control of arteriolar tone.
■ State the relative importance of local metabolic and neural control of
coronary blood flow.
■ Define systolic compression and indicate its relative importance to
blood flow in the endocardial and epicardial regions of the right and left
ventricular walls.
■ Describe the major mechanisms of blood flow control in skeletal muscle
and brain.

VASCULAR SMOOTH MUSCLE tractile activity of vascular smooth muscle cells, which are pres-
ent in the walls of all vessels except capillaries. Vascular smooth
The cardiovascular system must adjust the diameter of its vessels muscle is unique because it must maintain its vessel diameter in
to efficiently distribute the cardiac output among tissues with the face of the continuous distending pressure of the blood within
different current needs (the job of arterioles), and to regulate the it, and therefore sustain active tension for prolonged periods.
distribution of blood volume and cardiac filling (the job of veins). The basics of smooth muscle operation were presented in
Vascular diameter adjustments are made by regulating the con- Chapter 11. Here the focus is on the functional consequences of

263

Ch27_263-274.indd 263 11/26/10 11:51:15 AM


264 SECTION V Cardiovascular Physiology

influences on vascular smooth muscle that have particular rel- of basal tone is that it serves as a baseline from which arterio-
evance to the operation of the cardiovascular system. In general, lar tone can be regulated, either increased or decreased as nec-
these influences on vascular smooth muscle can be separated essary to meet the needs of changing situations. Another
into those that originate locally (and therefore have only local important consequence is that the basal tone of arterioles
consequences) and those that have global influences on vessels throughout the body collectively contributes directly to the
throughout the body. Examples of the latter are general changes total peripheral resistance (TPR) and thus to arterial blood
in autonomic nerve activity and changes in the blood concen- pressure in a resting individual.
trations of hormones that affect vascular smooth muscle. Such
influences have much different functional consequences on
LOCAL INFLUENCES ON ARTERIOLES
arterioles than on veins. Consequently, the operations of arteri-
oles and veins are described separately in this chapter.
Local Metabolic Influences
The arterioles that control flow through a given organ lie within
VASCULAR TONE the organ tissue itself. Thus, arterioles and the smooth muscle
in their walls are exposed to the chemical composition of the
Vascular tone is a term commonly used to characterize the
interstitial fluid of the organ they serve. The interstitial concen-
general contractile state of a vessel or a vascular region. The
trations of many substances reflect the balance between the
“vascular tone” of a region can be taken as an indication of
metabolic activity of the tissue and its blood supply. Interstitial
the “level of activation” of the individual smooth muscle cells
oxygen levels, for example, decrease when cells are using oxy-
in that region. An increase in arteriolar tone is automatically
gen faster than it is being supplied to the tissue by blood flow.
taken to imply a decrease in arteriolar vessel diameter, the
Conversely, interstitial oxygen levels increase whenever more
functional consequences of which are an increase in arteriolar
oxygen is delivered than is used by a tissue. In nearly all vascu-
resistance and a decrease in flow. The primary functional con-
lar beds, exposure to low oxygen reduces arteriolar tone and
sequence of an increase in venous tone is a decrease in venous
causes vasodilation, whereas high oxygen levels cause arterio-
volume and thus a peripheral-to-central shift of blood volume
lar vasoconstriction. Thus, a local feedback mechanism exists
that increases cardiac filling.
that automatically operates on arterioles to regulate blood flow
to a tissue in accordance with its metabolic needs.
CONTROL OF ARTERIOLAR TONE Many substances in addition to oxygen are present within
tissues and can affect the tone of vascular smooth muscle.
As described in Chapter 26, the blood flow through any organ When the metabolic rate of skeletal muscle is increased by
is determined largely by its vascular resistance, which is depen- exercise, for example, tissue levels of carbon dioxide, H+, and
dent primarily on the diameter of its arterioles. Consequently, K+ increase. These chemical alterations cause arteriolar dila-
an organ’s flow is controlled by factors that influence the arte- tion. In addition, with increased metabolic activity or oxygen
riolar smooth muscle tone. deprivation, cells in many tissues may release adenosine,
which is a potent vasodilator agent.
BASAL TONE At present, it is not known which of these (or possibly other)
metabolically related chemical alterations within tissues are
The arterioles in a healthy individual at rest have a certain level most important in the local metabolic control of blood flow. It
of basal tone that lies somewhere between complete relaxation is likely that arteriolar tone depends on the combined action of
and maximum possible constriction. A myriad of influences many factors.
on arteriolar smooth muscle contribute collectively to the Figure 27–1 summarizes current understanding of local
establishment of this basal tone. One important consequence metabolic control. Vasodilator factors enter the interstitial

Release proportional to tissue metabolism


Tissue cells

Removal rate proportional to blood flow


Vasodilator factors

FIGURE 27–1 Local metabolic vasodilator Blood flow


hypothesis. (Modified with permission from Mohrman
DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York:
Lange Medical Books/McGraw-Hill, 2006.) Arterioles Capillaries Veins
CHAPTER 27 Vascular Control 265

space from the tissue cells at a rate proportional to tissue transmural pressure. For example, a sudden increase in the
metabolism. These vasodilatory factors are removed from internal pressure within an arteriole produces first an initial
the tissue at a rate proportional to blood flow. Whenever tis- slight passive mechanical distention (slight because arterioles
sue metabolism is at a rate for which the blood flow is inade- are relatively thick-walled and muscular), and second, an
quate, the interstitial vasodilator concentrations build up and active constriction that, within seconds, may completely
cause the arterioles to dilate and blood flow to increase. The reverse the initial distention. A sudden decrease in transmural
process continues until blood flow has increased sufficiently to pressure elicits essentially the opposite response, that is, an
appropriately match the tissue metabolic rate and prevent fur- immediate passive decrease in diameter followed shortly by a
ther accumulation of vasodilator factors. The same system also decrease in active tone that returns the arteriolar diameter to
operates to reduce blood flow when it is greater than required near that which existed before the pressure change. The active
by the tissue metabolic activity because the interstitial concen- phase of such behavior is referred to as a myogenic response,
trations of metabolic vasodilator factors are decreased. because it seems to originate within the smooth muscle itself.
Local metabolic mechanisms are usually the most important The mechanism of the myogenic response is not known for
means of local blood flow control in most tissues. Individual certain, but stretch-sensitive ion channels on arteriolar vascu-
organs are therefore able to regulate their own blood flow in lar smooth muscle cells are likely candidates for involvement.
accordance with their metabolic needs. As indicated below, All arterioles have some normal distending pressure to
there are several other types of local influences on blood ves- which they probably are actively responding. Therefore, the
sels. However, many of these represent fine-tuning mecha- myogenic mechanism is likely to be a fundamentally impor-
nisms and may be important only in certain, usually tant factor in determining the basal tone of arterioles every-
pathological, situations. where. The myogenic response is potentially involved in the
vascular reaction to any cardiovascular disturbance that
Local Nonmetabolic Influences involves a change in arteriolar transmural pressure, as will be
discussed in the next section.
An ever-increasing number of local factors unrelated to tissue
metabolism have been shown to influence arterioles within an
organ. Table 27–1 contains a list of some of these more impor- FLOW RESPONSES CAUSED BY
tant factors and summarizes some of the information about
their actions. LOCAL MECHANISMS
Many of these factors exert their vascular effects by action on
In organs with a highly variable metabolic rate, such as skeletal
endothelial cells. Thus, endothelial cells can actively participate
and cardiac muscle, the blood flow closely follows the tissue
in the control of arteriolar diameter by producing local chemi-
metabolic rate. For example, skeletal muscle blood flow increases
cals that affect the tone of the surrounding smooth muscle cells.
within seconds of the onset of muscle exercise and returns to
The vasodilatory influence produced by endothelial cells is
control values shortly after exercise ceases. This phenomenon,
mediated by nitric oxide (NO). NO is produced within
which is illustrated in Figure 27–2A, is known as exercise or
endothelial cells from the amino acid, l-arginine, by the action
active hyperemia (hyperemia means high flow). It should be
of an enzyme, NO synthase, that is activated by an increase in
clear how active hyperemia could result from the local meta-
the intracellular level of Ca2+. NO is a small lipid-soluble mol-
bolic vasodilator feedback on arteriolar smooth muscle. As
ecule that, once formed, easily diffuses into adjacent smooth
mentioned previously, once initiated by local metabolic influ-
muscle cells where it causes relaxation by stimulating cGMP
ences on small resistance vessels, endothelial flow–dependent
production as mentioned in Chapter 11. Acetylcholine and
mechanisms may assist in propagating the vasodilation to larger
several other agents (including bradykinin, vasoactive intesti-
vessels upstream, which helps promote the delivery of blood to
nal peptide, and substance P) stimulate endothelial cell NO
the exercising muscle.
production because their receptors on endothelial cells are
Reactive or post-occlusion hyperemia is a greater-than-
linked to receptor-operated Ca2+ channels. Blood flow–related
normal blood flow that occurs transiently after the removal of
shear stresses on endothelial cells stimulate NO production
any restriction that has caused a period of lower-than-normal
through stretch-sensitive channels for Ca2+. This phenomenon
blood flow. The phenomenon is illustrated in Figure 27–2B.
may explain why exercise and increased blood flow through
For example, flow through an extremity is greater than normal
muscles of the lower leg can cause dilation of the blood-
for a period after a tourniquet is removed from the extremity.
supplying femoral artery at points far upstream from the exer-
Both local metabolic and myogenic mechanisms may be
cising muscle itself.
involved in producing reactive hyperemia. The magnitude and
duration of reactive hyperemia depend on the duration and
Transmural Pressure severity of the occlusion as well as the metabolic rate of the tis-
The passive elastic mechanical properties of arteries and veins sue. These findings are best explained by an interstitial accu-
and how changes in transmural pressure affect their diameters mulation of metabolic vasodilator substances during the period
were discussed in Chapter 26. The effect of transmural pres- of flow restriction. However, unexpectedly large flow increases
sure on arteriolar diameter is actually more complex because can follow arterial occlusions lasting only 1 or 2 seconds. These
arterioles respond both passively and actively to changes in may be best explained by a myogenic dilation response to the
266 SECTION V Cardiovascular Physiology

TABLE 27–1 Local influences on vascular smooth muscle.


Substance Description Vascular Response Other Information

Nitric oxide (NO), From endothelial cells in response to: Vasodilation Basal release may reduce normal net
endothelial-derived acetylcholine, vasoactive intestinal resting tone of vascular smooth muscle
relaxing factor (EDRF) peptide, substance P, bradykinin, shear locally throughout the body
stress, others
Acetylcholine Neurotransmitter—normal dilation Vasodilation and/or Important local effects on GI circulation
mediated by NO vasoconstriction (from enteric plexus neurons)
-Constriction occurs when
endothelium is absent/
damaged
Vasoactive intestinal Neurotransmitter (also a peptide Vasodilation GI neurotransmitter from enteric plexus
peptide (VIP) hormone)—action is mediated by NO neuron, causes smooth muscle relaxation,
promotes secretion in the gut

Substance P Neurotransmitter—action is mediated Vasodilation Important local roles in pain nociception,


by NO GI function, vomiting, skin circulation

Bradykinin Polypeptide formed from plasma protein Vasodilation -Increases vascular permeability
by action of enzyme, kallikrein—action is -Involved in pain mechanisms
mediated by NO

Shear stress Action is mediated by NO Vasodilation Dependent on flow velocity

Endothelial-derived Unknown factor from endothelial cells Vasodilation May be K+, cANP, electrogenic spread of
hyperpolarizing hyperpolarization, other possible factors
factor (EDHF)

Endothelin Polypeptide from endothelial cells Vasoconstriction Basal release may reduce normal net
resting tone of vascular smooth muscle
locally in tissues throughout the body

Prostacyclin (PGI2) Arachidonic acid (AA) metabolite of the Vasodilation Inflammatory responses, blocked by
cyclooxygenase (CO) pathway cyclooxygenase inhibitors such as aspirin

Thromboxane AA metabolite of the CO pathway (made Vasoconstriction Important for platelet aggregation and
by platelets) blood clotting, also blocked by aspirin

Other prostaglandins AA metabolites of the CO pathway Vasoconstriction Actions vary with the specific organ and
and/or vasodilation local conditions

Leukotrienes AA metabolites of the lipoxygenase Vasoconstriction Increase vascular permeability during


pathway and/or vasodilation inflammatory responses

Histamine Secretory granules of tissue mast cells Vasodilation Leads to an increase in vascular
and circulating basophils permeability, edema formation
-Involved in inflammatory and immune
reactions

reduced intravascular pressure and decreased stretch of the slightly increased blood flow because the increased driving
arteriolar walls during the period of occlusion. pressure is counteracted by a greater-than-normal vascular
Except when displaying active and reactive hyperemia, resistance. As with the phenomenon of reactive hyperemia,
nearly all organs tend to keep their blood flow constant despite blood flow autoregulation may be caused by both local meta-
variations in arterial pressure—that is, they display autoregu- bolic feedback mechanisms and myogenic mechanisms. The
lation of blood flow. As shown in Figure 27–3A, an abrupt arteriolar vasoconstriction responsible for the autoregulatory
increase in arterial pressure is normally accompanied by an response shown in Figure 27–3A, for example, may be partially
initial abrupt increase in organ blood flow that then gradually due to (1) a “washout” of metabolic vasodilator factors from
returns toward normal despite the sustained increase in arte- the interstitium by the excessive initial blood flow and (2) a
rial pressure. The initial increase in flow with increased pres- myogenic increase in arteriolar tone stimulated by the increase
sure is expected from the basic flow equation (Q̇ = Δ P / R). The in stretching forces that the increase in pressure imposes on
subsequent return of flow toward the normal level is caused by the vessel walls. There is also a tissue pressure hypothesis of
a gradual increase in active arteriolar tone and resistance to blood flow autoregulation for which it is assumed that an
blood flow. Ultimately, a new steady state is reached with only abrupt increase in arterial pressure causes transcapillary fluid
CHAPTER 27 Vascular Control 267

A A

Arterial pressure
Organ blood flow

Sustained pressure
Active hyperemia increase

Blood flow
Period of increased autoregulation

Organ blood flow


metabolic rate

B Steady state
Organ blood flow

Reactive hyperemia B

Autoregulatory
Steady-state Organ blood flow
range

Normal
Period of arrested blood flow

FIGURE 27–2 Organ blood flow responses caused by local


Normal
mechanisms: A) active and B) reactive hyperemia. (Modified with
permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York:
Lange Medical Books/McGraw-Hill, 2006.)

100 200
filtration and thus leads to a gradual increase in interstitial Mean arterial pressure (mm Hg)
fluid volume and pressure. Presumably the increase in extravas-
FIGURE 27–3 A and B) Autoregulation of organ blood flow.
cular pressure would cause a decrease in vessel diameter by (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology,
simple compression. This mechanism might be especially 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
important in organs such as the kidney and brain whose vol-
umes are constrained by external structures.
Although not illustrated in Figure 27–3A, autoregulatory of the vasculature. Sympathetic vasoconstrictor nerves are
mechanisms operate in the opposite direction in response to a the backbone of the system for controlling TPR and thus are
decrease in arterial pressure below the normal value. One essential participants in global cardiovascular tasks such as
important general consequence of local autoregulatory mecha- regulating arterial blood pressure.
nisms is that the steady-state blood flow in many organs tends Sympathetic vasoconstrictor nerves release norepinephrine
to remain near the normal value over quite a wide range of arte- from their terminal structures in amounts generally propor-
rial pressure. This is illustrated in the graph of Figure 27–3B. As tional to their electrical activity. Norepinephrine causes an
will be discussed later, the inherent ability of certain organs to increase in the tone of arterioles after combining with an
maintain adequate blood flow despite lower-than-normal arte- α1-adrenergic receptor on smooth muscle cells.
rial pressure is of considerable importance in situations such as Sympathetic vasoconstrictor nerves normally have a con-
hypotension (low arterial pressure) from blood loss. tinuous or tonic firing activity. Thus, arterioles have a certain
level of neurogenic tone as a normal component of their nor-
mal baseline state of contraction. When the firing rate of sym-
NEURAL INFLUENCES ON ARTERIOLES pathetic vasoconstrictor nerves is increased above normal,
arterioles constrict and cause organ blood flow to fall below
Sympathetic Vasoconstrictor Nerves normal. Conversely, vasodilation and increased organ blood
These neural fibers innervate arterioles in all systemic organs flow occurs when the normal tonic activity of sympathetic
and provide by far the most important means of reflex control vasoconstrictor nerves is reduced.
268 SECTION V Cardiovascular Physiology

Other Neural Influences neys to decrease renal excretion of water. Its role in body fluid
balance has some very important indirect influences on car-
Most blood vessels do not receive innervation from the para-
diovascular function, which will be discussed in more detail in
sympathetic division of the autonomic nervous system and
Chapter 29. Because it is such a potent vasoconstrictor agent,
systemic vascular resistance is not significantly influenced by
even the normally low levels of circulating vasopressin are
parasympathetic nerve activity. However, parasympathetic
likely to have some normal tonic effect on the basal tone of
vasodilator nerves, which release acetylcholine, are present
arterioles throughout the body. Moreover, abnormally high
in vessels of the brain and heart but their influence on arterio-
levels of vasopressin are clearly important in the intense arte-
lar tone in these organs appears to be inconsequential. Para-
riolar constriction that accompanies certain disturbances such
sympathetic vasodilator nerves are also present in the vessels
as severe blood loss through hemorrhage.
of the salivary glands, pancreas, gastric mucosa, and external
genitalia (where they are responsible for the vasodilation of
inflow vessels responsible for erection). Angiotensin II
Angiotensin II is a circulating polypeptide that regulates aldos-
terone release from the adrenal cortex as part of the system for
HORMONAL INFLUENCES controlling body sodium balance. This system, to be discussed in
ON ARTERIOLES greater detail in Chapter 29, is very important in blood volume
regulation. Angiotensin II is also a very potent vasoconstrictor
Circulating Catecholamines agent. Like vasopressin, even the normal low circulating level
of angiotensin II likely has a role in producing the normal
During activation of the sympathetic nervous system, cate-
basal tone of arterioles throughout the body. In addition, an
cholamines epinephrine and norepinephrine are released from
abnormally high blood level of angiotensin II seems to be an
the adrenal medulla into the bloodstream (see Chapter 65).
important contributing factor in certain forms of hypertension.
Under normal circumstances, the concentrations of these agents
in the blood are probably not high enough to cause significant
cardiovascular effects. However, circulating catecholamines
may have cardiovascular effects in situations (such as vigorous CONTROL OF VENOUS TONE
exercise or hemorrhagic shock) that involve increased activity of
the sympathetic nervous system. In general, the cardiovascular Recall that venules and veins are relatively large-diameter ves-
effects of greatly increased levels of circulating catecholamines sels that have little resistance to flow but do contain relatively
parallel the direct effects of sympathetic activation that have large amounts of blood. Therefore, venous tone or diameter
already been discussed; both epinephrine and norepinephrine has relatively little direct effect on the flow through organs.
can activate cardiac β1-adrenergic receptors to increase heart However, venous diameter does have a large effect on the frac-
rate and myocardial contractility and can activate vascular tion of total blood volume that is located in the periphery
α1-receptors to cause vasoconstriction. Recall that in addition versus centrally. Consequently, when one considers what
to the α1-receptors that mediate vasoconstriction, arterioles in a peripheral veins are doing, one should be thinking primarily
few organs also possess β2-adrenergic receptors that mediate about what the effects will be on central venous pressure and
vasodilation. Because vascular β2-receptors are more sensitive cardiac output.
to epinephrine than are vascular α1-receptors, moderately Veins contain vascular smooth muscle that is influenced by
increased levels of circulating epinephrine can cause vasodila- many of the same things that influence the vascular smooth
tion—whereas higher levels cause α1-receptor-mediated vaso- muscle of arterioles. Constriction of the veins (venoconstric-
constriction. Vascular β2-receptors are not innervated and tion) is largely mediated through activity of the sympathetic
therefore are not activated by norepinephrine released directly nerves that innervate them. As in arterioles, these sympathetic
from sympathetic vasoconstrictor nerves. The physiological nerves release norepinephrine, which interacts with α1-receptors
importance of these vascular β2-receptors is unclear because and produces an increase in venous tone and a decrease in ves-
adrenal epinephrine release occurs during periods of increased sel diameter. There are, however, several functionally impor-
sympathetic activity when arterioles would simultaneously be tant differences between veins and arterioles. Compared with
undergoing direct neurogenic vasoconstriction. arterioles, veins normally have little basal tone. Thus, veins are
normally in a dilated state. One important consequence of the
lack of basal venous tone is that vasodilator metabolites that
Vasopressin may accumulate in the tissue have little effect on veins.
The polypeptide hormone vasopressin (also known as antidi- Because of their thin walls, veins are much more susceptible
uretic hormone (ADH), plays an important role in extracel- to physical influences than are arterioles. The large effect of
lular fluid homeostasis and is released into the bloodstream internal venous pressure on venous diameter was discussed in
from the posterior pituitary gland in response to low blood Chapter 26 and is evident in the pooling of blood in the veins
volume and/or high extracellular fluid osmolarity (see of the lower extremities that occurs during prolonged standing
Chapter 45). Vasopressin acts on collecting ducts in the kid- (as will be discussed further in Chapter 30).
CHAPTER 27 Vascular Control 269

External compression is an important determinant of venous organs is very strongly regulated by sympathetic nerve activity.
volume. This is especially true of veins in skeletal muscle. Very Consequently, some organs are automatically forced to par-
high pressures are developed inside skeletal muscle tissue dur- ticipate in overall cardiovascular reflex responses to a greater
ing contraction and cause venous vessels to collapse. Because extent than are other organs. The overall plan seems to be that
veins and venules have one-way valves, the blood displaced in cardiovascular emergency, flow to the brain and heart will
from veins during skeletal muscle contraction is forced in the be preserved at the expense of everything else.
forward direction toward the right heart. In fact, rhythmic
skeletal muscle contractions can produce a considerable
pumping action, often called the skeletal muscle pump, which VASCULAR CONTROL IN
helps return blood to the heart during exercise.
SPECIFIC ORGANS
The details of vascular control in many specific organs are pre-
SUMMARY OF PRIMARY sented in several other sections of this book. Included below
VASCULAR CONTROL are descriptions of vascular control in some important organs
that are not covered elsewhere.
MECHANISMS
Certain general factors dominate the primary control of the VASCULAR CONTROL OF
peripheral vasculature when it is viewed from the standpoint
of overall cardiovascular system function; these influences are
CORONARY BLOOD FLOW
summarized in Figure 27–4. Basal tone, local metabolic vaso- The major right and left coronary arteries that serve the heart
dilator factors, and sympathetic vasoconstrictor nerves acting tissue are the first vessels to branch off the aorta. Thus, the driv-
through α1-receptors are the major factors controlling arterio- ing force for myocardial blood flow is the systemic arterial pres-
lar tone and therefore the blood flow through peripheral sure, just as it is for other systemic organs. Most of the blood
organs. Sympathetic vasoconstrictor nerves, internal pressure, that flows through the myocardial tissue returns to the right
and external compression are the most important influences atrium by way of a large cardiac vein called the coronary sinus.
on venous diameter and therefore on peripheral–central dis-
tribution of blood volume.
The flow in organs such as the brain, heart muscle, and skel- Local Metabolic Control
etal muscle is very strongly regulated by local metabolic con- As emphasized before, coronary blood flow is controlled pri-
trol, whereas the flow in the kidneys, skin, and splanchnic marily by local metabolic mechanisms and thus it responds
rapidly and accurately to changes in myocardial oxygen con-
sumption. In a resting individual, the myocardium extracts
Reflex influences Local influences 70–75% of the oxygen in the blood that passes through it, which
is more than any other organ does. Myocardial oxygen extrac-
tion cannot increase significantly from its resting value. Conse-
Basal tone quently, increases in myocardial oxygen consumption must be
Sympathetic accompanied by appropriate increases in coronary blood flow.
constrictor NE α
nerves
The issue of which metabolic vasodilator factors play the
Vasodilator dominant role in modulating the tone of coronary arterioles is
metabolites
unresolved. Many believe that adenosine, released from myo-
Arterioles cardial muscle cells in response to increased metabolic rate, may
be an important local coronary metabolic vasodilator influence.
Regardless of the specific details, myocardial oxygen consump-
tion is the most important influence on coronary blood flow.
Passive
distention
Sympathetic α P
constrictor NE Systolic Compression
nerves
External Large forces and/or pressures are generated within the myo-
P
compression cardial tissue during cardiac muscle contraction. Such
intramyocardial forces press on the outside of coronary vessels
Veins and cause them to collapse during systole. Because of this sys-
FIGURE 27–4 Primary influences on arterioles and veins. tolic compression and the associated collapse of coronary
NE, norepinephrine; α, alpha-adrenergic receptor; P, pressure. vessels, coronary vascular resistance is greatly increased dur-
(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, ing systole. The result, at least for much of the left ventricular
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) myocardium, is that coronary flow is lower during systole than
270 SECTION V Cardiovascular Physiology

Aortic pressure

Left ventricular pressure

Left coronary flow

0
FIGURE 27–5 Phasic flows in the left and
right coronary arteries in relation to aortic
and left ventricular pressures. (Modified with
permission from Mohrman DE, Heller LJ: Cardiovascular Right coronary flow
Physiology, 6th ed. New York: Lange Medical Books/
McGraw-Hill, 2006.) 0

during diastole, even though systemic arterial pressure (i.e., consumption by increasing heart rate and contractility. The
coronary perfusion pressure) is highest during systole. This is increased local metabolic vasodilator influence outweighs the
illustrated in the left coronary artery flow trace shown in Fig- concurrent neurogenic vasoconstrictor. Whether these coro-
ure 27–5. Systolic compression has much less effect on flow nary vasoconstrictor fibers might be functionally important in
through the right ventricular myocardium. This is because the certain pathological situations is an open question.
peak systolic intraventricular pressure is much lower for the
right heart than for the left, and the systolic compressional
forces in the right ventricular wall are correspondingly less VASCULAR CONTROL OF
than those in the left ventricular wall. SKELETAL MUSCLE BLOOD FLOW
Because the endocardial surface of the left ventricle is exposed
to intraventricular pressure (∼120 mm Hg during systole), and Because of the large mass of skeletal muscle, blood flow
the epicardial surface is exposed only to intrathoracic pressure through it is an important factor in overall cardiovascular
(∼0 mm Hg), systolic compressional forces on coronary vessels hemodynamics. Collectively, the skeletal muscles constitute
are greater in the endocardial layers of the left ventricular wall 40–45% of body weight—more than any other single body
than in the epicardial layers. Thus, the flow to the endocardial organ. Even at rest, about 15% of the cardiac output goes to
layers of the left ventricle is impeded more than the flow to epi- skeletal muscle, and during strenuous exercise skeletal muscle
cardial layers by systolic compression. Normally, the endocardial may receive more than 80% of the cardiac output. Resting
region of the myocardium can make up for the lack of flow dur- skeletal muscle has a high level of intrinsic vascular tone.
ing systole by a high flow in the diastolic interval. However, when Because of this high tone of smooth muscle in resistance ves-
coronary blood flow is limited—for example, by coronary dis- sels of resting skeletal muscles, the blood flow per gram of tis-
ease and stenosis—the endocardial layers of the left ventricle are sue is low when compared with that of other organs such as
often the first regions of the heart to have difficulty maintaining the kidneys. However, resting skeletal muscle blood flow is
a flow sufficient for their metabolic needs. Myocardial infarcts still substantially above that required to sustain its metabolic
(areas of tissue killed by lack of blood flow) occur most frequently needs. Resting skeletal muscles normally extract only 25–30%
in the endocardial layers of the left ventricle. of the oxygen delivered to them in arterial blood. Thus,
changes in the activity of sympathetic vasoconstrictor fibers
can reduce resting muscle blood flow without compromising
Neural Influences on Coronary Flow resting tissue metabolic processes.
Coronary arterioles are densely innervated with sympathetic Local metabolic control of arteriolar tone is the most impor-
vasoconstrictor fibers, yet when the activity of the sympathetic tant influence on blood flow through exercising muscle. A
nervous system increases, the coronary arterioles normally particularly important characteristic of skeletal muscle is its
vasodilate rather than vasoconstrict. This is because an very wide range of metabolic rates. During strenuous exercise,
increase in sympathetic tone increases myocardial oxygen the oxygen consumption rate of and oxygen extraction by
CHAPTER 27 Vascular Control 271

skeletal muscle tissue can reach the high values typical of the discussed in more detail when postural reflexes are considered
myocardium. In most respects, the factors that control blood in Chapter 30.)
flow to exercising muscle are similar to those that control cor-
onary blood flow. Local metabolic control of arteriolar tone is
very strong in exercising skeletal muscle, and muscle oxygen VASCULAR CONTROL OF
consumption is the most important determinant of its blood CEREBRAL BLOOD FLOW
flow. Blood flow in skeletal muscle can increase 20-fold during
a bout of strenuous exercise. Interruption of cerebral blood flow for more than a few sec-
Alterations in sympathetic neural activity can alter nonex- onds leads to unconsciousness. One rule of overall cardiovas-
ercising skeletal muscle blood flow. For example, maximum cular system function is that, in all situations, measures are
sympathetic discharge rates can decrease blood flow in a taken that are appropriate to preserve adequate blood flow to
resting muscle to less than one fourth its normal value, and, the brain. Cerebral blood flow is regulated almost entirely by
conversely, if all neurogenic tone is removed, resting skeletal local mechanisms. The brain as a whole has a nearly constant
muscle blood flow may double. This is a modest increase in rate of metabolism that, on a per gram basis, is nearly as high
flow compared with what can occur in an exercising skeletal as that of myocardial tissue. Flow through the cerebrum is
muscle. Nonetheless, because of the large mass of tissue autoregulated very strongly and is little affected by changes in
involved, changes in the vascular resistance of resting skeletal arterial pressure unless it falls below about 60 mm Hg. When
muscle brought about by changes in sympathetic activity are arterial pressure decreases below 60 mm Hg, brain blood flow
very important in the overall reflex regulation of arterial decreases proportionately. It is presently unresolved whether
pressure. metabolic mechanisms or myogenic mechanisms or both are
Alterations in sympathetic neural activity can influence involved in the phenomenon of cerebral autoregulation.
exercising skeletal muscle blood flow. As will be discussed in Local changes in cerebral blood flow may be influenced by
Chapter 72, the cardiovascular response to muscle exercise local metabolic conditions. Presumably because the overall
involves a general increase in sympathetic activity. This average metabolic rate of brain tissue shows little variation,
reduces flow to susceptible organs, which include nonexer- total brain blood flow is remarkably constant in most situa-
cising muscles. In exercising muscles, the increased sympa- tions. The cerebral activity in discrete locations within the
thetic vasoconstrictor nerve activity is not evident as outright brain, however, changes from situation to situation. As a result,
vasoconstriction but does limit the degree of metabolic vaso- blood flow to discrete regions is not constant but closely fol-
dilation. One important function that this seemingly coun- lows the local neuronal activity. The mechanisms responsible
terproductive process may serve is preventing an excessive for this strong local control of cerebral blood flow are as yet
reduction in TPR during exercise. Indeed, if arterioles in undefined, but H+, K+, oxygen, and adenosine seem most
most of the skeletal muscles in the body were allowed to likely to be involved.
dilate to their maximum capacity simultaneously, TPR would As in most organs, cerebral blood flow increases whenever
be so low that the heart could not possibly supply enough the partial pressure of carbon dioxide (Pco2) in arterial blood
cardiac output to maintain arterial pressure. increases and, conversely, cerebral blood flow decreases when-
Rhythmic contractions can increase venous return from ever Pco2 decreases below normal. This is the normal state of
exercising skeletal muscle. As in the heart, muscle contraction affairs in most tissues but it has important nonvascular conse-
produces large compressional forces within the tissue, which quences when it happens in the brain. For example, the dizzi-
can collapse vessels and impede blood flow. Strong, sustained ness, confusion, and even fainting that can occur when a
(tetanic) skeletal muscle contractions may actually stop mus- person hyperventilates (and “blows off ” CO2) are direct
cle blood flow. About 10% of the total blood volume is nor- results of cerebral vasoconstriction. It appears that cerebral
mally contained within the veins of skeletal muscle, and during arterioles respond not to changes in Pco2 but to changes in the
rhythmic exercise the skeletal muscle pump is very effective in extracellular H+ concentration (i.e., pH) caused by changes in
displacing blood from skeletal muscle veins. Valves in the Pco2. Cerebral arterioles also dilate whenever the partial pres-
veins prevent reverse flow back into the muscles. Blood dis- sure of oxygen (Po2) in arterial blood decreases significantly
placed from skeletal muscle into the central venous pool is an below normal values. However, higher-than-normal arterial
important factor in the hemodynamics of strenuous whole- blood Po2, such as that caused by pure oxygen inhalation, pro-
body exercise. duces little decrease in cerebral blood flow.
Veins in skeletal muscle can constrict in response to Sympathetic and parasympathetic neural influences on cere-
increased sympathetic activity. However, they are sparsely bral blood flow are minimal. Although cerebral vessels receive
innervated with sympathetic vasoconstrictor fibers, and the both sympathetic vasoconstrictor and parasympathetic vasodi-
rather small volume of blood that can be mobilized from skel- lator fiber innervation, cerebral blood flow is influenced very
etal muscle by sympathetic nerve activation is probably not of little by changes in the activity of either under normal circum-
much significance to total body hemodynamics. This is stances. Sympathetic vasoconstrictor responses may, however, be
in sharp contrast to the large displacement of blood from exer- important in protecting cerebral vessels from excessive passive
cising muscle by the muscle pump mechanism. (This will be distention following large, abrupt increases in arterial pressure.
272 SECTION V Cardiovascular Physiology

The blood–brain barrier refers to the tightly connected vas-


cular endothelial cells that severely restrict transcapillary move- cigarettes a day for more than 25 years, and eats a high-fat,
ment of all polar and many other substances. Because of this high-salt diet. He is overweight, pale, and sweaty and is
blood–brain barrier, the extracellular space of the brain repre- clutching his chest. His heart rate is 110 beats/min and his
sents a special fluid compartment in which the chemical com- blood pressure is 110/90 mm Hg. His medical record
position is regulated separately from that in the plasma and indicates that he has been treated with sublingual nitro-
general body extracellular fluid compartment. The extracellular glycerin for mild angina pectoris for several years and
compartment of the brain encompasses both interstitial fluid had been instructed about lifestyle changes. The angina
and cerebrospinal fluid (CSF) that surrounds the brain and has been getting more severe and required increasingly
spinal cord and fills the brain ventricles. The CSF is formed more nitroglycerin to achieve relief. This time, the nitro-
from plasma by selective secretion (not simple filtration) by spe- glycerin has not worked. An ECG indicates that he has a
cialized tissues, the choroid plexus, located within the cerebral myocardial infarction in the anterior wall of the left ven-
ventricles. These processes regulate the chemical composition tricle. He is taken immediately to the cardiac catheteriza-
of the CSF. The interstitial fluid of the brain takes on the chemi- tion laboratory and an angiogram reveals an almost
cal composition of CSF through free diffusional exchange. complete occlusion of his left anterior descending coro-
The blood–brain barrier serves to protect the cerebral cells nary artery. A stent is placed in the artery and blood flow
from ionic disturbances in the plasma because it is not very is restored to the ischemic tissue.
permeable to charged substances. Also, by exclusion and/or The condition experienced by this man occurs when-
endothelial cell metabolism, it prevents many circulating hor- ever coronary blood flow decreases below that required to
mones (and drugs) from influencing the parenchymal cells of meet the metabolic needs of the heart. The myocardium
the brain and the vascular smooth muscle cells in brain ves- becomes ischemic and pumping capability of the heart is
sels. Brain capillaries have a special carrier system for glucose impaired. The most common cause of coronary artery
and present no barrier to oxygen and carbon dioxide diffu- disease is atherosclerosis of the large coronary arteries
sion. Thus, the blood–brain barrier does not restrict nutrient and this man had several of the known risk factors (smok-
supply to the brain tissue. ing, obesity, high stress, poor diet, high blood cholesterol).
Localized lipid deposits called plaques develop within the
arterial walls and with severe disease may become large
VASCULAR INFLUENCES ON enough to permanently narrow the lumen of arteries. If
PULMONARY BLOOD FLOW the coronary artery narrowing (stenosis) is not too severe,
local metabolic vasodilator mechanisms may reduce arte-
See Chapter 34. riolar resistance sufficiently to compensate for the abnor-
mally increased coronary arterial resistance.
VASCULAR CONTROL OF RENAL Coronary artery disease can jeopardize cardiac func-
tion in several ways. (1) Ischemic muscle cells are electri-
BLOOD FLOW cally irritable and the danger of fibrillation is enhanced
See Chapter 40. (see Chapter 25) because ectopic pacemaker foci may
develop. (2) Platelet aggregation and clotting function
may be abnormal in atherosclerotic coronary arteries
VASCULAR CONTROL OF and the danger of thrombi or emboli formation is
SPLANCHNIC BLOOD FLOW enhanced (see Chapter 22). (3) Myocardial ischemia pro-
duces intense, debilitating chest pain called angina
See Chapter 49. pectoris. Anginal pain is often absent in individuals with
coronary artery disease when they are resting but is
VASCULAR CONTROL OF induced during physical exertion or emotional excite-
ment when sympathetic activity is increased and myo-
CUTANEOUS BLOOD FLOW cardial oxygen consumption is elevated.
Primary treatment of coronary artery disease includes
See Chapter 70.
lifestyle alterations and attempts to lower blood lipids by
dietary and pharmacological techniques. Treatment of
angina that is a result of coronary artery disease may first
involve quick-acting vasodilator drugs such as nitroglyc-
CLINICAL CORRELATION erin to provide relief during an anginal attack. These
“nitrate” drugs are NO donors and directly vasodilate coro-
A 58-year-old man comes to the emergency room com-
nary vessels to acutely increase coronary blood flow. In
plaining of weakness and severe chest pain. He is a sales-
addition to increasing myocardial oxygen delivery, nitrates
man in a high stress industry, has smoked two packs of
CHAPTER 27 Vascular Control 273

■ Sympathetic vasoconstrictor nerves provide the primary reflex


reduce myocardial oxygen demand by dilating systemic mechanisms for regulating both arteriolar and venous tone.
veins (reducing preload) and by decreasing arterial resis- ■ Sympathetic vasoconstrictor nerves release norepinephrine,
tance (reducing afterload). Second, β-adrenergic blocking which interacts with α1-adrenergic receptors on vascular
agents such as propranolol may be used to block the effects smooth muscle to induce vasoconstriction.
of cardiac sympathetic nerves on heart rate and contractility. ■ The relative importance of local metabolic versus reflex
These agents limit myocardial oxygen consumption and sympathetic control of arteriolar tone (and therefore blood
prevent it from increasing above the level that the compro- flow) varies from organ to organ.
mised coronary blood flow can sustain. Third, calcium ■ In many organs (such as brain, heart muscle, and exercising
channel blockers such as verapamil may be used to dilate skeletal muscle), blood flow normally closely follows metabolic
coronary and systemic blood vessels, and to lower blood rate because of local metabolic influences on arterioles.
pressure and heart rate. These drugs, which block entry of ■ In other organs (such as skin and kidneys), blood flow is
calcium into the vascular smooth muscle cell, interfere normally regulated more by sympathetic nerves than by
with normal excitation–contraction coupling. local metabolic conditions.
Invasive or surgical interventions may be used to treat
coronary artery stenosis. Fluoroscopic techniques com-
bined with radio-opaque contrast injections can be used STUDY QUESTIONS
to visualize the coronary arteries. A balloon-tipped cath- 1. Vascular smooth muscle differs from cardiac muscle in that it
eter can be threaded into the occluded region of the coro- A) contains no actin molecules.
nary artery and rapidly inflated to squeeze the plaque B) can be directly activated in the absence of action potentials.
against the vessel wall and improve the patency of the ves- C) is unresponsive to changes in intracellular calcium levels.
sel. This technique, called coronary angioplasty, may also D) is unresponsive to changes in membrane potentials.
be effective in opening occlusions produced by intravas- E) is unresponsive to changes in muscle length.
cular clots associated with acute myocardial infarction. A 2. Arteriolar constriction tends to do which of the following?
small, mesh tubular device called a stent is often implanted A) decrease total peripheral resistance
inside the vessel at the angioplasty site. This rigid implant B) decrease mean arterial pressure
has been shown to improve continued patency of the ves- C) decrease capillary hydrostatic pressure
sel over a longer period than angioplasty alone. If angio- D) increase transcapillary fluid filtration
plasty and stent placement is inappropriate or unsuccessful, E) increase blood flow through the capillary bed
coronary bypass surgery may be performed. The stenotic 3. When an organ responds to an increase in metabolic activity
coronary artery segments are bypassed by implanting with a decrease in arteriolar resistance, this is known as
parallel low-resistance pathways formed from either nat- A) active hyperemia.
ural (e.g., saphenous vein or mammary artery) or artifi- B) reactive hyperemia.
cial vessels. C) autoregulation of blood flow.
D) flow-dependent vasodilation.
E) metabolic vasoconstriction.
4. A particular vascular bed demonstrates the phenomenon
of autoregulation of blood flow. This means that
CHAPTER SUMMARY A) when flow increases, capillary pressure increases.
■ Continuous adjustments of vascular diameter are required to B) when metabolic activity increases, flow increases.
properly distribute the cardiac output to the various systemic C) when arterial pressure increases, arteriolar resistance
tissues (the role of arterioles) and maintain adequate cardiac increases.
filling (the role of veins). D) when blood flow is interrupted, arteriolar resistance
■ Vascular adjustments are made by changes in the tone of decreases.
vascular smooth muscle. E) when arterial pressure falls, sympathetic vasoconstriction
■ Vascular smooth muscle has many properties that make it occurs.
sensitive to a wide array of local and reflex stimuli and capable 5. Which of the following is most likely to increase coronary
of maintaining tone for long periods of time. blood flow?
■ The tone of arterioles, but not veins, can be strongly influenced A) decreased arterial pressure
by local vasodilator factors produced by local tissue metabo- B) decreased heart rate
lism. C) increased sympathetic neural activity
■ In abnormal situations (such as tissue injury or severe blood D) reduced left ventricular end-diastolic volume
volume depletion), certain local factors such as histamine and E) reduced left ventricular ejection fraction
bradykinin, and hormonal factors such as vasopressin and
angiotensin have significant vascular influences.
This page intentionally left blank
28
C H A P T E R

Venous Return and


Cardiac Output
David E. Mohrman and Lois Jane Heller

O B J E C T I V E S

■ Describe the overall arrangement of the systemic circulation


and identify the primary functional properties of each of its major
components.
■ Define mean circulatory filling pressure and state the primary factors
that determine it.
■ Define venous return and explain how it is distinguished from cardiac
output.
■ State why cardiac output and venous return must be equal in the
steady state.
■ List the factors that control venous return.
■ Describe the relationship between central venous pressure and venous
return and draw the normal venous return curve.
■ Define peripheral venous pressure.
■ List the factors that determine peripheral venous pressure.
■ Predict the shifts in the venous return curve that occur with altered blood
volume and altered venous tone.
■ Describe how the output of the left heart pump is matched to that of the
right heart pump.
■ Draw the normal venous return and cardiac output curves on a graph and
describe the significance of the point of curve intersection.
■ Predict how normal venous return, cardiac output, and central venous
pressure will be altered with any given combination of changes in cardiac
sympathetic tone, peripheral venous sympathetic tone, or circulating
blood volume.
■ Identify conditions that may result in abnormally high or low central venous
pressure.

venous side of this system is often conceptually separated into


INTERACTION OF two different compartments: (1) a large and diverse peripheral
SYSTEM COMPONENTS section (the peripheral venous compartment) and (2) a
smaller intrathoracic section that includes the venae cavae and
As illustrated in Figure 28–1, the systemic cardiovascular sys- the right atrium (the central venous compartment). Each of
tem is a closed hydraulic circuit that includes the heart, arter- the segments of this circuit has a distinctly different role to
ies, arterioles, capillaries, and veins. (Note: The pulmonary play in the overall operation of the system because of inherent
circuit and lymphatics are not included because they do not differences in anatomical volume, resistance to blood flow,
influence the major points to be made in this chapter.) The and compliance that are summarized in Table 28–1.

275

Ch28_275-284.indd 275 11/26/10 11:51:25 AM


276 SECTION V Cardiovascular Physiology

Th Arteries
ora
x

Arterioles

Ventricle

Atrium Capillaries
Central venous compartment

FIGURE 28–1 Major functionally distinct


Peripheral venous compartment
components of the systemic cardiovascular
circuit. (Modified with permission from Mohrman DE, Heller
LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical
Books/McGraw-Hill, 2006.)

Note especially the surprisingly high ventricular diastolic any internal pressure. In a 70-kg adult, this amount is 3.5 L, as
compliance of 24 mL/mm Hg in Table 28–1. This value indi- indicated by the total systemic circuit volume (V0) in Table
cates how sensitive the ventricular end-diastolic volume (and 28–1. Normally, however, the systemic circuit contains about
therefore stroke volume and cardiac output) is to small changes 4.5 L of blood and thus is somewhat inflated. From the total
in cardiac filling pressure (i.e., central venous pressure). systemic circuit compliance (C) given in Table 28–1, one can
Cardiac filling pressure is a crucial factor that determines how see that an extra 1,000 mL of blood would result in an internal
well the cardiovascular system functions. pressure of about 7 mm Hg (i.e., 1,000 mL/140 mL/mm Hg).
This theoretical pressure is called the mean circulatory filling
pressure and is the pressure that would exist throughout the
MEAN CIRCULATORY system in the absence of flow.
FILLING PRESSURE The two major factors that affect mean circulatory filling
pressure are the circulating blood volume and the state of
Imagine the heart arrested in diastole with no flow around the the peripheral venous vessel tone. In the latter case, look at
circuit shown in Figure 28–1. It will take a certain amount of Figure 28–1 and imagine how constriction of the vessels of the
blood just to fill the anatomical space contained by the sys- large venous compartment (increasing venous tone) will sig-
temic system without stretching any of its walls or developing nificantly increase pressure throughout the system. In con-

TABLE 28–1 Typical properties of the major components of the systemic cardiovascular circuit.a
Compartment V0 (mL) C (mL/mm Hg) R (mm Hg/(L/min))

Ventricle in diastole 30 24 0

Arteries 600 2 1

Arterioles 100 0 13

Capillaries 250 0 5

Peripheral venous compartment 2,500 110 1

Central venous compartment 80 4 0

Entire circuit 3,560 140 20


a
Values are for a normal, young, resting 70-kg adult. V0, anatomical volume of compartment at zero pressure: C, compliance of compartment; R, resistance to flow through
compartment.
Reproduced with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.
CHAPTER 28 Venous Return and Cardiac Output 277

Venous return Central venous compartment Cardiac output

FIGURE 28–2 Distinction between cardiac


output and venous return. (Modified with
permission from Mohrman DE, Heller LJ: Cardiovascular
Physiology, 6th ed. New York: Lange Medical Books/
Great veins in thorax and right atrium McGraw-Hill, 2006.)

trast, squeezing on arterioles (increasing arteriolar tone) will clinically useful information about the state of the circulatory
have a negligible effect on mean circulatory filling pressure system will be discussed.
because arterioles contain so little blood in any state. The other The central venous compartment corresponds roughly to
major components of the system (arteries and capillaries) do the volume enclosed by the right atrium and the great veins
not actively change their contained volume. in the thorax. Blood leaves the central venous compartment
by entering the right ventricle at a rate that is equal to the
FLOW-INDUCED DISTRIBUTION OF cardiac output. Venous return, in contrast, is the rate at
which blood returns to the thorax from the peripheral
BLOOD VOLUME AND PRESSURE vascular beds and thus is the rate at which blood enters the
central venous compartment. The important distinction
The presence of flow around the circuit does not change the
between venous return to the central venous compartment
total volume of blood in the system or the mean circulatory
and cardiac output from the central venous compartment is
filling pressure. The flow caused by cardiac pumping action
illustrated in Figure 28–2.
does, however, tend to shift some of the blood volume from
In any stable situation, venous return must equal cardiac
the venous side of the circuit to the arterial side. This causes
output or blood would gradually accumulate in either the cen-
pressures on the arterial side to increase above the mean cir-
tral venous compartment or the peripheral vasculature. How-
culatory pressure while pressures on the venous side
ever, there often are temporary differences between cardiac
decrease below it. Because veins are about 50 times more
output and venous return. Whenever such differences exist,
compliant than arteries (Table 28–1), the flow-induced
the volume of the central venous compartment must be chang-
decrease in venous pressure is only about 1/50th as large as
ing. Because the central venous compartment is enclosed by
the accompanying increase in arterial pressure. Thus,
elastic tissues, any change in central venous volume produces
flow or no flow, pressure in the peripheral venous compart-
a change in central venous pressure.
ment is normally quite close to the mean circulatory filling
As discussed in Chapter 24, the central venous pressure
pressure.
(i.e., cardiac filling pressure) has an extremely important posi-
tive influence on cardiac output (Starling’s law of the heart).
As explained below, central venous pressure has an equally
CENTRAL VENOUS PRESSURE: important negative effect on venous return. Thus, central
AN INDICATOR OF venous pressure is always automatically driven to a value that
makes cardiac output equal to venous return.
CIRCULATORY STATUS
The cardiovascular system must continuously adjust to meet
changing metabolic demands of the body. Because the cardio- INFLUENCE OF CENTRAL VENOUS
vascular system is a closed hydraulic loop, adjustments in any PRESSURE ON VENOUS RETURN
one part of the circuit will have pressure, flow, and volume
effects throughout the circuit. Because of the critical influence The important factors involved in the process of venous
of cardiac filling on cardiovascular function, the remainder of return can be summarized as shown in Figure 28–3A. Ana-
this chapter will focus on the factors that determine the pres- tomically the peripheral venous compartment is scattered
sure in the central venous compartment. In addition, the way throughout the systemic organs, but functionally it can be
in which measures of central venous pressure can provide viewed as a single vascular space that has a particular pressure
278 SECTION V Cardiovascular Physiology

A
Thorax

Intrathoracic pressure ≅
0 mm Hg
From
P PV ≅ P CV
capillaries Venous return Cardiac output
7 mm Hg

Peripheral venous compartment


Central venous
Venous resistance compartment

8
Venous return (L/min)

6
Ve
nou
s
fu
4 nc
tio
n
cu
rv
e
2

0 2 4 6 8 10
Central venous pressure (mm Hg)
FIGURE 28–3 Venous Return A) Factors influencing venous return. B) The venous function curve. (Modified with permission from Mohrman DE,
Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)

(PPV) at any instant. The normal operating pressure in the pressure decreases. There are two minor additional points to
peripheral venous compartment is usually very close to mean be made about this venous function curve. First, changes in
circulatory filling pressure. Moreover, the same factors that venous resistance can influence the slope of the venous func-
influence mean circulatory filling pressure have essentially tion curve but, in the example given, venous return will be
equal influences on peripheral venous pressure. Thus, 0 L/min when PCV = 7 mm Hg at any level of venous vascular
“peripheral venous pressure” can be viewed as essentially resistance. Second, if central venous pressure reaches very low
equivalent to “mean circulatory filling pressure.” The blood values and decreases below the intrathoracic pressure, the
flow between the peripheral venous compartment and veins in the thorax collapse and tend to limit venous return. In
the central venous compartment is governed by the basic flow the example of Figure 28–3, intrathoracic pressure is taken
equation Q̇ = Δ P/R, where ΔP is the pressure decrease between to be 0 mm Hg and the flat portion of the venous function
the peripheral and central venous compartments and R the curve indicates that lowering central venous pressure below
small resistance associated with the peripheral veins. In the 0 mm Hg produces no additional increase in venous return.
example in Figure 28–3, peripheral venous pressure is Just as a cardiac function curve shows how central venous
assumed to be 7 mm Hg. Thus, there will be no venous return pressure influences cardiac output, a venous function curve
when the central venous pressure (PCV) is also 7 mm Hg as shows how central venous pressure influences venous return.
shown in Figure 28–3B. (By convention, these relationships are plotted with the inde-
If the peripheral venous pressure remains at 7 mm Hg, pendent variable on the horizontal axis and the dependent vari-
decreasing central venous pressure will increase the pressure able on the vertical axis and they must be read in that sense.
difference across the venous pathway and consequently cause For example, Figure 28–3B says that increasing central venous
an increase in venous return to the central venous pool. This pressure tends to cause decreased venous return. Figure 28–3B
relationship is summarized by the venous function curve, does not imply that increasing venous return will tend to lower
which shows how venous return increases as central venous central venous pressure.)
CHAPTER 28 Venous Return and Cardiac Output 279

INFLUENCE OF PERIPHERAL
VENOUS PRESSURE ON VENOUS 10

RETURN In

Venous return (L/min)


8 cr
ea
As can be deduced from Figure 28–3A, it is the pressure dif- se
d
ference between the peripheral and central venous compart- bl
ood
6 Co
ments that determines venous return. Therefore, an increase nt vo
ro lum
in peripheral venous pressure can be just as effective in lv e
De en or
increasing venous return as a decrease in central venous 4 or cr ou ve
ve ea s no
pressure. no se f un us
us d b ct to
The two ways in which peripheral venous pressure can l
to oo
i o n ne
change were discussed in Chapter 26. First, because veins are 2 ne d cu
vo rv
lu e
elastic vessels, changes in the volume of blood contained within m
e
the peripheral veins alter the peripheral venous pressure.
0 2 4 6 8 10
Moreover, because the veins are much more compliant than
Central venous pressure (mm Hg)
any other vascular segment, changes in circulating blood vol-
ume produce larger changes in the volume of blood in the FIGURE 28–4 Effect of changes in blood volume and venous
veins than in any other vascular segment. For example, blood tone on venous function curves. (Modified with permission from Mohrman
DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/
loss by hemorrhage or loss of body fluids through severe
McGraw-Hill, 2006.)
sweating, vomiting, or diarrhea will decrease circulating blood
volume and significantly reduce the volume of blood con-
tained in the veins and thus decrease the peripheral venous
pressure. Conversely, transfusion, fluid retention by the kid-
ney, or transcapillary fluid reabsorption will increase circulat-
CENTRAL VENOUS PRESSURE
ing blood volume and venous blood volume. Whenever DETERMINES BOTH CARDIAC
circulating blood volume increases, so does peripheral venous
pressure. OUTPUT AND VENOUS RETURN
Recall from Chapter 27 that the second way that peripheral The significance of the fact that central venous pressure simul-
venous pressure can be altered is through changes in venous taneously affects both cardiac output and venous return can be
tone produced by increasing or decreasing the activity of sym- best seen by plotting the cardiac function curve (the Starling
pathetic vasoconstrictor nerves supplying the venous smooth curve) and the venous function curve on the same graph, as in
muscle. Peripheral venous pressure increases whenever the Figure 28–5.
activity of sympathetic vasoconstrictor fibers to veins increases.
In addition, an increase in any force compressing veins from
the outside has the same effect on the pressure inside veins as
an increase in venous tone. Thus, such things as muscle exer-
cise and wearing elastic stockings tend to increase peripheral
Cardiac output or Venous return (L/min)

venous pressure.
Whenever peripheral venous pressure is altered, the rela- 10
tionship between central venous pressure and venous return is
also altered. For example, whenever peripheral venous pres- 8
sure is increased by increases in blood volume or by sympa-
thetic stimulation, the venous function curve shifts upward Cardiac function curve
6
and to the right (Figure 28–4). This phenomenon can be most
easily understood by focusing first on the central venous pres-
sure at which there will be no venous return. When peripheral 4
venous pressure is 7 mm Hg, venous return is 0 L/min when Venous function curve
central venous pressure is 7 mm Hg. When peripheral venous 2
pressure is increased to 10 mm Hg, considerable venous return
occurs with a central venous pressure of 7 mm Hg, and venous
return stops only when central venous pressure is increased to 0 2 4 6 8 10
10 mm Hg. Thus, increasing peripheral venous pressure shifts Central venous pressure (mm Hg)
the whole venous function curve to the right. By similar logic, FIGURE 28–5 Interaction of cardiac output and venous
decreased peripheral venous pressure caused by blood loss or return through central venous pressure. (Modified with permission from
decreased sympathetic vasoconstriction of peripheral veins Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical
shifts the venous function curve to the left. Books/McGraw-Hill, 2006.)
280 SECTION V Cardiovascular Physiology

Central venous pressure, as defined earlier, is the filling


pressure of the right heart. Strictly speaking, this pressure

Cardiac output or Venous return (L /min)


directly affects only the stroke volume and output of the right
10
heart pump. In most contexts, however, “cardiac output”
implies the output of the left heart pump. How is it then, as has Normal cardiac function curve
often been previously implied, that central venous pressure 8
(the filling pressure of the right heart) profoundly affects car-
diac output (the output of the left heart)? The short answer is
6
that in the steady state, the right and left hearts have equal
outputs. (Since the right and left hearts always beat with iden-
tical rates, this implies that their stroke volumes must be equal 4 N
or
in the steady state.) The detailed answer is that changes in cen- Fu m
al
nc ve
tral venous pressure automatically cause essentially parallel tio n
2 n ou
changes in the filling pressure of the left heart (i.e., in left atrial cu s
rv
pressure). Consider, for example, the following sequence of e
consequences that a small step-wise increase in central venous
0 2 4 6 8 10
pressure has on a heart that previously was in a steady state:
Central venous pressure (mm Hg)
(1) Increased central venous pressure → (2) increased right
ventricular stroke volume via Starling’s law → (3) increased FIGURE 28–6 Families of cardiac function and venous
output of right heart → (4) right heart output temporarily function curves. Intersection points indicate equilibrium values
exceeds that of the left heart → (5) as long as this imbalance for cardiac output, venous return, and central venous pressure.
(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular
exists, blood accumulates in the pulmonary vasculature and
Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
raises pulmonary venous and left atrial pressure → (6)
increased left atrial pressure increases left ventricular stroke
volume via Starling’s law → (7) very quickly, a new steady state
will be reached when left atrial pressure has risen sufficiently Recall from Chapter 24 that cardiac output is affected by
to make left ventricular stroke volume exactly equal to the more than just cardiac filling pressure and that at any moment,
increased right ventricular stroke volume. the heart may be operating on any one of a number of cardiac
The major conclusion here is that left atrial pressure will function curves, depending on the existing level of cardiac
change in the correct direction to match left ventricular stroke sympathetic tone (see Figure 24–8). The family of possible
volume to the current right ventricular stroke volume. Conse- cardiac function curves may be plotted along with the family
quently, it is usually an acceptable simplification to say that of possible venous function curves, as shown in Figure 28–6.
central venous pressure affects cardiac output as if the heart At a particular moment, the existing influences on the heart
consisted only of a single pump as is shown in Figure 28–1. dictate the cardiac function curve on which it is operating, and
Note that in Figure 28–5, cardiac output and venous return similarly, the existing influences on peripheral venous pres-
are equal (at 5 L/min) only when the central venous pressure is sure dictate the venous function curve that applies. Thus, the
2 mm Hg. If central venous pressure were to decrease influences on the heart and on the peripheral vasculature
to 0 mm Hg, cardiac output would decrease (to 2 L/min) and determine where the cardiac and venous function curves
venous return would increase (to 7 L/min). With a venous intersect and thus what the central venous pressure and car-
return of 7 L/min and a cardiac output of 2 L/min, the volume diac output (and venous return) are in the steady state. In the
of the central venous compartment would necessarily be intact cardiovascular system, cardiac output can rise only
increasing and this would produce a progressively increasing when the point of intersection of the cardiac and venous func-
central venous pressure. In this manner, central venous pres- tion curves is raised. All changes in cardiac output are caused
sure would return to the original level (2 mm Hg) in a very by a shift in the cardiac function curve, a shift in the venous
short time. Moreover, if central venous pressure were to function curve, or both.
increase from 2 to 4 mm Hg, venous return would decrease (to The cardiac function and venous function curves are use-
3 L/min) and cardiac output would increase (to 7 L/min). This ful for understanding the complex interactions that occur in
would quickly reduce the volume of blood in the central the intact cardiovascular system. With the help of Figure
venous pool, and the central venous pressure would soon fall 28–7, let us consider, for example, what happens to the car-
back to the original level. The cardiovascular system automat- diovascular system when there is a significant loss of blood
ically adjusts to operate at the point where the cardiac and (hemorrhage). Assume that before the hemorrhage, sympa-
venous function curves intersect. thetic activity to the heart and peripheral vessels is normal,
Central venous pressure is always inherently driven to the as is the blood volume. Therefore, cardiac output is related to
value that makes cardiac output and venous return equal. central venous pressure as indicated by the “normal” cardiac
Cardiac output and venous return always stabilize at the level function curve in Figure 28–7. In addition, venous return is
where the cardiac function and venous function curves intersect. determined by central venous pressure as indicated by the
CHAPTER 28 Venous Return and Cardiac Output 281

the same venous function curve. Cardiac sympathetic nerves


do not affect the venous function curve. Venous return is
higher at point C than at point B, but the venous function
Cardiac output or Venous return (L /min)

Increased cardiac
10
sympathetic nerve activity curve has not shifted.
An additional compensatory mechanism evoked by blood
8 loss is increased activity of the sympathetic nerves leading
to veins. Recall that this increases peripheral venous pres-
Normal cardiac function curve sure and causes a rightward shift of the venous function
6 curve. Therefore, increased sympathetic activity to veins
D
A tends to shift the venous function curve, originally decreased
Venous constriction by blood loss, back toward normal. As a consequence of the
4 after hemorrhage
C increased peripheral venous tone and the shift to a more
B normal venous function curve, the cardiovascular operation
Normal venous
2
function curve shifts from point C to point D in Figure 28–7. Thus, periph-
eral venous constriction raises cardiac output by increasing
Hemorrhage
central venous pressure and moving upward along a fixed
0 2 4 6 8 10 cardiac function curve. It must be pointed out that separat-
Central venous pressure (mm Hg) ing the response to hemorrhage into distinct, progressive
FIGURE 28–7 Cardiovascular adjustments to hemorrhage.
steps (i.e., A to B to C to D) is only a conceptualization for
(Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology,
appreciating the individual effects of the different processes
6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) involved. In reality, the reflex venous and cardiac responses
occur simultaneously and so quickly that they will keep up
with the blood loss as it occurs. Thus, the actual response to
hemorrhage would follow nearly a straight line from point
“normal” venous function curve shown. The normal cardiac A to point D.
and venous function curves intersect at point A, so cardiac In summary, point D illustrates that normal cardiac output
output is 5 L/min and central venous pressure is 2 mm Hg in can be sustained in the face of blood loss by the combined
the normal state. When blood volume decreases due to hem- effect of peripheral and cardiac adjustments. Hemorrhage
orrhage, the peripheral venous pressure decreases and the is only one of the numerous potential disturbances to the car-
venous function curve is shifted to the left. In the absence of diovascular system. Plots such as those shown in Figure 28–7
any reflex responses, the cardiovascular system must switch are very useful for understanding the many disturbances to
its operation to point B because this is now the point at which the cardiovascular system and the ways by which they may
the cardiac function curve and the new venous function be compensated.
curve intersect. This automatically occurs because at the
moment of blood loss, the venous function curve is shifted
to the left and venous return decreases below cardiac output CLINICAL IMPLICATIONS OF
at the central venous pressure of 2 mm Hg. This is what leads
to the decrease in the central venous compartment’s volume ABNORMAL CENTRAL VENOUS
and pressure that causes the shift in operation from point A PRESSURES
to point B. By comparing points A and B in Figure 28–7,
note that blood loss itself decreases cardiac output and cen- Although there is no way to actually determine the position of
tral venous pressure by shifting the venous function curve. either cardiac function or venous function curves, important
In going from point A to point B, cardiac output decreases information about a patient’s circulatory status can be obtained
solely because of decreased filling pressure and Starling’s law from measures of central venous pressure. From what has been
of the heart. presented in this chapter, it is possible to conclude that a
Subnormal cardiac output evokes a number of compensa- patient with abnormally high central venous pressure must have
tory mechanisms to bring cardiac output back to more normal a depressed cardiac function curve, a right-shifted venous
levels. One of these is an increase in the activity of cardiac function curve, or both. Very high central venous pressures
sympathetic nerves that, as discussed in Chapter 24, causes a are a hallmark of patients with congestive heart failure because
shift to a cardiac function curve that is higher than normal. they have the combination of dysfunctional heart muscle
The effect of increasing cardiac sympathetic activity is illus- (depressed cardiac function curve) and excessive fluid volume
trated by a shift in cardiovascular operation from point B to (right-shifted venous function curve). Abnormally low central
point C. In itself, the increased cardiac sympathetic nerve venous pressures, on the other hand, could theoretically be
activity increases cardiac output (from 3 to 4 L/min) but causes caused by either an increased cardiac function curve or a left-
a further decrease in central venous pressure. This decrease in shifted venous function curve. The clinical reality is that
central venous pressure occurs because points B and C lie on abnormally low central venous pressures are invariably the
282 SECTION V Cardiovascular Physiology

result of a left shift of the venous function curve caused by


either low blood volume or lack of venous tone. pleura and parietal pleura. An echocardiogram revealed
Rough estimates of a patient’s central venous pressure can dilated cardiac chambers, thickened left ventricular wall,
be obtained by observing the external jugular veins. Because and a left ventricular ejection fraction of 0.35 (normal
gravity keeps veins in the head and neck collapsed when a nor- value >0.55) (see Chapter 25).
mal individual is upright, there is no distention (or retrograde She was diagnosed with chronic congestive heart failure
pulsations from atrial contractions) observed in these neck secondary to chronic hypertension. She was treated with
veins. Conversely, when fully recumbent, neck veins are full a diuretic to increase her urine output and relieve her
and pulsations are detected. If a normal individual is placed in symptoms of congestion and an angiotensin converting
a semi-recumbent position so that external jugular veins are enzyme (ACE) inhibitor to reduce her high blood pres-
positioned at 7 cm above the right atrium, the point between sure. Once her condition stabilizes, she will be treated
the collapsed venous segment and the filled segment can often with beta-adrenergic receptor blockers at a low dose to
be visualized. Abnormally high central venous pressure is decrease sympathetic stimulation of the heart.
associated with neck vein distention at a higher level (perhaps Chronic heart failure (CHF) exists whenever ventric-
even when the patient is upright). ular function is depressed through conditions that
Because of its diagnostic value in critical care situations, directly impair the mechanical performance of heart
central venous pressure is often monitored continuously via muscle such as (1) progressive coronary artery disease,
a catheter inserted in a peripheral vein and advanced cen- (2) sustained increase in cardiac afterload as that which
trally until its tip is in the central venous compartment (i.e., accompanies arterial hypertension or aortic valve steno-
near or in the right atrium). In some situations, it is desirable sis, (3) reduced functional muscle mass following myo-
to assess left atrial pressure, which is the filling pressure for cardial infarction, or (4) primary cardiomyopathy.
the left side of the heart. This is commonly done with a spe- Regardless of the precipitating cause, most forms of
cialized, flow-directed venous catheter with a small inflat- heart failure are associated ultimately with a reduced
able balloon at its tip to drag it with the blood flow through myocyte function. Systolic heart failure is associated
the right ventricle and pulmonic valve into the pulmonary with a left ventricular ejection fraction of less than 0.40.
artery. The balloon is then deflated and the cannula is This also implies that the heart is operating on a lower-
advanced further until it wedges into a terminal branch of than-normal cardiac function curve, that is, a reduced
the pulmonary vasculature. The pulmonary wedge pressure cardiac output at any given filling pressure. An example
recorded at this junction provides a useful estimate of left of the progression of events leading to CHF is well illus-
atrial pressure because there are no valves between the left trated by the cardiac output and venous function curves
atrium and the catheter tip. shown in Figure 28–8. Initially, normal cardiac output
and normal venous function curves will intersect at
point A with cardiac output of 5 L/min at a central
venous pressure of less than 2 mm Hg. With an abrupt
heart failure as might accompany a myocardial infarc-
CLINICAL CORRELATION tion and severe damage to the ventricular muscle, the
heart’s operation will shift to a much lower-than-normal
A 75-year-old woman visits her doctor complained of
cardiac output curve and the “equilibrium” will shift
increasing weakness and fatigue, shortness of breath on
from the normal point A to point B—that is, cardiac out-
minimal exertion, and a recent increase in body weight.
put decreases below normal while central venous pres-
She often has to get up at night to urinate and has noticed
sure increases above normal. The decreased cardiac
that her feet and ankles seem to be swollen. She reports
output leads to decreased arterial pressure and reflex
several episodes of waking up at night feeling like she could
activation of the cardiovascular sympathetic nerves.
not catch her breath until she got out of bed and stood at
Increased sympathetic nerve activity tends to (1) raise
her open window. She has been physically well until this
the cardiac function curve toward normal and (2)
condition developed over the last few months. She does
increase peripheral venous pressure through venous
not smoke or drink and takes no medicines except for an
constriction and thus raises the venous function curve
occasional aspirin or antacid. She is 5′3″ (160 cm), 146 lb
above normal. The heart’s operation will now shift from
(66 kg) with heart rate of 88 beats/min and blood pressure
point B to point C. Thus, the depressed cardiac output is
165/95 mm Hg (normal values 120–140/80–90 mm Hg).
substantially improved by the immediate consequences
Auscultation of the chest with a stethoscope indicates nor-
of increased sympathetic nerve activity. Note, however,
mal heart sounds but abnormal breath sounds with fine
that the cardiac output at point C is still below normal.
crackles heard over both lung bases late in expiration. An
The arterial pressure associated with cardiovascular
ECG indicates mild left ventricular hypertrophy and her
operation at point C is likely to be near normal, however,
chest x-ray shows an enlarged cardiac silhouette and pleu-
because higher-than-normal total peripheral resistance
ral effusion, an accumulation of fluid between the visceral
CHAPTER 28 Venous Return and Cardiac Output 283

↑↑ Fluid retention
Normal heart

12
Normal sympathetic activity

Cardiac output or Venous return (L/min)


↑ Fluid retention
10

↑ Venous tone ↑↑ Sympathetic activity


8
Failing
↑ Sympathetic
heart
Normal activity
6
Normal
sympathetic
A C activity
D E
4

2 B

0 2 4 6 8 10
Central venous pressure (mm Hg)
FIGURE 28–8 Cardiovascular alterations with compensated chronic systolic heart failure. (Modified with permission from Mohrman DE, Heller
LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)

will also accompany higher-than-normal sympathetic after reaching the compensated state even though sympa-
nerve activity. thetic activity may have returned to near-normal levels.
In the long term, cardiovascular operation cannot (Net fluid loss from this new steady state with expanded
remain at point C in Figure 28–8. Operation at point body fluid volume requires a period of less-than-normal
C involves higher-than-normal sympathetic activity, and sympathetic activity.)
this will inevitably cause a gradual increase in blood vol- Unfortunately, the consequences of fluid retention in
ume by mechanisms that will be described in Chapter 29. cardiac failure are not all beneficial. Note in Figure 28–8
Over several days, there is a progressive rise in the venous that fluid retention (C → D → E) will cause both periph-
function curve as a result of increased blood volume and, eral and central venous pressures to be much higher than
consequently, increased mean circulatory filling pressure. their normal values. Chronically high central venous
This will progressively shift the cardiovascular operating pressure causes chronically increased end-diastolic vol-
point from C to D to E. ume (cardiac dilation). Up to a point, cardiac performance
Note that increased fluid retention (C → D → E in is improved by increased cardiac filling volume through
Figure 28–8) causes a progressive increase in cardiac out- Starling’s law. Excessive cardiac dilation, however, can
put toward normal and simultaneously allows a reduction impair cardiac function because according to the law of
in sympathetic nerve activity toward the normal value. Laplace, increased total wall tension is required to gener-
Reduced sympathetic activity is beneficial for several rea- ate pressure within an enlarged ventricular chamber.
sons. First, decreased arteriolar constriction permits renal The high venous pressure associated with fluid retention
and splanchnic blood flow to return toward more normal also adversely affects organ function because high venous
values. Second, myocardial oxygen consumption may fall pressure produces transcapillary fluid filtration, edema for-
as sympathetic nerve activity falls, even though cardiac mation, and congestion (hence the commonly used term
output tends to increase. Recall that increased heart rate congestive heart failure). Left heart failure may lead to
and increased cardiac contractility greatly increase myo- pulmonary edema with dyspnea (shortness of breath) and
cardial oxygen consumption. Reduced myocardial oxygen respiratory crisis. Patients often complain of difficulty
consumption is especially beneficial in situations where breathing especially during the night (paroxysmal noctur-
inadequate coronary blood flow is the cause of the heart nal dyspnea). Being recumbent promotes a fluid shift from
failure. In any case, once a normal cardiac output has been the extremities into the central venous pool and lungs,
achieved, the individual is said to be in a “compensated” making the horizontal patient’s pulmonary problems worse.
state. The extracellular fluid volume remains expanded Such patients often sleep more comfortably when propped
284 SECTION V Cardiovascular Physiology

up. Right heart failure results in distended neck veins,


STUDY QUESTIONS
ankle edema, and fluid accumulation in the abdomen 1. In a severely dehydrated patient, you would expect to find
(ascites) with liver congestion and dysfunction. Plasma A) a depressed cardiac function curve.
volume expansion combines with abnormal liver function B) an increased mean circulatory filling pressure.
to reduce the concentration of plasma proteins by as much C) an increased central venous pressure.
as 30%. This reduction in plasma oncotic pressure further D) distended jugular veins.
contributes to the development of interstitial edema of con- E) decreased cardiac output.
gestive heart failure. 2. If you gave a blood transfusion to a patient who had recently
In the example shown in Figure 28–8, the depression in experienced a severe hemorrhage, you would expect
the cardiac output curve because of heart failure is only A) to expand arterial volume.
moderately severe. Thus, it is possible, through moderate B) to expand venous volume.
C) to decrease central venous pressure.
fluid retention, to achieve a new steady state with a nor-
D) to decrease the mean circulatory filling pressure.
mal cardiac output and essentially normal sympathetic E) to reduce cardiac output.
activity (point E). The situation at point E is relatively
3. Which of the following would directly (by themselves in the
stable because the stimuli for further fluid retention have absence of any compensatory responses) tend to decrease
been removed. If, however, the heart failure is more severe, central venous (cardiac filling) pressure?
the cardiac output curve may be so depressed that normal A) increased sympathetic nerve activity to only the heart
cardiac output cannot be achieved by any amount of fluid B) increased parasympathetic nerve activity to only the heart
retention. In these cases fluid retention is extremely C) increased blood volume
marked, as is the elevation in venous pressure, and the D) decreased total peripheral resistance
complications of congestion are very serious problems. E) immersion in water up to the waist
4. Which of the following will decrease the mean circulatory
filling pressure?
A) increased circulating blood volume
CHAPTER SUMMARY B) increased cardiac output
■ Mean circulatory filling pressure is a theoretical measure C) decreased arteriolar tone
of pressure in the systemic circuit when flow is stopped and D) decreased venous tone
is influenced primarily by blood volume and peripheral E) increased arterial stiffness
venous tone. 5. In a steady state, venous return will be greater than cardiac
■ Central venous pressure has a negative influence on venous output when
return that can be illustrated graphically as a venous function A) peripheral venous pressure is higher than normal.
curve. B) blood volume is higher than normal.
■ Peripheral venous pressure has a positive influence on venous C) heart rate is lower than normal.
return and can be elevated by increased blood volume and/or D) cardiac contractility is lower than normal.
increased venous tone. E) Never, because in a steady state venous return must
equal cardiac output.
■ Because of its opposing influences on cardiac output and
venous return, central venous pressure automatically attains a
value that makes cardiac output and venous return equal.
■ Central venous pressure gives clinically relevant information
about circulatory status.
■ Central venous pressure can be estimated noninvasively
by noting the fullness of a patient’s jugular veins.
29
C H A P T E R

Arterial Pressure
Regulation
David E. Mohrman and Lois Jane Heller

O B J E C T I V E S

■ Identify the sensory receptors, afferent pathways, central integrating


centers, efferent pathways, and effector organs that participate in the
arterial baroreceptor reflex.
■ State the location of the arterial baroreceptors and describe their operation.
■ Describe how changes in the afferent input from arterial baroreceptors
influence the activity of the sympathetic and parasympathetic preganglionic
fibers.
■ Describe how the sympathetic and parasympathetic outputs from the
medullary cardiovascular centers change in response to changes in arterial
pressure.
■ Diagram the chain of events that are initiated by the arterial baroreceptor
reflex to compensate for a change in arterial pressure.
■ Describe how inputs to the medullary cardiovascular centers from
cardiopulmonary baroreceptors, and arterial and central chemoreceptors
influence sympathetic activity, parasympathetic activity, and mean arterial
pressure.
■ Describe and indicate the mechanisms involved in the cerebral
ischemic response, the Cushing reflex, the alerting reaction, blushing,
vasovagal syncope, and the cardiovascular responses to emotion
and pain.
■ Describe baroreceptor adaptation.
■ Describe the influence of changes in body fluid volume on arterial pressure
and diagram the steps involved in this process.
■ Describe how mean arterial pressure is adjusted in the long term to that
which causes fluid output rate to equal fluid intake rate.

puts unnecessary demands on the heart and vessels. Arterial


REGULATION OF pressure is continuously monitored by sensors located within
ARTERIAL PRESSURE the body. Whenever arterial pressure varies from normal,
multiple reflex responses are initiated that cause the adjust-
Appropriate systemic arterial pressure is the single most ments in cardiac output and total peripheral resistance neces-
important requirement for the proper function of the cardio- sary to return arterial pressure to its normal value. In the short
vascular system. Without sufficient arterial pressure, the brain term (seconds), these adjustments are brought about by
and the heart do not receive adequate blood flow no matter changes in the activity of the autonomic nerves leading to the
what adjustments are made in their vascular resistance by local heart and peripheral vessels. In the long term (minutes to
control mechanisms. In contrast, excessive arterial pressure days), other mechanisms such as changes in cardiac output

285

Ch29_285-294.indd 285 11/26/10 11:51:35 AM


286 SECTION V Cardiovascular Physiology

brought about by changes in blood volume play an increas- nerves. The effector organs are the heart and peripheral
ingly important role in the control of arterial pressure. The blood vessels.
short- and long-term regulations of arterial pressure will be
discussed in order in this chapter.
Efferent Pathways
Previous chapters have discussed the many actions of the sym-
pathetic and parasympathetic nerves leading to the heart
SHORT-TERM REGULATION and blood vessels. For both systems, postganglionic fibers,
OF ARTERIAL PRESSURE whose cell bodies are in ganglia outside the central nervous
system, form the terminal link to the heart and vessels. The
ARTERIAL BARORECEPTOR REFLEX influences of these postganglionic fibers on key cardiovascular
variables are summarized in Figure 29–1.
The arterial baroreceptor reflex is the single most impor- The activity of the terminal postganglionic fibers of the
tant mechanism providing short-term regulation of arte- autonomic nervous system is determined by the activity of
rial pressure. Recall that the usual components of a reflex preganglionic fibers whose cell bodies lie within the central
pathway include sensory receptors, afferent pathways, inte- nervous system (see Chapter 19). In the sympathetic pathways,
grating centers in the central nervous system, efferent path- the cell bodies of the preganglionic fibers are located within
ways, and effector organs. As shown in Figure 29–1, the the spinal cord. These preganglionic neurons have spontane-
efferent pathways of the arterial baroreceptor reflex are the ous activity that is modulated by excitatory and inhibitory
cardiovascular sympathetic and cardiac parasympathetic inputs, which arise from centers in the brainstem and descend

Medulla
Spinal
cord rvlm
− ?? nts
+

− +
rn +
Sympathetic na
preganglionic
fibers
Parasympathetic
+
preganglionic
+ fibers
Ganglia

+
Ganglia

Arterial
+ + − baroreceptor
+
Pcv SV × HR = CO +
Central −
Pa = CO × TPR
venous Heart
pool

Systemic organs

Veins Arterioles
Ppv
TPR
+ +

FIGURE 29–1 Components of the arterial baroreceptor reflex pathway. nts, nucleus tractus solitarius; rvlm, rostral ventrolateral
medullary group; rn, raphe nucleus; na, nucleus ambiguus; ??, incompletely mapped integration pathways that may also involve structures
outside the medulla. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.)
CHAPTER 29 Arterial Pressure Regulation 287

in distinct excitatory and inhibitory spinal pathways. In the Central Integration


parasympathetic system, the cell bodies of the preganglionic
Much of the central integration involved in reflex regulation of
fibers are located within the brainstem. Their spontaneous
the cardiovascular system occurs in the medulla oblongata in
activity is modulated by inputs from adjacent centers in the
what are traditionally referred to as the medullary cardiovas-
brainstem.
cular centers. The neural interconnections between the dif-
fuse structures in this area are complex and not completely
Afferent Pathways mapped. Moreover, these structures appear to serve multiple
Sensory receptors, called arterial baroreceptors, are found in functions including respiratory control. What is known with a
abundance in the walls of the aorta and carotid arteries. Major fair degree of certainty is where the cardiovascular afferent
concentrations of these receptors are found near the arch of and efferent pathways enter and leave the medulla. As indi-
the aorta (the aortic baroreceptors) and at the bifurcation of cated in Figure 29–1, the afferent sensory information from
the common carotid artery into the internal and external the arterial baroreceptors enters the medullary nucleus trac-
carotid arteries on either side of the neck (the carotid sinus tus solitarius, where it is relayed via polysynaptic pathways to
baroreceptors). The receptors are mechanoreceptors that other structures in the medulla (and higher brain centers, such
sense arterial pressure indirectly from the degree of stretch of as the hypothalamus, as well). The cell bodies of the efferent
the elastic arterial walls. In general, increased stretch causes an vagal parasympathetic cardiac nerves are located primarily in
increased action potential generation rate by the arterial barore- the medullary nucleus ambiguus. The sympathetic autonomic
ceptors. Baroreceptors actually sense not only absolute stretch, efferent information leaves the medulla predominantly from
but also the rate of change of stretch. For this reason, both the the rostral ventrolateral medulla group of neurons (via an
mean arterial pressure and arterial pulse pressure affect barore- excitatory spinal pathway) or the raphe nucleus (via an inhib-
ceptor firing rate as indicated in Figure 29–2. The dashed itory spinal pathway). The intermediate processes involved in
curve shows how baroreceptor firing rate is affected by differ- the actual integration of the sensory information into appro-
ent levels of a steady arterial pressure. The solid curve indi- priate sympathetic and parasympathetic responses are not well
cates how baroreceptor firing rate is affected by the mean value understood. Much of this integration takes place within the
of a pulsatile arterial pressure. Note that the presence of pulsa- medulla, but higher centers such as the hypothalamus are
tions increases the baroreceptor firing rate at any given level of probably involved as well. In this context, knowing the details
mean arterial pressure. Note also that changes in mean arterial of the integration process is not as important as appreciating
pressure near the normal value of 100 mm Hg produce the the overall effects that changes in arterial baroreceptor activity
largest changes in baroreceptor discharge rate and there is very have on the activities of parasympathetic and sympathetic car-
little output at low pressures. diovascular nerves.
If arterial pressure remains above normal over a period of Several functionally important points about the central con-
several days, the arterial baroreceptor firing rate will gradually trol of the autonomic cardiovascular nerves are illustrated in
return toward normal. Thus, arterial baroreceptors are said to Figure 29–1. The major external influence on the cardiovascu-
adapt to long-term changes in arterial pressure. For this rea- lar centers comes from the arterial baroreceptors. Because the
son, the arterial baroreceptor reflex does not serve as a mecha- arterial baroreceptors are active at normal arterial pressures,
nism for the long-term regulation of arterial pressure. they supply a tonic input to the central integration centers.
As indicated in Figure 29–1, the integration process is such
that increased input from the arterial baroreceptors tends to
simultaneously: (1) inhibit the activity of the spinal sympa-
thetic excitatory tract, (2) stimulate the activity of the spinal
Max
sympathetic inhibitory tract, and (3) stimulate the activity of
Baroreceptor nerve activity

parasympathetic preganglionic nerves. Thus, an increase in


Pulsatile the arterial baroreceptor discharge rate (caused by increased
arterial pressure) causes a decrease in the tonic activity of car-
Steady
diovascular sympathetic nerves and a simultaneous increase
in the tonic activity of cardiac parasympathetic nerves. Con-
versely, decreased arterial pressure causes increased sympa-
thetic and decreased parasympathetic activity.

0 50 100 150 200 OPERATION OF THE ARTERIAL


Mean arterial pressure (mm Hg) BARORECEPTOR REFLEX
FIGURE 29–2 Effect of mean arterial pressure on
baroreceptor nerve activity. (Modified with permission from Mohrman DE, The arterial baroreceptor reflex is a continuously operating
Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/ control system that automatically makes adjustments to pre-
McGraw-Hill, 2006.) vent disturbances in the heart and/or vessels from causing
288 SECTION V Cardiovascular Physiology

↓ Mean arterial pressure (primary disturbance)

29

↓ Baroreceptor discharge

29
29 29
Medullary cardiovascular centers

↑ Sympathetic activity ↓ Parasympathetic activity

27
27 24
↑ Arteriolar tone ↑ Venous tone ↑ Cardiac contractility
27
27

↑ Blood volume
↑ Peripheral venous pressure
23
28 23
27 28
Transcapillary fluid reabsorption
+ −
Central venous pressure
26 24

↓ Capillary pressure 24

↑ Vasoconstriction
26 ↑ Stroke volume ↑ Heart rate
24
26 24
↑ Total peripheral resistance ↑ Cardiac output

26 26
↑ Mean arterial pressure (counteracting response)

FIGURE 29–3 Immediate cardiovascular adjustments caused by a decrease in arterial blood pressure. Circled numbers indicate the
chapter in which each interaction is discussed. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange
Medical Books/McGraw-Hill, 2006.)

large changes in mean arterial pressure. The arterial barore- important in some areas such as the kidney, the skin, and the
ceptor reflex mechanism acts to regulate arterial pressure in a splanchnic organs than in the brain and heart muscle. Thus,
negative feedback fashion as was described in Chapter 1. the reflex response to a decrease in arterial pressure may, for
Figure 29–3 shows many events in the arterial baroreceptor example, include a significant increase in renal vascular resis-
reflex pathway that occur in response to a disturbance of tance and a decrease in renal blood flow without changing the
decreased mean arterial pressure. All of the events shown in cerebral vascular resistance or blood flow. The peripheral vas-
Figure 29–3 have already been discussed, and each should be cular adjustments associated with the arterial baroreceptor
carefully examined (and reviewed if necessary) at this point reflex take place primarily in organs with strong sympathetic
because a great many of the interactions that are essential to vascular control.
understanding cardiovascular physiology are summarized in
this figure.
Note in Figure 29–3 that the overall response of the arterial OTHER CARDIOVASCULAR
baroreceptor reflex to the disturbance of decreased mean arte- REFLEXES & RESPONSES
rial pressure is to increase mean arterial pressure (i.e., the
response tends to counteract the disturbance). A disturbance In spite of the arterial baroreceptor reflex mechanism, large
of increased mean arterial pressure would elicit events oppo- and rapid changes in mean arterial pressure occur in certain
site to those shown in Figure 29–3 and produce a decrease in physiological and pathological situations. These reactions are
mean arterial pressure; again, the response tends to counteract caused by influences on the medullary cardiovascular centers
the disturbance. Recall that neural control of vessels is more other than those from the arterial baroreceptors. As outlined
CHAPTER 29 Arterial Pressure Regulation 289

in the following sections, these inputs to the medullary cardio- ably the cerebral ischemic response is initiated by chemore-
vascular centers arise from many types of peripheral and cen- ceptors located within the central nervous system. However, if
tral receptors as well as from “higher centers” in the central cerebral blood flow is severely inadequate for several minutes,
nervous system such as the hypothalamus and the cortex. the cerebral ischemic response wanes and is replaced by
An analogy was made between the arterial baroreceptor marked loss of sympathetic activity. This results when func-
reflex operating to control arterial pressure to a home heating tion of the nerve cells in the cardiovascular centers becomes
system acting to control room temperature (see Chapter 1). directly depressed by the unfavorable chemical conditions in
The temperature setting on the thermostat determines the set the cerebrospinal fluid.
point for temperature regulation. Most of the influences that Whenever intracranial pressure is increased—for example,
are about to be discussed influence arterial pressure as if they by increased cerebral spinal fluid (CSF) pressure or trauma-
changed the arterial baroreceptor reflex’s set point for pressure induced bleeding within the rigid cranium—there is a parallel
regulation. Consequently, the arterial baroreceptor reflex does increase in arterial pressure. This is called the Cushing reflex.
not resist most of these pressure disturbances but actually It can cause mean arterial pressures of more than 200 mm Hg
assists in producing them. in severe cases of increased intracranial pressure. The benefit
of the Cushing reflex is that it prevents collapse of cranial ves-
sels and thus preserves adequate brain blood flow in the face of
REFLEXES FROM RECEPTORS large increases in intracranial pressure. The mechanisms
IN HEART & LUNGS responsible for the Cushing reflex are not known but could
involve the central chemoreceptors. A hallmark of the Cush-
A host of mechanoreceptors and chemoreceptors that elicit ing reflex is acutely increased arterial pressure in spite of
reflex cardiovascular responses have been identified in the atria, accompanying bradycardia. It seems as if the short-term arte-
ventricles, coronary vessels, and lungs. The role of these cardio- rial baroreceptor reflex is attempting to counteract this distur-
pulmonary receptors in the control of the cardiovascular sys- bance by activating parasympathetic nerves to the SA node of
tem is, in most cases, incompletely understood, but they are the heart.
likely involved in many physiological and pathological states.
Cardiopulmonary baroreceptors (sometimes referred to as
low-pressure receptors) sense the pressure (or volume) in the CARDIOVASCULAR RESPONSES
atria and central venous pool. Increased central venous pres- ASSOCIATED WITH EMOTION
sure (or volume) causes activation of these receptors by stretch,
and elicits a reflex decrease in sympathetic activity. Decreased Cardiovascular responses are frequently associated with cer-
central venous pressure (or volume) produces the opposite tain states of emotion. These responses originate in the cere-
response. These cardiopulmonary baroreflexes normally exert a bral cortex and reach the medullary cardiovascular centers
tonic inhibitory influence on sympathetic activity. Alterations in through corticohypothalamic pathways. The least compli-
sympathetic activity evoked by increases or decreases in central cated of these responses is the blushing that is often detectable
venous pressure not only have short-term influences on arterial in individuals with lightly pigmented skin during states of
pressure, but also influence renal mechanisms that influence embarrassment. The blushing response involves a loss of sym-
blood volume and long-term regulation of arterial pressure. pathetic vasoconstrictor activity only to particular cutaneous
vessels, and this produces the blushing by allowing engorge-
ment of the cutaneous venous sinuses.
CHEMORECEPTOR REFLEXES Excitement or a sense of danger often elicits a complex
behavioral pattern called the alerting reaction (also called the
Low Po2, low pH, and/or high Pco2 levels in the arterial blood “defense” or “fight-or-flight” response). The alerting reac-
cause reflex increases in breathing and mean arterial pressure. tion involves a host of responses such as pupillary dilation and
These responses appear to be a result of increased activity of increased skeletal muscle tenseness that are generally appro-
arterial chemoreceptors, located in the carotid arteries and priate preparations for some form of intense physical activity.
the arch of the aorta, and central chemoreceptors, located The cardiovascular component of the alerting reaction is an
within the central nervous system. Chemoreceptors probably increase in blood pressure caused by a general increase in car-
play little role in the normal regulation of arterial pressure diovascular sympathetic nervous activity and a decrease in
because arterial blood Po2 and Pco2 are normally held very nearly cardiac parasympathetic activity.
constant by respiratory control mechanisms. See Chapter 38 Some individuals respond to situations of extreme stress by
for more details. fainting, a situation referred to clinically as vasovagal syn-
An extremely strong reaction called the cerebral ischemic cope. The loss of consciousness is due to decreased cerebral
response is triggered by inadequate blood flow (ischemia) to blood flow that is itself produced by a sudden dramatic loss of
the brain and can produce a more intense sympathetic vaso- arterial blood pressure that occurs as a result of a sudden loss
constriction and cardiac stimulation than is elicited by any of sympathetic tone and a simultaneous large increase in para-
other influence on the cardiovascular control centers. Presum- sympathetic tone and decrease in heart rate.
290 SECTION V Cardiovascular Physiology

CENTRAL COMMAND phasize that the arterial baroreceptors normally and


continuously supply the major input to the medullary cen-
The term central command is used to imply an input from the ters. The arterial baroreceptor input is shown as inhibitory
cerebral cortex to lower brain centers during voluntary muscle because an increase in arterial baroreceptor firing rate results
exercise. The concept is that the same cortical drives that initi- in a decrease in sympathetic output. (Decreased sympathetic
ate somatomotor (skeletal muscle) activity also simultaneously output should be taken to imply also a simultaneous increase
initiate cardiovascular (and respiratory) adjustments appro- in parasympathetic output that is not shown in this figure.)
priate to support that activity. In the absence of any other obvi- As is indicated in Figure 29–4, the nonarterial baroreceptor
ous causes, central command is at present the best explanation influences on the medullary cardiovascular centers fall into two
as to why both mean arterial pressure and respiration increase categories: (1) those that increase arterial pressure by raising the
during voluntary exercise. set point for the arterial baroreceptor reflex and thus cause an
increase in sympathetic activity and (2) those that decrease arte-
rial pressure by lowering the set point for the arterial barorecep-
REFLEX RESPONSES TO PAIN tor reflex and thus cause a decrease in sympathetic activity.
Pain can have either a positive or a negative influence on arte-
rial pressure. Generally, superficial or cutaneous pain causes
an increase in blood pressure in a manner similar to that asso- LONG-TERM REGULATION OF
ciated with the alerting response and perhaps over many of
the same pathways. Deep pain from receptors in the viscera or ARTERIAL PRESSURE
joints, however, often causes a cardiovascular response similar Long-term regulation of arterial pressure is a topic of clinical
to that which accompanies vasovagal syncope, that is, decreased relevance because of the prevalence of hypertension (sustained
sympathetic tone, increased parasympathetic tone, and a sig- excessive arterial blood pressure) in our society. The most
nificant decrease in blood pressure. This response may con- long-standing and generally accepted theory of long-term
tribute to the state of shock that often accompanies crushing pressure regulation is that it crucially involves the kidneys,
injuries and/or joint displacement. their sodium handling, and ultimately the regulation of blood
volume. This theory is sometimes referred to as the “renal-
SUMMARY MAP set point” or “fluid balance” model of long-term arte-
rial blood pressure control. In essence, this theory asserts that
The influences on the medullary cardiovascular centers in the long term, mean arterial pressure is whatever it needs to
that have been discussed in the preceding sections are sum- be to maintain an appropriate blood volume through arterial
marized in Figure 29–4. This figure is intended first to reem- pressure’s direct effects on renal function.

Response to exercise (central command)


Sense of danger (alerting/defense reaction)
Cerebral ischemic response
↑ Intracranial pressure (cushing reflex)
↓ PO2, ↑ PCO2 in arterial blood
↓ Central venous pressure (cardiopulmonary baroreflexes)
Cutaneous pain

Increase
set point

+
Arterial
Sympathetic Medullary baroreceptor

output centers input

Lower
FIGURE 29–4 Summary of the factors that set point
influence the set point of the arterial
baroreceptor reflex. (Modified with permission from Vasovagal syncope
Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New Deep pain
York: Lange Medical Books/McGraw-Hill, 2006.) ↑ Central venous pressure (cardiopulmonary baroreflexes)
CHAPTER 29 Arterial Pressure Regulation 291

FLUID BALANCE & pressure, hours or even days may be required before a change in
ARTERIAL PRESSURE urinary output rate produces a significant accumulation or loss
of total body fluid volume. What this fluid volume mechanism
Several key factors in the long-term regulation of arterial lacks in speed, however, it more than makes up for in persis-
blood pressure have already been considered. First is the fact tence. As long as there is any inequality between the fluid intake
that the baroreceptor reflex, however well it counteracts tem- rate and the urinary output rate, body fluid volume is changing
porary disturbances in arterial pressure, cannot effectively and this fluid volume mechanism has not completed its adjust-
regulate arterial pressure in the long term for the simple rea- ment of arterial pressure. The fluid volume mechanism is in
son that the baroreceptor firing rate adapts to prolonged equilibrium only when the urinary output rate equals the fluid
changes in arterial pressure. intake rate. (In the present discussion, assume that fluid intake
The second pertinent fact is that circulating blood volume rate represents that in excess of the obligatory fluid losses that
can influence arterial pressure because: ↑ blood volume → normally occur in the feces and by transpiration from the skin
and structures in the respiratory tract. The processes that regu-
↑ peripheral venous pressure → right shift of venous function
curve → ↑ central venous pressure → ↑ cardiac output → late thirst are not well understood but seem to involve many of
the same factors that influence urinary output.) In the long term,
↑ arterial pressure.
Arterial pressure has a profound influence on urinary out- the arterial pressure must be that which makes the urinary output
put rate and thus affects total body fluid volume. Because rate equal to the fluid intake rate.
blood volume is one of the components of the total body fluid, The arterial baroreceptor reflex is, of course, essential for
blood volume alterations accompany changes in total body counteracting rapid changes in arterial pressure. The fluid vol-
fluid volume. The mechanisms are such that a decrease in ume mechanism, however, determines the long-term level of
arterial pressure causes a decrease in urinary output rate and arterial pressure because it slowly overwhelms all other influ-
thus an increase in blood volume. But, as outlined in the pre- ences. Through adaptation, the baroreceptor mechanism
ceding sequence, increased blood volume tends to increase adjusts itself so that it operates to prevent acute changes in
arterial pressure. Thus, the complete sequence of events that blood pressure from the prevailing long-term level as deter-
are initiated by a decrease in arterial pressure can be listed as mined through fluid balance.
follows:
↓ Arterial pressure (disturbance) → ↓ urinary output rate
→ ↑ fluid volume → ↑ blood volume → ↑ cardiac output → EFFECT OF ARTERIAL PRESSURE
↑ arterial pressure (compensation). ON URINARY OUTPUT RATE
As indicated in Figure 29–5, both the arterial baroreceptor
reflex and this fluid volume mechanism are negative feedback As discussed above, an increase in arterial pressure normally
loops that regulate arterial pressure. Whereas the arterial barore- leads to an increase in urine output rate. Many mechanisms
ceptor reflex is very quick to counteract disturbances in arterial are involved in this phenomenon and are discussed in detail

SHORT-TERM LONG-TERM Fluid intake rate


Baroreceptor reflex Fluid balance

Blood volume

− − +
TPR CO

+ + +

Fluid Volume
Arterial pressure


Kidney
+ Urinary output rate FIGURE 29–5 Mechanisms of short- and
long-term regulations of arterial pressure. TPR,
total peripheral resistance; CO, cardiac output.
(Modified with permission from Mohrman DE, Heller LJ:
Cardiovascular Physiology, 6th ed. New York: Lange Medical
Books/McGraw-Hill, 2006.)
292 SECTION V Cardiovascular Physiology

ECG shows left deviation of the ventricular mean electri-


3 Heathy person
cal axis (−35°) (see Chapter 25).
A tentative diagnosis of chronic hypertension is made.
Urine output rate × normal

Hypertensive person He has access to blood pressure monitoring equipment at


home and was instructed to monitor his blood pressure

y
rap
2 daily for 1 week and to report his results to the doctor.

d
ate
he
At that time, a decision about therapeutic strategies will

ic t

tre
Un
ret
be made.
Normal fluid intake N Diu Systemic hypertension is defined as a chronic increase in
1 mean systemic arterial pressure above 140/90 mm Hg. It is
A
C an extremely common cardiovascular problem, affecting
B Restricted fluid intake more than 20% of the adult population of the Western
world. It has been established that hypertension increases
50 100 150 200 the risk of coronary artery disease, myocardial infarction,
Mean arterial pressure (mm Hg) heart failure, stroke, and many other serious cardiovascular
problems. Moreover, it has been clearly demonstrated that
FIGURE 29–6 Renal function curves in a normal person and
in a hypertensive person with and without antihypertensive
the risk of serious cardiovascular incidents is reduced by
therapy. (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular proper treatment of hypertension.
Physiology, 6th ed. New York: Lange Medical Books/McGraw-Hill, 2006.) In approximately 90% of cases, the primary abnor-
mality that produces high blood pressure is unknown.
(This condition is sometimes referred to as primary or
in Section 7. At this point, it is only important to recognize essential hypertension because the elevated level was
that, because of many synergistic influences, arterial pressure previously thought to be “essential” to drive the blood
has a huge positive effect on renal urine output rate as is indi- through the systemic circulation, particularly the brain.)
cated by the very steep slope of the relationship shown in Genetic factors contribute importantly to the develop-
Figure 29–6. In a steady state, urine output (plus fluid lost ment of hypertension (generally being more common
from the body by other means) is equal to fluid intake (point in males than females and in blacks than whites) and
N in this figure). At arterial pressures below point N, fluid environmental factors such as obesity, high-salt diets,
intake exceeds urinary output and bodily fluid volume will diabetes mellitus, and/or certain forms of psychological
necessarily be increasing. The opposite is true at arterial blood stress may either aggravate or precipitate hypertension
pressure higher than that at point N. Thus, a healthy person in susceptible individuals.
with a normal fluid intake rate will have, as a long-term aver- Structural changes in the left heart and arterial vessels
age, the arterial pressure associated with point N in Figure occur in response to hypertension. Early alterations
29–6. Because of the steepness of the curve shown in Figure include hypertrophy of muscle cells and thickening of the
29–6, even marked changes in fluid intake rate have minor walls of the ventricle and resistance vessels. Late changes
influences on the arterial pressure of a normal individual. associated with deterioration of function include increases
in connective tissue and increased tissue stiffness.
The established phase of hypertension is associated
with increased total peripheral resistance. Cardiac output
CLINICAL CORRELATION and/or blood volume may be increased during the early
developmental phase, but are usually normal after the
A 35-year-old African American male has come to the hypertension is established. The increased total periph-
doctor for a general physical exam. He has not been to see eral resistance associated with established hypertension
a physician for at least 10 years. At present, he has no spe- may be due to microvessel rarefaction (decrease in den-
cific complaints about health conditions, but admits to not sity), pronounced structural adaptations of the peripheral
getting as much exercise as he did while in his twenties. His vascular bed, increased basal vascular smooth muscle
father had a mild heart attack at 50 years of age, received a tone, increased sensitivity and reactivity of the vascular
coronary artery stent, and has been treated for hyperten- smooth muscle cells to external vasoconstrictor stimuli,
sion for the 15 years since that time. His mother has recently and/or diminished production and/or effect of endoge-
been diagnosed with type 2 diabetes mellitus. nous vasodilator substances (e.g., nitric oxide).
He is 5′11″ (180 cm), 240 lb (109 kg), and has a heart rate Chronic hypertension is not due to a sustained increase
of 64 beats/min and an arterial pressure of 150/92 mm Hg. in sympathetic vasoconstrictor neural discharge nor is it
Chest sounds and heart sounds are normal. All other due to a sustained increase of any blood-borne vasocon-
aspects of the physical exam are within normal ranges. An strictive factor. (Both neural and hormonal influences,
CHAPTER 29 Arterial Pressure Regulation 293

however, may help initiate primary hypertension.) Blood of the amount of saturated fats in the diet, and establish-
pressure–regulating reflexes (both the short-term arte- ment of a regular exercise program, may also help reduce
rial and cardiopulmonary baroreceptor reflexes and the blood pressure in certain individuals.
long-term, renal-dependent, pressure-regulating reflexes)
become adapted or “reset” to regulate blood pressure at a
higher-than-normal level. Disturbances in renal function
contribute importantly to the development and mainte- CHAPTER SUMMARY
nance of primary hypertension. Recall that, in the long ■ Arterial pressure is closely regulated to ensure adequate blood
term, arterial pressure can stabilize only at the level that flow to the tissues.
makes urinary output rate equal to fluid intake rate. As ■ The arterial baroreceptor reflex is responsible for regulating
shown by point N in Figure 29–6, this pressure is approx- arterial pressure in the short term on a second-to-second and
imately 100 mm Hg in a normal individual. All forms of moment-to-moment basis.
hypertension involve an alteration somewhere in ■ The arterial baroreceptor reflex involves the following: pressure
the chain of events by which changes in arterial pressure sensing by stretch-sensitive baroreceptor nerve endings in the
produce changes in urinary output rate such that the walls of arteries, neural integrating centers in the brainstem
renal function curve is shifted rightward as indicated in that adjust autonomic nerve activity in response to the pressure
Figure 29–6. Higher-than-normal arterial pressure is information they receive from the arterial baroreceptors, and
required to produce a normal urinary output rate in a responses of the heart and vessels to changes in autonomic
nerve activity.
hypertensive individual.
■ Overall, the arterial baroreflex operates such that increases
The untreated hypertensive individual in Figure 29–6
in arterial pressure lead to an essentially immediate decrease
would have a very low urinary output rate at the normal
in sympathetic nerve activity and a simultaneous increase in
mean arterial pressure of 100 mm Hg. With a normal
parasympathetic nerve activity (and vice versa).
fluid intake rate, this untreated hypertensive patient
■ The brainstem integrating centers also receive nonarterial
retains fluid to ultimately stabilize at point A (mean arte-
baroreceptor inputs that can raise or lower the set point for
rial pressure = 150 mm Hg). Baroreceptors adapt within short-term arterial pressure regulation.
days so that they will have a normal discharge rate at the
■ In the long term, arterial pressure is regulated by changes in
prevailing average arterial pressure. Thus, once the hyper- blood volume that come about because arterial pressure has
tensive individual has been at point A for a week or more, a strong influence on urinary output rate by the kidney.
even the baroreceptor mechanism will begin resisting
acute changes from the 150-mm Hg pressure level. In
certain hypertensive individuals, dietary salt restriction
produces a substantial reduction in blood pressure
STUDY QUESTIONS
because of the reduced requirement for water retention 1. In the normal operation of the arterial baroreceptor reflex, a
to osmotically balance the salt load. This effect is illus- cardiovascular disturbance that decreases mean arterial pressure
trated by a shift from point A to point B. The efficacy of will evoke a decrease in
lowering salt intake to lower arterial pressure depends A) urine output rate.
heavily on the slope of the renal function curve in the B) sympathetic nerve activity.
hypertensive individual. The arterial pressure of a nor- C) heart rate.
D) total peripheral resistance.
mal individual, for example, is affected only slightly by
E) myocardial contractility.
changes in salt intake because the normal renal function
2. Which one of the following, after all reflex adjustments are
curve is so steep. A second common treatment of hyper-
complete, will result in an increase in mean arterial pressure?
tension is diuretic therapy (see Chapters 44 and 46). The
A) low carbon dioxide levels in arterial blood
net effect of diuretic therapy is that the urinary output
B) increased intracranial pressure
rate for a given arterial pressure is increased, that is,
C) decreased cardiac filling pressure
diuretic therapy shifts the renal function curve upward. D) low blood volume
The combined result of restricted fluid intake and diuretic E) supraventricular tachycardia
therapy for the hypertensive individual of Figure 29–6 is 3. An individual has higher-than-normal mean arterial pressure
illustrated by point C. and lower-than-normal pulse rate. Which of the following is
A variety of antihypertensive pharmacological most likely to cause such a combination?
approaches are available that may include β-adrenergic A) low oxygen levels in arterial blood
blockers, angiotensin-converting enzyme inhibitors, B) anxiety
angiotensin II receptor blockers, and calcium channel C) exercise
blockers (see Section 7). Alterations in lifestyle, including D) significant blood loss
reduction of stress, decreases in caloric intake, limitation E) a drug that selectively stimulates alpha-adrenergic
receptors
294 SECTION V Cardiovascular Physiology

4. Which of the following best describes, first, the immediate direct 5. An increase in arterial baroreceptor firing rate will reflexly result in
effect and, second, the reflex cardiovascular consequences of A) an increase in vagal activity to the heart.
giving a normal person a drug that blocks beta1-adrenergic B) an increase in sympathetic activity to arterioles in the brain.
receptors? C) an increase in renal blood flow.
A) decreased heart rate, increased total peripheral resistance D) an increase in mean arterial pressure.
B) decreased ejection fraction, decreased total peripheral E) an increase in cardiac ejection fraction.
resistance
C) increased heart rate, increased urine production
D) increased cardiac output, decreased total peripheral
resistance
E) decreased end-diastolic volume, increased heart rate
30
C H A P T E R

Cardiovascular Responses
to Physiological Stress
Lois Jane Heller and David E. Mohrman

O B J E C T I V E S

■ Identify the primary disturbance(s) that any given normal homeostatic


disturbance (such as changes in body position) places on the cardiovascular
system.
■ List how any such given primary disturbance changes the influence on
the medullary cardiovascular centers from (1) arterial baroreceptors and
(2) other sources.
■ State what immediate reflex compensatory changes will occur in sympathetic
and parasympathetic nerve activities as a result of the altered influences on
the medullary cardiovascular centers.
■ Indicate what immediate compensatory changes will occur to influence
mean arterial pressure in response to any given primary disturbance,
including changes in: heart rate, cardiac contractility, stroke volume, arteriolar
tone, venous tone, peripheral venous pressure, central venous pressure, total
peripheral resistance, resistance in any major organ, blood flow through any
major organ, cutaneous blood flow, transcapillary fluid movement, and
long-term renal adjustments in urine output and total body fluid balance.
■ State how gravity influences arterial, venous, and capillary pressures at any
height above or below the heart in a standing individual.
■ Describe and explain the changes in central venous pressure and the changes
in transcapillary fluid balance and venous volume in the lower extremities
caused by standing upright.
■ Describe the operation of the “skeletal muscle pump” and explain how it
simultaneously promotes venous return and decreases capillary hydrostatic
pressure in the muscle vascular beds.
■ Identify the primary disturbances and compensatory responses evoked by
acute changes in body position.
■ Describe the chronic effects of long-term bed rest on cardiovascular variables.
■ List the cardiovascular consequences of respiratory activity.
■ Identify the major maternal cardiovascular adjustments that occur
during pregnancy.
■ Follow the pathway of blood flow through the fetal heart and describe the
changes that occur at birth.
■ Indicate the normal changes that occur in cardiovascular variables during
childhood.
■ Identify age-dependent changes that occur in cardiovascular variables
■ Describe gender-dependent differences in cardiovascular variables.

295

Ch30_295-304.indd 295 11/26/10 11:50:50 AM


296 SECTION V Cardiovascular Physiology

CARDIOVASCULAR RESPONSES EFFECT OF GRAVITY


TO PHYSIOLOGICAL STRESSES RESPONSES TO CHANGES
A wide variety of normal, everyday situations can disturb IN BODY POSITION
homeostasis within the cardiovascular system and can evoke
an equally wide variety of reflex responses. The key to Significant cardiovascular readjustments accompany changes in
understanding the cardiovascular adjustments in each situa- body position because gravity has an effect on pressures within
tion is to recall that the arterial baroreceptor reflex and renal the cardiovascular system. In the preceding chapters, the influ-
fluid balance mechanisms always act to blunt changes in ence of gravity was ignored and pressure differences between
arterial pressure. The overall result is that adequate blood various points in the systemic circulation were related only to
flow to the brain and the heart muscle is maintained in any flow and vascular resistance (Δ P = Q̇ R). As shown in Figure
circumstance. 30–1, this is approximately true only for a recumbent individual.
The cardiovascular alterations in the following example is In a standing individual, additional cardiovascular pressure dif-
produced by the combined effects of (1) the primary, direct ferences exist between the heart and regions that are not at heart
influences of the disturbance on the cardiovascular variables level. This is most important in the lower legs and feet of a stand-
and (2) the reflex compensatory adjustments that are trig- ing individual. As indicated in Figure 30–1B, intravascular pres-
gered by the primary disturbances. As you will see in later sures in the feet may be increased by 90 mm Hg simply from the
sections of this book, the general pattern of reflex adjustment weight of the blood in the arteries and veins leading to and from
is similar in all situations. Rather than trying to memorize the the feet. Note by comparing Figure 30–1A and B that standing
cardiovascular alterations that accompany each situation, the upright does not in itself change the flow through the lower
reader should strive to understand each response in terms of extremities, since gravity has the same effect on arterial and
the primary disturbances and reflex compensatory reactions venous pressures and thus does not change the arteriovenous
involved. pressure difference at any given level above or below the heart.

A RECUMBENT
P = 100 mm Hg Arteries P = 95 mm Hg

Arterioles
Heart Foot
Capillaries
Veins
P = 25 mm Hg

P = 0 mm Hg Valves P = 5 mm Hg

STANDING
Surface P = 100 mm Hg P = 0 mm Hg P =100 mm Hg
P = 0 mm Hg Heart level
P = 0 mm Hg
Lymphatics

Arteries
Veins

P = 95 mm Hg
1.5-m depth
P = 95 mm Hg P = 185 mm Hg P = 20 mm Hg P = 185 mm Hg
Foot level
P = 90 mm Hg Filtration P = 185 mm Hg P = 105 mm Hg P = 40 mm Hg
P = 115 mm Hg
Without compensation With sympathetic stimulation During skeletal muscle contraction Shortly after contraction
B C D E
FIGURE 30–1 Effect of gravity on vascular pressure (A and B) with compensatory influences of sympathetic stimulation (C) and
the skeletal muscle pump (D and E). (Modified with permission from Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/
McGraw-Hill, 2006.)
CHAPTER 30 Cardiovascular Responses to Physiological Stress 297

There are, however, two major direct effects of the increased ence the cardiovascular centers by lessening the normal input
pressure in the lower extremities that are shown in Figure 30–1B: from both the arterial and the cardiopulmonary baroreceptors.
(1) the increase in venous transmural pressure distends the The result of a decreased baroreceptor input to the cardiovas-
compliant peripheral veins and greatly increases peripheral cular centers will be reflex adjustments appropriate to increase
venous volume by as much as 500 mL in a normal adult and blood pressure—that is, decreased cardiac parasympathetic
(2) the increase in capillary transmural hydrostatic pressure nerve activity and increased activity of the cardiovascular sym-
causes a tremendously high transcapillary filtration rate. pathetic nerves as shown in the bottom part of Figure 30–2.
For reasons to be described, reflex activation of sympathetic Heart rate and cardiac contractility will increase, as will arterio-
nerves accompanies the transition from a recumbent to an lar and venous constriction in most systemic organs (brain and
upright position. However, Figure 30–1C shows how vasocon- heart excepted).
striction from sympathetic activation is only marginally effec- Heart rate and total peripheral resistance are greater when
tive in ameliorating the adverse effects of gravity on the lower an individual stands than when the individual is lying
extremities. Arteriolar constriction can cause a greater pressure down. Note that these particular cardiovascular variables are
drop across arterioles, but this has only a limited effect on capil- not directly influenced by standing but are changed by the
lary pressure because venous pressure remains extremely high. compensatory responses. Stroke volume and cardiac output,
Filtration will continue at a very high rate. In fact, the normal conversely, are usually decreased below their recumbent val-
cardiovascular reflex mechanisms alone are incapable of deal- ues during quiet standing despite the reflex adjustments that
ing with upright posture without the aid of the skeletal muscle tend to increase them. This is because the reflex adjustments
pump. A person who remained upright without intermittent do not quite overcome the primary disturbance on these vari-
contraction of the skeletal muscles in the legs would lose con- ables caused by standing. This is in keeping with the general
sciousness in 10–20 minutes because of the decreased brain dictum that short-term cardiovascular compensations never
blood flow that would stem from diminished central blood vol- completely correct the initial disturbance.
ume, stroke volume, cardiac output, and arterial pressure. Mean arterial pressure is often found to increase when a per-
The effectiveness of the skeletal muscle pump in counteract- son changes from the recumbent to the standing position. At first
ing venous blood pooling and edema formation in the lower glance, this is a violation of many rules of cardiovascular system
extremities during standing is illustrated in Figure 30–1D and E. operation. How can a compensation be more than complete?
The compression of vessels during skeletal muscle contraction Moreover, how is increased sympathetic activity compatible with
expels both venous blood and lymphatic fluid from the lower higher-than-normal mean arterial pressure in the first place? In
extremities (Figure 30–1D). Immediately after a skeletal muscle the case of standing, there are many answers to these apparent
contraction, both veins and lymphatic vessels are relatively puzzles. First, the average arterial baroreceptor discharge rate
empty because their one-way valves prevent the backflow of can actually decrease in spite of a small increase in mean arterial
previously expelled fluid (Figure 30–1E). Most important, the pressure if there is simultaneously a sufficiently large decrease in
weight of the venous and lymphatic fluid columns is temporar- pulse pressure. Second, the influence on the medullary cardio-
ily supported by the closed one-way valve leaflets. Consequently, vascular centers from cardiopulmonary receptors is interpreted
venous pressure is drastically lowered immediately after skeletal as a decrease in blood volume and may increase arterial pressure
muscle contraction and rises only gradually as veins refill with by mechanisms that increase the set point. Third, mean arterial
blood from the capillaries. Thus, capillary pressure and trans- pressure determined by sphygmomanometry from the arm of a
capillary fluid filtration rate are dramatically reduced for some standing individual overestimates the mean arterial pressure
period after a skeletal muscle contraction. Some transcapillary actually being sensed by the baroreceptors in the carotid sinus
fluid filtration is still present, but the increased lymphatic flow region of the neck because of gravitational effects.
resulting from the skeletal muscle pump is normally sufficient The kidney is especially susceptible to changes in sympathetic
to prevent noticeable edema formation in the feet. nerve activity (as will be discussed in Section 7). Consequently,
The actions of the skeletal muscle pump, however benefi- as shown in Figure 30–2, every reflex alteration in sympathetic
cial, do not completely prevent an increase in the average activity has influences on fluid balance that become important
venous pressure and blood pooling in the lower extremities on in the long term. Standing, which is associated with an increase
standing. Thus, assuming an upright position upsets the car- in sympathetic tone, ultimately results in an increase in fluid
diovascular system and elicits reflex cardiovascular adjust- volume. The ultimate benefit of this is that an increase in blood
ments, as shown in Figure 30–2. volume generally reduces the magnitude of the reflex altera-
As with all cardiovascular responses, the key to understand- tions required to tolerate upright posture.
ing the alterations associated with standing is to distinguish the
primary disturbances from the compensatory responses.
As shown in the top part of Figure 30–2, the immediate conse- RESPONSES TO LONG-TERM BED REST
quence of standing is an increase in both arterial and venous
pressures in the lower extremities. The latter causes a major The cardiovascular system of an individual who is sub-
redistribution of blood volume out of the central venous pool. jected to long-term bed rest undergoes a variety of adaptive
By the chain of events shown, the primary disturbances influ- changes. The most significant immediate change that occurs on
298 SECTION V Cardiovascular Physiology

Standing

↑ Venous pressure ↑ Arterial pressure


(lower extremities) (lower extremities)

↑ Capillary pressure
Central → Peripheral

Primary disturbances (uncompensated)


(lower extremities)
blood volume shift ↓ Blood volume

Capillary filtration
(lower extremities)
↓ Central venous pressure

Edema
↓ Stroke volume (lower extremities)

↓ Cardiac output
↓ Pulse
↓ Mean arterial pressure ↓ Firing rate of
pressure cardiopulmonary
baroreceptors

↓ Firing rate of
arterial baroreceptors
(increases set point)

Medullary cardiovascular centers


Compensatory responses

↓ Parasympathetic ↑ Sympathetic
activity activity

Heart ↑ Venous ↑ Arteriolar ↑ Fluid


FIGURE 30–2 Cardiovascular mechanisms ↑ ↑ Contractility
rate constriction constriction retention
involved when changing from a recumbent to
Heart Systemic organs Kidney
a standing position. (Modified with permission from
Mohrman DE, Heller LJ: Cardiovascular Physiology, 6th ed.
New York: Lange Medical Books/McGraw-Hill, 2006.) Immediate Long-term

assuming a recumbent position is a shift of fluid from the lower erance may take several days or even weeks. Efforts made to
extremities to the upper portions of the body. The consequences diminish the cardiovascular changes for the bedridden patient
of this shift include distention of the head and neck veins, facial may include intermittent sitting up or tilting the bed to lower
edema, nasal stuffiness, and decreases in calf girth and leg vol- the legs and trigger fluid retention mechanisms.
ume. In addition, the increase in central blood volume stimu-
lates the cardiopulmonary mechanoreceptors, which influence
renal function by neural and hormonal pathways to reduce
sympathetic drive and promote fluid loss. The individual begins
NORMAL CARDIOVASCULAR
to lose weight and, within just a few days, becomes hypo- ADAPTATIONS
volemic. When the bedridden patient initially tries to stand up,
the normal responses to gravity as described in Figure 30–2 are RESPONSES TO
not as effective, primarily because of the substantial decrease in RESPIRATORY ACTIVITY
circulating blood volume. Upon standing, blood shifts out of
the central venous pool into the peripheral veins, stroke vol- The physical processes associated with inhaling air into and
ume decreases, and the individual often becomes dizzy and exhaling air out of the lungs have major effects on venous return
may faint due to a dramatic decrease in blood pressure. This and cardiac output. The decrease in intrathoracic pressure with
phenomenon is referred to as orthostatic or postural hypoten- inspiration transiently increases the pressure gradient between
sion. Because there are other cardiovascular changes that may the peripheral and central venous pools and contributes to
accompany bed rest, complete reversal of this orthostatic intol- venous return from the systemic vascular beds to the right side
CHAPTER 30 Cardiovascular Responses to Physiological Stress 299

of the heart. Because the one-way valves in peripheral veins pre- uterus and developing placenta, but also the kidneys and
vent backflow during expiration, the thoracic pressure changes the gastrointestinal organs. However, one of the most strik-
constitute a respiratory pump (also called the thoracoabdomi- ing cardiovascular changes of pregnancy is the nearly
nal pump). Details of the effects of the respiratory pump on 50% increase in circulating blood volume. The placenta,
venous return in exercise and the effects of positive pressure ven- being a low-resistance organ added in parallel with the other
tilation on venous return and cardiac output are presented in systemic organs, reduces the overall systemic total peripheral
Chapters 34 and 72. A variety of complex signals from cardiopul- resistance by about 40%. Without the substantial increase in
monary receptors during the respiratory cycle contribute to a circulating blood volume to support cardiac filling, the nec-
respiratory-based cardiac arrhythmia that is mediated primarily essary increase in cardiac output to balance the decrease in
by changing activity of vagal efferents to the SA node. total peripheral resistance would not be possible and preg-
There are other instances when the cardiovascular effect of nancy would result in a substantial decrease in mean arterial
respiratory efforts has physiological consequences (i.e., yawn- pressure. At birth, the loss of the placenta contributes to an
ing, coughing, laughing). One of the more important situations abrupt increase of maternal total peripheral resistance back
occurs during the Valsalva maneuver, which is a forced expi- toward normal levels.
ration against a closed glottis. This maneuver is normally per-
formed by individuals during defecation (“straining at stool”),
or when attempting to lift a heavy object. The sustained increase FETAL CIRCULATION &
in intrathoracic pressure leads to decreases in venous return CHANGES AT BIRTH
and blood pressure, which evokes a compensatory reflex
increase in heart rate and peripheral vasoconstriction. (During During fetal development, the exchange of nutrients, gases,
this period, the red face and distended peripheral veins are and waste products between fetal and maternal blood occurs
indicative of high peripheral venous pressures.) At the cessa- in the placenta. This exchange occurs by diffusion between
tion of the maneuver, there is an abrupt decrease in pressure for separate fetal and maternal capillaries without any direct con-
a few heart beats due to the reduction of intrathoracic pressure. nection between the fetal and maternal circulations. From a
Venous blood then moves rapidly into the central venous pool; hemodynamic standpoint, the placenta represents a tempo-
stroke volume, cardiac output, and arterial pressure increase rary additional large systemic organ for both the fetus and the
rapidly; and a reflex bradycardia occurs. The combination of an mother. The fetal component of the placenta has a low vascu-
episode of high peripheral venous pressure followed by an epi- lar resistance and receives a substantial portion of the fetal car-
sode of high arterial pressure and pulse pressure is particularly diac output.
dangerous for people who are candidates for cerebral vascular Blood circulation in the developing fetus almost completely
accidents (strokes) because this combination may rupture a bypasses the collapsed, fluid-filled fetal lungs. Very little blood
weakened blood vessel. flows into the pulmonary artery because the vascular resis-
tance in the collapsed fetal lungs is very high. By the special
structural arrangements shown in Figure 30–3, the fetal right
CARDIOVASCULAR CHANGES and left hearts operate in parallel to pump blood through the
DURING PREGNANCY systemic organs and the placenta. As shown in Figure 30–3A,
fetal blood returning from the systemic organs and placenta
Pregnancy causes alterations in vascular structure and blood fills both the left and right hearts together because of an open-
flow to many maternal organs in order to support growth of ing in the intra-atrial septum called the foramen ovale. As
the developing fetus. These organs include not only the indicated in Figure 30–3B, most of the blood that is pumped

A B Ductus
arteriosus
Foramen
ovale
a
pv

pv
pa

vc la
FIGURE 30–3 Fetal circulation (A) during cardiac
ra
filling and (B) during cardiac ejection. pv, pulmonary
veins; la, left atrium; lv, left ventricle; rv, right ventricle;
ra, right atrium; vc, venae cavae; a, aorta; pa, pulmonary
lv rv artery. (Modified with permission from Mohrman DE, Heller
rv lv
LJ: Cardiovascular Physiology, 6th ed. New York: Lange Medical Books/
McGraw-Hill, 2006.)
300 SECTION V Cardiovascular Physiology

by the fetal right heart does not enter the constricted pulmo- cal axis orientation) that converts to the normal left ventricu-
nary circulation but rather is diverted into the aorta through a lar dominance during childhood.
vascular connection between the pulmonary artery and the Heart murmurs are also quite common in childhood and
aorta called the ductus arteriosus. have been reported to be present in as many as 50% of healthy
At birth, an abrupt decrease in pulmonary vascular resis- children. Most of these murmurs fall in the category of “inno-
tance occurs in the newborn with the onset of lung ventilation. cent” murmurs resulting from normal cardiac tissue vibra-
This is a partly a result of the introduction of oxygen into the tions, high flow through valves, and thin chest walls that make
airways and the decrease in hypoxic pulmonary vasoconstric- noises from the vasculature easy to hear. Less than 1% of them
tion as discussed in Chapter 34. This permits more blood result from various congenital heart defects.
to flow into the lungs from the pulmonary artery and tends to Growth and development of the vascular system parallels
lower pulmonary arterial pressure. Meanwhile, total systemic growth and development of the body with most of the local
vascular resistance of the newborn increases greatly because of and reflex regulatory mechanisms becoming operational
the interruption of flow through the placenta. This causes an shortly after birth.
increase in aortic pressure. The combination of the reduced
pulmonary and elevated systemic arterial pressure retards or
even reverses the flow through the ductus arteriosus. Through CARDIOVASCULAR CHANGES
mechanisms that are incompletely understood but clearly WITH NORMAL AGING
linked to an increase in blood oxygen tension, the ductus arte-
riosus gradually constricts and completely closes in a few hours Changes in cardiovascular function occur over the normal
to a few days. The circulatory changes that occur at birth tend human lifetime. These changes are generally associated with a
to simultaneously increase the pressure afterload on the left slowing of some of the basic processes and a reduction in the
ventricle and decrease that on the right ventricle. This indi- ability of the cardiovascular system as a whole to respond to
rectly causes left atrial pressure to increase above that in the various stresses. Details of the aging process are discussed in
right atrium so that the pressure gradient for flow through the Chapter 73.
foramen ovale is reversed. Reverse flow through the foramen Age-dependent cardiac alterations include: (1) a decrease in
ovale is, however, prevented by a flaplike valve that covers the the resting and maximum cardiac index, (2) a decrease in the
opening in the left atrium. Normally, the foramen ovale even- maximum heart rate, (3) an increase in the contraction and
tually is closed permanently by the growth of fibrous tissue. relaxation times of the heart muscle, (4) an increase in the myo-
cardial stiffness during diastole, (5) a decrease in number of
functioning myocytes, and (6) an accumulation of pigment in
PEDIATRIC CARDIOVASCULAR the myocardial cells. Age-dependent vascular changes include:
CHARACTERISTICS (1) a decrease in capillary density in some tissues, (2) a decrease
in arterial and venous compliance, and (3) an increase in total
Cardiovascular variables change significantly during the child- peripheral vascular resistance. These changes combine to pro-
hood. The normal neonate has, by adult standards, a high rest- duce the age-dependent increases in arterial pulse pressure
ing heart rate (average of 140 beats/min) and a low arterial and mean arterial pressure that were discussed in Chapter 26
blood pressure (average of 60/35 mm Hg). These values rap- (see Figure 26–10). The increases in arterial pressure impose a
idly change over the first year (to 120 beats/min and 100/65 greater afterload on the heart, and this may be partially respon-
mm Hg, respectively). By the time the child enters adoles- sible for the age-dependent decreases in cardiac index.
cence, these values are near adult levels. Arterial baroreceptor–induced responses to changes in
Pulmonary vascular resistance decreases precipitously at blood pressure are blunted with age. This is due in part to a
birth as described above and then continues to decline during decrease in afferent activity from the arterial baroreceptors
the first year, at which time pulmonary vascular pressures because of the age-dependent increase in arterial rigidity. In
resemble adult levels. These resistance changes appear to be due addition, the total amount of norepinephrine contained in
to a progressive remodeling of the microvascular arterioles from the sympathetic nerve endings of the myocardium decreases
thick-walled, small-diameter vessels to thin-walled, large-diam- with age, and the myocardial responsiveness to catecholamines
eter microvessels. Also as the lung grows, the number of alveoli declines. Thus, the efferent component of the reflex is also
and therefore pulmonary vessels increases. (At birth there are compromised. These changes may partially account for the
only about 20 million alveoli, compared to 300–450 million in apparent age-dependent sluggishness in the responses to pos-
adults. Most of the growth occurs in the first 8 years). tural changes and recovery from exercise.
It is noteworthy that distinct differences between right and
left ventricular mass and wall thickness develop only after EFFECT OF GENDER
birth. Presumably these differences arise because of a hyper-
trophic response of the left ventricle to the increased afterload There are a few well-documented gender-dependent differ-
it must assume at birth. Accordingly, the electrocardiogram of ences in the cardiovascular system. Compared with age-matched
children shows an early right ventricular dominance (electri- men, premenopausal women have a lower left ventricular mass
CHAPTER 30 Cardiovascular Responses to Physiological Stress 301

to body mass ratio, which may reflect a lower cardiac afterload


in women. This may result from their lower arterial blood pres- blood sample was obtained and his hematocrit was 34
sure, greater aortic compliance, and improved ability to induce (normal 41–53%). The most immediate problem was
vasodilatory mechanisms (such as endothelial-dependent flow- determined to be hemorrhagic hypovolemic shock and he
mediated vasodilation). These differences are thought to be was given a liter of blood. Within an hour, his heart rate
related to protective effects of estrogen and may account for the was 90 beats/min and his blood pressure was 115/85 mm
lowered risk of premenopausal women for developing cardio- Hg. He was still weak and pale but more alert. He had not
vascular disease. After menopause, these gender differences yet urinated and was very thirsty. Additional blood was
disappear. In fact, older women with ischemic heart disease infused and his vital signs returned to normal.
often have a worse prognosis than men. Circulatory shock exists whenever there is a severe reduc-
There are also gender-dependent differences in cardiac elec- tion in blood supply to the body tissues and the metabolic
trical properties. Women often have lower intrinsic heart rates needs of the tissues are not met. Even with all cardiovascular
and longer QT intervals than do men. They are at greater risk compensatory mechanisms activated, arterial pressure is
for developing long-QT syndrome and torsades de pointes. usually (though not always) low in shock. The shock state is
They are also twice as likely as men to have atrioventricular precipitated by one of the following two conditions:
nodal reentry tachycardias. (1) severely depressed myocardial function or (2) grossly
inadequate cardiac filling due to low mean circulatory fill-
ing pressure. The former situation is called cardiogenic
shock and the latter situation can result from a variety of
non-cardiac causes. These shock states are described in
CLINICAL CORRELATION Table 30–1 along with the primary disturbance on the car-
An elderly man was hunting with some friends when he diovascular system and the consequences on cardiac filling
inadvertently shot himself in the foot. This resulted in a pressure. The common primary disturbances in all forms of
significant loss of blood and, by the time he was brought shock lead to decreased mean arterial pressure and decreased
to the hospital, he was very weak and pale, his skin was arterial baroreceptor discharge rate. In the case of hypo-
cold and clammy, and he was somewhat confused. His volemic, anaphylactic, and septic shock, diminished activ-
heart rate was 105 beats/min, and blood pressure was ity of the cardiopulmonary baroreceptors due to a decrease
85/65 mm Hg. His breathing was rapid and shallow and in central venous pressure and/or volume acts on the med-
jugular venous pulses could not be observed when he was ullary cardiovascular centers to stimulate sympathetic out-
recumbent. An intravenous catheter advanced from a put. In the case of cardiogenic shock, central venous pressure
peripheral vein into his right atrium recorded central will increase, and in the case of neurogenic shock, central
venous pressure to be 0 mm Hg (normal 2–6 mm Hg). A venous pressure cannot be predicted because both cardiac

TABLE 30–1 Circulatory Shock


Effect on Central
Type of Shock Causes Primary Disturbance Venous Pressure

Cardiogenic -Myocardial infarction -Decreased cardiac output -Increased CVP


-Severe arrhythmia -Right shift of cardiac
-Abrupt valve malfunction function curve

Hypovolemic -Hemorrhage -Decreased circulating -Decreased CVP


-Severe burns blood volume
-Chronic vomiting or diarrhea
-Dehydration

Anaphylactic -Severe systemic allergic reaction associated -Decreased total peripheral -Decreased CVP
with release of histamine, prostaglandins, resistance, reduced venous tone
leukotrienes, bradykinin

Septic -Circulating infective agents releasing -Decreased total peripheral -Decreased CVP
vasodilator substances such as endotoxin resistance, reduced venous tone
(lipopolysaccharide) inducing
NO synthesis
Neurogenic -Reduced sympathetic and/or increased -Decreased cardiac output, -Variable effect on CVP because
parasympathetic activity (vasovagal total peripheral resistance, both cardiac output and venous
syncope) and venous tone return will decrease
302 SECTION V Cardiovascular Physiology

output and venous return are likely to be depressed. If arte- tissues (other than the heart and brain) despite nearly
rial pressure decreases below the autoregulatory range for normal arterial pressure. Intense sympathetic activation
cerebral blood flow (below about 60 mm Hg), perfusion of can lead to renal, splanchnic, or hepatic ischemic damage.
the brain begins to decrease, eliciting the cerebral ischemic If this ischemia is prolonged, self-reinforcing decompen-
response that causes the most intense of all signals to acti- satory mechanisms (positive feedback described in
vate sympathetic nerves. Chapter 1) will progressively drive arterial pressure down
Unless the primary disturbance precludes these com- and unless corrective measures are taken quickly, death
pensatory responses, the increase in sympathetic activity will ultimately result.
(and decrease in parasympathetic activity) will lead to an
increase in cardiac output (by increasing heart rate and
cardiac contractility), an increase in total peripheral resis-
tance (by generalized arteriolar constriction), and an CHAPTER SUMMARY
increase in peripheral venous tone (which will shift blood ■ Cardiovascular responses to physiological stresses should be
into the central venous pool). Many of the commonly rec- evaluated in terms of the initial effects of the primary distur-
ognized symptoms of shock (e.g., pallor, cold clammy bance and the subsequent effects of the reflex compensatory
skin, rapid heart rate, muscle weakness, venous constric- responses.
tion) are a result of greatly increased sympathetic nerve ■ Gravity, and hence body position, has a significant effect on
activity. When the immediate compensatory processes the cardiovascular system, and various reflex compensatory
are inadequate, the individual may also show signs of mechanisms are required to overcome venous pooling that
abnormally low arterial pressure, such as dizziness, con- accompanies the upright position.
fusion, or loss of consciousness. ■ Long-term bed rest causes decreases in circulating blood
Additional compensatory processes during a shock volume that contributes to orthostatic hypotension.
state may include (1) rapid, shallow breathing, which pro- ■ Cardiovascular characteristics are influenced by a variety of
motes venous return to the heart by action of the respira- conditions including respiratory activity, gender, pregnancy,
growth and development from the fetal period, through birth,
tory pump (see Chapter 72), (2) release of various
pediatric stages, adulthood, and old age.
powerful vasoconstrictor hormones such as epinephrine
(see Chapter 19), angiotensin II, and vasopressin
(see Chapter 45), which contribute to the increase in total
peripheral resistance, (3) a net shift of fluid from the
STUDY QUESTIONS
interstitial space into the vascular space due to the very 1. All of the following tend to occur when a person lies down.
low capillary hydrostatic pressure downstream of the Which one is the primary disturbance that causes all the others to
vasoconstricted arterioles, and (4) an increase in extracel- happen?
lular osmolarity (as a result of increased glycogenolysis in A) Heart rate will decrease.
the liver induced by epinephrine and norepinephrine) B) Stroke volume will decrease.
that will induce a shift of fluid from the intracellular space C) Sympathetic activity will decrease.
into the extracellular (including intravascular) space. The D) Parasympathetic activity will increase.
E) Central venous pressure will increase.
latter two processes result in a sort of autotransfusion
that can move as much as a liter of fluid into the vascular 2. A 35-year-old man has had a severe bout of the flu with vomiting
and diarrhea for several days. All of the following conditions
space in the first hour after the onset of the shock episode
would be expected to be present except
(see Chapter 26). This fluid shift accounts for the reduc-
A) orthostatic hypotension.
tion in hematocrit that is commonly observed in hemor-
B) increased cardiac preload.
rhagic shock. C) increased cardiac ejection fraction.
In addition to the immediate compensatory responses D) increased hematocrit.
described above, fluid retention mechanisms are evoked E) increased total peripheral vascular resistance.
that promote renal retention of fluid and an increase in 3. Total systemic peripheral vascular resistance of a newborn
circulating blood volume. These processes are described baby undergoes an abrupt and sustained increase at birth.
in detail in Chapter 45 and contribute to the replenish- This is because
ment of extracellular fluid volume within a few days of the A) circulating blood volume increases.
shock episode. B) the high-resistance lungs inflate.
If the primary disturbances are not corrected soon, the C) the low-resistance placental circulation is removed.
strong compensatory responses can reduce perfusion of D) sympathetic neural stimulation is increased.
E) cardiac output increases.
CHAPTER 30 Cardiovascular Responses to Physiological Stress 303

4. Vertical immersion to the chest in tepid water produces a diuresis 5. All of the following help maintain circulation during states
in many individuals. What mechanisms might account for this of hypovolemic shock except
phenomenon? A) an increase in heart rate.
A) an increase in sympathetic activity to the kidney B) rapid respiratory effort to promote venous return of blood
B) an increase in mean arterial pressure to the heart.
C) a shift of blood from the central to the peripheral C) vasoconstrictive contributions from increases in circulating
venous pool epinephrine.
D) decreased firing of arterial baroreceptors D) autotransfusion of interstitial fluid into capillary beds.
E) increased firing of the cardiopulmonary baroreceptors E) transcapillary filtration of plasma into interstitial space.
This page intentionally left blank
SECTION VI PULMONARY PHYSIOLOGY

31
C H A P T E R

Function and Structure


of the Respiratory System
Michael Levitzky

O B J E C T I V E S

■ Describe the exchange of oxygen and carbon dioxide with the atmosphere
and relate gas exchange to the metabolism of the tissues of the body.
■ List the functions of the lungs.
■ Describe functions and structures of the conducting airways,
the alveolar–capillary unit, and the chest wall.
■ Describe the central nervous system initiation of breathing and the
innervation of the respiratory muscles.

The main functions of the respiratory system are to obtain GAS EXCHANGE
oxygen from the external environment and supply it to the
cells, and to remove from the body the carbon dioxide pro- The exchange of carbon dioxide for oxygen takes place in the
duced by cellular metabolism. lungs. As shown in Figure 31–1, fresh air, containing oxygen,
The respiratory system is composed of the lungs, the is inspired into the lungs through the conducting airways.
conducting airways, the parts of the central nervous system The forces needed to cause the air to flow are generated by the
concerned with the control of the muscles of respiration, and respiratory muscles, acting on commands initiated by the
the chest wall. The chest wall consists of the muscles of central nervous system. At the same time, venous blood
respiration— the diaphragm, the intercostal muscles, and returning from the various body tissues is pumped into the
the abdominal muscles—and the rib cage. lungs by the right ventricle of the heart. This mixed venous
blood has a high carbon dioxide content and a low oxygen
content. In the pulmonary capillaries, carbon dioxide is
FUNCTIONS OF THE exchanged for oxygen. The blood leaving the lungs, which
RESPIRATORY SYSTEM now has a high oxygen content and a lower carbon dioxide
content, is distributed to the tissues of the body by the left side
The functions of the respiratory system include gas exchange, of the heart. During expiration, gas with a high concentration
acid–base balance, phonation, pulmonary defense and metab- of carbon dioxide is expelled from the body.
olism, and the handling of bioactive materials.

305

Ch31_305-312.indd 305 11/26/10 11:50:59 AM


306 SECTION VI Pulmonary Physiology

EXTERNAL ENVIRONMENT
High O2, Low CO2

O2

Inspiration
Expiration
CONDUCTING AIRWAYS

CO2

ALVEOLI
O2

CO2
PULMONARY
PULMONARY CAPILLARIES PULMONARY
ARTERY VEINS
Low O2 High O2
High CO2 Low CO2

LEFT
RIGHT ATRIUM
VENTRICLE
LEFT
RIGHT VENTRICLE
ATRIUM

Low O2 High O2
High CO2 Low CO2
VEINS AORTA

SYSTEMIC
CAPILLARIES
O2

CO2

METABOLIZING TISSUES

FIGURE 31–1 Schematic representation of gas exchange between the tissues of the body and the environment. (Modified with
permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

OTHER FUNCTIONS OF THE Phonation


RESPIRATORY SYSTEM Phonation is the production of sounds by the movement of air
through the vocal cords. Speech, singing, and other sounds are
Acid–Base Balance produced by the actions of the central nervous system control-
In the body, increases in carbon dioxide lead to increases in lers on the muscles of respiration, causing air to flow through
hydrogen ion concentration (and vice versa) because of the the vocal cords and the mouth. The physiology of speech is
following reaction: discussed in Chapter 21.

CO2 + H2O H2CO3 H+ + HCO3–


The respiratory system can therefore participate in acid– Pulmonary Defense Mechanisms
base balance by removing CO2 from the body. The central Each breath brings into the lungs a small sample of the local
nervous system has sensors for the CO2 and the hydrogen ion atmospheric environment. This may include microorganisms
levels in the arterial blood and in the cerebrospinal fluid that such as bacteria, dust, particles of silica or asbestos, toxic gases,
send information to the controllers of breathing. Acid–base smoke (cigarette and other types), and other pollutants. In
balance is discussed in greater detail in Chapter 37; the control addition, the temperature and humidity of the local atmo-
of breathing is discussed in Chapter 38. sphere can vary tremendously. As will be described below, as
CHAPTER 31 Function and Structure of the Respiratory System 307

air passes through the airways, it is warmed to body temper- ple, prostaglandins E1, E2, and F2α are nearly completely
ature and filtered to remove particulate matter. Most of the removed in a single pass through the lungs. On the other
particles in inspired air are removed before they reach the hand, prostaglandins A1, A2, and I2 (prostacyclin) are not
alveoli. The mechanisms by which these impurities are affected by the pulmonary circulation. Similarly, about 30% of
removed from the respiratory tract are described in the sec- the norepinephrine in mixed venous blood is removed by the
tion “Structure of the Respiratory System.” lung, but epinephrine is unaffected. It appears that some sub-
stances released into specific vascular beds for local effects are
Pulmonary Metabolism and inactivated or removed as they pass through the lungs, pre-
the Handling of Bioactive Materials venting them from entering the systemic circulation; other
The cells of the lung must metabolize substrates to supply substances, apparently intended for more general effects, are
energy and nutrients for their own maintenance. Some special- not affected.
ized pulmonary cells also produce substances necessary for
normal pulmonary function. These substances include pulmo-
nary surfactant, which is synthesized in type II alveolar epi-
STRUCTURE OF THE
thelial cells (described below) and released at the alveolar RESPIRATORY SYSTEM
surface. Surfactant plays an important role in reducing the
alveolar elastic recoil due to surface tension and in stabilizing THE AIRWAYS
the alveoli, as discussed later in Chapter 32. Histamine, lyso-
somal enzymes, prostaglandins, leukotrienes, platelet- Air enters the respiratory system through the nose or mouth.
activating factor, neutrophil and eosinophil chemotactic Air entering through the nose is filtered, warmed to body tem-
factors, and serotonin can be released from mast cells in the perature, and humidified as it passes through the nose and
lungs in response to conditions such as pulmonary embolism nasal turbinates. The upper airways (airways above the
(see Chapter 34) and anaphylaxis (an acute life-threatening trachea) are shown in Figure 31–2. The mucosa of the nose,
systemic allergic reaction). These substances may cause bron- the nasal turbinates, the oropharynx, and the nasopharynx
choconstriction or immune or inflammatory responses, or they have a rich blood supply and constitute a large surface area.
may initiate cardiopulmonary reflexes. Many substances are The nasal turbinates alone have a surface area said to be about
also produced by cells of the lung and released into the alveoli 160 cm2. As inspired air passes through these areas and con-
and airways, including mucus and other tracheobronchial tinues through the tracheobronchial tree, it is warmed to
secretions; surface enzymes, proteins, and other factors; and body temperature and humidified. This protective function is
immunologically active substances. These substances are pro- more effective if one is breathing through the nose than
duced by goblet cells, submucosal gland cells, Clara cells, and through the mouth.
macrophages. Substances produced by lung cells and released Because the olfactory receptors are located in the posterior
into the blood under various circumstances include bradyki- nasal cavity rather than in the trachea or alveoli, a person can
nin, histamine, serotonin, heparin, prostaglandins E2 and sniff to attempt to detect potentially hazardous gases or dan-
F2α, and the endoperoxides (prostaglandins G2 and H2). gerous material in the inspired air. This rapid, shallow inspira-
In addition, the pulmonary capillary endothelium contains tion brings gases into contact with the olfactory sensors
a great number of enzymes that can produce, metabolize, or without bringing them into the lung. Chapter 17 discusses the
modify naturally occurring vasoactive substances. For exam- physiology of olfaction (the sense of smell).





  


   

 
FIGURE 31–2 Schematic drawing of the
 

upper airways. (Reproduced with permission from Proctor
 
 
DF. Physiology of the upper airway. In: Fenn WO, Rahn H, eds.
 
Handbook of Physiology, sec 3: Respiration. Vol. 1. Washington,
 DC: American Physiological Society; 1964:309–345.)
308 SECTION VI Pulmonary Physiology

Number
of the distal portions of the tracheobronchial tree, even though
Name of branches of tubes the diameters of the individual airways are quite small.
in branch

Trachea 1
Structure of the Airways
The structure of the airways varies considerably, depending on
Bronchi 2
their location in the tracheobronchial tree. The trachea is a
Conducting zone

fibromuscular tube supported ventrolaterally by C-shaped car-


4 tilage and completed dorsally by smooth muscle. The cartilage
of the large bronchi is semicircular, like that of the trachea, but
8 as the bronchi enter the lungs, the cartilage rings disappear
Bronchioles 16 and are replaced by irregularly shaped cartilage plates. They
32 completely surround the bronchi and give the intrapulmonary
bronchi their cylindrical shape. These plates, which help sup-
Terminal bronchioles port the larger airways, diminish progressively in the distal
6 x 104 airways and disappear in airways about 1 mm in diameter. Air-
ways with no cartilage are termed bronchioles. Because the
Respiratory bronchioles bronchioles and alveolar ducts contain no cartilage support,
they are subject to collapse when compressed, as will be dis-
Respiratory zone

5 x 105 cussed later in this chapter. This tendency is partly opposed by


the attachment of the alveolar septa, containing elastic tissue,
Alveolar ducts to their walls, as seen in Figure 31–4, a scanning electron
micrograph of the alveolar–capillary surface (also shown sche-
matically in Figure 32–18). As the cartilage plates become
Alveolar sacs 8 x 106 irregularly distributed around distal airways, the muscular
layer completely surrounds these airways. The muscular layer
is intermingled with elastic fibers. As the bronchioles proceed
FIGURE 31–3 Schematic representation of airway branching
in the human lung. (Reproduced with permission from Weibel ER. toward the alveoli, the muscle layer becomes thinner, although
Morphometry of the Human Lung. Berlin; Springer; 1963.) smooth muscle can even be found in the walls of the alveolar
ducts. The outermost layer of the bronchiolar wall is sur-
rounded by dense connective tissue with many elastic fibers.
The entire respiratory tract, except for part of the pharynx, the
Air then passes through the glottis and the larynx and enters anterior third of the nose, and the respiratory units distal to the
the tracheobronchial tree. After passing through the conduct- terminal bronchioles, is lined with ciliated cells interspersed with
ing airways, the inspired air enters the alveoli, where it comes mucus-secreting goblet cells and other secretory cells. In the
into contact with the mixed venous blood in the pulmonary bronchioles, the goblet cells become less frequent and are
capillaries. replaced by another type of secretory cell, the Clara cell. The
Starting with the trachea, the air may pass through as few as ciliated epithelium, along with mucus secreted by glands along
10 or as many as 23 generations, or branchings, on its way to the airways and the goblet cells and the secretory products of the
the alveoli. The branchings of the tracheobronchial tree and its Clara cells, constitutes an important mechanism for the protec-
nomenclature are shown in Figure 31–3. Alveolar gas exchange tion of the lung called the mucociliary escalator.
units are denoted by the U-shaped sacs.
The first 16 generations of airways, the conducting zone,
contain no alveoli and thus are anatomically incapable of
Filtration and Removal of Inspired
gas exchange with the venous blood. They constitute the Particles by the Airways
anatomic dead space, which is discussed in Chapter 33. Filtration of Inspired Air Air passing through the nose is
Alveoli start to appear at the 17th to the 19th generations, in first filtered by passing through the nasal hairs, or vibrissae. This
the respiratory bronchioles, which constitute the transi- removes most particles larger than 10–15 μm in diameter. Most of
tional zone. The 20th to 22nd generations are lined with the particles greater than 10 μm in diameter are removed on im-
alveoli. These alveolar ducts and the alveolar sacs, which pact in the large surface area of the nasal septum and turbinates
terminate the tracheobronchial tree, are referred to as the (Figure 31–2). The inspired air stream changes direction abruptly
respiratory zone. at the nasopharynx so that many of these larger particles impact
The portion of the lung supplied by a primary respiratory the posterior wall of the pharynx. The tonsils and adenoids are
bronchiole is called an acinus. All of the airways of an acinus located near this impaction site, providing immunologic defense
participate in gas exchange. The numerous branchings of against biologically active material filtered at this point. Air enter-
the airways result in a tremendous total cross-sectional area ing the trachea contains few particles larger than 10 μm, and most
CHAPTER 31 Function and Structure of the Respiratory System 309

stimulation of receptors in the trachea. In either case, a deep in-


spiration is followed by a forced expiration against a closed glot-
tis. Pressure in the chest surrounding the lungs (intrapleural
pressure) may rise to more than 100 mm Hg during this phase
of the reflex. The glottis opens suddenly, and pressure in the air-
ways decreases rapidly, resulting in compression of the airways
and an explosive expiration, with linear airflow velocities said to
approach the speed of sound. Such high airflow rates through
the narrowed airways are likely to carry the irritant, along with
some mucus, out of the respiratory tract. In a sneeze, the expira-
tion is via the nose; in a cough, the expiration is via the mouth.
The cough or sneeze reflex is also useful in helping to move the
mucous lining of the airways toward the nose or mouth. The
term “cough” is not specific to this complete involuntary respi-
ratory reflex. Coughs can be initiated by many causes, including
postnasal drip from allergies or viral infections, asthma, gas-
troesophageal reflux, as an adverse effect of the very commonly
prescribed angiotensin-converting enzyme inhibitors, mucus
production from chronic bronchitis, infections, and other air-
way disorders. Voluntary coughs are not usually as pronounced
as the violent involuntary reflex described above.
Particles that are trapped in the mucus lining the airways
can be removed by the mucociliary escalator, which has an
estimated total surface area of 0.5 m2. The mucus is a complex
polymer of mucopolysaccharides. The mucous glands are
found mainly in the submucosa near the supporting cartilage
of the larger airways. In pathologic states, such as chronic
bronchitis, the number of goblet cells may increase and the
mucous glands may hypertrophy, resulting in greatly increased
FIGURE 31–4 Scanning electron micrograph of human lung
mucous gland secretion and increased viscosity of mucus.
parenchyma. A, alveolus; S, alveolar septa; D, alveolar duct; PK, pore
of Kohn; PA, small branch of the pulmonary artery. (Reproduced with The cilia lining the airways beat in such a way that the mucus
permission from Fishman AP, Elias JA: Fishman’s Pulmonary Diseases and Disorders, covering them is always moved up the airway, away from the
3rd ed. New York: McGraw-Hill, Health Professions Division, 1998.) alveoli and toward the pharynx. The mucous blanket appears
to be involved in the mechanical linkage between the cilia. The
cilia beat at frequencies between 600 and 900 beats/min, and
of these will impact mainly at the carina or within the bronchi. the mucus moves progressively faster as it travels from the
Sedimentation of most particles in the size range of 2–5 μm periphery. Several studies have shown that ciliary function is
occurs by gravity in the smaller airways, where airflow rates are inhibited or impaired by cigarette smoke.
extremely low. Thus, most of the particles between 2 and 10 μm The mucociliary escalator is an especially important mecha-
in diameter are removed by impaction or sedimentation and nism for the removal of inhaled particles that come to rest in the
become trapped in the mucus that lines the upper airways, airways. Material trapped in the mucus is continuously moved
trachea, bronchi, and bronchioles. Smaller particles and all upward toward the pharynx. This movement can be greatly
foreign gases reach the alveolar ducts and alveoli. Some smaller increased during a cough, as described previously. Mucus that
particles (0.1 μm and smaller) are deposited as a result of Brown- reaches the pharynx is usually swallowed, expectorated, or
ian motion due to their bombardment by gas molecules. The removed by blowing one’s nose. Patients who cannot clear their
other particles, between 0.1 and 0.5 μm in diameter, mainly stay tracheobronchial secretions (e.g., an intubated patient or a
suspended as aerosols, and about 80% of them are exhaled. patient who cannot cough adequately) continue to produce
secretions. If the secretions are not removed from the patient by
Removal of Filtered Material Filtered or aspirated mate- suction or other means, airway obstruction may develop.
rial trapped in the mucus that lines the respiratory tract can be
removed in several ways. Mechanical or chemical stimulation
of receptors in the nose, trachea, larynx, or elsewhere in the re- THE ALVEOLAR–CAPILLARY UNIT
spiratory tract may produce bronchoconstriction to prevent
deeper penetration of the irritant into the airways and may also The alveolar–capillary unit is the site of gas exchange in the
produce a cough or a sneeze. A sneeze results from stimulation lung. The alveoli, traditionally estimated to number about 300
of receptors in the nose or nasopharynx; a cough results from million in the adult (a more recent study calculated the mean
310 SECTION VI Pulmonary Physiology

the lung to injury. As type I alveolar epithelial cells are injured,


type II cells proliferate to reestablish a continuous epithelial
surface. Studies in animals have shown that these type II cells
can develop into type I cells after injury.
A cross-section of a pulmonary capillary is shown in the
transmission electron micrograph in Figure 31–6. An eryth-
rocyte is seen in cross-section in the lumen of the capillary.
Capillaries are formed by a single layer of squamous epithelial
cells that are aligned to form tubes. The nucleus of one of the
capillary endothelial cells can be seen in the micrograph.
The barrier to gas exchange between the alveoli and pulmonary
capillaries can also be seen in the figure. It consists of the alveolar
epithelium, the capillary endothelium, and the interstitial space
between them. Gases must also pass through the fluid lining the
alveolar surface (not visible in Figure 32–6) and the plasma in the
capillary. The barrier to diffusion is normally 0.2–0.5 μm thick.
Gas exchange by diffusion is discussed in Chapter 35.

FIGURE 31–5 Scanning electron micrograph of the surface and


cross-section of an alveolar septum. Capillaries (C) are seen sectioned
in the foreground, with erythrocytes (EC) within them. A, alveolus; D,
alveolar duct; PK, pore of Kohn; AR, alveolar entrance to duct; *,
connective tissue fibers. The encircled asterisk is at a junction of three
septa. (Reproduced with permission from Fishman AP, Elias JA: Fishman’s Pulmonary
Diseases and Disorders, 3rd ed. New York: McGraw-Hill, Health Professions Division, 1998.)

number of alveoli to be 480 million), are almost completely


enveloped in pulmonary capillaries. There may be as many as
280 billion pulmonary capillaries, or approximately 500–1,000
pulmonary capillaries per alveolus. The result of these stagger-
ing numbers of alveoli and pulmonary capillaries is a vast area
of contact between alveoli and pulmonary capillaries—
probably 50–100 m2 of surface area available for gas exchange
by diffusion. The alveoli are about 200–250 μm in diameter.
Figure 31–5 shows an even greater magnification of the site
of gas exchange than that shown in Figure 31–4.
The alveolar septum appears to be almost entirely composed
of pulmonary capillaries. Red blood cells (erythrocytes) can be
seen inside the capillaries at the point of section. Elastic and con-
nective tissue fibers, not visible in the figure, are found between
the capillaries in the alveolar septa. Also shown in these figures
are the pores of Kohn that are interalveolar communications.
The alveolar surface is mainly composed of a single thin
layer of squamous epithelial cells, the type I alveolar cells.
Interspersed among these are the larger cuboidal type II alveo-
lar cells that produce the fluid layer that lines the alveoli.
Although there are about twice as many type II cells as there
are type I cells in the human lung, type I cells cover 90–95% of
the alveolar surface, because the average type I cell has a much FIGURE 31–6 Transmission electron micrograph of a cross-
section of a pulmonary capillary. An erythrocyte (EC) is seen within the
larger surface area than the average type II cell does. A third
capillary. C, capillary; EN, capillary endothelial cell (note its large nucleus);
cell type, the free-ranging phagocytic alveolar macrophage, is EP, alveolar epithelial cell; IN, interstitial space; BM, basement membrane;
found in varying numbers in the extracellular lining of the FB, fibroblast processes; 2, 3, and 4, diffusion pathway through the
alveolar surface. These cells patrol the alveolar surface and alveolar–capillary barrier, the plasma, and the erythrocyte, respectively.
phagocytize inspired particles such as bacteria. The type II (Reproduced with permission from Weibel, E.R. : Morphometric estimation of pulmonary
alveolar epithelial cell also plays a major role in the response of diffusion capacity, I. Model and method. Respir Physiol 1970;11:54–75.)
CHAPTER 31 Function and Structure of the Respiratory System 311

Removal of Material from lining of the chest wall, the visceral and parietal pleura. Other
the Alveolar Surface muscles of respiration include the abdominal muscles, includ-
ing the rectus abdominis; the parasternal intercartilaginous
Inspired material that reaches the terminal airways and alveoli muscles; and the sternocleidomastoid and scalenus muscles.
may be removed in several ways, including ingestion by alveo-
lar macrophages, enzymatic destruction, entry into the lym-
phatics, and immunologic reactions. Inhaled particles engulfed THE CENTRAL NERVOUS SYSTEM
by alveolar macrophages may be destroyed by their lysosomes AND THE NEURAL PATHWAYS
(see Chapter 1). Most bacteria are digested in this manner.
Some material ingested by the macrophages, however, such as Another important component of the respiratory system is the
silica, is not degradable by the macrophages and may even central nervous system. Unlike cardiac muscle, the muscles of
be toxic to them. If the macrophages carrying such material respiration do not contract spontaneously. Each breath is initi-
are not removed from the lung, the material will be redepos- ated in the brain, and this message is carried to the respiratory
ited on the alveolar surface on the death of the macrophages. muscles via the spinal cord and the nerves innervating the
The mean life span of alveolar macrophages is believed to be respiratory muscles.
1–5 weeks. The main exit route of macrophages carrying such Spontaneous automatic breathing is generated by groups of
nondigestible material is migration to the mucociliary escala- neurons located in the medulla. This medullary respiratory
tor via the pores of Kohn and eventual removal through the center is also the final integration point for influences from
airways. Particle-containing macrophages may also migrate higher brain centers, for information from chemoreceptors in
from the alveolar surface into the septal interstitium, from the blood and cerebrospinal fluid, and for afferent information
which they may enter the lymphatic system or the mucociliary from neural receptors in the airways, joints, muscles, skin, and
escalator. Macrophage function has been shown to be inhib- elsewhere in the body. The control of breathing is discussed in
ited by cigarette smoke. Alveolar macrophages are also impor- Chapter 38.
tant in the lung’s immune and inflammatory responses. They
secrete many enzymes, arachidonic acid metabolites, immune
response components, growth factors, cytokines, and other CHAPTER SUMMARY
mediators that modulate the function of other cells, such as
■ The main function of the respiratory system is the exchange of
lymphocytes. oxygen from the atmosphere for carbon dioxide produced by
Some particles reach the mucociliary escalator because the the cells of the body.
alveolar fluid lining itself is slowly moving upward toward ■ Other functions of the respiratory system include participation
the respiratory bronchioles. Others penetrate into the inter- in the acid–base balance of the body, phonation, pulmonary
stitial space or enter the blood, where they are phagocytized defense, and metabolism.
by interstitial macrophages or blood phagocytes, or enter the
lymphatics. Particles may be destroyed or detoxified by
surface enzymes and factors in the serum and in airway
secretions.
STUDY QUESTIONS
1. The functions of the respiratory system include
A) gas exchange.
THE MUSCLES OF RESPIRATION B) acid–base balance.
AND THE CHEST WALL C) phonation.
D) pulmonary defense and metabolism.
The muscles of respiration and the chest wall are essential com- E) handling bioactive materials.
ponents of the respiratory system. The lungs are not capable of F) all of the above.
inflating themselves—the force for this inflation must be 2. Particulate matter in the inspired air that enters the airways or
supplied by the muscles of respiration. The chest wall must be alveoli may be removed by
intact and able to expand if air is to enter the alveoli normally. A) the mucociliary escalator.
The interactions among the muscles of respiration and the chest B) alveolar macrophages.
wall and the lungs are discussed in detail later in Chapter 32. C) surface enzymes.
D) the lymphatics.
The primary components of the chest wall include the rib
E) all of the above.
cage; the external and internal intercostal muscles and the dia-
phragm, which are the main muscles of respiration; and the
This page intentionally left blank
32
C H A P T E R

Mechanics of the
Respiratory System
Michael Levitzky

O B J E C T I V E S

■ Describe the generation of a pressure gradient between the atmosphere


and the alveoli.
■ Describe the passive expansion and recoil of the alveoli.
■ Define the mechanical interaction of the lung and the chest wall.
■ Describe the pressure–volume characteristics of the lung and the chest wall,
and predict changes in the compliance of the lung and the chest wall in
different physiologic and pathologic conditions.
■ State the roles of pulmonary surfactant and alveolar interdependence
in the recoil and expansion of the lung.
■ Define the functional residual capacity (FRC), and predict changes in FRC in
different physiologic and pathologic conditions.
■ Define airway resistance and list the factors that contribute to or alter the
resistance to airflow.
■ Describe the dynamic compression of airways during a forced expiration.
■ List the factors that contribute to the work of breathing.
■ Predict alterations in the work of breathing in different physiologic and
pathologic states.

Air, like other fluids, moves from a region of higher pressure ent between the atmosphere and the alveoli by normal
to one of lower pressure. Therefore, for air to be moved into or negative-pressure breathing. Air flows out of the lungs when
out of the lungs, a pressure difference between the atmosphere alveolar pressure is sufficiently greater than atmospheric
and the alveoli must be established. If there is no pressure gra- pressure to overcome the resistance to airflow offered by the
dient, no airflow will occur. conducting airways.
Under normal circumstances, inspiration is accomplished
by causing alveolar pressure to decrease below atmospheric
pressure. When the mechanics of breathing are being dis- GENERATION OF A PRESSURE
cussed, atmospheric pressure is conventionally referred to
as 0 cm H2O, so lowering alveolar pressure below atmo- GRADIENT BETWEEN THE
spheric pressure is known as negative-pressure breathing. ATMOSPHERE AND THE ALVEOLI
When a pressure gradient sufficient to overcome the resis-
tance to airflow offered by the conducting airways is estab- During normal negative-pressure breathing, alveolar pressure
lished between the atmosphere and the alveoli, air flows is made lower than atmospheric pressure. This is accomplished
into the lungs. It is also possible to cause air to flow into the by causing the muscles of inspiration to contract, which
lungs by increasing the pressure at the nose or mouth above increases the volume of the alveoli, thus lowering the alveolar
alveolar pressure. This positive-pressure ventilation is gen- pressure according to Boyle’s law: at constant temperature, the
erally used on patients unable to generate a pressure gradi- product of the pressure and the volume of a gas is constant.

313

Ch32_313-330.indd 313 12/15/10 11:46:29 AM


314 SECTION VI Pulmonary Physiology

atmospheric pressure: 0 cm H2O atmospheric pressure: 0 cm H2O


no air flow: atmospheric air flows in : atmospheric
pressure = alveolar pressure pressure>alveolar pressure

outward recoil force generated by


of chest wall inspiratory muscles

alveolar pressure:
0 cm H2O alveolar pressure:
1 cm H2O

intrapleural pressure: inward recoil


of alveoli intrapleural pressure:
5 cm H2O
8 cm H2O

transmural pressure 
0 cm H2O(5 cm H2O)5 cm H2O
transmural pressure 
1 cm H2O (8 cm H2O)7 cm H2O

END EXPIRATION DURING INSPIRATION

FIGURE 32–1 Representation of the interaction of the lung and chest wall. Left: At end expiration, the muscles of respiration are
relaxed. The inward elastic recoil of the lung is balanced by the outward elastic recoil of the chest wall. Intrapleural pressure is –5 cm H2O; alveolar
pressure is 0. The transmural pressure gradient across the alveolus is therefore 0 – (–5) cm H2O, or 5 cm H2O. Since alveolar pressure is equal to
atmospheric pressure, no airflow occurs. Right: During inspiration, contraction of the muscles of inspiration causes intrapleural pressure to
become more negative. The transmural pressure gradient increases and the alveoli are distended, decreasing alveolar pressure below atmospheric
pressure, which causes air to flow into the alveoli. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

The alveoli are not capable of expanding themselves. They fluid-filled pleural space, as seen on the left in Figure 32–1.
expand passively in response to an increased distending pres- There is normally no free gas in the intrapleural space, and the
sure across the alveolar wall. This increased transmural pres- lung is held against the chest wall by the thin layer of serous
sure gradient, generated by the muscles of inspiration, further intrapleural liquid, estimated to have a total volume of about
opens the highly distensible alveoli and thus decreases the 15–25 mL in the average adult.
alveolar pressure. The transmural pressure gradient is conven- Initially, before any airflow occurs, the pressure inside the
tionally calculated by subtracting the outside pressure (in this alveoli is the same as atmospheric pressure—by convention
case, the intrapleural pressure) from the inside pressure (in 0 cm H2O. Alveolar pressure is greater than intrapleural pres-
this case, the alveolar pressure). sure because it represents the sum of the intrapleural pressure
The pressure in the thin, liquid-filled space between the vis- plus the alveolar elastic recoil pressure:
ceral and parietal pleura is normally slightly less than atmo- Alveolar pressure =
spheric pressure, even when no inspiratory muscles are (1)
Intrapleural pressure + Alveolar elastic recoil pressure
contracting. This negative intrapleural pressure (sometimes
also referred to as negative intrathoracic pressure) of –3 to The muscles of inspiration act to increase the volume of the
–5 cm H2O is mainly caused by the mechanical interaction thoracic cavity. As the inspiratory muscles contract, expand-
between the lung and the chest wall. At the end of expiration, ing the thoracic volume and increasing the outward stress
when all the respiratory muscles are relaxed, the lung and the on the lung, the intrapleural pressure becomes more negative.
chest wall are acting on each other in opposite directions. The Therefore, the transmural pressure gradient tending to distend
lung is tending to decrease its volume because of the inward the alveolar wall (also called the transpulmonary pressure)
elastic recoil of the distended alveolar walls; the chest wall is increases as shown in Figure 32–1, and the alveoli enlarge pas-
tending to increase its volume because of its outward elastic sively. Increasing alveolar volume lowers alveolar pressure and
recoil. Thus, the chest wall is acting to hold the alveoli open in establishes the pressure gradient for airflow into the lung. In
opposition to their elastic recoil. Similarly, the lung is acting reality, only a small percentage of the total number of alveoli
by its elastic recoil to hold the chest wall in. Because of this are directly exposed to the intrapleural surface pressure, and at
interaction, the pressure is negative at the surface of the very first thought, it is difficult to see how alveoli located centrally
thin (about 10–30 μm in thickness at normal lung volumes), in the lung could be expanded by a more negative intrapleural
CHAPTER 32 Mechanics of the Respiratory System 315

Intrapleural Negative pressure breathing


pressure
(A)

Interdependence of alveolar units

Positive pressure ventilation

(B)

FIGURE 32–2 Structural interdependence of alveolar units. The pressure gradient across the outermost alveoli is transmitted
mechanically through the lung via the alveolar septa. The insets show the author’s idea of what might happen in negative-pressure breathing
and positive-pressure ventilation. In negative-pressure breathing (inset A), the mechanical stress would likely be transmitted from the more
exterior alveoli (those closest to the chest wall) to more interior alveoli, so the exterior alveoli might be more distended. In positive-pressure
ventilation (inset B), the lungs must push against the diaphragm and rib cage to move them. The outermost alveoli might be more compressed
than those located more interiorly. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

pressure. However, careful analysis has shown that the pres- volume. It is innervated by the two phrenic nerves, which are
sure at the pleural surface is transmitted through the alveolar motor nerves that leave the spinal cord at the third to the fifth
walls to more centrally located alveoli and small airways. This cervical segments.
structural interdependence of alveolar units is depicted in The muscle fibers of the diaphragm are inserted into the ster-
Figure 32–2. num and the six lower ribs and into the vertebral column by the
two crura. The other ends of these muscle fibers converge to
attach to the fibrous central tendon, which is also attached to
THE MUSCLES OF RESPIRATION the pericardium on its upper surface (Figure 32–3). During
normal quiet breathing, contraction of the diaphragm causes its
Inspiratory Muscles dome to descend 1–2 cm into the abdominal cavity, with little
The muscles of respiration are skeletal muscles and their activ- change in its shape. This elongates the thorax and increases its
ity is normally initiated by the nervous system.The muscles of volume. These small downward movements of the diaphragm
inspiration include the diaphragm, the external intercostal are possible because the abdominal viscera can push out against
muscles, and the accessory muscles of inspiration. The dia- the relatively compliant abdominal wall. During a deep inspira-
phragm is a large (about 250 cm2 in surface area), dome- tion, the diaphragm can descend as much as 10 cm. With such
shaped muscle that separates the thorax from the abdominal a deep inspiration, the limit of the abdominal wall to expand is
cavity. It is the primary muscle of inspiration. When a person reached, abdominal pressure increases, and the indistensible
is in the supine position, the diaphragm is responsible for central tendon becomes fixed against the abdominal contents.
about two thirds of the air that enters the lungs during normal After this point, contraction of the diaphragm against the fixed
quiet breathing (which is called eupnea). When a person is central tendon elevates the lower ribs as shown in the figure.
standing or seated in an upright posture, the diaphragm may Contraction of the external intercostal, parasternal inter-
be responsible for only about one third to one half of the tidal costal, and scalene muscles raises and enlarges the rib cage.
316 SECTION VI Pulmonary Physiology

Pleura

Rib cage
Mediastinum

Pericardium

Central tendon

Diaphragm

Crura

END EXPIRATION DEEP INSPIRATION


FIGURE 32–3 Illustration of the actions of diaphragmatic contraction in expanding the thoracic cavity. (Modified with permission from
Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

Figure 32–4 demonstrates how contraction of these muscles inspiratory muscles of the rib cage contracted alone, the dia-
increases the anteroposterior dimension of the chest as the phragm would be pulled upward into the thorax.
ribs rotate upward about their axes and also increases the trans- The accessory muscles of inspiration are not involved dur-
verse dimension of the lower portion of the chest. These mus- ing normal quiet breathing but may be called into play during
cles are innervated by nerves leaving the spinal cord at the 1st exercise, during the inspiratory phase of coughing or sneezing,
to the 11th thoracic segments. During inspiration, the dia- or in a pathologic state, such as asthma. Dyspnea, the feeling
phragm and inspiratory rib cage muscles contract simultane- that breathing is difficult, is probably often related to fatigue of
ously. If the diaphragm contracted alone, the rib cage muscles the inspiratory muscles. Other potential causes of dyspnea will
would be pulled inward (this is called retraction). If the be discussed in Chapter 38.

INSPIRATION ACTIVE EXPIRATION

Accessory muscles

External intercostals

Internal intercostals

Diaphragm

Abdominal muscles

Posterior Anterior
FIGURE 32–4 Illustration of the actions of contraction of the intercostal muscles, abdominal muscles, and accessory muscles.
(Modified with permission of the publisher from by Weibel ER: The Pathway for Oxygen. Cambridge, MA: Harvard University Press, p. 304. Copyright © 1984 by the President and
Fellows of Harvard College.)
CHAPTER 32 Mechanics of the Respiratory System 317

Expiratory Muscles in this chapter, and on pulmonary blood flow, which will be
discussed in Chapter 34.
Expiration is passive during normal quiet breathing, and no
respiratory muscles contract. As the inspiratory muscles relax,
the increased elastic recoil of the distended alveoli is sufficient
to decrease the alveolar volume and raise alveolar pressure SUMMARY OF THE EVENTS
above atmospheric pressure, establishing the pressure gradient OCCURRING DURING THE
for airflow from the lung. COURSE OF A BREATH
Although the diaphragm is usually considered to be com-
pletely relaxed during expiration, it is likely that some dia- The events occurring during the course of an idealized nor-
phragmatic muscle tone is maintained, especially when one is mal quiet breath (summarized in Table 32–1) are shown in
in the horizontal position. Figure 32–5. For the purpose of clarity, inspiration and expira-
Active expiration occurs during exercise, speech, singing, tion are considered to be of equal duration, although during
the expiratory phase of coughing or sneezing, and in patho- normal quiet breathing, the expiratory phase is longer than
logic states such as chronic bronchitis. The main muscles of the inspiratory phase.
expiration are the muscles of the abdominal wall and the inter- Initially, alveolar pressure equals atmospheric pressure, so no
nal intercostal muscles. When the abdominal muscles con- air flows into the lung. Intrapleural pressure is –5 cm H2O.
tract, they increase abdominal pressure and push the Contraction of the inspiratory muscles causes intrapleural pres-
abdominal contents against the relaxed diaphragm, forcing it sure to become more negative as the lungs are pulled open and
upward into the thoracic cavity. They also help depress the the alveoli are distended. As the alveoli are distended, the pres-
lower ribs and pull down the anterior part of the lower chest. sure inside them decreases below atmospheric pressure and air
Contraction of the internal intercostal muscles depresses the flows into the alveoli, as seen in the tidal volume panel. As the
rib cage downward in a manner opposite to the actions of the air flows into the alveoli, alveolar pressure returns to 0 cm H2O
external intercostals. Active expiration compresses the thorax and airflow into the lung ceases. At the vertical line, the inspira-
and causes positive intrapleural pressure. This has important tory effort ceases and the inspiratory muscles relax. Intrapleu-
effects on the respiratory system, which will be discussed later ral pressure becomes less negative, and the elastic recoil of the

TABLE 32–1 Events involved in a normal tidal breath.


Inspiration

1. Brain initiates inspiratory effort

2. Nerves carry the inspiratory command to the inspiratory muscles

3. Diaphragm (and/or external intercostal muscles) contracts

4. Thoracic volume increases as the chest wall expandsa

5. Intrapleural pressure becomes more negative

6. Alveolar transmural pressure gradient increases

7. Alveoli expand (according to their individual compliance curves) in response to the increased transmural pressure gradient.
This increases alveolar elastic recoil

8. Alveolar pressure falls below atmospheric pressure as the alveolar volume increases, thus establishing a pressure gradient for airflow

9. Air flows into the alveoli until alveolar pressure equilibrates with atmospheric pressure

Expiration (passive)

1. Brain ceases inspiratory command

2. Inspiratory muscles relax

3. Thoracic volume decreases, causing intrapleural pressure to become less negative and decreasing the alveolar transmural pressure gradientb

4. Decreased alveolar transmural pressure gradient allows the increased alveolar elastic recoil to return the alveoli to their preinspiratory volumes

5. Decreased alveolar volume increases alveolar pressure above atmospheric pressure, thus establishing a pressure gradient for airflow

6. Air flows out of the alveoli until alveolar pressure equilibrates with atmospheric pressure
a
Note that numbers 4–8 occur simultaneously.
b
Note that numbers 3–5 occur simultaneously.
Reproduced with permission from Levitzky MG, Cairo JM, Hall SM: Introduction to Respiratory Care. Philadelphia: WB Saunders and Company, 1990.
318 SECTION VI Pulmonary Physiology

100
–5

Intrapleural
cm H2O pressure 75
Expiration

Total lung volume, %


–10
+1 Inspiration
50
Alveolar
cm H2O 0 pressure

–1 25

+0.5

L/sec 0 Air flow


0 10 20 30 40
Transpulmonary pressure cm H2O
–0.5
FIGURE 32–6 Pressure–volume curve for isolated lungs.
0.5 (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
Change in New York: McGraw-Hill Medical, 2007.)
L lung volume

0 lungs from an animal or a cadaver and then graph the changes


in volume that occur for each change in transpulmonary pres-
Onset of End of End of sure to which the lungs are subjected (Figure 32–6).
inspiration inspiration expiration
Figure 32–6 shows that as the transpulmonary pressure
increases, the lung volume increases. Of course, this relation-
FIGURE 32–5 Volume, pressure, and airflow changes during
ship is not a straight line: the lung is composed of living tissue,
a single idealized respiratory cycle. Described in text. (Reproduced
with permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New
and although the lung distends easily at low lung volumes, at
York: McGraw-Hill, 2009.) high lung volumes the distensible components of alveolar
walls have already been stretched, and large increases in trans-
pulmonary pressure yield only small increases in volume. The
alveolar walls (which is increased at the higher lung volume) is lung also is difficult to distend at very low lung volumes
allowed to compress the alveolar gas. This raises alveolar pres- because some alveoli may be collapsed and extra energy is
sure above atmospheric pressure so that air flows out of the necessary to reopen them.
lung until an alveolar pressure of 0 cm H2O is restored. At this The slope between two points on a pressure–volume curve is
point, airflow ceases until the next inspiratory effort. known as the compliance. Compliance is defined as the change
in volume divided by the change in pressure. Lungs with high
compliance have a steep slope on their pressure–volume curves,
PRESSURE–VOLUME that is, a small change in distending pressure will cause a large
change in volume. It is important to remember that compliance
RELATIONSHIPS IN THE is the inverse of elasticity, or elastic recoil. Compliance denotes
the ease with which something can be stretched or distorted;
RESPIRATORY SYSTEM elasticity refers to the tendency for something to oppose stretch
The relationship between changes in the pressure distending or distortion, as well as to its ability to return to its original con-
the alveoli and changes in lung volume is important to under- figuration after the distorting force is removed.
stand because it dictates how the lung inflates with each breath. There are several other interesting things to note about an
As mentioned before, the alveolar-distending pressure is often experiment like that illustrated in Figure 32–6. The curve
referred to as the transpulmonary pressure. obtained is the same whether the lungs are inflated with posi-
tive pressure (by forcing air into the trachea) or with negative
pressure (by suspending the lung, except for the trachea, in a
COMPLIANCE OF THE LUNG closed chamber and pumping out the air around the lung). So
AND THE CHEST WALL when the lung alone is considered, only the transpulmonary
pressure is important, not how the transpulmonary pressure is
The pressure–volume characteristics of the lung can be generated. A second feature of the curve in Figure 32–6 is that
inspected in several ways. One of the simplest is to remove the there is a difference between the pressure–volume curve for
CHAPTER 32 Mechanics of the Respiratory System 319

inflation and the curve for deflation, as shown by the arrows. Δ Lung volume ΔV
Compliance = =
Such a difference is called hysteresis. One possible explana- Δ (P alv – P ip) Δ P tp
tion for this hysteresis is the stretching on inspiration and the
compression on expiration of the surfactant that lines the air–
liquid interface in the alveoli (discussed later in this chapter). Increased
compliance
Another is that some alveoli or small airways may open on
inspiration (“recruitment”) and close on expiration (“dere-
cruitment”) as noted above. Some researchers believe that lung
Normal
volume changes primarily by recruitment and derecruitment compliance

Lung volume (ml)


of alveoli rather than by volume changes of individual alveoli.
Finally, it can be helpful to think of each alveolus as having its
own pressure–volume curve like that shown in the figure.
Decreased
Clinical Evaluation of the Compliance compliance

of the Lung and the Chest Wall


The compliance of the lung and the chest wall provides very
useful data for the clinical evaluation of a patient’s respiratory
system because many diseases or pathologic states affect the
compliance of the lung, the chest wall, or both. The lung and 0 Transpulmonary pressure (P tp)
the chest wall are physically in series with each other, and (P alv – P ip)
therefore their compliances add as reciprocals:
FIGURE 32–7 Representative static pulmonary compliance
1
____________ curve for normal lungs; lungs with low compliance, for example,
=
Total compliance (2) lungs with fibrosis; and lungs with high compliance, for example,
1
_________________ 1
+ ____________________ lungs with emphysema. (Reproduced with permission from Widmaier EP, Raff
Compliance of the lung Compliance of the chest wall H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
Compliances in parallel add directly. Therefore, both lungs
together are more compliant than either one alone.
The compliance curve for the lungs can be generated by the compliance of the lungs because it destroys the alveolar
having the patient take a very deep breath and exhale in stages, septal tissue that normally opposes lung expansion.
stopping periodically for pressure and volume determinations. The compliance of the chest wall is decreased in obese peo-
During these determinations, no airflow is occurring; alveolar ple, for whom moving the diaphragm downward and the rib
pressure therefore equals atmospheric pressure, 0 cm H2O. cage up and out is much more difficult. Musculoskeletal disor-
Similar measurements can be made as the patient inhales in ders that lead to decreased mobility of the rib cage, such as
stages from a low lung volume to a high lung volume. Such kyphoscoliosis, also decrease the chest wall compliance.
curves are called static compliance curves because all mea- Because they must generate greater transpulmonary pres-
surements are made when no airflow is occurring. The com- sures to breathe in the same volume of air, people with
pliance of the chest wall is normally obtained by determining decreased compliance of the lungs must do more work
the compliance of the total system and the compliance of the to inspire than those with normal pulmonary compliance.
lungs alone and then calculating the compliance of the chest Similarly, more muscular work must be done when chest wall
wall according to the above formula. Dynamic compliance compliance is decreased.
assesses pressure–volume characteristics during the breath.
Representative static compliance curves for the lungs are
shown in Figure 32–7. Note that these curves correspond to ELASTIC RECOIL OF THE LUNG
the expiratory curve in Figure 32–6. Many pathologic states
shift the curve to the right; that is, for any increase in transpul- So far, the elastic recoil of the lungs has been discussed as
monary pressure, there is a smaller increase in lung volume. A though it were only due to the elastic properties of the pulmo-
proliferation of connective tissue called fibrosis may occur in nary parenchyma itself. However, there is another component
sarcoidosis or after chemical or thermal injury to the lungs. of the elastic recoil of the lung—the surface tension at the air–
This will make the lungs less compliant, or “stiffer,” and liquid interface in the alveoli.
increase alveolar elastic recoil. Similarly, pulmonary vascular Surface tension forces occur at any gas–liquid interface and
engorgement or areas of collapsed alveoli (atelectasis) also are generated by the cohesive forces between the molecules of
make the lung less compliant. Other conditions that interfere the liquid. These cohesive forces balance each other within
with the lung’s ability to expand (such as the presence of air, the liquid phase but are unopposed at the surface of the liq-
excess fluid, or blood in the intrapleural space) will decrease uid. Surface tension causes water to bead and form droplets,
the measured compliance of the lungs. Emphysema increases and a liquid to shrink to form the smallest possible surface
320 SECTION VI Pulmonary Physiology

Saline Air
200

150
Volume (mL)

T P T T  Pr
100

50
T
Resolved direction of tension
0 FIGURE 32–9 Relationship between the pressure inside a
4 8 12 16 20
Pressure (cm H2O) distensible sphere, such as an alveolus, and its wall tension.
FIGURE 32–8 Pressure–volume curves for excised cat lungs
(Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New
York: McGraw-Hill Medical, 2007.)
inflated with air or saline. (Modified from Radford EP. Recent studies of
mechanical properties of mammalian lungs. In: Remington JW. Tissue Elasticity.
Washington: American Physiological Society; 1957.)
If two alveoli of different sizes are connected by a common
airway (Figure 32–10) and the surface tension of the two alve-
area. The role of the surface tension forces in the elastic recoil oli is equal, then according to Laplace’s law, the pressure in the
of the lung can be demonstrated in an experiment shown in small alveolus is greater than that in the larger alveolus and the
Figure 32–8. smaller alveolus will empty into the larger alveolus. If surface
In this experiment, a pressure–volume curve for an excised tension is independent of surface area, the smaller the alveolus
lung was first generated with air inflation, so an air–liquid on the right becomes, the higher the pressure in it.
interface was present in the lung, and surface tension forces Thus, if the lung were composed of interconnected alveoli of
contributed to alveolar elastic recoil. Then, all the gas was different sizes (which it is) with a constant surface tension at
removed from the lung, and it was inflated again, this time the air–liquid interface, it would be inherently unstable, with a
with saline instead of with air. In this situation, surface tension tendency for smaller alveoli to collapse into larger ones. This is
forces were absent because there was no air–liquid interface. usually not the case, which is fortunate because collapsed alve-
The elastic recoil was due only to the lung tissue itself. Note oli require very great distending pressures to reopen, partly
that there is no hysteresis with saline inflation. Whatever because of the cohesive forces at the liquid–liquid interface of
causes the hysteresis appears to be related to surface tension in collapsed alveoli. At least two factors cause the alveoli to be
the lung. The curve at left (saline inflation) therefore repre- more stable than this prediction based on constant surface
sents the elastic recoil due to only the lung tissue itself; the
curve at right demonstrates the recoil due to both the lung tis-
sue and the surface tension forces. The difference between the
two curves is the recoil due to surface tension forces.
The demonstration of the large role of surface tension forces
in the recoil pressure of the lung led to consideration of how
surface tension affects the alveoli. One traditional way of
thinking about this has been to consider the alveolus to be a
sphere hanging from the airway, as in Figure 32–9. The rela- P1 r T
T P2
tionship between the pressure inside the alveolus and the wall 2r
tension of the alveolus would then be given by Laplace’s law
(units in brackets):
T
2 × tension [dyn/cm] P1 
Pressure [dyn/cm ] = _______________
2
(3) r
Radius [cm]
T
This can be rearranged as follows: P2 
2r
Pr
T = __
2
(4) FIGURE 32–10 Schematic representation of two alveoli of
different sizes connected to a common airway. If the surface
where T is the wall tension, P the pressure inside the alveolus, tension is the same in both alveoli, then the smaller alveolus will have
and r the radius of the alveolus. a higher pressure and will empty into the larger alveolus. (Modified with
The surface tension of most liquids (such as water) is con- permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill
stant and not dependent on the area of the air–liquid interface. Medical, 2007.)
CHAPTER 32 Mechanics of the Respiratory System 321

tension. The first factor is a substance called pulmonary sur- the alveoli and the tendency for spontaneous atelectasis to
factant, which is produced by specialized alveolar cells, and occur because of a lack of pulmonary surfactant. Exogenous
the second is the structural interdependence of the alveoli. pulmonary surfactant is now administered directly into the
airway of neonates with infant respiratory distress syndrome.
In summary, pulmonary surfactant helps decrease the work
Pulmonary Surfactant of inspiration by lowering the surface tension of the alveoli,
Pulmonary surfactant decreases the elastic recoil due to surface thus reducing the elastic recoil of the lung and making the
tension, thereby increasing the compliance of the lungs above lung more compliant. Surfactant also helps stabilize the alveoli
that predicted by an air–water interface and decreasing by lowering even further the surface tension of smaller alveoli,
the inspiratory work of breathing. Pulmonary surfactant has a equalizing the pressure inside alveoli of different sizes.
second major effect. It decreases the surface tension of smaller
alveoli. This helps equalize alveolar pressures throughout the
lung (so the end-expiratory pressure of all the alveoli is 0 cm Alveolar Interdependence
H2O and the situation depicted in Figure 32–10 does not occur) A second factor tending to stabilize the alveoli is their mechan-
and to stabilize alveoli. Pulmonary surfactant is a complex con- ical interdependence, which was discussed at the beginning of
sisting of about 85–90% lipids and 10–15% proteins. The lipid this chapter. Alveoli do not hang from the airways like a
portion is about 85% phospholipid, approximately 75% of “bunch of grapes” (the translation of the Latin word “acinus”),
which is dipalmitoyl phosphatidylcholine. This complex is and they are not spheres. They are mechanically interdepen-
produced by type II alveolar epithelial cells (described above). dent polygons with flat walls shared by adjacent alveoli. Alve-
Pulmonary surfactant appears to be continuously produced in oli are normally held open by the chest wall pulling on the
the lung, but it is also continuously cleared from the lung. Some outer surface of the lung, as shown in Figure 32–2. If an alveo-
pulmonary surfactant is taken back into the type II cells lus were to begin to collapse, it would increase the stresses on
(reuptake), where it is recycled and secreted again, or it is the walls of the adjacent alveoli, which would tend to hold it
degraded and used to synthesize other phospholipids. Surfactant open. This process would oppose a tendency for isolated alve-
is also cleared from the alveoli by alveolar macrophages, absorp- oli with a relative lack of pulmonary surfactant to collapse
tion into the lymphatics, or migration up to the small airways spontaneously. Conversely, if a whole subdivision of the lung
and the mucociliary escalator (discussed in Chapter 31). Type (such as a lobule) has collapsed, as soon as the first alveolus is
II alveolar epithelial cells may also help remove liquid from the reinflated, it helps to pull other alveoli open by its mechanical
alveolar surface by actively pumping sodium and water from interdependence with them. Thus, both pulmonary surfactant
the alveolar surface into the interstitium. and the mechanical interdependence of the alveoli help stabi-
The clinical consequences of a lack of functional pulmonary lize the alveoli and oppose alveolar collapse (atelectasis).
surfactant occur in several conditions. Surfactant is not pro-
duced by the fetal lung until about the fourth month of gesta-
tion, and it may not be fully functional until the seventh month MECHANICAL INTERACTION OF
or later. Prematurely born infants who do not have functional THE LUNG AND CHEST WALL
pulmonary surfactant experience great difficulty in inflating
their lungs, especially on their first breaths. Even if their alveoli The inward elastic recoil of the lung normally opposes the
are inflated for them with positive-pressure ventilation, the ten- outward elastic recoil of the chest wall, and vice versa. If the
dency toward spontaneous collapse is great because their alve- integrity of the lung–chest wall system is disturbed by break-
oli are much less stable without pulmonary surfactant. ing the seal of the chest wall (e.g., by a penetrating knife
Therefore, the lack of functional pulmonary surfactant in a pre- wound), the inward elastic recoil of the lung is no longer
maturely born neonate may be a major factor in the infant opposed by the outward recoil of the chest wall, and their
respiratory distress syndrome. Pulmonary surfactant may also interdependence ceases. Lung volume decreases, and alveoli
be important in maintaining the stability of small airways. have a much greater tendency to collapse, especially if air
Alveolar hypoxia or hypoxemia (low oxygen in the arterial moves in through the wound (causing a pneumothorax) until
blood), or both, may lead to a decrease in surfactant produc- intrapleural pressure equalizes with atmospheric pressure and
tion or an increase in surfactant destruction. This condition abolishes the transpulmonary pressure gradient. At this point,
may be a contributing factor in the acute respiratory distress nothing is tending to hold the alveoli open and their elastic
syndrome (also known as adult respiratory distress syndrome recoil is causing them to collapse. Similarly, the chest wall
or “shock lung syndrome”) that can occur in patients after tends to expand because its outward recoil is no longer
trauma or surgery. One approach to maintain patients with opposed by the inward recoil of the lung.
acute or infant respiratory distress syndrome is to ventilate When the lung–chest wall system is intact and the respiratory
their lungs with positive-pressure ventilators and to keep their muscles are relaxed, the volume of gas left in the lungs is deter-
alveolar pressure above atmospheric pressure during expira- mined by the balance of these two forces. The volume of gas
tion (this is known as positive end-expiratory pressure in the lungs at the end of a normal tidal expiration, when no
[PEEP]). This process opposes the increased elastic recoil of respiratory muscles are actively contracting, is known as the
322 SECTION VI Pulmonary Physiology

functional residual capacity (FRC). For any given situation, the The resistance to airflow is analogous to electrical resistance
FRC will be that lung volume at which the outward recoil of the in that resistances in series are added directly:
chest wall is equal and opposite to the inward recoil of the lungs.
Rtot = R1 + R2 + … (7)
If the lung volume increases above the FRC, the increased
inward elastic recoil of the lung exceeds the decreased outward Resistances in parallel are added as reciprocals:
elastic recoil of the chest wall. At high lung volumes (above 1 1 __
about 70% of the total lung capacity [TLC]), the chest wall
___ = __ + 1 +…
Rtot R1 R2
(8)
also has inward elastic recoil. Therefore, at high lung volumes, Airflow, like that of other fluids, can occur as either laminar or
the elastic recoil of both the lung and chest wall are inward. At turbulent flow, as was discussed in Chapter 26 (see Figure 26–6).
lung volumes below the FRC, the outward recoil of the chest When a fluid such as air is in laminar flow through rigid, smooth
wall is greater than the reduced inward recoil of the lungs. bore tubes, its behavior is governed by Poiseuille’s law, as was
A change from the upright to the supine position decreases discussed in Chapter 22.
FRC. The reason for this decrease of about 30% is the effect of Turbulent flow tends to occur if airflow is high, gas density
gravity on the mechanics of the chest wall, especially the dia- is high, the tube radius is large, or all three conditions exist.
phragm. When standing up or sitting, the contents of the True laminar flow probably occurs only in the smallest air-
abdomen are being pulled away from the diaphragm by grav- ways, where the linear velocity of airflow is extremely low. Lin-
ity. When lying down, the abdominal contents are pushing ear velocity (cm/s) is equal to the flow (cm3/s) divided by the
inward against the relaxed diaphragm. This decreases the cross-sectional area. The sum of the cross-sectional areas of
overall outward recoil of the chest wall and decreases the lung the smallest airways is very large, so the linear velocity of air-
volume at which the outward recoil of the chest wall is equal flow is very low. The airflow in the trachea and larger airways
and opposite to the inward recoil of the lungs. is usually either turbulent or a mixture of laminar and turbu-
lent flow.
AIRWAY RESISTANCE
Several factors besides the elastic recoil of the lungs and the DISTRIBUTION OF
chest wall must be overcome to move air into or out of the AIRWAY RESISTANCE
lungs. These factors are primarily the frictional resistance of
the lung and chest wall tissue, and the frictional resistance of About 25–40% of the total resistance to airflow is located in
the airways to the flow of air. Pulmonary tissue resistance is the upper airways: the nose, nasal turbinates, oropharynx,
caused by the friction encountered as the lung tissues move nasopharynx, and larynx. Resistance is higher when one
against each other as the lung expands. The airway resistance breathes through the nose than when one breathes through
plus the pulmonary tissue resistance is often referred to as the the mouth.
pulmonary resistance. Pulmonary tissue resistance normally The vocal cords open slightly during normal inspirations
contributes about 20% of the pulmonary resistance, with air- and close slightly during expirations. During deep inspira-
way resistance responsible for the other 80%. Pulmonary tis- tions, they open widely. The muscles of the oropharynx also
sue resistance can be increased in such conditions as pulmonary contract during normal inspiration; this dilates and stabilizes
sarcoidosis and fibrosis. Because airway resistance is the major the upper airway. During deep forced inspirations, the devel-
component of the total resistance and because it can increase opment of negative pressure could cause the upper airway to
tremendously both in healthy people and in those suffering be pulled inward and partly or completely obstruct airflow.
from various diseases, the remainder of this chapter will con- Reflex contraction of these pharyngeal dilator muscles nor-
centrate on airway resistance. mally keeps the airway open.
The component with the highest individual resistance of the
LAMINAR, TURBULENT, AND tracheobronchial tree is obviously the smallest airway, which
has the smallest radius. Nevertheless, because the smallest air-
TRANSITIONAL FLOW ways are arranged in parallel, their resistances add as recipro-
cals, so that the total resistance to airflow offered by the
As was discussed in Chapter 22, the relationship among
numerous small airways is extremely low during normal, quiet
pressure, flow, and resistance is:
breathing. Therefore, the greatest resistance to airflow usually
Pressure difference = Flow × Resistance (5) resides in the medium-sized bronchi.
Therefore, we have:
Pressure difference [cm H O]
Resistance = _____________________
2
(6) CONTROL OF BRONCHIAL
Flow [L/s]
SMOOTH MUSCLE
This means that resistance is a meaningful term only during
flow. When airflow is considered, the units of resistance are The smooth muscle of the airways from the trachea down to
usually cm H2O/[L/s]. the alveolar ducts is under the control of efferent fibers of the
CHAPTER 32 Mechanics of the Respiratory System 323

TABLE 32–2 Active control of the airways.


100
Constrict
Parasympathetic stimulation

Airway resistance (% maximum)


Acetylcholine
Histamine 75
Leukotrienes
Thromboxane A2
Serotonin
α-Adrenergic agonists 50
Decreased Pco2 in small airways

Dilate
Sympathetic stimulation (β2 receptors) 25
Circulating β2 agonists
Nitric oxide
Increased Pco2 in small airways RV TLC
0
Decreased Po2 in small airways 0 2 4 6 8
Lung volume (L)

autonomic nervous system (see Chapter 19). Stimulation of FIGURE 32–11 Relationship between lung volume and
the cholinergic parasympathetic postganglionic nerves airway resistance. Total lung capacity (TLC) is at right; residual
causes constriction of bronchial smooth muscle as well as volume (RV) is at left. (Reproduced with permission from Kibble J, Halsey CR:
increased glandular mucus secretion. The preganglionic fibers The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)

travel in the vagus nerve. Stimulation of the adrenergic sym-


pathetic nerves causes dilation of bronchial and bronchiolar
smooth muscle as well as inhibition of glandular secretion. resistance, even with so many parallel pathways. To increase
This dilation of the airway smooth muscle is mediated by beta2 lung volume, a person breathing normally takes a “deep
(β2) receptors, which predominate in the airways. Selective breath,” that is, makes a strong inspiratory effort. This effort
stimulation of the alpha (α) receptors with pharmacologic causes intrapleural pressure to become much more negative
agents causes bronchoconstriction. Adrenergic transmitters than –7 or –10 cm H2O in a normal, quiet breath. The trans-
carried in the blood may be as important as those released mural pressure gradient across the wall becomes much more
from the sympathetic nerves in causing bronchodilation. The positive, and small airways are distended.
bronchial smooth muscle is normally under greater parasym- A second reason for the decreased airway resistance at
pathetic tone than sympathetic tone. higher lung volumes is that the traction on the small airways
Inhalation of chemical irritants, smoke, or dust; stimulation increases. As shown in the schematic drawing in Figure 32–12,
of the arterial chemoreceptors; and substances such as hista- the small airways traveling through the lung form attachments
mine cause constriction of the airways. Decreased CO2 in the to the walls of alveoli. As the alveoli expand during the course
branches of the conducting system causes a local constriction of a deep inspiration, the elastic recoil in their walls increases;
of the smooth muscle of the nearby airways; increased CO2 or this elastic recoil is transmitted to the attachments at the air-
decreased O2 causes a local dilation. This may help balance way, pulling it open.
ventilation and perfusion (see Chapter 35). Many other sub-
stances can have direct or indirect effects on airway smooth
muscle (Table 32–2). Leukotrienes usually cause bronchocon-
striction, as do some prostaglandins.

LUNG VOLUME AND


AIRWAY RESISTANCE
AIRWAY Traction on airway by elastic
Airway resistance decreases with increasing lung volume, as recoil of alveolar septa
shown in Figure 32–11. There are two reasons for this rela-
tionship; both mainly involve the small airways that, as
described in this chapter, have little or no cartilaginous sup-
port. The small airways are therefore rather distensible and
also compressible. Thus, the transmural pressure gradient
across the wall of the small airways is an important determi- FIGURE 32–12 Representation of “traction” of the alveolar
nant of the radius of the airways: since resistance is inversely septa on a small distensible airway. Compare this figure with the
proportional to the radius to the fourth power, changes in the picture of the alveolar duct in Figure 31–4. (Modified with permission from
radii of small airways can cause dramatic changes in airway Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)
324 SECTION VI Pulmonary Physiology

DYNAMIC COMPRESSION OF AIRWAYS pressure gradient would generate very high rates of airflow.
However, the airways are not uniformly rigid and the smallest
Airway resistance is extremely high at low lung volumes, as airways, which have no cartilaginous support and rely on the
shown in Figure 32–11. To achieve low lung volumes, a person traction of alveolar septa to help keep them open, may be com-
must make a forced expiratory effort by contracting the mus- pressed or may even collapse. Whether or not they actually
cles of expiration, mainly the abdominal and internal intercos- collapse depends on the transmural pressure gradient across
tal muscles. This effort generates a positive intrapleural the walls of the smallest airways.
pressure, which can be as high as 120 cm H2O during a maxi- The situation during a normal passive expiration at the same
mal forced expiratory effort. (Maximal inspiratory intrapleu- lung volume (note the same alveolar elastic recoil pressure) is
ral pressures can be as low as –80 cm H2O.) shown in the left part of Figure 32–13. The transmural pres-
The effect of this high positive intrapleural pressure on the sure gradient across the smallest airways is +1 – (–8) cm H2O
transmural pressure gradient during a forced expiration can = +9 cm H2O tending to hold the airway open. During the
be seen at right in Figure 32–13, a schematic drawing of a sin- forced expiration at right, the transmural pressure gradient is
gle alveolus and airway. 30 – 25 cm H2O, or only 5 cm H2O holding the airway open.
The muscles of expiration are generating a positive intra- The airway may then be slightly compressed, and its resistance
pleural pressure of +25 cm H2O. Pressure in the alveolus is to airflow will be even greater than that during the passive
higher than intrapleural pressure because of the alveolar elas- expiration. This increased resistance during a forced expira-
tic recoil pressure of +10 cm H2O, which, together with intra- tion is called dynamic compression of airways.
pleural pressure, gives an alveolar pressure of +35 cm H2O. Consider what occurs during a maximal forced expiration.
The alveolar elastic recoil pressure decreases at lower lung vol- As the expiratory effort is increased to attain a lower lung
umes because the alveolus is not as distended. In the figure, a volume, intrapleural pressure is getting more and more posi-
gradient has been established from the alveolar pressure of tive, and more and more dynamic compression will occur.
+35 cm H2O to the atmospheric pressure of 0 cm H2O. If the Furthermore, as lung volume decreases, there will be less
airways were rigid and incompressible, the large expiratory alveolar elastic recoil pressure and the difference between

0 0

0.25 3

8 25 5 25
8
10
0.5
15
8 8 25 25 Contraction of
20 internal
intercostals and
25 accessory muscles
8 8 25 25 of expiration
30
1

10 10 10 10


2 35

10 10 10 10


8 8 25 25

Relaxed diaphragm pushed up by


abdominal muscle contraction

PASSIVE EXPIRATION FORCED EXPIRATION

FIGURE 32–13 Schematic diagram illustrating dynamic compression of airways and the equal pressure point hypothesis during
a forced expiration. Left: Passive (eupneic) expiration. Intrapleural pressure is –8 cm H2O, alveolar elastic recoil pressure is +10 cm H2O, and
alveolar pressure is +2 cm H2O. Right: Forced expiration at the same lung volume. Intrapleural pressure is +25 cm H2O, alveolar elastic recoil
pressure is +10 cm H2O, and alveolar pressure is +35 cm H2O. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill
Medical, 2007.)
CHAPTER 32 Mechanics of the Respiratory System 325

alveolar pressure and intrapleural pressure will decrease. At


any instant during a forced expiration, there is a point along
the airways where the pressure inside the airway is just equal
to the pressure outside the airway. At that point (the “equal
pressure point”), the transmural pressure gradient is 0 (note
the arrows in Figure 32–13). Above that point, the transmu-

PA  Ppl ( Pel)
ral pressure gradient is negative: the pressure outside the air- Intrapleural Dynamic
way is greater than the pressure inside it, and the airway will pressure compression
collapse if cartilaginous support or alveolar septal traction is
insufficient to keep it open. As the forced expiratory effort
continues, the equal pressure point is likely to move down the Alveolar Traction
airway from larger to smaller airways. This movement hap-
elastic recoil
pens because, as the muscular effort increases, intrapleural
pressure increases and because, as lung volume decreases,
alveolar elastic recoil pressure decreases. As the equal pres-
sure point moves down the airway, dynamic compression
Alveolar Pel Pel
increases and the airways begin to collapse. This airway clo- elastic PA
sure can be demonstrated only at especially low lung volumes recoil Pel Pel
in healthy subjects, but the closing volume may occur at
higher lung volumes in patients with high lung compliance as
in emphysema. The closing volume will be discussed in
Chapter 33.
During a passive expiration, the pressure gradient for air-
flow (ΔP in ΔP = V̇ R) is simply alveolar pressure minus atmo- FIGURE 32–14 Representation of the effects of alveolar
spheric pressure. But if dynamic compression occurs, the elastic recoil on airflow during a forced expiration. When dynamic
effective pressure gradient is alveolar pressure minus intrapleu- compression occurs, alveolar elastic recoil helps to oppose it by
traction on the small airways. The alveolar elastic recoil pressure
ral pressure (which equals the alveolar elastic recoil pressure)
becomes the effective driving pressure for airflow from the lung. PA,
because intrapleural pressure is greater than atmospheric pres- alveolar pressure; Ppl, intrapleural pressure; Pel, the alveolar elastic
sure and because intrapleural pressure can exert its effects on recoil pressure. (Modified with permission from Levitzky MG: Pulmonary
the compressible portion of the airways. Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)
Thus, during a forced expiration, when intrapleural pressure
becomes positive and dynamic compression occurs, the effective
driving pressure for airflow from the lung is the alveolar elastic
tance during expiration will be emphasized because that factor
recoil pressure. Alveolar elastic recoil is also important in oppos-
is of interest in patients with lung disease.
ing dynamic compression of the airways because of its role in the
traction of the alveolar septa on small airways, as shown in
Figure 32–12. The effects of alveolar elastic recoil on airflow Forced Vital Capacity
during a forced expiration are illustrated in Figure 32–14. One way of assessing expiratory airway resistance is to look at
the results of a forced expiration into a spirometer, as shown in
Figure 32–15. This measurement is called a forced vital capac-
ASSESSMENT OF AIRWAY RESISTANCE ity (FVC). The vital capacity (VC) is the volume of air a sub-
ject is able to expire after a maximal inspiration to the TLC.
The resistance to airflow cannot be measured directly but must An FVC means that a maximal expiratory effort was made
be calculated from the pressure gradient and airflow during a during this maneuver.
breath: In an FVC test, a person makes a maximal inspiration to the
ΔP TLC. After a moment, he or she makes a maximal forced expi-
R = ___ (9)
V̇ ratory effort, blowing as much air as possible out of the lungs.
The above formula is an approximation because it presumes At this point, only a residual volume (RV) of air is left in the
that all airflow is laminar, which is not true. But there is a sec- lungs. (The lung volumes will be described in detail in the next
ond problem: how can the pressure gradient be determined? chapter.) This procedure takes only a few seconds, as can be
To know the pressure gradient, the alveolar pressure—which seen on the time scale.
also cannot be measured directly—must be known. Alveolar The part of the curve most sensitive to changes in expira-
pressure can be calculated using a body plethysmograph, an tory airway resistance is the first second of expiration. The
expensive piece of equipment described in the next chapter, volume of air expired in the first second of expiration (the
but this procedure is not often done. Instead, airway resistance FEV1, or forced expiratory volume in 1 second), especially
is usually assessed indirectly. The assessment of airway resis- when expressed as a ratio with the total amount of air expired
326 SECTION VI Pulmonary Physiology

7
6
5 Normal

Volume (L)
RV
4 FEV1 / FVC  80%

3 FEV1  3.6 L Obstruction


RV
FEV1 / FVC  50% FVC  4.5 L
2
FVC  3.0 L
1 FEV1  1.5 L

TLC 0 1 2 3 4 5 6
Time (s)

FIGURE 32–15 Forced vital capacity (FVC) maneuver using a rolling seal spirometer. FVCs from a normal subject and from a patient
with obstructive disease. FEV1, forced expiratory volume in the first second. Note that the total lung capacity (TLC) is at the bottom of the curves
and the residual volumes (RVs) are at the top; volume therefore refers to the volume exhaled into the spirometer in the bottom trace. The time
scale is from left to right. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

during the FVC, is a good index of expiratory airway resis- Obstructive diseases interfere with airflow; restrictive diseases
tance. In normal subjects, the FEV1/FVC is greater than restrict the expansion of the lung.
0.80; that is, at least 80% of the FVC is expired in the first Figure 32–17 shows that both obstruction and restriction can
second. A patient with an airway obstruction would be cause a decrease in the maximal flow rate that the patient can
expected to have an FEV1/FVC far below 0.80, as shown in attain, the peak expiratory flow (PEF; shown in Figure 32–16),
Figure 32–16, which shows FVC curves obtained from a but that this decrease occurs for different reasons. Restrictive
commonly used rolling seal spirometer. Note that the TLC is lung diseases, which usually entail increased alveolar elastic
at the bottom left, and the RVs are at the top right. The time recoil, may have decreased PEF because the TLC (and thus the
scale is left to right. Note the calculations of the FEV1 to VC) is decreased. The effort-independent part of the curve is
FVC ratios for a healthy person and for one with airway similar to normal lungs. In fact, the FEV1/FVC is usually nor-
obstruction. mal or even above normal since both the FEV1 and FVC are
Figure 32–15 clearly shows that elevated airway resistance decreased because the lung has a low volume and because alve-
takes time to overcome. olar elastic recoil pressure may be increased. On the other hand,
with obstructive diseases, the PEF and FEV1/FVC are both low.
Obstructive diseases—such as asthma, bronchitis, and
Flow–Volume Curves emphysema—are often associated with high lung volumes,
Flow–volume curves are also used to assess airway resis- which is helpful because the high volumes increase the alveo-
tance. A family of flow–volume curves such as that depicted in lar elastic recoil pressure. The RV may be greatly increased if
Figure 32–16 is obtained by having a subject make repeated airway closure occurs at relatively high lung volumes. A sec-
expiratory maneuvers with different degrees of effort. Flow ond important feature of the flow–volume curve of a patient
rates are plotted against lung volume for expiratory efforts of with obstructive disease is the effort-independent portion of
different intensities. There are two interesting points about the curve, which is depressed inward: flow rates are low for
this family of curves. At high lung volumes, the airflow rate is any relative volume.
effort-dependent, which can be seen in the left-hand portion Flow–volume curves are very useful in assessing obstruc-
of the curves. As the subject exhales with greater effort, flow tions of the upper airways and the trachea. Flow–volume
rate increases. At low lung volumes, however, the expiratory loops can help distinguish between fixed obstructions (those
efforts of different initial intensities all merge into the same not affected by the inspiratory or expiratory effort) and vari-
effort-independent curve, as seen in the right-hand portion able obstructions (changes in the transmural pressure gradi-
of the curve. This difference is because intrapleural pressures ent caused by the inspiratory or expiratory effort result in
high enough to cause dynamic compression are necessary to changes in the cross-sectional area of the obstruction). If the
attain very low lung volumes, no matter what the initial expi- obstruction is variable, flow–volume loops can demonstrate
ratory effort. Also, at low lung volumes there is less alveolar whether the obstruction is extrathoracic or intrathoracic
elastic recoil, so there is less traction on the same airways and (Figure 32–18). A fixed obstruction affects both expiratory
a smaller pressure gradient for airflow. and inspiratory airflow (Figure 32–18A). Both the expiratory
The maximal flow–volume curve is used as a diagnostic and inspiratory flow–volume curves are truncated, with
tool, as shown in Figure 32–17, because it helps distinguish decreased peak expiratory and peak inspiratory flows. The
between two major classes of pulmonary diseases—airway flow–volume loop is unable to distinguish between a fixed
obstructive diseases and restrictive diseases, such as fibrosis. extrathoracic and a fixed intrathoracic obstruction, which
CHAPTER 32 Mechanics of the Respiratory System 327

10 Peak expiratory Maximal curve


flow

Effort independence

Effort dependence
Expiration

5
Airflow (L/s)

TLC RV
0
Inspiration

Maximal curve

10
Volume (L)
FIGURE 32–16 Flow–volume curves of varying intensities, demonstrating effort dependence at high lung volumes and effort
independence at low lung volumes. Note that there is no effort independence in inspiration. The peak expiratory flow (PEF) is labeled for the
maximal expiratory curve. TLC, total lung capacity; RV, residual volume. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York:
McGraw-Hill Medical, 2007.)

would usually be determined with a bronchoscope. Fixed However, during a forced inspiration, the pressure inside the
obstructions can be caused by foreign bodies or by scarring upper airway decreases below atmospheric pressure, and
that makes a region of the airway too stiff to be affected by the unless the stability of the upper airway is maintained by reflex
transmural pressure gradient. contraction of the pharyngeal muscles or by other structures,
During a forced expiration, the cross-sectional area of a the cross-sectional area of the upper airway will decrease.
variable extrathoracic obstruction increases as the pressure Therefore, the inspiratory flow–volume curve is truncated
inside the airway increases (Figure 32–18B). The expiratory with variable extrathoracic obstructions. Variable extratho-
flow–volume curve is therefore nearly normal or not affected. racic obstructions can be caused by tumors, fat deposits,
328 SECTION VI Pulmonary Physiology

15
Normal

12

Restrictive disease
Airflow (L/s) 9
Obstructive disease

0
9 8 7 6 5 4 3 2 1 0
Lung volume (L)
FIGURE 32–17 Maximal expiratory flow–volume curves representative of obstructive and restrictive diseases. (Modified with
permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

A. Expiration

Flow
TLC RV

Inspiration Expiration Inspiration

Fixed (intra- or extrathoracic)


B. expiration
Flow

TLC RV

Paw < Patm Paw > Patm

Inspiration Expiration Inspiration

Variable extrathoracic
FIGURE 32–18 Inspiratory and
C. Expiration
expiratory flow–volume curves representing
the patterns in: A) fixed intrathoracic or
extrathoracic obstruction; B) variable
TLC RV
extrathoracic obstruction; C) variable
Flow

intrathoracic obstruction. TLC, total lung


capacity; RV, residual volume; Paw, airway
pressure; Patm, atmospheric pressure; Ppl,
intrapleural pressure. (Modified with permission Paw > Ppl Paw < Ppl
from Burrows B, Knudson RJ, Quan SF, Kettel LJ:
Respiratory Disorders: A Pathophysiologic Approach, Inspiration Expiration Inspiration
2nd ed. Copyright © 1983 by Year Book Medical
Publishers, Chicago.) Variable intrathoracic
CHAPTER 32 Mechanics of the Respiratory System 329

weakened or flabby pharyngeal muscles (as in obstructive


sleep apnea), paralyzed vocal cords, enlarged lymph nodes, or
THE WORK OF BREATHING
inflammation. The major points discussed in this chapter can be summa-
During a forced expiration, positive intrapleural pressure rized by considering the work of breathing. The work done
decreases the transmural pressure gradient across a variable in breathing is proportional to the pressure change times the
intrathoracic tracheal obstruction, decreasing its cross- volume change. The volume change is the volume of air
sectional area and decreasing the PEF (Figure 32–18C). moved into and out of the lung—the tidal volume. The pres-
During a forced inspiration, as large negative intrapleural sure change is the change in transpulmonary pressure neces-
pressures are generated, the transmural pressure gradient sary to overcome the elastic work of breathing and the
across the variable intrathoracic obstruction increases and resistive work of breathing. The elastic work of breathing is
its cross-sectional area increases. Thus, the inspiratory the work done to overcome the elastic recoil of the chest wall
flow–volume curve is nearly normal or not affected. Variable and the pulmonary parenchyma and the work done to over-
intrathoracic obstructions of the trachea are most com- come the surface tension of the alveoli. Restrictive diseases
monly caused by tumors. are those diseases in which the elastic work of breathing is
increased. For example, the work of breathing is elevated in
obese patients (who have decreased outward chest wall elastic
CLINICAL CONSEQUENCES OF recoil) and in patients with pulmonary fibrosis or a relative
INCREASED AIRWAY RESISTANCE AND lack of pulmonary surfactant (who have increased elastic
DECREASED ALVEOLAR COMPLIANCE recoil of the alveoli). The resistive work of breathing is the
work done to overcome the tissue resistance and the airway
As discussed at the beginning of this chapter, the lung has mil- resistance. The tissue resistance may be elevated in conditions
lions of small airways and hundreds of millions of alveoli. If we such as sarcoidosis, asbestosis, or silicosis. Elevated airway
think of a pair of hypothetical alveoli supplied by the same resistance is much more common and occurs in obstructive
airway, we can consider the time courses of their changes in diseases such as asthma, bronchitis, and emphysema; upper
volume in response to an abrupt increase in airway pressure airway obstruction; and accidental aspiration of foreign
(a “step” increase). If the resistances and compliances of the objects. Normally, most of the resistive work is that done to
two units were equal, the two alveoli would fill with identical overcome airway resistance. The resistive work of breathing
time courses to identical volumes. If the resistances were equal, can be very great during a forced expiration, when dynamic
but the compliance of one were half that of the other, then the compression occurs. This is especially true in patients who
two alveoli would fill with nearly identical time courses but the already have elevated airway resistance during normal, quiet
less compliant one would receive only half the volume received breathing. For example, in patients with emphysema, a dis-
by the other. If the compliances of the two units were equal but ease that attacks and obliterates alveolar walls, the work of
one was supplied by an airway with twice the resistance to breathing can be tremendous because of the destruction of
airflow of the one supplying the other, the two units would the elastic tissue support of their small airways, which allows
ultimately fill to the same volume. However, the one supplied dynamic compression to occur unopposed. Also, the
by the airway with elevated resistance will fill more slowly than decreased elastic recoil of alveoli leads to a decreased pressure
the other because of its elevated resistance. This difference gradient for expiration.
means that at high breathing frequencies, the one that fills
more quickly will receive a larger volume of air per breath; the
one that fills more slowly will receive less ventilation per
breath. CLINICAL CORRELATION
Now let us extrapolate this two-unit situation to a lung with
A 26-year-old man comes to the emergency department
millions of airways supplying hundreds of millions of alveoli.
because of sudden dyspnea (a feeling that breathing is dif-
In a patient with small airways disease, many alveoli may be
ficult, also called “shortness of breath”) and pain in the
supplied by airways with higher resistance to airflow than
upper part of the left side of his chest. He has no history of
normal. These alveoli are sometimes referred to as “slow alve-
any medical problems. He is 183-cm (6′2″) tall and weighs
oli” or alveoli with long “time constants.” As the patient
about 63.5 kg (140 lb). Blood pressure is 125/80 mm Hg,
increases the breathing frequency, the slowest alveoli will not
heart rate is 90/min, and respiratory rate is 22/min
have enough time to fill. As the frequency increases, more and
(usually12–15/min in a healthy adult). There are no breath
more slow alveoli will drop out.
sounds on the left side of his chest, which is hyperresonant
This can also be a problem during positive-pressure ventila-
(louder and more hollow-sounding) to percussion (the
tion. There may be enough time for alveoli to fill because air is
physician tapping on the chest with his or her fingers).
forced into them by the ventilator. However, because expira-
The patient has a pneumothorax. Air has entered the
tion is passive, there may not be enough time for the alveoli to
pleural space on the left side of his chest and he is unable to
empty, resulting in overinflation, especially of more compliant
expand his left lung. Therefore, there are no breath sounds
alveoli, causing lung injury.
330 SECTION VI Pulmonary Physiology

■ During forced expiration, when intrapleural pressure be-


on the left side of his chest and it is hyperresonant to per- comes positive, small airways are compressed (dynamic
cussion. In this case, the pneumothorax is a primary spon- compression) and may even collapse.
taneous pneumothorax because it occurred suddenly, and ■ The two main components of the work of breathing are the
is not attributable to an underlying pulmonary disease (sec- elastic recoil of the lungs and chest wall and the resistance
ondary spontaneous pneumothorax) or trauma (traumatic to airflow.
pneumothorax). The inability to ventilate his left lung,
combined with pain and anxiety, explains his high respira-
tory rate, as will be discussed in Chapters 33 and 38. STUDY QUESTIONS
Primary spontaneous pneumothorax is most common in
1. In a normal healthy adult at the functional residual capacity
tall, thin males between 10 and 30 years of age, although
the reason for this is not known. It is believed to occur A) alveolar pressure is greater than atmospheric pressure.
B) alveolar pressure is less than atmospheric pressure.
when overexpanded alveoli (“blebs”) rupture, perhaps as a
C) the inward recoil of the lungs is equal and opposite to
result of a cough or sneeze.
the outward recoil of the chest wall.
If the pneumothorax is mild and the patient is not in too D) intrapleural pressure is positive.
much distress, it may resolve without treatment other than E) the alveolar transmural pressure gradient is negative.
observation. More severe pneumothorax is treated by 2. Which of the following would be expected to cause increased
inserting a catheter or chest tube through the skin and static lung compliance (i.e., shift the pulmonary pressure–
intercostal muscles into the pleural space to allow removal volume curve upward and to the left)?
of the air by external suction. A) a relative lack of functional pulmonary surfactant
A tension pneumothorax is a potentially life-threatening B) diffuse interstitial alveolar fibrosis
disorder that most commonly occurs as a result of trauma C) pulmonary vascular congestion
or lung injury. Air enters the pleural space on inspiration D) emphysema
but cannot leave on expiration, progressively increasing E) diffuse alveolar collapse
intrapleural pressure above atmospheric. This can com- 3. The compliance of the lungs is
press the structures on the affected side of the chest A) greater at low lung volumes than it is at high lung volumes.
(e.g., blood vessels, heart, etc.) and eventually the struc- B) in parallel with the compliance of the chest wall.
tures on the other side of the chest as well. C) increased in a person after surgical removal of one lobe
of the lung.
D) increased in a person with pulmonary interstitial fibrosis.
E) less than the compliance of a single lobe of the lung.
CHAPTER SUMMARY 4. During a forced expiration to the residual volume
■ A pressure gradient between the atmosphere and the alveoli A) intrapleural pressure becomes more negative.
must be established to move air into or out of the alveoli. B) alveolar elastic recoil is increasing.
C) the outward recoil of the chest wall is increasing.
■ During inspiration, alveoli expand passively in response to an
D) intrapleural pressure is greater than alveolar pressure.
increased transmural pressure gradient; during normal quiet
E) airflow remains dependent on expiratory effort.
expiration, the elastic recoil of the alveoli returns them to their
original volume. 5. Which of the following will likely decrease the work of breathing?
■ The volume of gas in the lungs at the end of a normal tidal A) doubling the tidal volume at the same breathing frequency
expiration (the FRC), when no respiratory muscles are actively B) breathing through the mouth instead of the nose
contracting, is determined by the balance point of the inward C) doubling the breathing frequency at the same tidal volume
recoil of the lungs and the outward recoil of the chest wall. D) breathing through a 1-cm diameter, 3-ft long tube
E) gaining 100 lb of body weight
■ At the FRC, intrapleural pressure is negative because the pleural
liquid is between the opposing forces of the inward recoil of the 6. The resistance to airflow in a normal healthy person would
lungs and the outward recoil of the chest wall. be greatest
■ Alveoli are more compliant (and have less elastic recoil) at low A) during a eupneic inspiration.
volumes; alveoli are less compliant (and have more elastic B) during a eupneic expiration.
recoil) at high volumes. C) during a forced inspiration.
D) during a forced expiration.
■ Pulmonary surfactant increases alveolar compliance and helps
E) at the functional residual capacity.
prevent atelectasis by reducing surface tension in the alveoli.
33
C H A P T E R

Alveolar Ventilation
Michael Levitzky

O B J E C T I V E S

■ Define alveolar ventilation.


■ Define the standard lung volumes.
■ Predict the effects of alterations in lung and chest wall mechanics, due to
normal or pathologic processes, on the lung volumes.
■ Define anatomic dead space and relate the anatomic dead space and the
tidal volume to alveolar ventilation.
■ Calculate alveolar ventilation.
■ Define and calculate physiologic and alveolar dead space.
■ Predict the effects of alterations of alveolar ventilation on alveolar carbon
dioxide and oxygen levels.
■ Describe and explain the regional differences in alveolar ventilation found in
the normal lung.
■ Define the closing volume.
■ Predict how changes in pulmonary mechanics affect the closing volume.

Alveolar ventilation is the exchange of gas between the size, and are normally expressed as the body temperature and
alveoli and the external environment. It is the process by ambient barometric pressure and saturated with water vapor
which oxygen is brought into the lungs from the atmo- (BTPS).
sphere and by which the carbon dioxide carried into the
lungs in the mixed venous blood is expelled from the body.
Although alveolar ventilation is usually defined as the vol- THE STANDARD LUNG
ume of fresh air entering the alveoli per minute, a similar VOLUMES AND CAPACITIES
volume of alveolar air leaving the body per minute is
implicit in this definition. There are four standard lung volumes and four standard
lung capacities, which consist of two or more of the standard
lung volumes (Figure 33–1).
THE LUNG VOLUMES
The volume of gas in the lungs at any instant depends on the The Tidal Volume
mechanics of the lungs and chest wall and the activity of the The tidal volume (VT) is the volume of air entering or leaving
muscles of inspiration and expiration. The size of the lungs the nose or mouth per breath. During normal, quiet breathing
depends the height and weight or body surface area, as well as (eupnea), the VT of a 70-kg adult is about 500 mL per breath,
age and sex. Therefore, the lung volumes are usually compared but this volume can increase substantially, for example, during
with “predicted” lung volumes matched to age, sex, and body exercise.

331

Ch33_331-340.indd 331 11/26/10 11:50:04 AM


332 SECTION VI Pulmonary Physiology

maximal inspiration

INSPIRATORY RESERVE
INSPIRATORY
VOLUME (IRV)
CAPACITY (IC)
2.5 L
3.0 L
VITAL
CAPACITY (VC)
TOTAL LUNG TIDAL VOLUME (VT) 4.5 L
CAPACITY (TLC) 0.5 L resting volume
6.0 L EXPIRATORY RESERVE
VOLUME (ERV)
FUNCTIONAL 1.5 L
RESIDUAL
FIGURE 33–1 The standard lung volumes and CAPACITY (FRC)
3.0 L maximal expiration
capacities. Typical values for a 70-kg adult are shown. RESIDUAL VOLUME (RV)
(Modified with permission from Levitzky MG: Pulmonary 1.5 L
Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.) no air in lungs

The Residual Volume between the inward elastic recoil of the lungs and the outward
elastic recoil of the chest wall, as discussed in Chapter 32.
The residual volume (RV) is the volume of gas remaining in
During exercise, the FRC may be lower than the relaxation
the lungs after a maximal forced expiration. It is determined
volume because of active contraction of the expiratory mus-
by the force generated by the muscles of expiration and the
cles. The FRC, as seen in Figure 33–1, consists of the RV plus
inward elastic recoil of the lungs as they oppose the outward
the ERV and is therefore about 3 L in a healthy 70-kg adult.
elastic recoil of the chest wall. Dynamic compression of the
airways during the forced expiratory effort may also be an
important determinant of the RV as airway collapse occurs The Inspiratory Capacity
and traps gas in the alveoli. The RV of a healthy 70-kg adult is
The inspiratory capacity (IC) is the volume of air that is
about 1.5 L, but it can be much greater in emphysema, a lung
inhaled into the lungs during a maximal inspiratory effort that
disease with increased compliance in which inward alveolar
begins at the end of a normal tidal expiration (the FRC). It is
elastic recoil is diminished and airway collapse and gas trap-
therefore equal to the VT plus the IRV, as shown in Figure 33–1.
ping occur. The RV is important to a healthy person because it
The IC of a healthy 70-kg adult is about 3 L.
prevents the lungs from collapsing at very low lung volumes.
Such collapsed alveoli would require great inspiratory efforts
to reinflate. The Total Lung Capacity
The total lung capacity (TLC) is the volume of air in the lungs
The Expiratory Reserve Volume after a maximal inspiratory effort. It is determined by the
strength of contraction of the inspiratory muscles and the
The expiratory reserve volume (ERV) is the volume of gas
inward elastic recoil of the lungs and the chest wall. The TLC
that is expelled from the lungs during a maximal forced expi-
is the sum of all four lung volumes: the RV, the VT, the IRV, and
ration that starts at the end of a normal tidal expiration. It is
the ERV. It is about 6 L in a healthy 70-kg adult.
therefore determined by the difference between the functional
residual capacity (FRC, see below) and the RV. The ERV is
about 1.5 L in a healthy 70-kg adult. The Vital Capacity
The vital capacity (VC), discussed in Chapter 32, is the vol-
The Inspiratory Reserve Volume ume of air expelled from the lungs during a maximal forced
The inspiratory reserve volume (IRV) is the volume of gas that expiration starting after a maximal forced inspiration. It is
is inspired into the lungs during a maximal forced inspiration therefore equal to the TLC minus the RV, or about 4.5 L in a
starting at the end of a normal tidal inspiration. It is determined healthy 70-kg adult. The VC is also equal to the sum of the VT
by the strength of contraction of the inspiratory muscles, the and the IRV and ERV. It is determined by the factors that
inward elastic recoil of the lung and the chest wall, and the start- determine the TLC and RV.
ing point, which is the FRC plus the VT . The IRV of a healthy
70-kg adult is about 2.5 L.
MEASUREMENT OF THE
LUNG VOLUMES
The Functional Residual Capacity
The FRC is the volume of gas remaining in the lungs at the end Measurement of the lung volumes is important clinically because
of a normal tidal expiration. It represents the balance point many pathologic states can alter specific lung volumes or their
CHAPTER 33 Alveolar Ventilation 333

relationships to one another. The lung volumes, however, can eter can be determined. The spirometer can therefore measure
also change for normal physiologic reasons. Changing from a only the lung volumes that the subject can exchange with it. As
standing to a supine posture decreases the FRC because gravity is the case with many pulmonary function tests, the subject
is no longer pulling the abdominal contents away from the dia- must be conscious and cooperative and understand the instruc-
phragm. This decreases the outward elastic recoil of the chest tions for performing the test. Figure 33–3 shows that the VT, IRV,
wall, as noted in Chapter 32. Determination of the lung volumes ERV, IC, and VC can all be measured with a spirometer (as can
can be useful diagnostically in differentiating between two the forced expiratory volume in 1 second [FEV1], forced vital
major types of pulmonary disorders—the restrictive diseases capacity [FVC], and the FEV1/FVC, as discussed in Chapter 32).
and the obstructive diseases. Restrictive diseases such as alveo- The RV, the FRC, and the TLC, however, cannot be determined
lar fibrosis reduce the compliance of the lungs, increase elastic with a spirometer because the subject cannot exhale all the gas in
recoil, and lead to compressed lung volumes (Figure 33–2). The the lungs into the spirometer.
VT may even be decreased, with a corresponding increase in
breathing frequency, to minimize the work of breathing. Measurement of Lung Volumes
Obstructive diseases such as emphysema and chronic bron-
chitis cause increased resistance to airflow. Airways may become
Not Measurable with Spirometry
completely obstructed because of mucous plugs and because of The lung volumes not measurable with spirometry can be
the high intrapleural pressures generated to overcome the ele- determined by the nitrogen-washout technique, the helium-
vated airway resistance during a forced expiration. This is espe- dilution technique, and body plethysmography. The FRC is
cially a problem in emphysema, in which destruction of alveolar usually determined, and RV (which is equal to FRC minus
septa leads to decreased elastic recoil of the alveoli and less ERV) and the TLC (which is equal to VC plus RV) are then
radial traction, which normally help hold small airways open. calculated from volumes obtained by spirometry.
For these reasons, the RV, the FRC, and the TLC may be greatly
increased in obstructive diseases, as seen in Figure 33–2. The Nitrogen-washout technique
VC and ERV are usually decreased. The breathing frequency In the nitrogen-washout technique, the person breathes 100%
may be decreased to reduce the work expended overcoming the oxygen through a one-way valve to wash all of the nitrogen out
airway resistance, with a corresponding increase in the VT. of the alveoli. The expired gas is collected and the volume of
nitrogen washed out of the person’s lungs is calculated. The
total volume of nitrogen in the person’s lungs at the beginning
Spirometry of the test can thus be determined. Nitrogen constitutes about
The spirometer is a simple device for measuring gas volumes. 80% of the person’s initial lung volume, so multiplying the ini-
As the person breathes in and out through a mouthpiece (a nose tial nitrogen volume by 1.25 gives the person’s initial lung vol-
clip prevents airflow via the nose) and a tube connected to the ume. If the test starts at the end of a normal tidal expiration,
spirometer, the volumes of gas entering and leaving the spirom- the volume determined is the FRC.

IRV
IC
VC

VT
TLC
IRV ERV
IC

VT VC FIGURE 33–2 Illustration of typical


TLC alterations in the lung volumes and capacities
IC IRV
in restrictive and obstructive diseases. The
FRC
ERV VC pattern shown for obstructive diseases is more
RV
characteristic for emphysema and asthma than
VT
for chronic bronchitis. IC, inspiratory capacity;
FRC TLC
TLC, total lung capacity; FRC, functional residual
ERV capacity; IRV, inspiratory reserve volume; VT, tidal
RV FRC volume; ERV, expiratory reserve volume; RV,
RV residual volume; VC, vital capacity. (Modified with
permission from Levitzky MG: Pulmonary Physiology, 7th
normal restrictive obstructive ed. New York: McGraw-Hill Medical, 2007.)
334 SECTION VI Pulmonary Physiology

SPIROMETER TRACE (SPIROGRAM)

Maximal inspiration

Inspiratory reserve volume

Inspiratory
Vital capacity

volume (L)
capacity

Tidal
volume
FIGURE 33–3 Determination of the
tidal volume, vital capacity, inspiratory
capacity, inspiratory reserve volume,
Expiratory reserve volume
and expiratory reserve volume from a Maximal expiration
spirometer trace. (Modified with permission
from Levitzky MG: Pulmonary Physiology, 7th ed.
New York: McGraw-Hill Medical, 2007.) Time (s)

Helium-dilution technique The body plethysmograph makes use of Boyle’s law: for a
The helium-dilution technique makes use of the following closed container at a constant temperature, the pressure times
relationship: if the total amount of a substance dissolved in a the volume of a gas mixture is constant. The body plethysmo-
volume is known and its concentration can be measured, the graph is an airtight chamber large enough that the patient can sit
volume in which it is dissolved can be determined. inside it and breathe through a mouthpiece and tubing. The
Helium is used for this test because it is not taken up by the patient breathes in for an instant against a closed airway and the
pulmonary capillary blood, so the total amount of helium used pressures at the mouth and in the plethysmograph are moni-
in the test does not change during the test. The person breathes tored. As the patient breathes in against the closed airway, the
in and out of a spirometer filled with a mixture of helium and chest expands and the pressure measured in the plethysmograph
oxygen. When an equilibrium is reached, the concentration of increases because the volume of air in the plethysmograph
helium is the same in the lungs as it is in the spirometer, and decreases by the amount the chest volume increased. The pres-
the test is stopped at the end of a normal tidal expiration, in sure measured at the mouth decreases as the patient breathes in
other words, at the FRC. against a closed airway. Boyle’s law is used to calculate the
The calculated increase in the volume of distribution of changes in volumes of the body plethysmograph and the lungs
helium therefore represents the lung volume. Since it may take to determine the FRC.
several minutes for the helium concentration to equilibrate
between the lungs and the spirometer, CO2 is absorbed from
the system and oxygen is added to the spirometer at the rate ANATOMIC DEAD SPACE AND
at which it is used by the person. Both the nitrogen-washout
and helium-dilution methods can be used on unconscious ALVEOLAR VENTILATION
patients.
The volume of air entering and leaving the nose or mouth per
minute, the minute volume, is not equal to the volume of air
Body plethysmography entering and leaving the alveoli per minute. Alveolar ventilation
A problem common to both the nitrogen-washout technique is less than the minute volume because the last part of each inspi-
and the helium-dilution technique is that neither can measure ration remains in the conducting airways and does not reach the
trapped gas because nitrogen trapped in alveoli supplied by alveoli. Similarly, the last part of each expiration remains in the
closed airways cannot be washed out and because the helium conducting airways and is not expelled from the body.
cannot enter alveoli supplied by closed airways. Furthermore, The anatomic dead space is illustrated in Figure 33–4. When
if the lungs have many alveoli served by airways with high a person breathes in a tidal volume of 500 mL, not all the air
resistance to airflow (the “slow alveoli” discussed at the end of reaches the alveoli: the final 150 mL of the inspired air remains
Chapter 32), it may take a long time for all the nitrogen to in the conducting airways. The volume of gas reaching the alve-
wash out of the lungs or for the inspired and expired helium oli is equal to the volume inspired minus the volume of the ana-
concentrations to equilibrate. In such patients, measurements tomic dead space, in this case 500 – 150 mL, or 350 mL. During
of the lung volumes with a body plethysmograph are much expiration, the first gas breathed out is inspired air that remained
more accurate because they do include trapped gas. in the anatomic dead space; the last 150 mL is alveolar gas that
CHAPTER 33 Alveolar Ventilation 335

150 ml

Tidal volume = 500 ml


350 ml

Volume in Anatomic
conducting dead space = 150 ml Conducting
airways left over 150 ml 150 ml
airways
from preceding
breath
350 ml
FIGURE 33–4 Illustration of the anatomic dead
Alveolar gas
space. Of a 500-mL tidal volume, 150 mL remains in the
150 ml
conducting airways and does not participate in gas
exchange; only 350 mL enters the alveoli. (Reproduced with
permission from Widmaier EP, Raff H, Strang KT: Vander’s Human
Physiology, 11th ed. McGraw-Hill, 2008.)

remains in the anatomic dead space. Therefore, for any respira- Estimation of Anatomic Dead Space
tory cycle, not all the tidal volume reaches the alveoli because
For a healthy subject, the anatomic dead space can be esti-
the last part of each inspiration and each expiration remains in
mated by referring to a table of standard values matched to
the dead space. The relationship for the VT breathed in and out
sex, age, height, and weight or body surface area. The anatomic
through the nose or mouth, the dead space volume (VD), and
dead space is not measured clinically; a reasonable estimate of
the volume of gas entering and leaving the alveoli per breath
anatomic dead space is 1 mL of dead space per pound (2.2 kg)
(VA) is:
of ideal body weight.
VT = VD + VA (1)
or Physiologic Dead Space:
VA = VT – VD (2) The Bohr Equation
The alveolar ventilation (per minute) can be determined by The air in the anatomic dead space may not be the only
multiplying both sides of the above equation by the breathing inspired air that does not participate in gas exchange. The
frequency (f) in breaths per minute: alveolar dead space is the volume of gas that enters unper-
fused alveoli per breath. Alveolar dead space is therefore alve-
f (VA) = f (VT ) – f (VD ) (3) oli that are ventilated but not perfused with pulmonary
Thus, for f = 12 breaths/min in the above example, we have: capillary blood. No gas exchange occurs in these alveoli for
physiologic, rather than anatomic, reasons. A healthy person
4,200[mL/min] = 6,000[mL/min] – 1,800[mL/min] (4) has little or no alveolar dead space, but a person with a low
.
The alveolar ventilation (VA) in liters per minute is equal to cardiac output might have significant alveolar dead space, for
.
the minute volume (VE) minus reasons explained in Chapter 34.
. the volume wasted ventilating
the dead space per minute (VD): The Bohr equation permits the determination of the sum of
. . . the anatomic and the alveolar dead space. The anatomic dead
VA = VT – VD (5)
space plus the alveolar dead space is known as the physiologic
. The dot over the letter V indicates per minute. The symbol dead space:
VE is used because expired gas is usually collected. There is a
Physiologic dead space =
difference between the volume of gas inspired and the volume (6)
Anatomic dead space + Alveolar dead space
of gas expired because as air is inspired, it is warmed to body
temperature and humidified and also because normally less The Bohr equation makes use of a simple concept: any
carbon dioxide is produced than oxygen is consumed. measurable volume of carbon dioxide found in the mixed
expired gas must come from alveoli that are both ventilated
and perfused because there are negligible amounts of carbon
MEASUREMENT OF ALVEOLAR dioxide in inspired air. Inspired air remaining in the anatomic
VENTILATION dead space or entering unperfused alveoli will leave the body
as it entered (except for having been warmed to body tem-
Alveolar ventilation cannot be measured directly but must be perature and humidified), contributing little or no carbon
determined from the VT, the breathing frequency, and the dead dioxide to the mixed expired air, as shown in the following
space ventilation, as noted in the previous section. Bohr equation:
336 SECTION VI Pulmonary Physiology

50
End-tidal

40

Pco2 (mm Hg)


30
FIGURE 33–5 Partial pressure of carbon dioxide at
the mouth during a breath. During inspiration, the Pco2
rapidly decreases to near zero (0.3 mm Hg). The first
expired gas comes from the anatomic dead space and 20
therefore also has a Pco2 near zero. After exhalation of a
mixture of gas from alveoli and anatomic dead space,
the gas expired is a mixture from all ventilated alveoli. 10
The slope of the alveolar plateau normally rises slightly
because the alveolar Pco2 increases a few mm Hg between
inspirations. The last alveolar gas expired before 0
inspiration is called end-tidal. (Modified with permission 0 1 2 3 4 5
from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Expiration starts Inspiration starts
Medical, 2007.) Time (s)

VDco Paco2 – PEco2


____2
= _________ (7) space, so their physiologic dead space is equal to their ana-
VT Paco2
tomic dead space. Situations in which alveoli are ventilated but
where VDco2 is the dead space for CO2 (the physiologic dead not perfused include those in which portions of the pulmo-
space), VT the tidal volume, Paco2 the arterial partial pressure nary vasculature have been occluded by blood clots or other
of carbon dioxide, and PEco2 the mixed expired partial pres- material in the venous blood (pulmonary emboli), those in
sure of carbon dioxide. which there is low venous return leading to low right ventricu-
The arterial Pco2 must be determined from an arterial blood lar output (hemorrhage), and those in which alveolar pressure
sample; the Pco2 of the collected mixed expired gas can be is high (positive-pressure ventilation with positive end-
determined with a CO2 meter. After the physiologic dead space expiratory pressure). These will be discussed in greater detail
is calculated using the Bohr equation, the estimated anatomic in Chapter 34.
dead space can be subtracted from it to calculate the alveolar The anatomic dead space can be altered by bronchocon-
dead space. striction, which decreases VD; bronchodilation, which
The CO2 meter can also used to estimate the mean alveo- increases VD; or traction or compression of the airways, which
lar Pco2 by analyzing the gas expelled at the end of a normal increases and decreases VD, respectively.
tidal expiration, the “end-tidal CO2” (Figure 33–5). In a
person with significant alveolar dead space, however, the
estimated alveolar Pco2 obtained in this fashion may not
reflect the Pco2 of alveoli that are ventilated and perfused ALVEOLAR VENTILATION &
because some of this mixed end-tidal gas comes from unper- ALVEOLAR OXYGEN AND CARBON
fused alveoli. This gas dilutes the CO2 coming from alveoli
that are both ventilated and perfused. There is, however, an DIOXIDE LEVELS
equilibrium between the Pco2 of perfused alveoli and their
end-capillary Pco2 (see Chapter 35 for detailed discussion), The levels of oxygen and carbon dioxide. in alveolar gas are
so that in patients without significant venous-to-arterial determined .by the alveolar ventilation (VA), the oxygen con-
shunts, the arterial Pco2 represents the mean Pco2 of the per- sumption
. (VO2) of the body, and the carbon dioxide produc-
fused alveoli. tion (VCO2 )of the body.
If the arterial Pco2 is greater than the mixed alveolar Pco2
determined by sampling the end-tidal CO2, then the physio-
logic dead space is probably greater than the anatomic dead PARTIAL PRESSURES OF
space, that is, a significant arterial–alveolar CO2 difference RESPIRATORY GASES
means that there is significant alveolar dead space; a person
with no alveolar dead space has an arterial–end-tidal CO 2 dif- According to Dalton’s law, in a gas mixture, the pressure
ference of zero. As already noted, this difference is determined exerted by each individual gas is independent of the pres-
from the Pco2 from an arterial blood gas sample and from the sures of other gases in the mixture. The partial pressure of a
end-tidal Pco2 . Young healthy people have no alveolar dead particular gas is equal to its fractional concentration times
CHAPTER 33 Alveolar Ventilation 337

the total pressure of all the gases in the mixture. Thus, for TABLE 33–1 Partial pressures in mm Hg of oxygen,
any gas in a mixture (gas1), its partial pressure is given as carbon dioxide, nitrogen, and water vapor in dry air,
follows: inspired air, alveolar air, and expired air at a barometric
Pgas = Total gas (%) × Ptot (8) pressure of 760 mm Hg.
1

Oxygen constitutes 20.93% of dry atmospheric air. At a PO2 PCO2 PN2 PH2O
standard barometric pressure of 760 mm Hg, we have:
Dry air 159 0.3 601 0
PO2 = 0.2093 × 760 mm Hg = 159 mm Hg (9)
Inspired air 149 0.3 564 47
Carbon dioxide constitutes only about 0.04% of dry atmo- Alveolar air 104 40 569 47
spheric air, so we have:
Expired air 120 27 566 47
PCO2 = 0.0004 × 760 mm Hg = 0.3 mm Hg (10)
As air is inspired through the upper airways, it is warmed
and humidified, as discussed in Chapter 31. The partial pres-
sure of water vapor is a relatively constant 47 mm Hg at body inspiration. Expired air is a mixture of about 350 mL of alve-
temperature, so the humidification of 1 L of dry gas in a closed olar air and 150 mL of air from the dead space. Therefore,
container at 760 mm Hg would increase its total pressure to the Po2 of mixed expired air is higher than alveolar Po2 and
760 + 47 mm Hg = 807 mm Hg. In the body, the gas will lower than the inspired Po2, or approximately 120 mm Hg.
expand, according to Boyle’s law, so that 1 L of gas at 760 mm Similarly, the Pco2 of mixed expired air is much higher than
Hg is diluted by the added water vapor. The Po2 of inspired air, the inspired Pco2 but lower than the alveolar Pco2 , or about 27
or PIo2 (saturated with water vapor at a standard barometric mm Hg. The expected partial pressures of oxygen, carbon
pressure), then is equal to the fractional concentration of dioxide, nitrogen, and water vapor in dry air, inspired air,
inspired oxygen (FIo2) times the barometric pressure minus the alveolar air, and expired air at an atmospheric pressure of
water vapor pressure: 760 mm Hg are shown in Table 33–1.

PIo2 = FIo2 (PB – PH2O) (11)


where PB is the barometric pressure and PH2O the water vapor ALVEOLAR VENTILATION
pressure. Then we have: AND CARBON DIOXIDE
0.2093(760 – 47) mm Hg = 149 mm Hg (12)
The concentration of carbon dioxide in the alveolar gas is, as
The Pco2 of inspired air (PIco2 ) is equal to FIco2 (PB – PH2O) or already discussed, dependent on the alveolar ventilation and
0.0004(760 – 47) mm Hg = 0.29 mm Hg (rounded up to 0.3 on the rate of carbon dioxide production by the body (and its
mm Hg). delivery to the lung in the mixed venous blood).. The volume
Alveolar gas is composed of 2.5–3.0 L of gas already in the of carbon dioxide expired. per unit of time (VEco2 ) is equal to
lungs at the FRC and approximately 350 mL per breath enter- the alveolar ventilation (VA ) times the alveolar fractional con-
ing and leaving the alveoli. About 300 mL of oxygen is con- centration of CO2 (FAco2 ). No carbon dioxide comes from the
tinuously diffusing from the alveoli into the pulmonary dead space:
capillary blood per minute at rest and is being replaced by . .
VEco2 = VA FAco2 (13)
alveolar ventilation. Similarly, about 250 mL of carbon dioxide
is diffusing from the mixed venous blood in the pulmonary Similarly, the fractional concentration of carbon dioxide in
capillaries into the alveoli per minute and is then removed by the alveoli is directly proportional
. to the carbon dioxide pro-
alveolar ventilation. (The Po2 and Pco2 of mixed venous blood duction by the body (Vco2) and inversely proportional to the
are about 40 and 45–46 mm Hg, respectively.) Therefore, the alveolar ventilation:
.
partial pressures of oxygen and carbon dioxide in the alveolar Vco
air are determined by the alveolar ventilation, pulmonary cap- FAco2 ∝ ___
. 2
(14)
VA
illary perfusion, oxygen consumption, and carbon dioxide
Since FAco2 (PB – PH2O) = PAco2 , we have:
production. Alveolar ventilation is normally adjusted by the .
V
respiratory control center in the brain to keep mean arterial ___
PAco2 ∝ co. 2 (15)
and alveolar Pco2 at about 40 mm Hg (see Chapter 38). Mean VA
alveolar Po2 is about 104 mm Hg (usually considered to be 100 In healthy people, alveolar Pco2 is in equilibrium with arterial
mm Hg for convenience). Pco2 (Paco2 ). Thus, if alveolar ventilation is doubled (and car-
The alveolar Po2 increases by 2–4 mm Hg with each nor- bon dioxide production is unchanged), then the alveolar and
mal tidal inspiration and decreases slowly until the next arterial Pco2 are reduced by one half. If alveolar ventilation is
inspiration. Similarly, the alveolar Pco2 decreases 2–4 mm Hg cut in half, then alveolar and arterial Pco2 will double. This can
with each inspiration and increases slowly until the next be seen in the upper part of Figure 33–6.
338 SECTION VI Pulmonary Physiology

150 REGIONAL DISTRIBUTION OF


ALVEOLAR VENTILATION
100 As previously discussed, a 70-kg person has about 2.5–3.0 L of
PACO2
(mm Hg) gas in the lungs at the FRC. Each breath brings about 350 mL
of fresh gas into the alveoli and removes about 350 mL of alve-
50 olar air from the lung. Studies performed on healthy subjects
standing or seated upright have shown that alveoli in the lower
regions of the lungs receive more ventilation per unit volume
0 than do those in the upper regions of the lung. The lower
2 4 6 8 10 regions of the lung are relatively better ventilated than the
Alveolar ventilation (L/min) upper regions of the lung.
If a similar study is done on a subject lying on his or her left
side, the regional differences in ventilation between the ana-
150 tomic upper, middle, and lower regions of the lung disappear,
although there is better relative ventilation of the left lung than
of the right lung. The regional differences in ventilation are
100 therefore mainly a result of the effects of gravity, with regions
of the lung lower with respect to gravity (the “dependent”
PAO2
regions) relatively better ventilated than those regions above
(mm Hg)
them (the “nondependent” regions).
50
The explanation for these regional differences in ventilation
is regional differences in intrapleural pressure. In Chapter 32,
the intrapleural surface pressure was discussed as if it were
0 uniform throughout the thorax, which is not the case. The
2 4 6 8 10
Alveolar ventilation (L/min) intrapleural surface pressure is less negative in the lower, grav-
ity-dependent regions of the thorax than it is in the upper,
FIGURE 33–6 Predicted alveolar gas tensions for different nondependent regions. There is a gradient of the intrapleural
levels of alveolar ventilation. (Modified with permission from Nunn JF: surface pressure such that for every centimeter of vertical dis-
Applied Respiratory Physiology, 4th ed. 1993. Reprinted by permission of Elsevier
placement down the lung (from nondependent to dependent
Science Limited.)
regions), the intrapleural surface pressure increases by about
+0.2 to +0.5 cm H2O. This gradient is caused by gravity and by
ALVEOLAR VENTILATION AND OXYGEN mechanical interactions between the lung and the chest wall.
The influence of this gradient of intrapleural surface pres-
As alveolar ventilation increases, the alveolar Po2 will also sure on regional alveolar ventilation can be explained by
increase. Doubling alveolar ventilation, however, cannot double predicting its effect on the transpulmonary pressure gradients
PAo2 in a person whose alveolar Po2 is already 104 mm Hg in upper and lower regions of the lung. In the left side of
because the highest PAo2 one could possibly achieve breathing Figure 33–7, alveolar pressure is assumed to be zero in both
air at sea level is the inspired Po2 of about 149 mm Hg. The alve- regions of the lung at the FRC, as discussed in Chapter 32.
olar Po2 can be calculated by using the alveolar air equation: Since the intrapleural pressure is more negative in upper
PAco regions of the lung than it is in lower regions of the lung, the
PAo2 = PIo2 – ____
R
+F2
(16) transpulmonary pressure (alveolar minus intrapleural) is
.
Vco2 greater in upper regions of the lung than it is in lower regions
where R is the respiratory exchange ratio ( ___
. ) and F a small
Vo2 of the lung. Because the alveoli in upper regions of the lung are
correction factor.
subjected to greater distending pressures than those in more
As already noted, PIo2 = FIo2(PB – PH2O). F is usually ignored.
dependent regions of the lung, they have greater volumes than
Therefore, we have:
the alveoli in more dependent regions.
PA
PAo2 = FIo2(PB – PH2O)– ____
co 2
(17) It is this difference in volume that leads to the difference in
R
ventilation between alveoli located in dependent and nonde-
As alveolar ventilation increases, the alveolar Pco2 decreases, pendent regions of the lung. This can be seen on the hypo-
bringing the alveolar Po2 closer to the inspired Po2, as can be thetical pressure–volume curve shown on the right side of
seen in the lower part of Figure 33–6. Note that the alveolar Po2 Figure 33–7. This curve is similar to the pressure–volume
obtained using the alveolar air equation is a calculated ideal- curve for a whole lung shown in Figure 32–6, except that this
ized average alveolar Po2. It represents what alveolar Po2 should curve is drawn with the pressure–volume characteristics of
be, not necessarily what it is. single alveoli in mind. The abscissa is the transpulmonary
CHAPTER 33 Alveolar Ventilation 339

pressure (alveolar pressure minus intrapleural pressure). The volumes, so they have relatively more ventilation of nonde-
ordinate is the volume of the alveolus expressed as a percent of pendent alveoli.
its maximum.
Because of the greater transpulmonary pressure, the alveo-
lus in the upper region of the lung has a greater volume than THE CLOSING VOLUME
the alveolus in a more gravity-dependent region of the lung. At
the FRC, the alveolus in the upper part of the lung is on a less During a forced expiration, the lung volume at which airway
steep portion of the alveolar pressure–volume curve (i.e., it is closure begins to occur is known as the closing capacity; the
less compliant) in Figure 33–7 than is the more compliant volume of air exhaled from the time the first airways close
alveolus in the lower region of the lung. Therefore, any change until the subject reaches the RV and can exhale no more air is
in the transpulmonary pressure during a normal respiratory called the closing volume. (The terms are often used inter-
cycle will cause a greater change in volume in the alveolus in changeably.) Because people with emphysema have dimin-
the lower, gravity-dependent region of the lung than it will in ished alveolar elastic recoil to provide traction on the airways
the alveolus in the nondependent region of the lung, as shown and help get air out of the alveoli during a forced expiration,
by the arrows in the figure. Because the alveoli in the lower they have very high closing capacities. That is, their airways
parts of the lung have a greater change in volume per inspira- begin to close at high lung volumes, trapping gas in the affected
tion and per expiration, they are better ventilated than those alveoli. They learn to breathe at higher lung volumes to opti-
alveoli in nondependent regions (during eupneic breathing mize their elastic recoil. As discussed in Chapter 73, even
from the FRC). healthy people lose alveolar elastic recoil as they age, resulting
A second effect of the intrapleural pressure gradient in a in higher closing capacities.
person seated upright is on regional static lung volume, as is
evident from the above discussion. At the FRC, most of the
alveolar air is in upper regions of the lung because those alve-
oli have larger volumes. Most of the ERV is also in upper por- CLINICAL CORRELATION
tions of the lung. On the other hand, most of the IRV and IC A 38-year-old man with an obvious curvature of the spine
are in lower regions of the lung. Even at low lung volumes, the in the coronal and sagittal planes is seen by a pulmonolo-
upper alveoli are larger in volume than are the lower gravity- gist because of dyspnea that has gotten worse during
dependent alveoli. They therefore constitute most of the RV. the last few months. He is 163-cm (5′4″) tall and weighs
Patients with emphysema have greatly decreased alveolar 61.2 kg (135 lb). Blood pressure is 135/95 mm Hg, heart
elastic recoil, leading to high FRCs, extremely high RVs, and rate is 80/min, and his respiratory rate is tachypnic at
airway closure in dependent parts of the lung even at high lung

FRC
100
Pleural pressure (cm H2O)

8.5
80

35 cm

60
Volume (%)

1.5
40

20

0
10 0 10 20 30 40
Transpulmonary pressure (cm H2O)

FIGURE 33–7 Effect of the pleural surface pressure gradient on the distribution of inspired gas at the functional residual capacity
(FRC). (Modified with permission from Milic-Emili J: Pulmonary statics. In: Widdicombe JG, ed. MTP International Review of Sciences: Respiratory Physiology. London, England:
Butterworth; 1974:105–137.)
340 SECTION VI Pulmonary Physiology

25 breaths/min. His respiratory muscle strength appears


CHAPTER SUMMARY
to be normal. The pulmonologist orders pulmonary func- ■ Alveolar ventilation is less than the volume of air entering or
tion tests and an arterial blood gas (with reference ranges leaving the nose or mouth per minute (the minute volume)
because the last part of each inspiration remains in the
in parenthesis) with the following results:
conducting airways (the anatomic dead space).
TLC: 45% of predicted;
■ Alveoli that are ventilated but not perfused constitute alveolar
VC: 40% of predicted;
dead space.
RV: 75% of predicted;
■ The physiologic dead space is the sum of the anatomic dead
FRC: 50% of predicted;
space and the alveolar dead space.
FEV1: 40% of predicted;
FVC: 40% of predicted; ■ At constant carbon dioxide production, alveolar Pco2 is
approximately inversely proportional to alveolar ventilation;
FEV1/FVC: 80% of predicted;
alveolar Po2 is calculated using the alveolar air equation.
arterial Po2: 75 mm Hg (80–100 mm Hg);
■ At or near the FRC, alveoli in lower regions of the upright lung
arterial Pco2: 46 mm Hg (35–45 mm Hg);
are relatively better ventilated than those in upper regions of
arterial pH: 7.38 (7.35–7.45).
the lung.
The patient has kyphoscoliosis that is a lateral curva-
ture of the spine (scoliosis) as well as a sagittal curvature
(kyphosis). It can be congenital; secondary to many disor- STUDY QUESTIONS
ders, including muscular dystrophy, poliomyelitis, spina
1. Which of the following conditions are reasonable explanations
bifida, and cerebral palsy; or it may be idiopathic (of for a functional residual capacity that is significantly less than
unknown cause). Kyphoscoliosis results in decreased predicted?
compliance of the rib cage with much less outward recoil A) third trimester of pregnancy
of the chest wall at low thoracic volumes and much greater B) pulmonary fibrosis
inward recoil at higher volumes. Kyphoscoliosis is there- C) obesity
fore a restrictive disease. D) emphysema
It is difficult for the patient to breathe in and, as a E) all of the above
result, his inspiratory work of breathing is increased. It F) A, B, and C
explains his increased resting respiratory rate (normally 2–5. An unconscious patient’s ventilation is maintained with
12–15 breaths/min) because taking smaller tidal volumes positive-pressure ventilation with a tidal volume of 450 mL and
at an increased breathing frequency decreases his work a rate of 10 breaths/min. She weighs 100 lb. Her arterial Pco2 is
of breathing. The effects of the changes in the mechanics 42 mm Hg, her end-tidal Pco2 is 35 mm Hg, and her mixed expired
of his thorax can be seen in the lung volumes and capac- Pco2 is 28 mm Hg.
ities determined in this patient (see Figure 33–2). His 2. What is her minute volume?
FRC is low because, with less outward recoil of his chest A) 350 mL/min
wall, the balance point between the outward recoil of the B) 1,000 mL/min
chest wall and the inward recoil of his lungs occurs at a C) 3,500 mL/min
lower lung volume. His TLC is low because his ability to D) 4,500 mL/min
inhale maximally is severely impaired. His RV is also E) 5,500 mL/min
lower than predicted, but not as much as the TLC, 3. What is her alveolar ventilation?
because his ability to exhale is not as impaired. His VC, A) 350 mL/min
FVC, and FEV1 are all lower than predicted because his B) 1,000 mL/min
TLC is very low—he cannot exhale very much because C) 3,500 mL/min
D) 4,500 mL/min
he is unable to inhale very much. On the other hand, this
E) 5,500 mL/min
patient does not have airway obstruction. Although both
4. What is her physiologic dead space?
his FEV1 and FVC are low, the FEV1/FVC is within the
normal range. The blood gases demonstrate that the A) 50 mL
B) 100 mL
increased work of breathing has resulted in decreased
C) 150 mL
alveolar ventilation. His arterial Pco2 is high and his arte-
D) 200 mL
rial Po2 is low. E) 300 mL
Treatment of patients with kyphoscoliosis is aimed at
5. What is her alveolar dead space?
improving alveolar ventilation, for example, with nonin-
A) 50 mL
vasive mechanical ventilation at night. Orthopedic sur-
B) 100 mL
gery to help correct the problem may be effective in some C) 150 mL
patients. D) 200 mL
E) 300 mL
34
C H A P T E R

Pulmonary Perfusion
Michael Levitzky

O B J E C T I V E S

■ Compare and contrast the bronchial circulation and the pulmonary


circulation.
■ Describe the anatomy of the pulmonary circulation, and explain its
physiologic consequences.
■ Compare and contrast the pulmonary circulation and the systemic
circulation.
■ Describe and explain the effects of lung volume on pulmonary vascular
resistance.
■ Describe and explain the effects of elevated intravascular pressures on
pulmonary vascular resistance.
■ List the neural and humoral factors that influence pulmonary vascular
resistance.
■ Describe the effect of gravity on pulmonary blood flow.
■ Describe the interrelationships of alveolar pressure, pulmonary arterial
pressure, and pulmonary venous pressure and their effects on the regional
distribution of pulmonary blood flow.
■ Predict the effects of alterations in alveolar pressure, pulmonary arterial
and venous pressures, and body position on the regional distribution of
pulmonary blood flow.
■ Describe hypoxic pulmonary vasoconstriction and discuss its role in localized
and widespread alveolar hypoxia.
■ Describe the causes and consequences of pulmonary edema.

The lung receives blood flow via both the bronchial circula- About 280 billion pulmonary capillaries supply the approxi-
tion and the pulmonary circulation. Bronchial blood flow mately 300–480 million alveoli, resulting in a potential surface
constitutes a very small portion of the output of the left area for gas exchange estimated to be 50–100 m2. As was shown
ventricle and supplies part of the tracheobronchial tree with in Figure 31—5, the alveoli are completely enveloped in pul-
systemic arterial blood. Pulmonary blood flow constitutes monary capillaries.
the entire output of the right ventricle and supplies the lung
with the mixed venous blood draining all the tissues of the
body. It is this blood that undergoes gas exchange with the
alveolar air in the pulmonary capillaries. Because the right and
THE BRONCHIAL CIRCULATION
left ventricles are arranged in series after birth, pulmonary The bronchial arteries supply arterial blood to the tracheo-
blood flow is approximately equal to 100% of the output of the bronchial tree and to other structures of the lung down to the
left ventricle (the cardiac output). level of the terminal bronchioles. They also provide blood flow

341

Ch34_341-352.indd 341 11/26/10 11:50:35 AM


342 SECTION VI Pulmonary Physiology

CONDUCTING AIRWAYS

ALVEOLI

PULMONARY
PULMONARY 12 8
CAPILLARIES PULMONARY
ARTERY
(Mean  15) VEINS
25/8

VENTRICLE ATRIUM
25/0 5
RIGHT LEFT
ATRIUM VENTRICLE
2 120/0
120/80
VEINS AORTA
(Mean  100)
SYSTEMIC
15 CAPILLARIES 30

TISSUES

FIGURE 34–1 Pressures, expressed in mm Hg, in the systemic and pulmonary circulations. (Modified with permission from Levitzky MG:
Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

to other thoracic structures. Lung structures distal to the ter- branches. The pulmonary artery rapidly subdivides into ter-
minal bronchioles, including the respiratory bronchioles, alve- minal branches that have thinner walls and greater internal
olar ducts, alveolar sacs, and alveoli, receive oxygen directly by diameters than do corresponding branches of the systemic
diffusion from the alveolar air and nutrients from the mixed arterial tree. There is much less vascular smooth muscle in the
venous blood in the pulmonary circulation. The bronchial cir- walls of the vessels of the pulmonary arterial tree, and there
culation may be important in the “air conditioning” of inspired are no highly muscular vessels that correspond to the systemic
air (discussed in Chapter 31). arterioles. The pulmonary arterial tree rapidly subdivides over
The blood flow in the bronchial circulation constitutes about a short distance, ultimately branching into the approximately
2% of the output of the left ventricle. Blood pressure in the bron- 280 billion pulmonary capillaries, where gas exchange occurs.
chial arteries is the same as that in the other systemic arteries.
This is much higher than the blood pressure in the pulmonary
arteries (Figure 34–1). The venous drainage of the bronchial cir- PULMONARY VASCULAR
culation is atypical. Although some of the bronchial venous
blood enters the azygos and hemiazygos veins, a substantial RESISTANCE
portion of bronchial venous blood enters the pulmonary veins.
The blood in the pulmonary veins has undergone gas exchange The thin walls and small amount of smooth muscle found in
with the alveolar air—that is, the pulmonary veins contain the pulmonary arteries have important physiologic conse-
“arterial” blood. Therefore, the bronchial venous blood entering quences. The pulmonary vessels offer much less resistance to
the pulmonary venous blood is part of the normal anatomic blood flow than do the systemic arterial vessels. They are also
right-to-left shunt, which will be discussed in Chapter 35. much more distensible and compressible than systemic arterial
vessels. These factors lead to much lower intravascular pres-
sures than those found in the systemic arteries. The pulmo-
THE PULMONARY CIRCULATION nary vessels are located in the thorax and are subject to alveolar
and intrapleural pressures that can change greatly, ranging
The walls of the vessels of the pulmonary circulation are much from as low as −80 cm H2O during a maximal inspiratory
thinner than corresponding parts of the systemic circulation. effort to more than 100 cm H2O during a maximal forced
This is particularly true of the main pulmonary artery and its expiration. Therefore, factors other than the tone of the
CHAPTER 34 Pulmonary Perfusion 343

pulmonary vascular smooth muscle may have profound effects Another consequence of the pressure differences between
on pulmonary vascular resistance (PVR). the systemic and pulmonary circulations is that the workload
PVR cannot be measured directly but an approximation can and metabolic demand of the left ventricle is much greater
be calculated using the Poiseuille equation, as was discussed in than that of the right ventricle. The difference in wall thick-
Chapter 22. For the pulmonary circulation, the PVR is equal to ness of the left and right ventricles of the adult is a reminder of
the mean pulmonary artery pressure (Pa, the upstream pressure) the much greater workload of the left ventricle.
minus the mean left atrial pressure (the downstream pressure), The relatively small amounts of vascular smooth muscle,
divided by pulmonary blood flow (the cardiac output). However, low intravascular pressures, and high distensibility of the pul-
the mean left atrial pressure may not be the effective downstream monary circulation lead to a much greater importance of
pressure for the calculation of PVR under all lung conditions extravascular effects (passive factors) on PVR. Gravity, body
(see the section on zones of the lung later in this chapter). position, lung volume, alveolar and intrapleural pressures,
Because the right and left circulations are in series, the out- intravascular pressures, and right ventricular output all can
puts of the right and left ventricles must be approximately equal have profound effects on PVR without any alteration in the
to each other. (If they are not, blood and fluid will build up in tone of the pulmonary vascular smooth muscle.
the lungs or periphery.) If the two outputs are the same and the
measured pressure drops across the systemic circulation and
the pulmonary circulation are about 98 and 10 mm Hg, respec- LUNG VOLUME AND PULMONARY
tively (see Figure 34–1), then the PVR must be about one tenth VASCULAR RESISTANCE
that of the total peripheral resistance (TPR). TPR is some-
times called systemic vascular resistance (SVR). For distensible–compressible vessels, the transmural pressure
gradient is an important determinant of the vessel diameter
(see discussion of airway resistance in Chapter 32). As the
DISTRIBUTION OF PULMONARY transmural pressure gradient (which is equal to pressure inside
VASCULAR RESISTANCE minus pressure outside) increases, the vessel diameter increases
and resistance decreases; as the transmural pressure decreases,
The distribution of PVR can be estimated by the decrease in the vessel diameter decreases and the resistance increases.
pressure across each of the three major components of the pul- Negative transmural pressure gradients lead to compression or
monary vasculature: the pulmonary arteries, the pulmonary cap- even collapse of the vessel.
illaries, and the pulmonary veins. In Figure 34–1, the resistance Two different groups of pulmonary vessels must be consid-
is fairly evenly distributed among the three components. At rest, ered when the effects of lung volume changes on PVR are
about one third of the resistance to blood flow is located in the analyzed—namely, the alveolar and extra-alveolar vessels
pulmonary arteries, about one third is located in the pulmonary (Figure 34–2).
capillaries, and about one third is located in the pulmonary veins.
This is in contrast to the systemic circulation, in which about
70% of the resistance to blood flow is located in the systemic
arteries, mostly in the highly muscular systemic arterioles.

CONSEQUENCES OF DIFFERENCES IN ALVEOLUS

PRESSURE BETWEEN THE SYSTEMIC


AND PULMONARY CIRCULATIONS “extraalveolar” “alveolar”
The left ventricle must maintain a relatively high mean arterial
pressure because such high pressures are necessary to overcome
hydrostatic forces and pump blood to the brain. The apices of the
ALVEOLUS
lungs are a much shorter distance above the right ventricle, so
such high pressures are unnecessary. The high arterial pressure
in the systemic circulation allows the redistribution of left ven-
tricular output and the control of blood flow to different tissues. During inspiration
In the pulmonary circulation, redistribution of right ventricular
output is usually unnecessary because all alveolar–capillary units FIGURE 34–2 Illustration of alveolar and extra-alveolar
pulmonary vessels during an inspiration. The alveolar vessels
that are participating in gas exchange are performing the same
(pulmonary capillaries) are exposed to the expanding alveoli and
function. The pressure is low and the small amount of smooth
elongated. The extra-alveolar vessels, here shown exposed to the
muscle in the pulmonary vessels (which is in large part respon- intrapleural pressure, expand as the intrapleural pressure becomes
sible for the low pressure head) makes such local redistributions more negative and as radial traction increases during the inspiration.
unlikely. An exception to this will be described in the section (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
“Hypoxic Pulmonary Vasoconstriction.” New York: McGraw-Hill Medical, 2007.)
344 SECTION VI Pulmonary Physiology

As lung volume increases during a normal negative-pressure other, the resistances of the alveolar and extra-alveolar vessels
inspiration, the alveoli increase in volume. While the alveoli are additive at any lung volume. Thus, the effect of changes in
expand, the vessels between them, mainly pulmonary capillar- lung volume on the total PVR gives the U-shaped curve seen
ies, are elongated. As these vessels are stretched, their diame- in Figure 34–3. PVR is lowest near the functional residual
ters decrease, just as stretching a rubber tube causes its capacity and increases at both high and low lung volumes.
diameter to narrow. Resistance to blood flow through the alve- Also note that during mechanical positive-pressure ventila-
olar vessels increases as the alveoli expand because the alveolar tion, alveolar pressure (PA) and intrapleural pressure are positive
vessels are longer and because their radii are smaller. At high during inspiration. In this case, both the alveolar and extra-
lung volumes, then, the resistance to blood flow offered by the alveolar vessels are compressed as lung volume increases.
alveolar vessels increases; at low lung volumes, the resistance
to blood flow offered by the alveolar vessels decreases.
One group of the extra-alveolar vessels, the larger arteries RECRUITMENT AND DISTENTION
and veins, is exposed to the intrapleural pressure. As lung vol-
ume is increased by making the intrapleural pressure more During exercise, cardiac output can increase several-fold with-
negative, the transmural pressure gradient of the larger arteries out a correspondingly great increase in mean pulmonary
and veins increases and they distend. Another factor tending artery pressure. Although the mean pulmonary artery pres-
to decrease the resistance to blood flow offered by the extra- sure does increase, the increase is only a few millimeters of
alveolar vessels at higher lung volumes is radial traction by the mercury, even if cardiac output has doubled or tripled. Since
connective tissue and alveolar septa holding the larger vessels the pressure drop across the pulmonary circulation is propor-
in place in the lung. (Look at the small branch of the pulmo- tional to the cardiac output times the PVR (i.e., ΔP = Q̇ × R),
nary artery at the bottom of Figure 31–4.) Thus, at high lung this must indicate a decrease in PVR.
volumes (attained by normal negative-pressure breathing), the Like the effects of lung volume on PVR, this decrease
resistance to blood flow offered by the extra-alveolar vessels appears to be passive—that is, it is not a result of changes in the
decreases. During a forced expiration to low lung volumes, tone of pulmonary vascular smooth muscle caused by neural
however, intrapleural pressure becomes very positive. Extra- mechanisms or humoral agents. In fact, a decrease in PVR in
alveolar vessels are compressed, and as the alveoli decrease in response to increased blood flow or even an increase in perfu-
size, they exert less radial traction on the extra-alveolar vessels. sion pressure can be demonstrated in a vascularly isolated per-
The resistance to blood flow offered by the extra-alveolar ves- fused lung, as was used to obtain the data summarized in
sels therefore increases (see left side of Figure 34–3). Figure 34–4. (Note that the graph has blood pressure on the
Because the alveolar and extra-alveolar vessels may be x-axis; blood flow has a similar effect.) Increasing the left atrial
thought of as two groups of resistances in series with each pressure also decreases PVR.
There are two different mechanisms that can explain
this decrease in PVR in response to elevated blood flow and

Stretching of pulmonary
Pulmonary vascular resistance

capillaries
Pulmonary vascular resistance

Compression of extra- Recruitment & distention


alveolar vessels of pulmonary capillaries

FRC = Lowest PVR

Lung volume
Pulmonary blood flow
FIGURE 34–3 The effects of lung volume on pulmonary
vascular resistance (PVR). PVR is lowest near the functional residual FIGURE 34–4 The effect of pulmonary blood flow (or blood
capacity (FRC) and increases at both high and low lung volumes pressure) on pulmonary vascular resistance. Increased pulmonary
because of the combined effects on the alveolar and extra-alveolar artery blood pressure or pulmonary blood flow decreases pulmonary
vessels. (Reproduced with permission from Kibble J, Halsey CR: The Big Picture, vascular resistance. (Reproduced with permission from Kibble J, Halsey CR: The
Medical Physiology. New York: McGraw-Hill, 2009.) Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)
CHAPTER 34 Pulmonary Perfusion 345

Recruitment Distention
FIGURE 34–5 Illustration of the mechanisms by which increased mean pulmonary artery pressure may decrease pulmonary
vascular resistance. The upper figure shows a group of pulmonary capillaries, some of which are perfused. At left, the previously unperfused
capillaries are recruited (opened) by the increased perfusion pressure. At right, the increased perfusion pressure has distended those vessels
already open. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

perfusion pressure: recruitment and distention (Figure 34–5). may decrease alveolar dead space. Derecruitment caused by
As indicated in the diagram, at resting cardiac outputs, not all low right ventricular output or high alveolar pressures
the pulmonary capillaries are perfused. A substantial number decreases the surface area for gas exchange and may increase
of capillaries are probably unperfused because of hydrostatic alveolar dead space.
effects that will be discussed later in this chapter. Others may
be unperfused because they have a relatively high critical
opening pressure. That is, these vessels, because of their high CONTROL OF PULMONARY
vascular smooth muscle tone or other factors such as positive VASCULAR SMOOTH MUSCLE
alveolar pressure, require a higher perfusion pressure than
that solely necessary to overcome hydrostatic forces. Under Pulmonary vascular smooth muscle is responsive to both neu-
normal circumstances, it is not likely that the critical opening ral and humoral influences. These produce active alterations
pressures for pulmonary blood vessels are very great because in PVR, as opposed to the passive factors discussed in the pre-
they have so little smooth muscle. Increasing blood flow vious section. A final passive factor, gravity, will be discussed
increases the mean pulmonary artery pressure, which opposes later in this chapter. The main passive and active factors that
hydrostatic forces and exceeds the critical opening pressure in influence PVR are summarized in Tables 34–1 and 34–2.
previously unopened vessels. This series of events opens new The pulmonary vasculature is innervated by both sympa-
parallel pathways for blood flow, which lowers the PVR. This thetic and parasympathetic fibers of the autonomic nervous
opening of new pathways is called recruitment. Note that system. The innervation of pulmonary vessels is relatively
decreasing the cardiac output or pulmonary artery pressure sparse in comparison with that of systemic vessels, and the
can result in a derecruitment of pulmonary capillaries. autonomic nervous system has much less influence on the pul-
As perfusion pressure increases, the transmural pressure monary vessels. There is relatively more innervation of the
gradient of the pulmonary blood vessels increases, causing larger vessels and less of the smaller, more muscular vessels.
distention of the vessels. This increases their radii and decreases There appears to be no innervation of vessels smaller than
their resistance to blood flow. 30 μm in diameter, with little innervation of intrapulmonary
Both recruitment and distention cause the decreased PVR veins and venules.
with elevated perfusion pressure or blood flow. Note that Sympathetic stimulation of the innervation of the pulmonary
recruitment increases the surface area for gas exchange and vasculature may increase PVR or decrease the distensibility of
346 SECTION VI Pulmonary Physiology

TABLE 34–1 Passive influences on pulmonary vascular resistance.


Effect on Pulmonary
Cause Vascular Resistance Mechanism

Increased lung volume (above FRC) Increases Lengthening and compression of alveolar vessels

Decreased lung volume (below FRC) Increases Compression of and less traction on
extra-alveolar vessels

Increased pulmonary artery pressure, increased left Decreases Recruitment and distention
atrial pressure, increased pulmonary blood volume,
increased cardiac output

Gravity, body position Decreases in gravity-dependent Hydrostatic effects lead to recruitment and distention
regions of the lungs

Increased (more positive) interstitial pressure Increases Compression of vessels

Increased blood viscosity Increases Viscosity directly increases resistance

Positive-pressure ventilation

Increased alveolar pressure Increases Compression and derecruitment of alveolar vessels

Positive intrapleural pressure Increases Compression of extra-alveolar vessels; compression of


vena cava decreases pulmonary blood flow and leads
to derecruitment
FRC, functional residual capacity.

the larger vessels. Stimulation of the parasympathetic innerva- are also pulmonary vasoconstrictors, as is endothelin. Alveo-
tion of the pulmonary vessels generally causes vasodilation. lar hypoxia and hypercapnia also cause pulmonary vasocon-
The catecholamines epinephrine and norepinephrine striction, as will be discussed later in this chapter.
both increase PVR when injected into the pulmonary circula- Acetylcholine, the β-adrenergic agonist isoproterenol, nitric
tion. Histamine, found in the lung in mast cells, is also a oxide (NO), and certain prostaglandins, such as PGE1 and
pulmonary vasoconstrictor. Certain prostaglandins and PGI2 (prostacyclin), are pulmonary vasodilators.
related substances, such as PGF2α, PGE2, and thromboxane,

TABLE 34–2 Active influences on pulmonary


THE REGIONAL DISTRIBUTION OF
vascular resistance. PULMONARY BLOOD FLOW: THE
Increase Decrease ZONES OF THE LUNG
Stimulation of sympathetic Stimulation of parasympathetic Gravity is another important passive factor affecting local PVR
innervation (may have greater innervation (if vascular tone is and the relative perfusion of different regions of the lung. The
effect by decreasing large-vessel already elevated)
interaction of the effects of gravity and extravascular pressures
distensibility)
may have a profound influence on the relative perfusion of dif-
Norepinephrine, epinephrine Acetylcholine ferent areas of the lung.
α-Adrenergic agonists β-Adrenergic agonists

PGF2α, PGE2 PGE1 THE REGIONAL DISTRIBUTION


Thromboxane Prostacyclin (PGI2) OF PULMONARY BLOOD FLOW
Endothelin Nitric oxide
If a radioactive substance such as the gas xenon (133Xe) is dis-
Angiotensin Bradykinin
solved in saline and infused into the venous blood, it can be
Histamine (primarily a used to determine regional pulmonary blood flow. The greater
pulmonary venoconstrictor) the radioactivity measured over a specific region, the greater the
Alveolar hypoxia blood flow. A pattern like that shown in Figure 34–6 is observed
Alveolar hypercapnia
in a healthy person seated upright or standing up. There is
greater blood flow per unit volume (per alveolus) to lower
Low pH of mixed venous blood regions of the lung than to upper regions of the lung. Note that
PG, prostaglandin. the test was made with the subject at the total lung capacity.
CHAPTER 34 Pulmonary Perfusion 347

THE INTERACTION OF GRAVITY


150 AND EXTRAVASCULAR PRESSURE:
TLC
THE ZONES OF THE LUNG
Blood flow/alveolus ( %)

When the pulmonary artery pressure is low, the uppermost


100 regions of the lung receive no blood flow. Perfusion of the lung
ceases at the point at which alveolar pressure (PA) is just equal
to pulmonary artery pressure (Pa). Above this point, there is
no perfusion because alveolar pressure exceeds pulmonary
50 artery pressure, and the transmural pressure across capillary
walls is negative. Below this point, perfusion per unit volume
increases steadily with increased distance down the lung.
bottom rib 2 Thus, under circumstances in which alveolar pressure is
0 greater than pulmonary artery pressure in the upper parts of
20 15 10 5 0
Lung distance (cm below rib 2) the lung, no blood flow occurs in that region, and the region is
referred to as being in zone 1, as shown in Figure 34–7. (Note
FIGURE 34–6 Relative blood flow per alveolus (100% = that in this figure blood flow is on the x-axis and that distance
perfusion of each alveolus if all were perfused equally) versus up the lung is on the y-axis.) Any zone 1, then, is alveoli that
distance from the bottom of the lung in a human seated upright.
are ventilated but not perfused. It is alveolar dead space. For-
Measurement of regional blood flow was determined using an
intravenous injection of 133Xe. TLC, total lung capacity. (Modified with
tunately, during normal, quiet breathing in a person with a
permission from Hughes JM, Glazier JB, Maloney JE, West JB. Effect of lung volume
normal cardiac output, pulmonary artery pressure, even in the
on the distribution of pulmonary blood flow in man. Respir Physiol. 1968;4(1):58–72.) uppermost regions of the lung, is greater than alveolar pres-
sure, so there is no zone 1.
The lower portion of the lung in Figure 34–7 is said to be in
zone 3. In this region, the pulmonary artery pressure and the
If the subject lies down, this pattern of regional perfusion is
pulmonary vein pressure (Pv) are both greater than alveolar
altered so that perfusion to the anatomically upper and lower
pressure. The driving pressure (ΔP) for blood flow through the
portions of the lung is roughly evenly distributed, but blood
lung in this region is pulmonary artery pressure minus pulmo-
flow per unit volume is still greater in the more gravity-
nary vein pressure. Note that this driving pressure stays con-
dependent regions of the lung. For example, if the subject were
stant as one moves further down the lung in zone 3 because
to lie down on the left side, the left lung would receive more
the hydrostatic pressure effects are the same for both the arter-
blood flow per unit volume than would the right lung. Exercise,
ies and the veins.
which increases the cardiac output, increases the blood flow per
unit volume to all regions of the lung, but the perfusion gradi-
ent persists so that there is still relatively greater blood flow per
unit volume in more gravity-dependent regions of the lung.
The reason for this gradient of regional perfusion of the
lung is gravity. The pressure at the bottom of a column of a Zone 1
liquid is proportional to the height of the column times the PA>Pa>Pv
density of the liquid times gravity, so the intravascular pres-
sures in more gravity-dependent portions of the lung are
greater than those in upper regions. Because the pressures are Zone 2
greater in the more gravity-dependent regions of the lung, the PA Pa>PA>Pv
resistance to blood flow is lower in lower regions of the lung Pa Pv
owing to more recruitment or distention of vessels in these
Distance
regions. It is therefore not only gravity, but also the character-
istics of the pulmonary circulation that cause the increased
blood flow to more gravity-dependent regions of the lung.
After all, the same hydrostatic effects occur to an even greater Zone 3
extent in the systemic circulation, but the thick walls of the Pa>Pv>PA
systemic arteries are not affected. Blood flow
There is also considerable heterogeneity in pulmonary
FIGURE 34–7 The zones of the lung. The effects of gravity
blood flow at any vertical distance up the lung, that is, there and alveolar pressure on the perfusion of the lung. Described in text.
may be significant variations in pulmonary blood flow within (Modified with permission from West, J.B., Dollery, C.T., Naimark, A.: Distribution of
a given horizontal plane of the lung. These variations are blood flow in isolated lung: Relation to vascular and alveolar pressures. J Appl
caused by local factors and mechanical stresses. Physiol 1964;19:713–724.)
348 SECTION VI Pulmonary Physiology

The middle portion of the lung in Figure 34–7 is in zone smooth muscle cells to depolarize, allowing calcium to enter
2. In zone 2, pulmonary artery pressure is greater than alve- the cells. This, in turn, causes them to contract.
olar pressure, so blood flow does occur. Nevertheless, The hypoxic pulmonary vasoconstriction response is
because alveolar pressure is greater than pulmonary vein graded—constriction begins to occur at alveolar Po2’s of
pressure, the effective driving pressure for blood flow is pul- approximately 100 mm Hg and increases until PAo2 decreases
monary artery pressure minus alveolar pressure in zone 2. to about 20–30 mm Hg.
(This is analogous to the situation described in Chapter 32: If an area of the lung becomes hypoxic because of airway
during a forced expiration, the driving pressure for airflow obstruction or if localized atelectasis occurs, any mixed venous
is equal to alveolar pressure minus intrapleural pressure.) blood flowing to that area will undergo little or no gas exchange
Notice that in zone 2 (at right in Figure 34–7), the increase (Figure 34–8) and will mix with blood draining well-ventilated
in blood flow per distance down the lung is greater than it is areas of the lung as it enters the left atrium. This mixing will
in zone 3. This is because the upstream driving pressure, the lower the overall arterial Po2. The hypoxic pulmonary vasocon-
pulmonary artery pressure, increases according to the striction diverts mixed venous blood flow away from poorly
hydrostatic pressure increase, but the effective downstream ventilated areas of the lung by locally increasing vascular resis-
pressure, alveolar pressure, is constant throughout the lung tance, as shown in Figure 34–8C. Therefore, mixed venous blood
at any instant. is sent to better-ventilated areas of the lung (Figure 34–8D). The
To summarize: in zone 1, PA > Pa > Pv , and there is no blood problem with hypoxic pulmonary vasoconstriction is that it is
flow; in zone 2, Pa > PA > Pv, and the effective driving pressure not a very strong response because there is so little smooth mus-
for blood flow is Pa – PA; in zone 3, Pa > Pv > PA, and the driving cle in the pulmonary vasculature. Very high pulmonary artery
pressure for blood flow is Pa – Pv. pressures can interfere with hypoxic pulmonary vasoconstric-
It is important to realize that the boundaries between the tion, as can other physiologic disturbances, such as alkalosis.
zones are dependent on physiologic conditions—they are not In hypoxia of the whole lung, such as might be encountered
fixed anatomic landmarks. Alveolar pressure changes during at high altitude or in hypoventilation, hypoxic pulmonary
the course of each breath. During eupneic breathing these vasoconstriction occurs throughout the lung. Even this may be
changes are only a few centimeters of water, but they may be useful in increasing gas exchange because greatly increasing
much greater during speech, exercise, and other conditions. A the pulmonary artery pressure recruits previously unperfused
patient on a positive-pressure ventilator with positive end- pulmonary capillaries. This increases the surface area available
expiratory pressure (PEEP) may have substantial amounts of for gas diffusion and improves the matching of ventilation and
zone 1 because alveolar pressure is always high. After a hemor- perfusion, as will be discussed in the next chapter. On the other
rhage or during general anesthesia, pulmonary blood flow and hand, such a whole-lung hypoxic pulmonary vasoconstriction
pulmonary artery pressure are low and zone 1 conditions are increases the workload on the right ventricle, and the high pul-
also likely. During exercise, cardiac output and pulmonary monary artery pressure may overwhelm hypoxic pulmonary
artery pressure increase and any existing zone 1 will be vasoconstriction in some parts of the lung, increase the capil-
recruited to zone 2. The boundary between zones 2 and 3 will lary hydrostatic pressure in those vessels, and lead to pulmo-
move upward as well. Changes in lung volume also affect the nary edema (see the next section of this chapter).
regional distribution of pulmonary blood flow and will there- Alveolar hypercapnia (increased carbon dioxide) also
fore affect the boundaries between zones. Finally, changes in causes pulmonary vasoconstriction. Note that both hypoxic
body position alter the orientation of the zones with respect to and hypercapnic pulmonary vasoconstriction are opposite to
the anatomic locations in the lung, but the same relationships what occurs in the systemic circulation.
exist with respect to gravity and alveolar pressure.
PULMONARY EDEMA
HYPOXIC PULMONARY Pulmonary edema is the extravascular accumulation of fluid
VASOCONSTRICTION in the lung. This pathologic condition may be caused by one or
more physiologic abnormalities, but the result is inevitably
Alveolar hypoxia (low alveolar Po2) or atelectasis (collapsed impaired gas transfer. As the edema fluid builds up, first in the
alveoli) causes an active vasoconstriction in the pulmonary circu- interstitium and later in alveoli, diffusion of gases—particularly
lation. The site of vascular smooth muscle constriction appears to oxygen—decreases.
be in the arterial (precapillary) vessels very close to the alveoli. The capillary endothelium is much more permeable to water
The mechanism of hypoxic pulmonary vasoconstriction is and solutes than is the alveolar epithelium. Edema fluid there-
not completely understood. The response occurs locally, that fore accumulates in the interstitium before it accumulates in
is, only in the area of the alveolar hypoxia. Connections to the the alveoli.
central nervous system are not necessary. Hypoxia may act As discussed in Chapter 26, the Starling equation describes
directly on pulmonary vascular smooth muscle to produce the movement of liquid across the capillary endothelium:
hypoxic pulmonary vasoconstriction. Hypoxia inhibits an
outward potassium current, which causes pulmonary vascular Q̇f = Kf [(Pc – Pis) – σ(πpl – πis)] (1)
CHAPTER 34 Pulmonary Perfusion 349

O2 ⴝ 150 mm Hg O2 ⴝ 150 mm Hg
CO2 ⴝ 0 mm Hg CO2 ⴝ 0 mm Hg

O2 ⴝ 100 mm Hg Decreased O2
CO2 ⴝ 40 mm Hg Increased CO2

O2 ⴝ 40 mm Hg O2 ⴝ 100 mm Hg O2 ⴝ 40 mm Hg Decreased O2
CO2 ⴝ 45 mm Hg CO2 ⴝ 40 mm Hg CO2 ⴝ 45 mm Hg Increased CO2

A C

O2 ⴝ 150 mm Hg
CO2 ⴝ 0 mm Hg

O2 ⴝ 100 mm Hg Decreased O2
CO2 ⴝ 40 mm Hg Increased CO2
Decreased O2
Increased CO2

O2 ⴝ 40 mm Hg Decreased O2
CO2 ⴝ 45 mm Hg Increased CO2

B D
FIGURE 34–8 Illustration of the physiologic function of hypoxic pulmonary vasoconstriction (HPV). A) Normal alveolar–capillary
unit. B) Perfusion of a hypoventilated alveolus results in blood with a decreased Po and an increased Pco entering the left atrium. C) HPV
2 2
increases the resistance to blood flow to the hypoventilated alveolus. D) This diverts blood flow away from the hypoventilated alveolus to
better-ventilated alveoli, thus helping to maintainV̇/Q̇ matching. HPV, hypoxic pulmonary vasoconstriction; V̇/Q̇ = ventilation–perfusion ratio.
(Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

where Q̇f is the net flow of fluid, Kf the capillary filtration phatic drainage. The pulmonary capillary hydrostatic pressure
coefficient (this describes the permeability characteristics of often increases as a result of problems in the left side of the
the membrane to fluids), Pc the capillary hydrostatic pres- circulation, such as infarction of the left ventricle, left ventricu-
sure, Pis the hydrostatic pressure of the interstitial fluid, σ the lar failure, or mitral stenosis. As left atrial pressure and pulmo-
reflection coefficient (this describes the ability of the mem- nary venous pressure increase because of accumulating blood,
brane to prevent extravasation of solute particles), πpl the col- the pulmonary capillary hydrostatic pressure also increases.
loid osmotic (oncotic) pressure of the plasma, and πis the Other causes of increased pulmonary capillary hydrostatic
colloid osmotic pressure of the interstitial fluid. pressure include overzealous administration of intravenous
The components of the Starling equation are very useful in fluids and diseases that occlude the pulmonary veins.
understanding the potential causes of pulmonary edema, even The interstitial hydrostatic pressure of the lung is in the
though only the plasma colloid osmotic pressure (πpl) can be range of –5 to –7 mm Hg when a healthy person is at FRC.
measured clinically. Conditions that would decrease (i.e., make it more negative)
the interstitial pressure would increase the tendency for pul-
monary edema to develop. These appear to be limited mainly
CONDITIONS THAT MAY LEAD to potential actions of the health care worker, such as rapid
TO PULMONARY EDEMA evacuation of chest fluids or treatment of a pneumothorax.
Situations that increase alveolar surface tension, for example,
Infections, circulating or inhaled toxins, oxygen toxicity, and when decreased amounts of pulmonary surfactant are present,
other factors that destroy the integrity of the capillary endothe- could also make the interstitial hydrostatic pressure more neg-
lium and increase its permeability lead to localized or general- ative and increase the tendency for the formation of pulmo-
ized pulmonary edema. nary edema. Note that as fluid accumulates in the interstitium,
The pulmonary capillary hydrostatic pressure is estimated the interstitial hydrostatic pressure increases, which helps
to be about 10 mm Hg under normal conditions. If the capil- limit further fluid extravasation.
lary hydrostatic pressure increases dramatically, the filtration Any situation that permits more solute to leave the capillar-
of fluid across the capillary endothelium will increase greatly, ies, such as a decreased reflection coefficient, will lead to more
and enough fluid may leave the capillaries to exceed the lym- fluid movement out of the vascular space.
350 SECTION VI Pulmonary Physiology

Decreases in the colloid osmotic pressure of the plasma, than are necessary for gas exchange at rest: previously
which helps retain fluid in the capillaries, may lead to pulmo- unopened capillaries will be recruited. Obviously, no gas
nary edema. Plasma colloid osmotic pressure, normally in the exchange can occur distal to a particle embedded in and
range of 25–28 mm Hg, decreases with low plasma protein obstructing a capillary, so this mechanism is limited by the
concentration or overadministration of certain intravenous ability of the lung to remove such filtered material. If particles
solutions. On the other hand, increased colloid osmotic pres- are experimentally suspended in venous blood and are then
sure in the interstitium will pull fluid from the capillaries. trapped in the pulmonary circulation, the diffusing capacity
Any fluid that makes its way into the pulmonary intersti- (see Chapter 35) usually decreases for 4–5 days and then
tium must be removed by the lymphatic drainage of the lung. returns to normal. The mechanisms for removal of material
The volume of lymph flow from the human lung is capable of trapped in the pulmonary capillary bed include lytic enzymes
increasing as much as 10-fold under pathologic conditions. It in the vascular endothelium, ingestion by macrophages, and
is only when this large safety factor is overwhelmed that pul- penetration to the lymphatic system. Patients on cardiopul-
monary edema occurs. Conditions that block the lymphatic monary bypass do not have the benefit of this pulmonary cap-
drainage of the lung, such as tumors or scars, may predispose illary filtration, and blood administered to these patients must
patients to pulmonary edema. be filtered for them.
Pulmonary edema can be associated with head injury, her- The colloid osmotic pressure of the plasma proteins nor-
oin overdose, and high altitude. The causes of the edema for- mally exceeds the pulmonary capillary hydrostatic pressure.
mation in these conditions are not known, although This tends to pull fluid from the alveoli into the pulmonary
high-altitude pulmonary edema may be caused by high pul- capillaries and keep the alveolar surface free of liquids other
monary artery pressures secondary to the hypoxic pulmonary than pulmonary surfactant. Water taken into the lungs is rap-
vasoconstriction. idly absorbed into the blood. This protects the gas exchange
function of the lungs and opposes transudation of fluid from
the capillaries to the alveoli. As noted in Chapter 31, type II
alveolar epithelial cells may also actively pump sodium and
NONRESPIRATORY FUNCTIONS OF water from the alveolar surface into the interstitium.
THE PULMONARY CIRCULATION Drugs or chemical substances that readily pass through the
alveolar–capillary barrier by diffusion or by other means rap-
The pulmonary circulation, strategically located between the idly enter the systemic circulation. The lungs are frequently
systemic veins and arteries, is well suited for several functions used as a route of administration of drugs and for anesthetic
not directly related to gas exchange. The entire cardiac output gases, such as halothane and nitrous oxide. Aerosolized drugs
passes over the very large surface area of the pulmonary capil- intended for the airways only, such as the bronchodilator iso-
lary bed, allowing the lungs to act as a site of blood filtration proterenol and anti-inflammatory corticosteroids, may rap-
and storage, as well as for the metabolism of vasoactive con- idly pass into the systemic circulation, where they may have
stituents of the blood, as was discussed in Chapter 31. clinically significant effects. The effects of isoproterenol, for
A typical adult male has a pulmonary blood volume of about example, could include cardiac stimulation and vasodilation.
500 mL, which allows the pulmonary circulation to act as a
reservoir for the left ventricle. If left ventricular output is tran-
siently greater than systemic venous return, left ventricular
output can be maintained for a few strokes by drawing on
blood stored in the pulmonary circulation. CLINICAL CORRELATION
Because virtually all mixed venous blood must pass through A 60-year-old man who had a left ventricular myocardial
the pulmonary capillaries, the pulmonary circulation acts as a infarction 3 months ago returns to the cardiologist
filter, protecting the systemic circulation from materials that because of dyspnea on exertion but not at rest, a cough
enter the blood. The particles filtered, which may enter the productive of frothy fluid after exercise, and orthopnea
circulation as a result of natural processes, trauma, or thera- (easier breathing in the upright than recumbent posi-
peutic measures, may include small fibrin or blood clots, fat tion). At rest, his heart rate is 105/min, blood pressure is
cells, bone marrow, detached cancer cells, gas bubbles, aggluti- 120/90 mm Hg, and his respiratory rate is increased at
nated erythrocytes (especially in sickle cell disease), masses of 20/min. His chest radiograph shows evidence of edema in
platelets or leukocytes, and debris from stored blood or intra- gravity-dependent lung regions.
venous solutions. If these particles were to enter the arterial The patient does not have dyspnea (the feeling of diffi-
side of the systemic circulation, they might occlude vascular cult breathing or “shortness of breath”) at rest and his
beds with no other source of blood flow. This occlusion would blood pressure is within the normal range. His heart rate
be particularly disastrous if it occurred in the blood supply to at rest is slightly above the normal range (50–100/min;
the central nervous system or the heart. tachycardia) and his respiratory rate is high (normally
The lung can perform this very valuable service because 12–15/min; tachypnea). He does have orthopnea.
there are many more pulmonary capillaries present in the lung
CHAPTER 34 Pulmonary Perfusion 351

■ There is more blood flow in lower regions of the lung than


He had a left ventricular myocardial infarction 3 months in upper regions. The effects of pulmonary artery pressure,
ago and the damaged heart muscle has been replaced with pulmonary vein pressure, and alveolar pressure on pulmonary
scar tissue that cannot contract. Although his left ventri- blood flow are described as the zones of the lung.
cle can generate a sufficient stroke volume at rest, it can- ■ Alveolar hypoxia can cause constriction of precapillary
not match the increased right ventricular output during pulmonary vessels, diverting blood flow away from poorly
exercise, leading to increased left atrial pressure. Because ventilated or unventilated alveoli.
there are no valves between the left atrium and the pul-
monary veins and capillaries, pulmonary capillary hydro-
static pressure increases. Filtration of fluid from the STUDY QUESTIONS
capillaries into the pulmonary interstitium increases suf- 1. Compared to the systemic circulation, the pulmonary circulation
ficiently to exceed the ability of the pulmonary lymphatic has
drainage to remove it, resulting in interstitial edema and A) greater arterial pressure.
then alveolar edema. B) less distensible vessels.
The dyspnea results from several factors. Pulmonary C) a more evenly distributed vascular resistance to blood flow
vascular congestion (excess blood in the pulmonary among its arteries, capillaries, and veins.
blood vessels) decreases the compliance of the lungs. D) greater control of vascular resistance by the autonomic
Interstitial and alveolar edema increases the alveolar– nervous system.
capillary barrier for gas diffusion. This is particularly a E) greater total vascular resistance.
problem for oxygen diffusion, as will be discussed in the 2. Which of the following would likely increase pulmonary vascular
next chapter. Stretch receptors in the pulmonary circula- resistance?
tion respond to pulmonary vascular congestion and the A) inhaling from the FRC to the TLC
arterial chemoreceptors respond to low arterial Po2, both B) exhaling from the FRC to the RV
contributing to the sensation of dyspnea, as will be dis- C) breathing 10% O2–90% N2 for 10 minutes
D) decreasing the cardiac output from 5 to 2.5 L/min
cussed in Chapter 38. He breathes more easily in the
E) all of the above
upright position because the edema fluid collects in lower
3. In zone 2 of the lung
regions of the lungs, allowing better gas exchange in upper
parts of the lungs. A) alveolar pressure > pulmonary arterial pressure > pulmo-
nary venous pressure.
B) pulmonary arterial pressure > alveolar pressure > pulmo-
nary venous pressure.
C) pulmonary arterial pressure > pulmonary venous pressure >
alveolar pressure.
CHAPTER SUMMARY D) the effective pressure gradient for blood flow is pulmonary
■ Compared with the systemic arteries, the pulmonary arteries arterial pressure minus pulmonary venous pressure.
have much less vascular smooth muscle and therefore offer E) there is no blood flow.
much less resistance to blood flow. Pulmonary arteries are 4. Compared to the pulmonary circulation, the bronchial
more distensible, and because their intravascular pressures are circulation has
lower, more compressible than systemic arteries. The vascular A) more total blood flow.
transmural pressure gradient is therefore an important B) higher mean arterial pressure.
determinant of PVR. C) more distensible arteries.
■ Although pulmonary vascular smooth muscle can actively D) more even distribution of vascular resistance among
contract or relax in response to neural and humoral influences, the arteries, capillaries, and veins.
passive factors play a more important role in determining PVR E) lower arterial Po2.
than they do in determining SVR. 5. Which of the following is least likely to cause pulmonary edema?
■ PVR is usually lowest at the functional residual capacity and A) mitral stenosis
increases at higher and at lower lung volumes. B) two liters of rapidly administered intravenous saline
■ PVR usually decreases with increases in pulmonary blood flow, solution
pulmonary artery pressure, left atrial pressure, or pulmonary C) increased left atrial pressure
capillary blood volume because of distention of already open D) rapid administration of very negative intrapleural pressure
blood vessels, recruitment of previously unopened vessels, or to alleviate a pneumothorax
both. E) positive pressure ventilation
This page intentionally left blank
35
C H A P T E R

Ventilation–Perfusion
Relationships and
Respiratory Gas Exchange
Michael Levitzky

O B J E C T I V E S

■ Predict the consequences of mismatched ventilation and perfusion.


■ Explain the regional differences in the matching of ventilation and perfusion
of the normal upright lung.
■ Predict the consequences of the regional differences in the ventilation and
perfusion of the normal upright lung.
■ Define diffusion, and distinguish it from bulk flow.
■ State Fick’s law for diffusion.
■ Distinguish between perfusion limitation and diffusion limitation of gas
transfer in the lung.
■ Describe the diffusion of oxygen from the alveoli into the blood, and carbon
dioxide from the blood to the alveoli.
■ Define the diffusing capacity and discuss its measurement.

Alveolar ventilation and pulmonary perfusion have been dis- 0.8–1.2. However, ventilation and perfusion must be matched
cussed in the previous chapters in this section. The respiratory on the alveolar–capillary level for optimal gas exchange
gases must diffuse through the alveolar–capillary barrier for to occur and the V· /Q· for the whole lung is really of interest
gas exchange to occur. For optimal diffusion, the alveolar ven- only as an approximation of the situation in all the alveolar–
tilation must be matched to the pulmonary perfusion. capillary units of the lung.

CONSEQUENCES OF HIGH AND LOW


VENTILATION–PERFUSION VENTILATION–PERFUSION RATIOS
RELATIONSHIPS
Oxygen is delivered to the alveolus by alveolar ventilation, is
Alveolar ventilation brings oxygen into the lung and removes removed from the alveolus as it diffuses into the pulmonary
carbon dioxide from it. Similarly, the mixed venous blood capillary blood, and is carried away by blood flow. Similarly,
brings carbon dioxide into the lung and takes up alveolar oxy- carbon dioxide is delivered to the alveolus in the mixed venous
gen. The alveolar Po2 and Pco2 are thus determined by the rela- blood and diffuses into the alveolus in the pulmonary capillary.
tionship between alveolar ventilation and perfusion. It is removed from the alveolus by alveolar ventilation. As will
Alterations in the ratio of ventilation to perfusion, called the be discussed later in this chapter, at resting cardiac outputs, the
V·a /Q· c for alveolar ventilation/pulmonary capillary blood flow diffusion of both oxygen and carbon dioxide is normally lim-
(or just V· /Q· ), will result in changes in the alveolar Po2 and Pco2, ited by pulmonary perfusion. The alveolar partial pressures of
as well as in gas delivery to or removal from the lung. both oxygen and carbon dioxide are therefore determined by
Alveolar ventilation is normally about 4–6 L/min and pul- the V· /Q· . If the V· /Q· in an alveolar–capillary unit increases, the
monary blood flow (which is equal to cardiac output) has a delivery of oxygen relative to its removal will increase, as will
similar range, so the V· /Q· for the whole lung is in the range of the removal of carbon dioxide relative to its delivery. Alveolar

353

Ch35_353-362.indd 353 11/29/10 4:39:10 PM


354 SECTION VI Pulmonary Physiology

O2  150 mm Hg O2  150 mm Hg
CO2  0 mm Hg CO2  0 mm Hg
B. A. C.

O2  40 mm Hg O2  100 mm Hg O2  150 mm Hg
CO2  45 mm Hg CO2  40 mm Hg CO2  0 mm Hg

O2  40 mm Hg O2  40 mm Hg O2  40 mm Hg O2  100 mm Hg
CO2  45 mm Hg CO2  45 mm Hg CO2  45 mm Hg CO2  40 mm Hg

Mixed venous blood Inspired air


0 Decreasing VA/QC Normal Increasing VA/QC 
· · · ·
FIGURE 35–1 The effect of changes in the ventilation–perfusion ratio on the alveolar PO2 and PCO2. A) Normal VA / QC. B) VA / QC = 0.
· ·
C) VA / QC is infinite. Curved arrows denote direction of blood flow. (Modified with permission from West JB. Ventilation/Blood Flow and Gas Exchange. 5th ed.
Oxford: Blackwell; 1990.)

Po2 will therefore increase, and alveolar Pco2 will decrease. If the Units B and C represent the two extremes of a continuum of
V·/Q· in an alveolar–capillary unit decreases, the removal of oxy- ventilation–perfusion ratios. The V· /Q· ratio of a particular
gen relative to its delivery will increase and the delivery of car- alveolar–capillary unit can fall anywhere along this continuum,
bon dioxide relative to its removal will increase. Alveolar Po2 as shown at the bottom of Figure 35–1. The alveolar Po2 and Pco2
will therefore decrease, and alveolar Pco2 will increase. of such units will therefore fall between the two extremes
Figure 35–1 shows the consequences of alterations in the rela- shown in the figure: units with low V· /Q· ratios will have rela-
tionship of ventilation and perfusion on hypothetical alveolar– tively low Po2 and high Pco2; units with high V·/Q· ratios will have
capillary units. Unit A has a normal V·/Q· . Inspired air enters the relatively high Po2 and low Pco2. This is demonstrated graphi-
alveolus with a Po2 of about 150 mm Hg and a Pco2 of nearly 0 mm
Hg. Mixed venous blood enters the pulmonary capillary with a
Po2 of about 40 mm Hg and a Pco2 of about 45 mm Hg. This results
• •
in an alveolar Po2 of about 100 mm Hg and an alveolar Pco2 of 40 Shunt [V/Q = 0]
mm Hg. The partial pressure gradient for oxygen diffusion from Alveolar PO2 = Venous PO2 = 40
alveolus to pulmonary capillary is thus about 100 – 40, or 60 mm 50 • •
Decreasing V/Q
Hg; the partial pressure gradient for CO2 diffusion from pulmo- Normal values
nary capillary to alveolus is about 45 – 40, or 5 mm Hg. PO2 = 100
40 PCO2 = 40
The airway supplying unit B has become completely
occluded. Its V· /Q· is zero. As time goes on, the air trapped in
Alveolar PCO2 (mm Hg)

the alveolus equilibrates by diffusion with the gas dissolved in 30


the mixed venous blood entering the alveolar–capillary unit.
No gas exchange can occur, and any blood perfusing this alve- Increasing
• •
20 V/Q
olus will leave it exactly as it entered it. Unit B is therefore act-
ing as a right-to-left shunt.
The blood flow to unit C is blocked by a pulmonary embolus, 10 • •
and unit C is therefore completely unperfused. It has an infinite Deadspace [V/Q = ∞]
V·/Q· . Because no oxygen can diffuse from the alveolus into pul- Alveolar PO2 = inspired PO2 = 150
monary capillary blood and because no carbon dioxide can enter 0
0 10 20 30 40 50 60 70 80 90 100 110120 130 140 150
the alveolus from the blood, the Po2 of the alveolus is approxi-
Alveolar PO2 (mm Hg)
mately 150 mm Hg and its Pco2 is approximately zero; that is, the
gas composition of this unperfused alveolus is the same as that of FIGURE 35–2 The ventilation–perfusion ratio line on an
inspired air. Unit C is alveolar dead space. If unit C were unper- · ·
O2–CO2 diagram. Unit with a VA / QC of zero has the PO2 and PCO2 of
fused because its alveolar pressure exceeded its precapillary pres- · ·
mixed venous blood; a unit with an infinite VA / QC has the PO2 and PCO2
sure (rather than because of an embolus), then it would also of inspired air. (Reproduced with permission from Kibble J, Halsey CR: The Big
correspond to part of zone 1, as discussed in Chapter 34. Picture, Medical Physiology. New York: McGraw-Hill, 2009.)
CHAPTER 35 Ventilation–Perfusion Relationships and Respiratory Gas Exchange 355

cally in an O2–CO2 diagram such as that seen in Figure 35–2. the areas of absolute shunt (including the anatomic shunts)
The diagram shows the results of mathematical calculations of and the shuntlike areas into a single conceptual group. The
alveolar Po2 and Pco2 for V· /Q· ratios between zero (for mixed resulting ratio of shunt flow to the cardiac output, often
venous blood) and infinity (for inspired air). The resulting referred to as the venous admixture, is the part of the car-
curve is known as the ventilation–perfusion ratio line. This diac output that would have to be perfusing absolutely
simple O2–CO2 diagram can be modified to include correction unventilated alveoli to cause the systemic arterial oxygen
lines for other factors, such as the respiratory exchange ratios content obtained from a patient. A much larger portion of
of the alveoli and the blood or the dead space. The position of the cardiac output could be overperfusing poorly ventilated
the V· /Q· ratio line is altered if the partial pressures of the alveoli and yields the same ratio:
inspired gas or mixed venous blood are altered.
Q· s _______
__ Ccʹ –Ca
= o2 o2 (1)
Q· t Ccʹo2 –C –V o2
TESTING FOR MISMATCHED where Q·t represents the total pulmonary blood flow per minute
VENTILATION & PERFUSION (i.e., the cardiac output), Q· S represents the amount of blood flow
per minute entering the systemic arterial blood without receiv-
Several methods can demonstrate the presence or location of ing any oxygen (the “shunt flow”), Cao2 equals oxygen content
areas of the lung with mismatched ventilation and perfusion. of arterial blood (see Chapter 36) in milliliters of oxygen per
These methods include calculations of the physiologic shunt, 100 mL of blood, and C–V o and Ccʹo equal the oxygen content of
2 2
and the physiologic dead space, differences between the alveo- the mixed venous blood and the end-capillary oxygen content
lar and arterial Po2 and Pco2, and lung scans after inhaled and (the oxygen content in the blood at the end of the ventilated and
intravenously administered 133Xe or 99mTc. perfused pulmonary capillaries), respectively.
The shunt fraction is usually multiplied by 100% so that the
Physiologic Shunts and shunt flow is expressed as a percentage of the cardiac output.
The arterial and mixed venous oxygen contents can be
the Shunt Equation determined if blood samples are obtained from a systemic
A right-to-left shunt is the mixing of venous blood that has artery and from the pulmonary artery (for mixed venous
not been oxygenated (or not fully oxygenated) into the arterial blood), but the oxygen content of the blood at the end of the
blood. The physiologic shunt, which corresponds to the phys- pulmonary capillaries with well-matched ventilation and per-
iologic dead space, consists of the anatomic shunts plus the fusion is, of course, impossible to measure directly. This must
intrapulmonary shunts. The intrapulmonary shunts can be be calculated from the alveolar air equation and the patient’s
absolute shunts, or they can be “shuntlike states,” that is, areas hemoglobin concentration.
of low ventilation–perfusion ratios in which alveoli are under-
ventilated and/or overperfused. Anatomic shunts consist of
systemic venous blood entering the left ventricle without hav- Physiologic Dead Space
ing entered the pulmonary vasculature. In a healthy adult, The use of the Bohr equation to determine the physiologic
about 2–5% of the cardiac output, including venous blood from dead space was discussed in Chapter 33. If the anatomic dead
the bronchial veins, the thebesian veins, and the pleural veins, space is subtracted from the physiologic dead space, the result
enters the left side of the circulation directly without passing (if there is a difference) is alveolar dead space, or areas of infi-
through the pulmonary capillaries. Pathologic anatomic nite V· /Q·. Alveolar dead space also results in an arterial–
shunts such as right-to-left intracardiac shunts can also occur. alveolar CO2 difference (or arterial–end-tidal CO2
Mixed venous blood perfusing pulmonary capillaries associ- difference), that is, the end-tidal Pco2 is normally equal to the
ated with totally unventilated or collapsed alveoli constitutes an arterial Pco2. An arterial Pco2 greater than the end-tidal Pco2 usu-
absolute shunt (like the anatomic shunts) because no gas ally indicates the presence of alveolar dead space.
exchange occurs as the blood passes through the lung. Alveolar–
capillary units with low V·a / Q·c also act to lower the arterial oxy-
gen content because blood draining these units has a lower Po2 ALVEOLAR–ARTERIAL
than blood from units with well-matched ventilation and perfu- OXYGEN DIFFERENCE
sion. Increasing the percentage of inspired oxygen (FIO2) does
not significantly increase the arterial Po2 of patients with absolute The alveolar and arterial Po2 are often treated as though they are
intrapulmonary shunts or “shuntlike areas” because the pulmo- equal. However, the arterial Po2 is usually a few mm Hg less than
nary capillary blood that flows to unventilated or very poorly the alveolar Do2. This normal alveolar–arterial oxygen differ-
ventilated alveoli is not exposed to alveolar air. ence, the (A –a)Do2, is caused by the normal anatomic shunt,
The shunt equation conceptually divides all alveolar– some degree of ventilation–perfusion mismatch (see later in
capillary units into two groups: those with well-matched this chapter), and diffusion limitation in some parts of the lung.
ventilation and perfusion and those with ventilation– Of these, V· /Q· mismatch is usually the most important, with a
perfusion ratios of zero. Thus, the shunt equation combines small contribution from shunts and very little from diffusion
356 SECTION VI Pulmonary Physiology

TABLE 35–1 Causes of increased alveolar–arterial average alveolar Po2 that represents what alveolar Po2 should be,
oxygen difference. not necessarily what it is.

Increased right-to-left shunt


Anatomic
Intrapulmonary REGIONAL DIFFERENCES IN
Increased ventilation–perfusion mismatch VENTILATION–PERFUSION
Impaired diffusion RATIOS AND THEIR
Increased inspired partial pressure of oxygen
CONSEQUENCES
Decreased mixed venous partial pressure of oxygen
The regional variations in ventilation in the normal upright
Shift of oxyhemoglobin dissociation curve
lung were discussed in Chapter 33. They are summarized on
Adapted with permission from Marshall BE, Wyche MQ, Jr. Hypoxemia during and the left side of Figure 35–3. The right side of Figure 35–3 shows
after anesthesia. Anesthesiology. 1972;37(2):178–209.
that the more gravity-dependent regions of the lung also
receive more blood flow per unit volume than do the upper
limitation. Larger-than-normal differences between the alveolar regions of the lung, as discussed in Chapter 34.
and arterial Po2 may indicate significant ventilation–perfusion
mismatch; however, increased alveolar–arterial oxygen differ-
ences (Table 35–1) can also be caused by anatomic or intrapul- REGIONAL DIFFERENCES IN THE
monary shunts, diffusion block, low mixed venous Po2, breathing VENTILATION–PERFUSION RATIOS
higher-than-normal oxygen concentrations, or shifts of the IN THE UPRIGHT LUNG
oxyhemoglobin dissociation curve (also see Table 37–7).
The alveolar–arterial Po2 difference is normally about 5–15 Simplified graphs of the gradients of ventilation and perfusion
mm Hg in a young healthy person breathing room air at sea from the bottom to the top of normal upright lungs are shown
level. It increases with age because of the progressive decrease plotted on the same axes in Figure 35–4. The ventilation–
in arterial Po2 that occurs with aging (Chapter 73). The normal perfusion ratio was then calculated for several locations.
alveolar–arterial Po2 difference increases by about 20 mm Hg Figure 35–4 shows that even though the lower regions of the
between the ages of 20 and 70. lung receive both better ventilation and better perfusion than
Note that the “alveolar” Po2 used in determining the alveo- do the upper portions of the lung, the gradient of perfusion
lar–arterial oxygen difference is the PAo calculated using the
2
from the bottom of the lung to the top is greater than the gra-
alveolar air equation. As noted in Chapter 33, it is an idealized dient of ventilation. Because of this, the ventilation–perfusion

Ventilation Perfusion
Intrapleural pressure Lower intravascular
more negative pressures
Greater transmural Less recruitment,
pressure gradient distention
Alveoli larger, less Higher resistance
compliant Less blood flow
Less ventilation

Intrapleural pressure Greater vascular


FIGURE 35–3 Summary of regional less negative
Smaller transmural
pressures
More recruitment,
differences in ventilation (left) and perfusion pressure gradient distention
(right) in the normal upright lung. (Modified with Alveoli smaller, more Lower resistance
permission from Levitzky MG: Pulmonary Physiology, 7th ed. compliant Greater blood flow
New York: McGraw-Hill Medical, 2007.) More ventilation
CHAPTER 35 Ventilation–Perfusion Relationships and Respiratory Gas Exchange 357

temperatures. Gases move in both directions during diffusion,


3.0
but the area of higher partial pressure, because of its greater
• •
V/Q number of molecules per unit volume, has proportionately
more random “departures.” Thus, the net movement of gas is
dependent on the partial pressure difference between the two
Perfusion
areas. In a static situation, diffusion continues until no partial
2.0

Ventilation or perfusion
pressure differences exist for any gases in the two areas; in the
lungs, oxygen and carbon dioxide continuously enter and leave
V/Q ratio

Ventilation
the alveoli, so such an equilibrium does not take place.
• •

1.0 FICK’S LAW FOR DIFFUSION


• •
Oxygen is brought into the alveoli by bulk flow through the
Low V/Q conducting airways. When air flows through the conducting
physiologic
at lung P(A–a)O2 gradient
bases airways during inspiration, the linear velocity of the bulk flow
decreases as the air approaches the alveoli. This is because the
0
Base Distance up Apex total cross-sectional area increases dramatically in the distal
the lung portions of the tracheobronchial tree. By the time the air
(from base to apex)
reaches the alveoli, bulk flow probably ceases, and further gas
FIGURE 35–4 Distribution of ventilation and perfusion and movement occurs by diffusion. Oxygen then moves through
ventilation–perfusion ratio down the upright lung. (Reproduced with the gas phase in the alveoli according to its own partial pres-
permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New York: sure gradient. The distance from the alveolar duct to the
McGraw-Hill, 2009.) alveolar–capillary interface is usually less than 1 mm. Oxygen
then diffuses through the alveolar–capillary interface. It must
first, therefore, move from the gas phase to the liquid phase,
ratio is relatively low in more gravity-dependent regions of the according to Henry’s law, which states that the amount of a
lung and higher in upper regions of the lung. The effects of the gas absorbed by a liquid with which it does not combine
regional differences in V· /Q· on the alveolar Po2 and Pco2 can chemically is directly proportional to the partial pressure of
be predicted from Figure 35–2: the upper regions should have the gas to which the liquid is exposed and the solubility of the
a relatively high Po2 and a low Pco2; the lower regions should gas in the liquid. Oxygen must dissolve in and diffuse through
have a relatively low Po2 and a high Pco2. the thin layer of pulmonary surfactant, the alveolar epithe-
This means that the oxygen content of the blood draining the lium, the interstitium, and the capillary endothelium, as was
upper regions is higher and the carbon dioxide content is lower shown in Figure 31–6 (step 2, near the arrow). It must then
than that of the blood draining the lower regions. However, diffuse through the plasma (step 3), where some remains dis-
these contents are based on milliliters of blood (see Chapter 36), solved and the majority enters the erythrocyte and combines
and there is much less blood flow to the uppermost sections with hemoglobin (step 4). The blood then carries the oxygen
than there is to the bottom sections. Therefore, even though the out of the lung by bulk flow and distributes it to the other
uppermost sections have the highest V·/Q· and Po2 and the lowest tissues of the body, as was shown in Figure 31–1. At the tissues,
Pco2, there is more gas exchange in the more basal sections. oxygen diffuses from the erythrocyte through the plasma, cap-
illary endothelium, interstitium, tissue cell membrane, and
cell interior and into the mitochondrial membrane. The pro-
DIFFUSION OF GASES cess is almost entirely reversed for carbon dioxide.
The factors that determine the rate of diffusion of gas
Diffusion of a gas occurs when there is a net movement of
through the alveolar–capillary barrier are described by Fick’s
molecules from an area in which that particular gas exerts a
law for diffusion, shown as follows in a simplified form:
high partial pressure to an area in which it exerts a lower par-
AD(P1 – P2)
tial pressure. Movement of a gas by diffusion is therefore dif- V·gas = _________
T
(2)
ferent from the movement of gases through the conducting
airways, which occurs by “bulk flow” (mass movement or where V· gas is the volume of gas diffusing through the tissue
convection). In bulk flow, gas movement results from differ- barrier per time (mL/min), A the surface area of the barrier
ences in total pressure, and molecules of different gases move available for diffusion, D the diffusion coefficient, or diffusiv-
together along the total pressure gradient. In diffusion, each of ity, of the particular gas in the barrier, T the thickness of the
the different gases moves according to its own individual par- barrier or the diffusion distance, and P1 – P2 the partial pres-
tial pressure gradient. Gas transfer during diffusion occurs by sure difference of the gas across the barrier.
random molecular movement. It is therefore dependent on That is, the volume of gas per unit of time moving across the
temperature because molecular movement increases at higher alveolar–capillary barrier is directly proportional to the area of
358 SECTION VI Pulmonary Physiology

the barrier, the diffusivity, and the difference in concentration through the alveolar–capillary barrier before carbon dioxide
between the two sides, but is inversely proportional to the bar- retention due to diffusion impairment occurs.
rier thickness. The factors that limit the movement of a gas through the
The surface area of the blood–gas barrier is believed to be at alveolar–capillary barrier, as described by Fick’s law for diffu-
least 70 m2 in a healthy average-sized adult at rest. That is, sion, can be arbitrarily divided into three components: the dif-
about 70 m2 of the potential surface area is both ventilated and fusion coefficient, the surface area and thickness of the
perfused at rest. If more capillaries are recruited, as in exercise, alveolar–capillary membrane, and the partial pressure gradi-
the surface area available for diffusion increases; if venous ent across the barrier for each particular gas. The diffusion
return decreases, for example, because of hemorrhage, or if coefficient, as discussed in the previous section, is dependent
alveolar pressure is increased by positive-pressure ventilation, on the physical properties of the gases and the alveolar–
then capillaries may be derecruited and the surface area avail- capillary membrane. The surface area and thickness of the
able for diffusion may decrease. membrane are physical properties of the barrier, but they can
The thickness of the alveolar–capillary diffusion barrier is be altered by changes in the pulmonary capillary blood vol-
only about 0.2–0.5 μm. This barrier thickness can increase in ume, the cardiac output or the pulmonary artery pressure, or
interstitial fibrosis or interstitial edema, thus interfering with changes in lung volume. The partial pressure gradient of a gas
diffusion. Diffusion probably increases at higher lung volumes (across the barrier) is the final major determinant of its rate of
because as alveoli are stretched, the diffusion distance decreases diffusion. The partial pressure of a gas in the mixed venous
slightly (and also because small airways subject to closure may blood and in the pulmonary capillaries is just as important a
be open at higher lung volumes). factor as its alveolar partial pressure in determining its rate of
The diffusivity, or diffusion constant, for a gas is directly diffusion.
proportional to the solubility of the gas in the diffusion barrier
and is inversely proportional to the square root of the molecu- Diffusion Limitation
lar weight (MW) of the gas:
An erythrocyte and its attendant plasma spend an average of
Solubility about 0.75–1.2 seconds inside the pulmonary capillaries at
D α _______
____ (3)
√MW resting cardiac outputs. Figure 35–5 shows the calculated
Because oxygen is less dense than carbon dioxide, it should change with time in the partial pressures in the blood of three
diffuse 1.2 times as fast as carbon dioxide, but the solubility of gases: oxygen, carbon monoxide, and nitrous oxide. These are
carbon dioxide in the liquid phase is about 24 times that of shown in comparison to the alveolar partial pressures for each
oxygen, so carbon dioxide diffuses 20 times more rapidly gas, as indicated by the dotted line. This alveolar partial pres-
through the alveolar–capillary barrier than does oxygen. For sure is different for each of the three gases, and it depends on
this reason, patients develop problems in oxygen diffusion its concentration in the inspired gas mixture and on how rap-

Alveolar
partial
pressure

N2O

FIGURE 35–5 Calculated changes in the


Partial pressure in plasma

O2
partial pressures of carbon monoxide, nitrous
oxide, and oxygen in the blood as it passes
through a functional pulmonary capillary. There
are no units on the ordinate because the scale is
different for each of the three gases, depending on
the alveolar partial pressure of each gas. The
abscissa is in seconds, indicating the time the blood
has spent in the capillary. At resting cardiac outputs,
blood spends an average of 0.75 of a second in a
pulmonary capillary. The alveolar partial pressure of
each gas is indicated by the dotted line. Note that CO
the partial pressures of nitrous oxide and oxygen
equilibrate rapidly with their alveolar partial 0
0 0.25 0.50 0.75
pressure. (Modified with permission from Comroe JH: The Time in capillary (s)
Lung; Clinical Physiology and Pulmonary Function Tests, 2nd ed.
Chicago: Year Book Medical Publishers, 1962.) Enter capillary Leave capillary
CHAPTER 35 Ventilation–Perfusion Relationships and Respiratory Gas Exchange 359

idly it is removed by the pulmonary capillary blood. The sche- diffusion of both carbon monoxide and nitrous oxide may
matic is drawn as though all three gases were administered increase as the surface area for diffusion increases.)
simultaneously, but this is not necessarily the case. Consider
each gas as though it were acting independently of the others.
The partial pressure of carbon monoxide in the pulmonary
Diffusion of Oxygen
capillary blood rises very slowly compared with that of the As can be seen in Figure 35–5, the time course for oxygen trans-
other two gases in the figure if a low inspired concentration of fer falls between those for carbon monoxide and nitrous oxide.
carbon monoxide is used for a very short time. However, if the The partial pressure of oxygen rises fairly rapidly (it starts at the
content of carbon monoxide (in milliliters of carbon monoxide Po2 of the mixed venous blood, about 40 mm Hg, rather than at
per milliliter of blood) were measured simultaneously, it would zero), and equilibration with the alveolar Po2 of about 100 mm Hg
be rising very rapidly. The reason for this rapid rise is that car- occurs within about 0.25 of a second, or about one third of the
bon monoxide combines chemically with the hemoglobin in time the blood is in the pulmonary capillary at typical resting
the erythrocytes. The affinity of carbon monoxide for hemo- cardiac outputs. Oxygen moves easily through the alveolar–
globin is about 210 times that of oxygen for hemoglobin. The capillary barrier and into the erythrocytes, where it combines
carbon monoxide that is chemically combined with hemoglo- chemically with hemoglobin. The partial pressure of oxygen
bin does not contribute to the partial pressure of carbon mon- rises more rapidly than the partial pressure of carbon monox-
oxide in the blood because it is no longer physically dissolved in ide. Nonetheless, the oxygen chemically bound to hemoglobin
it. Therefore, the partial pressure of carbon monoxide in the (and therefore no longer physically dissolved) exerts no partial
pulmonary capillary blood does not come close to the partial pressure, so the partial pressure gradient across the alveolar–
pressure of carbon monoxide in the alveoli during the time capillary membrane is initially well maintained and oxygen
that the blood is exposed to the alveolar carbon monoxide. transfer occurs. The chemical combination of oxygen and
The partial pressure gradient across the alveolar–capillary bar- hemoglobin, however, occurs rapidly (within hundredths of a
rier for carbon monoxide is thus well maintained for the entire second), and at the normal alveolar partial pressure of oxygen,
time the blood spends in the pulmonary capillary. The diffu- the hemoglobin becomes nearly saturated with oxygen very
sion of carbon monoxide is therefore limited only by its diffu- quickly, as will be discussed in the next chapter. As this hap-
sivity in the barrier and by the surface area and thickness of pens, the partial pressure of oxygen in the blood rises rapidly to
the barrier. Carbon monoxide transfer from the alveolus to the that in the alveolus, and from that point, no further oxygen
pulmonary capillary blood is referred to as diffusion-limited transfer from the alveolus to the equilibrated blood can occur.
rather than perfusion-limited. Therefore, under the conditions of normal alveolar Po2 and rest-
ing cardiac output, oxygen transfer from alveolus to pulmonary
capillary is perfusion-limited.
Perfusion Limitation During exercise, blood moves through the pulmonary capil-
The partial pressure of nitrous oxide in the pulmonary capil- lary much more rapidly than it does at resting cardiac outputs.
lary blood equilibrates very rapidly with the partial pressure of In fact, the blood may stay in the pulmonary capillary an average
nitrous oxide in the alveolus because nitrous oxide moves of only about 0.25 of a second during strenuous exercise. Oxy-
through the alveolar–capillary barrier very easily and because gen transfer into the blood per time will be greatly increased
it does not combine chemically with the hemoglobin in the because there is little or no perfusion limitation of oxygen trans-
erythrocytes. After only about 0.1 of a second of exposure of fer. (Indeed, that part of the blood that stays in the capillary less
the pulmonary capillary blood to the alveolar nitrous oxide, than the average may be subjected to diffusion limitation of oxy-
the partial pressure gradient across the alveolar–capillary bar- gen transfer.) Of course, total oxygen transfer is also increased
rier has been abolished. From this point on, no further nitrous during exercise because of recruitment of previously unperfused
oxide transfer occurs from the alveolus to the blood in the cap- capillaries, which increases the surface area for diffusion, and
illary that has already equilibrated with the alveolar nitrous because of better matching of ventilation and perfusion. A per-
oxide partial pressure; during the last 0.6–0.7 of a second, no son with an abnormal alveolar–capillary barrier due to a fibrotic
net diffusion occurs between the alveolus and the blood as it thickening or interstitial edema may approach diffusion limita-
travels through the pulmonary capillary. Blood just entering tion of oxygen transfer at rest and may have a serious diffusion
the capillary at the arterial end will not be equilibrated with the limitation of oxygen transfer during strenuous exercise. A per-
alveolar partial pressure of nitrous oxide, so nitrous oxide can son with an extremely abnormal alveolar–capillary barrier might
diffuse into the blood at the arterial end. The transfer of nitrous have diffusion limitation of oxygen transfer even at rest.
oxide is therefore perfusion-limited. Nitrous oxide transfer
from a particular alveolus to one of its pulmonary capillaries
can be increased by increasing the cardiac output and thus Diffusion of Carbon Dioxide
reducing the amount of time the blood stays in the pulmonary The equilibration of the partial pressure of carbon dioxide in
capillary after equilibration with the alveolar partial pressure the pulmonary capillary blood with that of the alveolus in a
of nitrous oxide has occurred. (Because increasing the cardiac healthy person with a mixed venous partial pressure of car-
output may recruit previously unperfused capillaries, the total bon dioxide of 45 mm Hg and an alveolar partial pressure of
360 SECTION VI Pulmonary Physiology

carbon dioxide of 40 mm Hg is about 0.25 of a second, or TABLE 35–2 Conditions that decrease the
about the same as that for oxygen. This may seem surprising, diffusing capacity.
considering that the diffusivity of carbon dioxide is about
20 times that of oxygen, but the partial pressure gradient is Thickening of the barrier
normally only about 5 mm Hg for carbon dioxide, whereas it Interstitial or alveolar edema
Interstitial or alveolar fibrosis
is about 60 mm Hg for oxygen. Carbon dioxide transfer is Sarcoidosis
therefore also normally perfusion-limited, although it may Scleroderma
be diffusion-limited in a person with an abnormal alveolar– Decreased surface area
capillary barrier. Emphysema
Tumors
Low cardiac output
MEASUREMENT OF Low pulmonary capillary blood volume

DIFFUSING CAPACITY Decreased uptake by erythrocytes


Anemia
Low pulmonary capillary blood volume
It is often useful to determine the diffusion characteristics of a
patient’s lungs during their assessment in the pulmonary func- Ventilation–perfusion mismatch
tion laboratory. It may be particularly important to determine Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
whether an apparent impairment in diffusion is a result of per- New York: McGraw-Hill Medical, 2007.

fusion limitation or diffusion limitation.


The diffusing capacity is the rate at which oxygen or carbon
monoxide is absorbed from the alveolar gas into the pulmo-
nary capillaries (in milliliters per minute) per unit of partial CLINICAL CORRELATION
pressure gradient (in millimeters of mercury). It is usually
measured with very low concentrations of carbon monoxide A 40-year-old man with a broken leg in a cast because of
because carbon monoxide transfer from alveolus to capillary is a skiing injury and no history of respiratory problems
diffusion-limited as was discussed previously in this chapter. suddenly has difficulty breathing and complains of chest
The mean partial pressure of oxygen or carbon monoxide is, pain. He is brought to the hospital. In the emergency
as already discussed, affected by their chemical reactions with department, his breathing is observed to be rapid and
hemoglobin, as well as by their transfer through the alveolar– shallow. His heart rate is 120/min and his arterial blood
capillary barrier. For this reason, the diffusing capacity of the pressure is 80/60 mm Hg. His respiratory rate is 25/min.
lung is influenced by both the diffusing capacity of the mem- A chest x-ray and an electrocardiogram (ECG) are per-
brane and the reaction with hemoglobin. The amount of formed on the patient to help determine the cause of his
hemoglobin in the lung is dependent on the hemoglobin con- chest pain and dyspnea. The ECG shows no abnormalities
centration in the blood and the amount of blood in the pulmo- indicative of myocardial ischemia (insufficient blood
nary capillaries—the pulmonary capillary blood volume. flow to the heart muscle) or myocardial infarction (injury
Diffusion through the alveolus is normally very rapid and usu- of the heart muscle) such as ST segment or T-wave abnor-
ally can be disregarded, although it could be a major factor in malities (see Chapter 23). The chest x-ray shows no
a person with pulmonary edema. abnormalities indicative of pneumonia, atelectasis (col-
Several different methods are used clinically to measure the lapsed alveoli), or pneumothorax (air between the inside
carbon monoxide diffusing capacity (the DLCO) and involve of the chest wall and the outside of the lung). An arterial
both single-breath and steady-state techniques, sometimes blood sample is obtained from the patient while he was
during exercise. breathing room air to determine his arterial blood gases
The DLCO is decreased in diseases associated with interstitial (arterial Po2, arterial Pco2, and arterial pH). His arterial Po2
or alveolar fibrosis, such as sarcoidosis, scleroderma, and asbes- was 70 mm Hg (normal >90); his arterial Pco2 was
tosis, or with conditions causing interstitial or alveolar pulmo- 30 mm Hg (normal range is 35–45); his pH was 7.50 (nor-
nary edema, as indicated in Table 35–2. It is also decreased in mal range is 7.35–7.45).
conditions causing a decrease in the surface area available for The patient has a pulmonary embolus, most likely as a
diffusion, such as emphysema, tumors, a low cardiac output, or result of blood clotting in his immobilized leg. Flow of
a low pulmonary capillary blood volume, as well as in condi- venous blood in the broken leg is impaired by the cast
tions leading to ventilation–perfusion mismatch, which effec- and the lack of muscle contraction to enhance venous
tively decreases the surface area available for diffusion. return from his leg to his heart. Stasis (low or absent
The carbon monoxide diffusing capacity can be very useful flow) of blood often leads to clotting (thrombosis). When
in assessing patients with chronic obstructive pulmonary dis- thrombosis occurs in nonsuperficial veins such as those
ease (COPD). A low DLCO distinguishes patients whose disor- in the leg, it is called deep venous thrombosis (DVT).
der is primarily emphysema from those whose disorder is The thrombus can break loose and be carried to the right
primarily chronic bronchitis. The DLCO can also be helpful in side of the heart and enter the pulmonary arterial tree,
assessing patients with restrictive diseases.
CHAPTER 35 Ventilation–Perfusion Relationships and Respiratory Gas Exchange 361

Pco2; nonetheless, there is normally more gas exchange in lower


where it can block blood flow to part of the lung. This is regions of the lung because they receive more blood flow.
called a pulmonary embolus, in this case a thromboem- ■ The volume of gas per unit of time moving across the
bolus. Pulmonary emboli can be life-threatening if they alveolar–capillary barrier is directly proportional to the area
occlude a significant fraction of the pulmonary vascular of the barrier, the diffusivity of the gas in the barrier, and the
bed. The region of the lung with occluded blood flow cre- difference in concentration of the gas between the two sides
ates alveolar dead space (ventilated but not perfused) of the barrier, but is inversely proportional to the barrier
that contributes nothing to gas exchange. The patient’s thickness.
end-tidal Pco2 decreases because it contains air coming ■ If the partial pressure of a gas in the plasma equilibrates
from unperfused alveoli that contribute no carbon diox- with the alveolar partial pressure of the gas within the
ide to the exhaled air. The arterial Pco2 is therefore greater amount of time the blood is in the pulmonary capillary, its
transfer is perfusion-limited; if equilibration does not occur
than the end-tidal (“alveolar”) Pco2. The diffusing capac-
within the time the blood is in the capillary, its transfer is
ity of the patient is decreased because of decreased sur- diffusion-limited.
face area for gas exchange. Occlusion of pulmonary
vessels is likely to increase pulmonary vascular resistance,
increase pulmonary artery pressure, and increase right STUDY QUESTIONS
ventricular work. Blood flow to the left side of the patient’s
1. An otherwise normal person is brought to the emergency
heart decreases, which explains his low systemic blood
department after having accidentally aspirated a foreign body
pressure. His tachycardia is likely a result of the response into the right main-stem bronchus, partially occluding it.
of his baroreceptor reflex to his low blood pressure, and Which of the following is/are likely to occur?
the pain and anxiety he is experiencing. His tachypnea is A) The right lung’s alveolar Po2 will be lower and its alveolar
explained by the influence of receptors in his lungs (which Pco will be higher than those of the left lung.
2
will be described in Chapter 38) and the pain and anxiety. B) The calculated shunt fraction will increase.
The tachypnea resulted in hyperventilation causing his C) Blood flow to the right lung will decrease.
arterial Pco2 to decrease below the normal range and his D) The arterial Po2 will decrease.
arterial pH to exceed the normal range (see discussion of E) All of the above.
uncompensated respiratory alkalosis in Chapter 37). His 2. A healthy person lies down on her right side and breathes
low arterial Po2 is a result of the occlusion of pulmonary normally. Her right lung, in comparison to her left lung, will
vessels forcing blood flow to poorly ventilated alveoli. be expected to have a
Treatment of patients with pulmonary emboli (some- A) lower alveolar Po2 and a higher alveolar Pco2.
times called pulmonary embolism) depends on the sever- B) lower blood flow per unit volume.
ity of the disorder. Anticoagulants are used to prevent C) less ventilation per unit volume.
further clotting, thrombolytic drugs are used to break clots D) higher ventilation–perfusion ratio.
E) larger alveoli.
down, intravenous catheters with deployable filters can be
used to remove the emboli, and large life-threatening 3. Which of the following conditions or circumstances is expected
to increase the diffusing capacity (DL) of the lungs?
emboli may be removed surgically (embolectomy).
A) changing from the supine to the upright position
B) exercise
C) emphysema
D) anemia
CHAPTER SUMMARY E) low cardiac output due to blood loss
■ Ventilation and perfusion must be matched on the alveolar– F) diffuse interstitial fibrosis of the lungs
capillary level for optimal gas exchange. 4. If the pulmonary capillary partial pressure of a gas equilibrates with
■ Ventilation–perfusion ratios close to 1.0 result in alveolar Po2 of that in the alveolus before the blood leaves the capillary (assume
approximately 100 mm Hg and Pco2 close to 40 mm Hg; the gas is diffusing from the alveolus to the pulmonary capillary)
ventilation–perfusion ratios greater than 1.0 increase the Po2 A) its transfer is said to be perfusion-limited.
and decrease the Pco2; ventilation–perfusion ratios less than 1.0 B) its transfer is said to be diffusion-limited.
decrease the Po2 and increase the Pco2. C) increasing the cardiac output will not increase the amount
■ Alveolar dead space and intrapulmonary shunt represent the of the gas diffusing across the alveolar–capillary barrier.
two extremes of ventilation–perfusion ratios, infinite and zero, D) increasing the alveolar partial pressure of the gas will not
respectively. increase the amount of the gas diffusing across the
alveolar–capillary barrier.
■ The ventilation–perfusion ratios in lower regions of the normal
E) recruiting additional pulmonary capillaries will not increase
upright lung are lower than 1.0, resulting in lower Po2 and
the amount of the gas diffusing across the alveolar–capillary
higher Pco2; the ventilation–perfusion ratios in upper parts of
barrier.
the lung are greater than 1.0, resulting in higher Po2 and lower
This page intentionally left blank
36
C H A P T E R

Transport of Oxygen and


Carbon Dioxide
Michael Levitzky

O B J E C T I V E S

■ State the relationship between the partial pressure of oxygen in the


blood and the amount of oxygen physically dissolved in the blood.
■ Describe the chemical combination of oxygen with hemoglobin and
the oxyhemoglobin dissociation curve.
■ Define hemoglobin saturation, oxygen-carrying capacity, and oxygen
content.
■ State the physiologic consequences of the shape of the oxyhemoglobin
dissociation curve.
■ List the physiologic factors that can influence the oxyhemoglobin
dissociation curve, and predict their effects on oxygen transport by
the blood.
■ State the relationship between the partial pressure of carbon dioxide in the
blood and the amount of carbon dioxide physically dissolved in the blood.
■ Describe the transport of carbon dioxide as carbamino compounds with
blood proteins.
■ Explain how most of the carbon dioxide in the blood is transported
as bicarbonate.
■ Describe the carbon dioxide dissociation curve for whole blood.

TRANSPORT OF OXYGEN normal arterial blood with a Po2 of approximately 100 mm Hg


contains only about 0.003 mL O2/mL of blood, or 0.3 mL
BY THE BLOOD O2/100 mL of blood. (Blood oxygen content is conventionally
expressed in milliliters of oxygen per 100 mL of blood, also
Oxygen is transported both physically dissolved in blood and called volume percent.) The physically dissolved oxygen in
chemically combined to the hemoglobin in the erythrocytes. the blood therefore cannot meet the metabolic demand for
Much more oxygen is normally transported combined with oxygen, even at rest.
hemoglobin than is physically dissolved in the blood. Without
hemoglobin, the cardiovascular system could not supply suf-
ficient oxygen to meet tissue demands. CHEMICALLY COMBINED
WITH HEMOGLOBIN
PHYSICALLY DISSOLVED The Structure of Hemoglobin
At a temperature of 37°C, 1 mL of plasma contains 0.00003mL Hemoglobin is a complex molecule with a tetrameric struc-
O2/(mm Hg Po2). Whole blood contains a similar amount of ture consisting of four linked polypeptide chains (globin),
dissolved oxygen per milliliter because oxygen dissolves in the each of which is attached to a protoporphyrin (heme) group.
fluid of the erythrocytes in about the same amount. Therefore, Each heme group has a ferrous (Fe2+) iron atom at its center

363

Ch36_363-374.indd 363 11/26/10 10:11:07 AM


364 SECTION VI Pulmonary Physiology

and can bind a molecule of oxygen (or carbon monoxide), so The reaction of hemoglobin and oxygen is conventionally
the tetrameric hemoglobin molecule can combine chemically written as follows:
with four oxygen molecules (or eight oxygen atoms). Varia-
Hb + O2 HbO2 (2)
tions in the amino acid sequences of the four globin subunits
Deoxyhemoglobin Oxyhemoglobin
may have important physiologic consequences. Normal adult
hemoglobin (HbA) consists of two alpha (α) chains, each of
which has 141 amino acids, and two beta (β) chains, each of
which has 146 amino acids. Fetal hemoglobin (HbF), which
HEMOGLOBIN AND THE
consists of two α chains and two gamma (γ) chains, has a PHYSIOLOGIC IMPLICATIONS
higher affinity for oxygen than does HbA. Synthesis of β chains
normally begins about 6 weeks before birth, and HbA usually OF THE OXYHEMOGLOBIN
replaces almost all the HbF by the time an infant is 4 months DISSOCIATION CURVE
old. Other, abnormal hemoglobin molecules may be produced
by genetic substitution of a single amino acid for the normal The equilibrium point of the reversible reaction of hemoglobin
one in an α or β chain or (rarely) by alterations in the structure and oxygen is dependent on how much oxygen the hemoglobin
of heme groups. These alterations may produce changes in the in blood is exposed to. This corresponds directly to the partial
affinity of the hemoglobin for oxygen, change the physical pressure of oxygen (Po2) in the plasma under the conditions in
properties of hemoglobin, or alter the interaction of hemoglo- the body. Thus, the Po2 of the plasma determines the amount of
bin and other substances that affect its combination with oxy- oxygen that binds to the hemoglobin in the erythrocytes.
gen, such as 2,3-bisphosphoglycerate (2,3-BPG) (discussed
later in this chapter). More than 120 abnormal variants of nor-
THE OXYHEMOGLOBIN
mal HbA have been demonstrated in patients. The best known
of these, hemoglobin S, is present in sickle cell disease. Hemo- DISSOCIATION CURVE
globin S tends to polymerize and crystallize in the cytosol of
One way to express the proportion of oxygen that is bound to
the erythrocyte when it is not combined with oxygen. This
hemoglobin is as percent saturation. This is equal to the con-
polymerization and crystallization decreases the solubility of
tent of oxygen in the blood (minus that part physically dis-
hemoglobin S within the erythrocyte and changes the shape of
solved) divided by the oxygen-carrying capacity of the
the cell from the normal biconcave disk to a crescent or “sickle”
hemoglobin in the blood times 100%:
shape. A sickled cell is more fragile than a normal cell. In addi-
tion, the cells have a tendency to stick to one another, which O bound to Hb
% Hb saturation = _____________
2
× 100% (3)
increases blood viscosity and also favors thrombosis or block- O2 capacity of Hb
age of blood vessels. Note that the oxygen-carrying capacity of an individual
depends on the amount of hemoglobin in the blood. The blood
oxygen content also depends on the amount of hemoglobin
Chemical Reaction of present (as well as on the Po2). Both content and capacity are
expressed as milliliters of oxygen per 100 mL of blood. On the
Oxygen and Hemoglobin other hand, the percent hemoglobin saturation expresses only
Hemoglobin rapidly combines reversibly with oxygen. It is the a percentage and not an amount or volume of oxygen; “percent
reversibility of the reaction that allows oxygen to be released to saturation” is not interchangeable with “oxygen content.” For
the tissues; if the reaction did not proceed easily in both direc- example, two patients might have the same percent of hemo-
tions, hemoglobin would be of little use in delivering oxygen globin saturation, but if one has a lower blood hemoglobin
to satisfy metabolic needs. The reaction is very fast, with a concentration because of anemia, he or she will have a lower
half-time of 0.01 of a second or less. Each gram of hemoglobin blood oxygen content.
is capable of combining with about 1.39 mL of oxygen under The relationship between the Po2 of the plasma and the per-
optimal conditions, but under normal circumstances, some cent of hemoglobin saturation can be expressed graphically as
hemoglobin exists in forms such as methemoglobin (in which the oxyhemoglobin dissociation curve. An oxyhemoglobin
the iron atom is in the ferric state) or is combined with carbon dissociation curve for normal blood is shown in Figure 36–1.
monoxide, in which case the hemoglobin cannot bind oxygen. The oxyhemoglobin dissociation curve is really a plot of
For this reason, the oxygen-carrying capacity of hemoglobin how the availability of one of the reactants, oxygen (expressed
is conventionally considered to be 1.34 mL O2/(g Hb), that is, as the Po2 of the plasma), affects the reversible chemical reac-
each gram of hemoglobin, when fully saturated with oxygen, tion of oxygen and hemoglobin. The product, oxyhemoglobin,
binds 1.34 mL of oxygen. Therefore, a person with 15 g Hb/ is expressed as percent saturation—really a percentage of the
100 mL of blood has an oxygen-carrying capacity of 20.1 mL maximum for any given amount of hemoglobin.
O2/100 mL of blood: As can be seen in Figure 36–1, the relationship between Po2
15 g Hb
__________ 1.34 mL O 20.1 mL O2 (1) and HbO2 is not linear; it is a sigmoid (S-shaped) curve, steep
× ________2 = __________
100 mL blood g Hb 100 mL blood at the lower Po2 and nearly flat when the Po2 is above 70 mm Hg.
CHAPTER 36 Transport of Oxygen and Carbon Dioxide 365

100

Hemoglobin saturation ( %)
80

60
50%

40

20

0
0 20 40 60 80 100 120 140 160
P50
Partial pressure of oxygen (mm Hg)

FIGURE 36–1 A typical “normal” adult oxyhemoglobin dissociation curve for blood at 37°C with a pH of 7.40 and a PCO2 of
40 mm Hg. The P50 is the partial pressure of oxygen at which hemoglobin is 50% saturated with oxygen. (Modified with permission from Levitzky MG:
Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

It is this S shape that is responsible for several very important Oxygen physically dissolved at a Po2 of 40 mm Hg is given
physiologic properties of the reaction of oxygen and hemoglo- as follows:
bin. The reason that the curve is S-shaped and not linear is 0.003 mL O2 0.12 mL O2
that it is actually a plot of four reactions rather than one, that ______________________ × 40 mm Hg = __________ (6)
100 mL blood Po2 (in mm Hg) 100 mL blood
is, each of the four subunits of hemoglobin can combine with
one molecule of oxygen. Total blood oxygen content at a Po2 of 40 mm Hg (37°C,
The reactions of the four subunits of hemoglobin with oxy- pH 7.4) is given as follows:
gen do not occur simultaneously. Instead they occur sequen- 15.08 mL O2 0.12 mL O2 15.2 mL O2
__________ + __________ = __________
tially in four steps, with an interaction between the subunits 100 mL blood 100 mL blood 100 mL blood (7)
occurring in such a way that during the successive combina- Bound to Hb Physically dissolved Total
tions of the subunits with oxygen, each combination facilitates
the next. Similarly, dissociation of oxygen from hemoglobin As the blood passes through the pulmonary capillaries, it
subunits facilitates further dissociations. The dissociation equilibrates with the alveolar Po2 of about 100 mm Hg. At a Po2
curve for a single monomer of hemoglobin is far different of 100 mm Hg, hemoglobin is about 97.4% saturated with oxy-
from that for the tetramer (see Figure 36–4C). gen, as seen in Figure 36–1. This corresponds to 19.58 mL
O2/100 mL of blood bound to hemoglobin plus 0.3 mL
O2/100 mL of blood physically dissolved, or a total oxygen con-
Loading Oxygen in the Lung tent of 19.88 mL O2/100 mL of blood.
Oxygen bound to hemoglobin at a Po2 of 100 mm Hg (37°C,
Mixed venous blood entering the pulmonary capillaries pH 7.4) is given as follows:
normally has a Po2 of about 40 mm Hg. At a Po2 of 40 mm Hg,
hemoglobin is about 75% saturated with oxygen, as seen in 20.1 mL O2
___________ 19.58 mL O
× 97.4% = __________
2

Figure 36–1. Assuming a blood hemoglobin concentration of 100 mL blood 100 mL blood (8)
Capacity % saturation Content
15 g Hb/100 mL of blood, this corresponds to 15.08 mL
O2/100 mL of blood bound to hemoglobin plus an additional Oxygen physically dissolved at a Po2 of 100 mm Hg is given
0.12 mL O2/100 mL of blood physically dissolved, or a total as follows:
oxygen content of approximately 15.2 mL O2/100 mL of 0.003 mL O2 0.3 mL O2
______________________ × 100 mm Hg = __________ (9)
blood. 100 mL blood Po2 (in mm Hg) 100 mL blood
Oxygen-carrying capacity is given as follows:
15 g Hg 1.34 mL O 20.1 mL O2 Total blood oxygen content at a Po2 of 100 mm Hg (37°C,
__________ × ________2 = __________ (4)
100 mL blood g Hb 100 mL blood pH 7.4) is given as follows:
Oxygen bound to hemoglobin at a Po2 of 40 mm Hg (37°C, 19.58 mL O2
__________ 0.3 mL O2
__________ 19.88 mL O2
__________
+ =
pH 7.4) is given as follows: 100 mL blood 100 mL blood 100 mL blood (10)
Bound to Hb Physically dissolved Total
20.1 mL O2
__________ 15.08 mL O2
× 75% = ___________ (5) Thus, in passing through the lungs, each 100 mL of blood
100 mL blood 100 mL blood
Capacity % saturation Content has loaded (19.88 – 15.20) mL O2, or 4.68 mL O2. Assuming a
366 SECTION VI Pulmonary Physiology

cardiac output of 5 L/min, this means that approximately curve. High temperature, low pH, high Pco2, and elevated levels
234 mL O2 is loaded into the blood per minute: of 2,3-BPG all shift the oxyhemoglobin dissociation curve to
46.8 mL O2 ________
5 L blood ________
_______ 234 mL O the right; that is, for any particular Po2, there is less oxygen
× = min 2 (11)
min liter blood chemically combined with hemoglobin at higher tempera-
Note that the oxyhemoglobin dissociation curve is rela- tures, lower pH, higher Pco2, and elevated levels of 2,3-BPG.
tively flat when Po2 is greater than approximately 70 mm Hg. The rightward shift represents a decreased affinity of hemo-
This is very important physiologically because it means that globin for oxygen.
there is only a small decrease in the oxygen content of blood The effects of blood pH and Pco2 on the oxyhemoglobin dis-
equilibrated with a Po2 of 70 mm Hg instead of 100 mm Hg. In sociation curve are shown in Figure 36–2A and B. Low pH and
fact, the curve shows that at a Po2 of 70 mm Hg, hemoglobin high Pco2 both shift the curve to the right. High pH and low
is still approximately 94.1% saturated with oxygen. This con- Pco2 both shift the curve to the left. These two effects often
stitutes an important safety factor because a patient with a occur together. The influence of pH (and Pco2) on the oxyhe-
relatively low alveolar or arterial Po2 of 70 mm Hg (owing to moglobin dissociation curve is referred to as the Bohr effect.
hypoventilation or intrapulmonary shunt, for example) is The Bohr effect will be discussed in greater detail at the end of
still able to load adequate oxygen into the blood. A quick cal- this chapter.
culation shows that at 70 mm Hg, the total blood oxygen con- Figure 36–2C shows the effects of blood temperature on the
tent is approximately 19.12 mL O2/100 mL of blood compared oxyhemoglobin dissociation curve. High temperatures shift the
with the 19.88 mL O2/100 mL of blood at a Po2 of 100 mm Hg. curve to the right; low temperatures shift the curve to the left.
These calculations show that Po2 is often a more sensitive At very low blood temperatures, hemoglobin has such a high
diagnostic indicator of the status of a patient’s respiratory sys- affinity for oxygen that it does not release the oxygen, even at
tem than the arterial oxygen content. Of course, the oxygen very low Po2. 2,3-BPG (also called 2,3-diphosphoglycerate, or
content is more important physiologically to the patient. 2,3-DPG) is produced by erythrocytes during their normal gly-
Because hemoglobin is approximately 97.4% saturated at a colysis and is present in fairly high concentrations within red
Po2 of 100 mm Hg, increasing the alveolar Po2 above 100 mm Hg blood cells (about 15 mmol/(g Hb)). 2,3-BPG binds to the
can add little additional oxygen to hemoglobin (only about hemoglobin in erythrocytes, which decreases the affinity of
0.52 mL O2/100 mL of blood at a hemoglobin concentration of hemoglobin for oxygen. Higher concentrations of 2,3-BPG
15 g/100 mL of blood). Hemoglobin is fully saturated with therefore shift the oxyhemoglobin dissociation curve to the
oxygen at a Po2 of about 250 mm Hg. right, as shown in Figure 36–2D. It has been demonstrated that
more 2,3-BPG is produced during chronic hypoxic conditions,
Unloading Oxygen at the Tissues thus shifting the dissociation curve to the right and allowing
As blood passes from the arteries into the systemic capillaries, it more oxygen to be released from hemoglobin at a particular
is exposed to lower Po2 and oxygen is released by the hemoglo- Po2. Very low levels of 2,3-BPG shift the curve far to the left, as
bin. The Po2 in the capillaries varies from tissue to tissue, being shown in the figure. This means that blood deficient in 2,3-BPG
very low in some (e.g., myocardium) and relatively higher in does not unload much oxygen. Blood stored in blood banks for
others (e.g., renal cortex). As can be seen in Figure 36–1, the as little as 1 week has been shown to have very low levels of
oxyhemoglobin dissociation curve is very steep in the range of 2,3-BPG. Use of banked blood in patients may result in
40–10 mm Hg. This means that a small decrease in Po2 can result decreased oxygen unloading to the tissues unless steps are
in a substantial further dissociation of oxygen and hemoglobin, taken to restore the normal levels of 2,3-BPG.
unloading more oxygen for use by the tissues. At a Po2 of 40 mm As blood enters metabolically active tissues, it is exposed
Hg, hemoglobin is about 75% saturated with oxygen, with a to an environment different from that found in the arterial
total blood oxygen content of 15.2 mL O2/100 mL of blood (at tree. The Pco2 is higher, the pH is lower, and the temperature
15 g Hb/100 mL of blood). At a Po2 of 20 mm Hg, hemoglobin is is also higher than that of the arterial blood. The curve
only 32% saturated with oxygen. The total blood oxygen content shown in Figure 36–1 is for blood at 37°C, with a pH of 7.4
is only 6.49 mL O2/100 mL of blood, a decrease of 8.71 mL and a Pco2 of 40 mm Hg. Blood in metabolically active tissues
O2/100 mL of blood for only a 20-mm Hg decrease in Po2. and therefore the venous blood draining them are no longer
The unloading of oxygen at the tissues is also facilitated by subject to these conditions because they have been exposed
other physiologic factors that can alter the shape and position to a different environment. Because low pH, high Pco2,
of the oxyhemoglobin dissociation curve. These include increased 2,3-BPG, and higher temperature all shift the oxy-
the pH, Pco2, temperature of the blood, and concentration of hemoglobin dissociation curve to the right, they all can help
2,3-BPG in the erythrocytes. unload oxygen from hemoglobin at the tissues. On the other
hand, as the venous blood returns to the lung and CO2 leaves
the blood (which increases the pH), the affinity of hemoglo-
INFLUENCES ON THE OXYHEMOGLOBIN bin for oxygen increases as the curve shifts back to the left,
DISSOCIATION CURVE as shown in Figure 36–3. Note that the effects of pH, Pco2,
and temperature shown in Figure 36–2 have a more pro-
Figure 36–2 shows the influence of alterations in temperature, found effect on enhancing the unloading of oxygen at the
pH, Pco2, and 2,3-BPG on the oxyhemoglobin dissociation tissues than they do interfering with its loading at the lungs.
CHAPTER 36 Transport of Oxygen and Carbon Dioxide 367

100 100
20°
37°
43°
0
80 7.6 80

0
Hemoglobin saturation ( %)

Hemoglobin saturation (%)


7.4

0
pH

7.2
pH


pH

60 60

40 40

20 20

0 0
0 20 40 60 80 100 0 20 40 60 80 100
A. PO (mm Hg) C.
2 PO2 (mm Hg)

100 100
Hemoglobin saturation ( %)

80

-BPG
80

G
BP
Hemoglobin saturation (%)

,3

PG
,3-
60
PCO  20 mm Hg
No 2

-B
al 2

2,3
2

rm

ed
PCO  40 mm Hg 60 No
Add
40 2

PCO2  80 mm Hg

20 40

0
0 20 40 60 80 100 120 140 160
20
B. PO (mm Hg)
2

0
0 20 40 60 80 100
D. PO (mm Hg)
2

FIGURE 36–2 The effects of pH (A), PCO2 (B), temperature (C), and 2,3-BPG (D) on the oxyhemoglobin dissociation curve. (Modified with
permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

A convenient way to discuss shifts in the oxyhemoglobin example, anemia secondary to erythrocyte loss does not
dissociation curve is the P50, shown in Figures 36–1 and 36–3. affect the combination of oxygen and hemoglobin for the
The P50 is the Po2 at which 50% of the hemoglobin present in remaining erythrocytes. It is the amount of hemoglobin that
the blood is in the deoxyhemoglobin state and 50% is in the decreases, not the percent saturation or even the arterial
oxyhemoglobin state. At a temperature of 37°C, a pH of 7.4, Po2. The arterial content of oxygen, however, in milliliters of
and a Pco2 of 40 mm Hg, normal human blood has a P50 of 26 oxygen per 100 mL of blood, is reduced, as shown in
or 27 mm Hg. If the oxyhemoglobin dissociation curve is Figure 36–4A, because the decreased amount of hemoglobin
shifted to the right, the P50 increases. If it is shifted to the left, per 100 mL of blood decreases the oxygen-carrying capacity
the P50 decreases. of the blood.
Carbon monoxide has a much greater affinity for hemoglo-
bin than does oxygen, as discussed in Chapter 35. It can there-
Other Factors Affecting Oxygen Transport fore effectively block the combination of oxygen with
Most forms of anemia (low blood hemoglobin concentra- hemoglobin because oxygen cannot be bound to iron atoms
tion or low number of red blood cells) do not affect the oxy- already combined with carbon monoxide. Carbon monoxide
hemoglobin dissociation curve if the association of oxygen has a second deleterious effect: it shifts the oxyhemoglobin
and hemoglobin is expressed as percent saturation. For dissociation curve to the left. Thus, carbon monoxide can
368 SECTION VI Pulmonary Physiology

a to oxidant drugs, or it can be found congenitally in patients


100
with hemoglobin M. Iron atoms in the Fe3+ state will not com-
bine with oxygen.
80 As already discussed in this chapter, variants of the normal
HbA may have different affinities for oxygen. HbF in red
Hemoglobin saturation (%)

blood cells has a dissociation curve to the left of that for


HbA, as shown in Figure 36–4B. Fetal Po2 is much lower than
60 v-
in the adult; the curve is located properly for its operating
50% range. Furthermore, HbF’s greater affinity for oxygen relative
to the maternal hemoglobin promotes transport of oxygen
40
across the placenta by maintaining the diffusion gradient.
The shape of the HbF curve in blood appears to be a result of
the fact that 2,3-BPG has little effect on the affinity of HbF
20
for oxygen.
Myoglobin (Mb), a heme protein that occurs naturally in
muscle cells, consists of a single polypeptide chain attached to
0 a heme group. It can therefore combine chemically with a sin-
0 20 40 60 80 100
P50 PO2 (mm Hg) gle molecule of oxygen and is similar structurally to a single
FIGURE 36–3 Oxyhemoglobin dissociation curves for arterial subunit of hemoglobin. As can be seen in Figure 36–4C, the
and venous blood. The venous curve is shifted to the right because hyperbolic dissociation curve of Mb (which is similar to that
the pH is lower and the PCO2 (and possibly the temperature) is higher. of a single hemoglobin subunit) is far to the left of that of nor-
The rightward shift results in a higher P50 for venous blood. a, arterial mal HbA; that is, at lower Po2, much more oxygen remains
point (P 2 = 100mm Hg); –v, mixed venous point (P 2 = 40mm Hg).
O O bound to Mb. Mb can therefore store oxygen in skeletal mus-
(Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: cle. As blood passes through the muscle, oxygen leaves hemo-
McGraw-Hill Medical, 2007.)
globin and binds to Mb. It can be released from the Mb when
conditions within muscle cause lower tissue Po2.
Cyanosis is not really an influence on the transport of oxy-
prevent the loading of oxygen into the blood in the lungs and gen but rather is a sign of poor transport of oxygen. It occurs
can also interfere with the unloading of oxygen at the tissues. when more than 5 g Hb/100 mL of arterial blood is in the
This can be seen in Figure 36–4A. deoxy state. It is a bluish purple discoloration of the skin, nail
Carbon monoxide is particularly dangerous for several rea- beds, and mucous membranes, and its presence is indicative of
sons. A person breathing very low concentrations of carbon an abnormally high concentration of deoxyhemoglobin in the
monoxide can slowly reach life-threatening levels of carboxy- arterial blood. Its absence, however, does not exclude hypox-
hemoglobin (COHb) in the blood because carbon monoxide emia because an anemic patient with hypoxemia may not have
has such a high affinity for hemoglobin. The effect is cumula- sufficient hemoglobin to appear cyanotic.
tive. What is worse is that a person breathing carbon monox-
ide is not aware of doing so—the gas is colorless, odorless, and
tasteless and does not elicit any reflex coughing or sneezing, TRANSPORT OF CARBON
increase in ventilation, or feeling of difficulty in breathing.
Smoking and living in urban areas cause small amounts of DIOXIDE BY THE BLOOD
COHb to be present in the blood of healthy adults. A non-
Carbon dioxide is carried in the blood in physical solution,
smoker who lives in a rural area may have only about 1%
chemically combined to amino acids in blood proteins, and as
COHb; a smoker who lives in an urban area may have 5–8%
bicarbonate ions. About 200–250 mL of carbon dioxide is pro-
COHb in the blood.
duced by the tissue metabolism each minute in a resting 70-kg
Hemoglobin within erythrocytes can rapidly scavenge nitric
person and must be carried by the venous blood to the lung for
oxide (NO). NO can react with oxyhemoglobin to form meth-
removal from the body. At a cardiac output of 5 L/min, each
emoglobin and nitrate or react with deoxyhemoglobin to form
100 mL of blood passing through the lungs must therefore
a hemoglobin–NO complex. In addition, hemoglobin may act
unload 4–5 mL of carbon dioxide.
as a carrier for NO, in the form of S-nitrosothiol, on the cysteine
residues on the β-globin chain. This is called s-nitrosohemo-
globin (SNO-Hb). When hemoglobin binds oxygen, the for- PHYSICALLY DISSOLVED
mation of this S-nitrosothiol is enhanced; when hemoglobin
releases oxygen, NO could be released. Thus, in regions where Carbon dioxide is about 20 times more soluble in the plasma
the Po2 is low, NO—a potent vasodilator—could be released. (and inside the erythrocytes) compared to oxygen. As a result,
Methemoglobin is hemoglobin with iron in the ferric (Fe3+) about 5–10% of the total carbon dioxide transported by the
state. It can be caused by nitrite poisoning or by toxic reactions blood is carried in physical solution.
CHAPTER 36 Transport of Oxygen and Carbon Dioxide 369

20 100
Normal blood
O2 bound to hemoglobin, mL O2/100 mL

16 80

Mb

nit
bu
su
60

A
12

Hb
Saturation (%)

Hb
50% CO Hb

40
8
Anemia
(6 g Hb/100 mL blood)

20
4

0
0 0 20 40 60 80 100
0 20 40 60 80 100
A. C. PO (mm Hg)
PO2 (mm Hg) 2

100

80
Hemoglobin saturation (%)

HbF

60
A
Hb

40 FIGURE 36–4 Other physiologic factors influencing oxygen


transport and storage. A) The effects of carbon monoxide and
anemia on the carriage of oxygen by hemoglobin. Note that the
ordinate is expressed as the volume of oxygen bound to hemoglobin
20 in milliliters of oxygen per 100 mL of blood. B) A comparison of the
oxyhemoglobin dissociation curves for normal adult hemoglobin
(HbA) and fetal hemoglobin (HbF). C) Dissociation curves for normal
0 HbA, a single monomeric subunit of hemoglobin (Hb subunit), and
0 20 40 60 80 100 myoglobin (Mb). (Modified with permission from Levitzky MG: Pulmonary
B. PO2 (mm Hg) Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

About 0.0006mL CO2/(mm Hg Pco2) will dissolve in 1 mL CARBAMINO COMPOUNDS


of plasma at 37°C. One hundred milliliters of plasma or
whole blood at a Pco2 of 40 mm Hg, therefore, contains about Carbon dioxide can combine chemically with the terminal
2.4 mL CO2 in physical solution. Figure 36–5 shows that the amine groups in blood proteins, forming carbamino com-
total CO2 content of whole blood is about 48 mL CO2/100 pounds. The reaction occurs rapidly; no enzymes are necessary.
mL of blood at 40 mm Hg, so approximately 5% of the car- Note that a hydrogen ion is released when a carbamino com-
bon dioxide carried in the arterial blood is in physical solu- pound is formed:
tion. Similarly, multiplying 0.06mL CO2/100mL of blood/
(mm Hg Pco2) times a venous Pco2 of 45 mm Hg shows that H H
about 2.7 mL CO2 is physically dissolved in the mixed venous
blood. The total carbon dioxide content of venous blood is R N + CO2 R N + H+
about 52.5 mL CO2/100 mL of blood; a little more than 5% of
H COO–
the total carbon dioxide content of venous blood is in physi- Terminal Carbamino
cal solution. amine group compound
370 SECTION VI Pulmonary Physiology

70

Whole blood carbon dioxide content, mL CO2 /100 mL blood


HbO 2
0%
60 HbO 2
70%
v- bO 2
%H
50 97.5
a

40

30

20

FIGURE 36–5 Carbon dioxide dissociation curves 10


for whole blood (37°C) at different oxyhemoglobin
saturations. Note that the ordinate is whole blood CO2 content
in milliliters of CO2 per 100 mL of blood. a, arterial point; –v, 0
0 10 20 30 40 50 60 70 80
mixed venous point. (Modified with permission from Levitzky MG:
PCO (mm Hg)
Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.) 2

Because the protein found in greatest concentration in the dissociation of carbonic acid, thus allowing the reaction to
blood is the globin of hemoglobin, most of the carbon diox- continue. This will be discussed in detail in the last section of
ide transported in this manner is bound to amino acids of this chapter.
hemoglobin. Deoxyhemoglobin can bind more carbon diox-
ide as carbamino groups than can oxyhemoglobin. Therefore,
as the hemoglobin in the venous blood enters the lung THE CARBON DIOXIDE
and combines with oxygen, it releases carbon dioxide from
its terminal amine groups. About 5–10% of the total car- DISSOCIATION CURVE
bon dioxide content of blood is in the form of carbamino
The carbon dioxide dissociation curve for whole blood is
compounds.
shown in Figure 36–5. Within the normal physiologic range
of Pco2, the curve is nearly a straight line, with no steep or
BICARBONATE flat portions. The carbon dioxide dissociation curve for
whole blood is shifted to the right at greater levels of oxyhe-
The remaining 80–90% of the carbon dioxide transported by moglobin and shifted to the left at greater levels of deoxyhe-
the blood is carried as bicarbonate ions. This is made possible moglobin. This is known as the Haldane effect. The Haldane
by the following reaction: effect allows the blood to load more carbon dioxide at the
tissues, where there is more deoxyhemoglobin, and unload
CO2 + H2O Carbonic anhydrase H2CO3 H+ + HCO3– (12) more carbon dioxide in the lungs, where there is more oxy-
hemoglobin.
Carbon dioxide can combine with water to form carbonic The Bohr and Haldane effects are both explained by the fact
acid, which then dissociates into a hydrogen ion and a bicar- that deoxyhemoglobin is a weaker acid than oxyhemoglobin;
bonate ion. that is, deoxyhemoglobin more readily accepts the hydrogen
Very little carbonic acid is formed by the association of ion liberated by the dissociation of carbonic acid, thus permit-
water and carbon dioxide without the presence of the enzyme ting more carbon dioxide to be transported in the form of
carbonic anhydrase because the reaction occurs so slowly. bicarbonate ion. This is referred to as the isohydric shift. Con-
Carbonic anhydrase, which is present in high concentration versely, the association of hydrogen ions with the amino acids
in erythrocytes (but not in plasma), makes the reaction pro- of hemoglobin lowers the affinity of hemoglobin for oxygen,
ceed about 13,000 times faster. (Note that the product of the thus shifting the oxyhemoglobin dissociation curve to the
carbonic anhydrase reaction is actually not carbonic acid, but right at low pH or high Pco2. The following relationship can
a bicarbonate ion and a proton—see Chapter 47.) Hemoglo-
therefore be written:
bin also plays an integral role in the transport of carbon diox-
ide because it can accept the hydrogen ion liberated by the H+Hb + O2 H+ + HbO2 (13)
CHAPTER 36 Transport of Oxygen and Carbon Dioxide 371

A. IN THE TISSUES

TISSUE PLASMA ERYTHROCYTE

Dissolved

CO2 CO2 CO2 Dissolved

H2O H2O  CO2

CAPILLARY WALL
Carbonic anhydrase CO2
H2CO3

HCO3 HCO3  H

Cl Cl
H  HbO2

O2 O2 O2  HHb HHb  CO2

Carbamino compounds

B. IN THE LUNG

ALVEOLUS PLASMA ERYTHROCYTE

Dissolved

CO2 CO2 CO2 Dissolved

H2O H2O  CO2


CAPILLARY WALL

Carbonic anhydrase CO2


H2CO3

HCO3 HCO3  H
Cl Cl
H  HbO2

O2 O2 O2  HHb HHb  CO2

Carbamino compounds

FIGURE 36–6 Schematic representation of uptake and release of carbon dioxide and oxygen at the tissues (A) and in the lung (B).
Note that small amounts of carbon dioxide can form carbamino compounds with blood proteins other than hemoglobin and may also be
hydrated in trivial amounts in the plasma to form carbonic acid and then bicarbonate (not shown in diagram). The circles represent the
bicarbonate–chloride exchange carrier protein. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

These effects can be seen in the schematic diagrams of oxy- deoxyhemoglobin in the erythrocytes and the deoxyhemo-
gen and carbon dioxide transport shown in Figure 36–6. globin is able to accept the hydrogen ions liberated by the dis-
At the tissues, the Po2 is decreased and the Pco2 is increased. sociation of carbonic acid and the formation of carbamino
Carbon dioxide dissolves in the plasma, and some diffuses compounds. The hydrogen ions released by the dissociation
into the erythrocyte. Some of this carbon dioxide dissolves in of carbonic acid and the formation of carbamino compounds
the cytosol, some forms carbamino compounds with hemo- bind to specific amino acid residues on the globin chains and
globin, and some is hydrated by carbonic anhydrase to form facilitate the release of oxygen from hemoglobin (the Bohr
carbonic acid. At low Po2, there are substantial amounts of effect). Bicarbonate ions diffuse out of the erythrocyte
372 SECTION VI Pulmonary Physiology

through the cell membrane much more readily than do


hydrogen ions. Because more bicarbonate ions than hydrogen output, and blood pressure. However, all of these
ions leave the erythrocyte, electrical neutrality is maintained responses were not sufficient to increase his blood pres-
by the exchange of chloride ions for bicarbonate ions by the sure or his cardiac output to normal levels, as he contin-
bicarbonate–chloride carrier protein. This is the “chloride ued to lose blood. The decreased cardiac output and
shift.” Small amounts of water also move into the cell to main- increased vascular resistance to most vascular beds
tain the osmotic equilibrium. resulted in decreased tissue perfusion (including his skin,
At the lung, the Po2 is increased and the Pco2 is decreased. As explaining his pale appearance). This ischemia resulted
oxygen combines with hemoglobin, the hydrogen ions that in production of lactic acid causing hydrogen ion stimu-
were taken up when it was in the deoxyhemoglobin state are lation of the arterial chemoreceptors (see Chapters 37
released. They combine with bicarbonate ions, forming car- and 38), which explains his tachypnea (high respiratory
bonic acid. This breaks down into carbon dioxide and water. rate). He was hyperventilating in compensation as dem-
At the same time, carbon dioxide is also released from the car- onstrated by the hypocapnia. As he continued to lose
bamino compounds. Carbon dioxide then diffuses out of the blood, his blood pressure was no longer sufficient to pro-
red blood cells and plasma and into the alveoli. A chloride vide adequate cerebral blood flow and he lost conscious-
shift opposite in direction to that in the tissues also occurs to ness and showed signs of circulatory shock.
maintain electrical neutrality. Administration of normal saline temporarily increased
blood volume, but diluted his erythrocytes, decreasing his
hematocrit, hemoglobin concentration, oxygen-carrying
capacity, and arterial oxygen content, even if his alveolar
and arterial partial pressures of oxygen were normal.
CLINICAL CORRELATION Mixed venous Po2 would decrease as tissues extracted as
An 18-year-old man is brought by ambulance to the emer- much oxygen as possible from the arterial blood. Renal
gency department about 35 minutes after being shot in the and endocrine responses to hemorrhage also would occur,
leg. He is conscious, although disoriented and in pain, and as will be discussed in Sections 7 and 9.
appears pale. Heart rate is 150/min and his arterial blood In the emergency department, his treatment would be
pressure is 80/60 mm Hg. He is breathing spontaneously aimed at stopping blood loss and restoring cardiac output
with a high respiratory rate of 26/min. During the trip to and blood pressure with matched packed red blood cells
hospital, the wound was stabilized and he received 2 L of (red blood cells after most of the plasma and other cells
normal saline (0.9% NaCl in water) solution intravenously. have been removed from whole blood) to restore his oxy-
In the emergency department, he continues to lose gen carrying capacity.
blood while the physicians attempt to stop the hemor-
rhage. As his arterial blood pressure continues to
decrease to 60/45 mm Hg, he is given 2 additional liters
of saline. His hematocrit decreases to 21% (normal range
CHAPTER SUMMARY
40–50%), corresponding to a hemoglobin concentration ■ Blood normally carries a small amount of oxygen physically
of 7 g/100 mL of blood (normal range 13–18 g/100 mL dissolved in the plasma and a large amount chemically
combined to hemoglobin: only the physically dissolved oxygen
blood). His respiratory rate increases to 40/min.
contributes to the partial pressure, but the partial pressure of
Results of blood gas analysis (see Chapter 37) from an
oxygen determines how much combines chemically with
arterial blood sample show an arterial Po2 of 95 mm Hg, an hemoglobin.
arterial Pco2 of 28 mm Hg (normal range 35–45 mm Hg),
■ The oxyhemoglobin dissociation curve describes the reversible
and an arterial pH of 7.30 (normal range 7.35–7.45) despite reaction of oxygen and hemoglobin to form oxyhemoglobin; it
the hypocapnia. He becomes agitated and loses conscious- is relatively flat at a Po2 above approximately 70 mm Hg and is
ness. He is intubated (a tube inserted into trachea) and very steep at a Po2 in the range of 20–40 mm Hg.
mechanically ventilated via the endotracheal tube. ■ Decreased pH, increased Pco2, increased temperature, and
The patient’s decreased blood volume led to decreased increased 2,3-BPG concentration of the blood all shift the
venous return, decreased cardiac output, and decreased oxyhemoglobin dissociation curve to the right.
systemic blood pressure. Decreased firing of the barore- ■ Blood normally carries small amounts of carbon dioxide
ceptors in the carotid sinuses and aortic arch decreased physically dissolved in the plasma and chemically combined to
parasympathetic stimulation of the heart and increased blood proteins as carbamino compounds and a large amount in
sympathetic stimulation of the heart, arterioles, and the the form of bicarbonate ions.
veins. This resulted in increased heart rate and myocar- ■ Deoxyhemoglobin favors the formation of carbamino
dial contractility; increased arteriolar tone; and decreased compounds, and it promotes the transport of carbon dioxide
venous compliance to enhance venous return, cardiac as bicarbonate ions by buffering hydrogen ions formed by
the dissociation of carbonic acid.
CHAPTER 36 Transport of Oxygen and Carbon Dioxide 373

STUDY QUESTIONS 3. Which of the following should increase the P50 of the
oxyhemoglobin dissociation curve?
1. An otherwise healthy person has lost enough blood to decrease A) hypercapnia
the hemoglobin concentration from 15 to 12 g/100 mL blood. B) acidosis
Which of the following would be expected to decrease? C) increased blood levels of 2,3-BPG
A) arterial Po2 D) increased body temperature
B) blood oxygen-carrying capacity E) all of the above
C) arterial hemoglobin saturation 4. Most of the carbon dioxide in the blood is transported
D) arterial oxygen content
A) as bicarbonate.
E) B and D.
B) as carbamino compounds.
2. A woman’s hemoglobin concentration is 10 g of hemoglobin per C) physically dissolved in the plasma.
100 mL of blood. If her hemoglobin is 90% saturated with oxygen D) physically dissolved inside erythrocytes.
at an arterial Po2 of 80 mm Hg, what is her total arterial oxygen
content, including physically dissolved oxygen?
A) 10.72 mL O2/100 mL blood
B) 10.96 mL O2/100 mL blood
C) 12.06 mL O2/100 mL blood
D) 12.30 mL O2/100 mL blood
E) 13.40 mL O2/100 mL blood
This page intentionally left blank
37
C H A P T E R

Acid–Base Regulation
and Causes of Hypoxia
Michael Levitzky

O B J E C T I V E S

■ Define acids, bases, and buffers.


■ List the buffer systems available in the human body.
■ State the normal ranges of arterial pH, PCO2, and bicarbonate concentration,
and define alkalosis and acidosis.
■ List the potential causes of respiratory acidosis and alkalosis and metabolic
acidosis and alkalosis.
■ Discuss the respiratory mechanisms that help compensate for acidosis
and alkalosis.
■ Evaluate blood gas data to determine acid–base status.
■ Classify and explain the causes of tissue hypoxia.

The respiratory and renal systems maintain the balance of strong acids or bases are added; that is, the changes in hydro-
acids and bases in the body. This chapter will introduce the gen ion concentration that occur when a strong acid or base is
major concepts of the respiratory system’s contribution to added to a buffer system are much smaller than those that
acid–base balance; Chapter 47 addresses the renal system con- would occur if the same amount of acid or base were added to
tribution to acid–base balance and includes a more detailed pure water or another nonbuffer solution.
discussion of the basic chemistry of acid–base physiology, buf- The hydrogen ion activity of pure water is about 1.0 ×
fers, and the chemistry of the CO2–bicarbonate system. 10–7 mol/L. By convention, solutions with hydrogen ion activi-
ties above 10–7 mol/L are considered to be acid; those with
hydrogen ion activities below 10–7 are considered to be alka-
INTRODUCTION TO line. The range of hydrogen ion concentrations or activities in
ACID–BASE CHEMISTRY the body is normally from about 10–1 for gastric acid to about
10–8 for the most alkaline pancreatic secretion. This wide range
An acid can be simply defined as a substance that can donate of hydrogen ion activities necessitates the use of the more con-
a hydrogen ion (a proton) to another substance and a base as venient pH scale. The pH of a solution is the negative loga-
a substance that can accept a hydrogen ion from another sub- rithm of its hydrogen ion activity. With the exception of the
stance. A strong acid is a substance that is completely or highly concentrated gastric acid, in most instances in the body,
almost completely dissociated into a hydrogen ion and its cor- the hydrogen ion activity is about equal to the hydrogen ion
responding or conjugate base in dilute aqueous solution; a concentration.
weak acid is only slightly ionized in aqueous solution. In gen- The pH of arterial blood is normally close to 7.40, with a
eral, a strong acid has a weak conjugate base and a weak acid normal range considered to be about 7.35–7.45. An arterial
has a strong conjugate base. The strength of an acid or a base pH (pHa) less than 7.35 is considered acidemia; a pHa greater
should not be confused with its concentration. than 7.45 is considered alkalemia. The underlying condition
A buffer is a mixture of substances in aqueous solution characterized by hydrogen ion retention or by loss of bicar-
(usually a combination of a weak acid and its conjugate base) bonate or other bases is referred to as acidosis; the underlying
that can resist changes in hydrogen ion concentration when condition characterized by hydrogen ion loss or retention of

375

Ch37_375-384.indd 375 11/29/10 4:40:09 PM


376 SECTION VI Pulmonary Physiology

TABLE 37–1 The pH scale. ventilation. Carbonic acid is therefore said to be a volatile acid
because it can be converted into a gas and then removed from
pH Concentration (nmol/L) an open system such as the body. Very great amounts of car-
6.90 126
bon dioxide can be removed from the lungs by alveolar venti-
lation: under normal circumstances, about 15,000–25,000
7.00 100 mmol of carbon dioxide is removed via the lungs daily.
7.10 79 A much smaller quantity of fixed or nonvolatile acids is
7.20 63
also normally produced during the course of the metabolism
of foodstuffs. The fixed acids produced by the body include
7.30 50 sulfuric acid, which originates from the oxidation of sulfur-
7.40 40 containing amino acids such as cysteine; phosphoric acid from
7.50 32
the oxidation of phospholipids and phosphoproteins; hydro-
chloric acid, which is produced during the conversion of
7.60 25 ingested ammonium chloride to urea and by other reactions;
7.70 20 and lactic acid from the anaerobic metabolism of glucose.
7.80 16
Other fixed acids may be ingested accidentally or formed in
abnormally large quantities by disease processes, such as the
Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
New York: McGraw-Hill Medical, 2007. acetoacetic and butyric acid formed during diabetic ketoaci-
dosis (see Chapter 66). About 70 mEq of fixed acids is nor-
mally removed from the body each day (about 1 mEq/kg/body
base is referred to as alkalosis. Under pathologic conditions, weight per day); the range is 50–100 mEq. A vegetarian diet
the extremes of arterial blood pH have been noted to range as may produce significantly less fixed acid and may even result
high as 7.8 and as low as 6.9. These correspond to hydrogen in no net production of fixed acids. The removal of fixed acids
ion concentrations as seen in Table 37–1 (hydrogen ion con- is accomplished mainly by the kidneys, as will be discussed in
centrations are expressed as nanomoles [10–9 mol/L] for conve- Chapter 47. Some may also be removed via the gastrointestinal
nience). tract. Fixed acids normally represent only about 0.2% of the
Note that the pH scale is “inverted” by the negative sign and total body acid production.
is also logarithmic as it is defined. An increase in pH repre- The body contains a variety of substances that can act as
sents a decrease in hydrogen ion concentration. In fact, an buffers in the physiologic pH range. These include bicarbon-
increase of only 0.3 pH units indicates that the hydrogen ion ate, phosphate, and proteins in the blood, the interstitial fluid,
concentration was cut in half. and inside cells (discussed in greater detail in Chapter 47). The
Hydrogen ions are the most reactive cations in body fluids, isohydric principle states that all the buffer pairs in a homo-
and they interact with negatively charged regions of other geneous solution are in equilibrium with the same hydrogen
molecules, such as those of body proteins. Interactions of ion concentration. For this reason, all the buffer pairs in the
hydrogen ions with negatively charged functional groups of plasma behave similarly, so that the detailed analysis of a single
proteins can lead to marked changes in protein structural con- buffer pair, like the bicarbonate buffer system, can reveal a
formations with resulting alterations in the behavior of the great deal about the chemistry of all the plasma buffers.
proteins. An example of this was already seen in Chapter 36, The main buffers of the blood are bicarbonate, phosphate,
where hemoglobin was noted to combine with less oxygen at a and proteins. The bicarbonate buffer system consists of the
lower pH (the Bohr effect). Alterations in the structural con- buffer pair of the weak acid, carbonic acid, and its conjugate
formations and charges of protein enzymes affect their activi- base, bicarbonate. The ability of the bicarbonate system to
ties, with resulting alterations in the functions of body tissues. function as a buffer of fixed acids in the body is largely due to
Extreme changes in the hydrogen ion concentration of the the ability of the lungs to remove carbon dioxide from the
body can result in loss of organ system function and may be body. In a closed system, bicarbonate would not be nearly as
fatal. effective.
Under normal circumstances, cellular metabolism is the At a temperature of 37°C, about 0.03 mmol of carbon diox-
main source of acids in the body. These acids are the waste ide per mm Hg of Pco2 will dissolve in a liter of plasma. (Note
products of substances ingested as foodstuffs. The greatest that the solubility of CO2 was expressed as milliliters of CO2
source of hydrogen ions is the carbon dioxide produced as one per 100 mL of plasma in Chapter 36.) Therefore, the carbon
of the end products of the oxidation of glucose and fatty acids dioxide dissolved in the plasma, expressed as millimoles per
during aerobic metabolism. The hydration of carbon dioxide liter, is equal to 0.03 x Pco2 . At body temperature in the plasma,
results in the formation of carbonic acid, which then can dis- the equilibrium of the reaction is such that there is roughly
sociate into a hydrogen ion and a bicarbonate ion, as discussed 1,000 times more carbon dioxide physically dissolved in the
in Chapter 36. This process is reversed in the pulmonary capil- plasma than there is in the form of carbonic acid. The dis-
laries, and CO2 then diffuses through the alveolar–capillary solved carbon dioxide is in equilibrium with the carbonic acid,
barrier into the alveoli, from which it is removed by alveolar though, so both the dissolved carbon dioxide and the carbonic
CHAPTER 37 Acid–Base Regulation and Causes of Hypoxia 377

acid are considered as the undissociated HA in the Hender- each value of pH and bicarbonate ion concentration, there is a
son–Hasselbalch equation (see Chapter 47) for the bicarbon- single corresponding Pco2 on the graph. Conversely, for any
ate system: particular pH and Pco2, only one bicarbonate ion concentra-
[HCO3–]p tion will satisfy the Henderson–Hasselbalch equation. If the
pH = pK + log ___________
[CO + H CO ]
(1) Pco2 is held constant, for example, at 40 mm Hg, an isobar line
2 2 3
can be constructed, connecting the resulting points as the pH
where [HCO3−]p stands for plasma bicarbonate concentration.
is varied. The representative isobars shown in Figure 37–1 give
The concentration of carbonic acid is negligible, so we have:
an indication of the potential alterations of acid–base status
[HCO3–]p
pH = pK′ + log _______ (2) when alveolar ventilation is increased or decreased. If every-
0.03 P co2 thing else remains constant, hypoventilation leads to acidosis;
where pK′ is the pK of the HCO3−–CO2 system in blood. hyperventilation leads to alkalosis.
The pK′ of this system at physiologic pH values and at 37°C The bicarbonate buffer system is a poor buffer for carbonic
is 6.1. Therefore, at a pHa of 7.40 and an arterial Pco2 of 40 mm acid. The presence of hemoglobin makes blood a much better
Hg, we have: buffer. The buffer value of plasma in the presence of hemoglobin
[HCO3–]p is four to five times that of plasma separated from erythrocytes.
7.40 = 6.1 + log _________ (3) Therefore, the slope of the normal in vivo buffer line shown in
1.2 mmol/L
Figures 37–1 is mainly determined by the nonbicarbonate buf-
Therefore, the arterial plasma bicarbonate concentration is
fers present in the body. The phosphate buffer system mainly
about 24 mmol/L (the normal range is 23–28 mmol/L) because
consists of the buffer pair of the dihydrogen phosphate (H2PO4−)
the logarithm of 20 is equal to 1.3.
and the monohydrogen phosphate (HPO42−) anions.
Note that the term total CO2 refers to the dissolved carbon
Although several potential buffering groups are found on
dioxide (including carbonic acid) plus the carbon dioxide
proteins, only one large group has pK in the pH range
present as bicarbonate.
encountered in the blood. These are the imidazole groups in
A useful way to display the interrelationships among the
the histidine residues of the peptide chains. The protein pres-
variables of pH, Pco2, and bicarbonate concentration of the
ent in the greatest quantity in the blood is hemoglobin. As
plasma, as expressed by the Henderson–Hasselbalch equation,
already noted, deoxyhemoglobin is a weaker acid than is
is the pH–bicarbonate diagram shown in Figure 37–1.
oxyhemoglobin. Thus, as oxygen leaves hemoglobin in the
As can be seen from Figure 37–1, pH is on the abscissa of
tissue capillaries, the imidazole group removes hydrogen
the pH–bicarbonate diagram, and the plasma bicarbonate
ions from the erythrocyte interior, allowing more carbon
concentration in millimoles per liter is on the ordinate. For
dioxide to be transported as bicarbonate. This process is
reversed in the lungs.
The bicarbonate buffer system is the major buffer found in
[H ] (nmol/L) the interstitial fluid, including the lymph. The phosphate buffer
100 80 70 60 50 40 30 20 16 pair is also found in the interstitial fluid. The volume of the
40
A interstitial compartment is much larger than that of the plasma,
r

so the interstitial fluid may play an important role in buffering.


ba

35
iso

The extracellular portion of bone contains very large depos-


r
ba
Hg

iso

its of calcium and phosphate salts, mainly in the form of


mm

r
ba
Hg
[HCO3]p (mmol/L)

30 B
iso

hydroxyapatite. In an otherwise healthy adult, where bone


80

Hg
m


growth and resorption are in a steady state, bone salts can buf-
60

m
2
CO

m


fer hydrogen ions in chronic acidosis. Chronic buffering of


P

25 r
40

C ba
2
CO

iso


hydrogen ions by the bone salts may therefore lead to demin-


P

Hg
2
CO
P

20 m eralization of bone.
D 0m

2 The intracellular proteins and organic phosphates of most
E P CO
2 cells can function to buffer both fixed acids and carbonic acid.
15 Normal buffer line
Of course, buffering by the hemoglobin in erythrocytes is
intracellular.
10
7.0 7.1 7.2 7.3 7.4 7.5 7.6 7.7 7.8
pH

FIGURE 37–1 The pH–bicarbonate diagram with PCO isobars.


2
ACIDOSIS AND ALKALOSIS
Note the hydrogen ion concentration in nanomoles per liter at the
top of the figure corresponding to the pH values on the abscissa. Acid–base disorders can be divided into four major categories:
Points A to E correspond to different pH values and bicarbonate respiratory acidosis, respiratory alkalosis, metabolic acidosis,
concentrations all falling on the same PCO isobar. (Modified with and metabolic alkalosis. These primary acid–base disorders
2
permission of the University of Chicago Press from Davenport HW: The ABC of may occur singly (“simple”) or in combination (“mixed”) or
Acid–Base Chemistry, 6th ed. 1974.) may be altered by compensatory mechanisms.
378 SECTION VI Pulmonary Physiology

RESPIRATORY ACIDOSIS TABLE 37–2 Common causes of respiratory acidosis.


The arterial Pco2 is normally maintained at or near 40 mm Hg Depression of the respiratory control centers
Anesthetics
(normal range is 35–45 mm Hg) by the mechanisms that reg- Sedatives
ulate breathing. Sensors exposed to the arterial blood and to Opiates
the cerebrospinal fluid provide the central controllers of Brain injury or disease
breathing with the information necessary to regulate the arte- Severe hypercapnia, hypoxia
rial Pco2 at or near 40 mm Hg (see Chapter 38). Any short-term Neuromuscular disorders
alterations (i.e., those which occur without renal compensa- Spinal cord injury
tion) in alveolar ventilation that result in an increase in alveo- Phrenic nerve injury
Poliomyelitis, Guillain–Barré syndrome, etc.
lar and therefore also in arterial Pco2 tend to lower the pHa,
Botulism, tetanus
resulting in respiratory acidosis. This can be appreciated by Myasthenia gravis
examining the Pco2 = 60 and 80 mm Hg isobars in Figure 37–1. Administration of curarelike drugs
The pHa at any Paco2 depends on the bicarbonate and other Diseases affecting the respiratory muscles
buffers present in the blood. Pure changes in arterial Pco2 Chest wall restriction
caused by changes in ventilation travel along the normal in Kyphoscoliosis
vivo buffer line (Figures 37–1 and 37–2). Pure uncompen- Extreme obesity
sated respiratory acidosis would correspond with point C in Lung restriction
Figure 37–2 (at the intersection of an elevated Pco2 isobar and Pulmonary fibrosis
the normal buffer line). Sarcoidosis
In respiratory acidosis, the ratio of bicarbonate to CO2 Pneumothorax, pleural effusions, etc.
decreases. Yet, as can be seen at point C in Figure 37–2, in Pulmonary parenchymal diseases
uncompensated primary (simple) respiratory acidosis, the Pneumonia, etc.
absolute plasma bicarbonate concentration does increase Pulmonary edema
somewhat because of the buffering of some of the hydrogen Airway obstruction
ions liberated by the dissociation of carbonic acid by nonbi- Chronic obstructive pulmonary disease
carbonate buffers. Upper airway obstruction
Any impairment of alveolar ventilation can cause respira- Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
tory acidosis. As shown in Table 37–2, depression of the New York: McGraw-Hill Medical, 2007.
respiratory centers in the medulla (see Chapter 38) by anes-
thetic agents, narcotics, hypoxia, central nervous system dis-
ease or trauma, or even greatly increased PaCo2 itself results in processes, drugs or toxins, or dysfunctions or deformities of
hypoventilation and respiratory acidosis. Interference with the respiratory muscles or the chest wall can result in respi-
the neural transmission to the respiratory muscles by disease ratory acidosis. Restrictive, obstructive, and obliterative dis-
eases of the lungs can also result in respiratory acidosis.

40 RESPIRATORY ALKALOSIS
Metabolic alkalosis
and respiratory acidosis F
35
Uncompensated Alveolar ventilation in excess of that needed to keep pace with
metabolic alkalosis
D body’s carbon dioxide production results in alveolar and arte-
Metabolic alkalosis rial Pco2 below 35 mm Hg. Such hyperventilation leads to respi-
[HCO3]p (mmol/L)

30 Uncompensated
respiratory acidosis
E ratory alkalosis. Uncompensated primary respiratory alkalosis
Metabolic alkalosis
C
and respiratory alkalosis
results in movement to a lower Pco2 isobar along the normal buf-
25 Metabolic acidosis A fer line, as seen at point B in Figure 37–2. The decreased Paco2
and respiratory acidosis
I Normal buffer line shifts the equilibrium of the series of reactions describing car-
Metabolic
20 G acidosis
bon dioxide hydration and carbonic acid dissociation to the left.
Uncompensated B This results in a decreased arterial hydrogen ion concentration,
metabolic acidosis Uncompensated
respiratory alkalosis increased pH, and a decreased plasma bicarbonate concentra-
15
Metabolic acidosis H tion. The ratio of bicarbonate to carbon dioxide increases.
and respiratory alkalosis
The causes of respiratory alkalosis include anything leading
10
7.0 7.1 7.2 7.3 7.4 7.5 7.6 7.7 7.8 to hyperventilation. As shown in Table 37–3, hyperventilation
pH syndrome, a psychological dysfunction of unknown cause,
FIGURE 37–2 Acid–base paths in vivo. (Modified with permission of results in chronic or recurrent episodes of hyperventilation and
the University of Chicago Press from Davenport HW: The ABC of Acid–Base Chemistry, respiratory alkalosis. Drugs, hormones (such as progesterone),
6th ed. 1974.) toxic substances, central nervous system diseases or disorders,
CHAPTER 37 Acid–Base Regulation and Causes of Hypoxia 379

TABLE 37–3 Common causes of respiratory alkalosis. TABLE 37–4 Common causes of metabolic acidosis.
Central nervous system Ingested drugs or toxic substances
Anxiety Methanol
Hyperventilation syndrome Ethanol
Inflammation (encephalitis, meningitis) Salicylates
Cerebrovascular disease Ethylene glycol
Tumors Ammonium chloride

Drugs or hormones Loss of bicarbonate ions


Salicylates Diarrhea
Progesterone Pancreatic fistulas
Renal dysfunction
Bacteremias, fever
Lactic acidosis
Pulmonary diseases Hypoxemia
Acute asthma Anemia, carbon monoxide
Pulmonary vascular diseases (pulmonary embolism) Shock (hypovolemic, cardiogenic, septic, etc.)
Severe exercise
Overventilation with mechanical ventilators Acute respiratory distress syndrome (ARDS)

Hypoxia; high altitude Ketoacidosis


Diabetes mellitus
Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
New York: McGraw-Hill Medical, 2007. Alcoholism
Starvation

Inability to excrete hydrogen ions


bacteremias, fever, overventilation by mechanical ventilators
Renal dysfunction
(or the clinician), or ascent to high altitude may all result in
respiratory alkalosis. Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
New York: McGraw-Hill Medical, 2007.

METABOLIC ACIDOSIS
Metabolic acidosis may be thought of as nonrespiratory
as a consequence of the ingestion, infusion, or excessive renal
acidosis. It can be caused by the ingestion, infusion, or
reabsorption of bases such as bicarbonate. Figure 37–2 shows
production of a fixed acid; decreased renal excretion of
that primary uncompensated metabolic alkalosis results in an
hydrogen ions; the movement of hydrogen ions from the
upward movement along the Pco2 = 40 mm Hg isobar to point
intracellular to the extracellular compartment; or the
D, that is, a net gain of buffer establishes a new blood–buffer
loss of bicarbonate or other bases from the extracellular com-
line higher than and parallel to the normal blood–buffer line.
partment. As can be seen in Figure 37–2, primary uncom-
Pco2 is unchanged, hydrogen ion concentration is decreased,
pensated metabolic acidosis results in a downward movement
and the ratio of bicarbonate concentration to carbon dioxide is
along the Pco2 = 40 mm Hg isobar to point G, that is, a net loss
increased.
of buffer establishes a new blood–buffer line lower than and
As shown in Table 37–5, loss of gastric juice by vomiting
parallel to the normal blood–buffer line. Pco2 is unchanged,
results in a loss of hydrogen ions and may cause metabolic
hydrogen ion concentration is increased, and the ratio of
alkalosis. Excessive ingestion of bicarbonate or other bases
bicarbonate concentration to CO2 is decreased.
(e.g., stomach antacids) or overinfusion of bicarbonate by
As shown in Table 37–4, ingestion of methyl alcohol or sali-
the clinician may cause metabolic alkalosis. In addition,
cylates can cause metabolic acidosis by increasing the fixed
acids in the blood. (Salicylate poisoning—for example, aspirin
overdose—causes both metabolic acidosis and later respira-
tory alkalosis.) Diarrhea can cause significant bicarbonate TABLE 37–5 Common causes of metabolic alkalosis.
losses, resulting in metabolic acidosis. Renal dysfunction can
lead to an inability to excrete hydrogen ions, as well as an Loss of hydrogen ions
inability to reabsorb bicarbonate ions, as will be discussed in Vomiting
Gastric fistulas
the next section. True “metabolic” acidosis may be caused by Diuretic therapy
an accumulation of lactic acid in severe hypoxemia or shock
and by diabetic ketoacidosis. Treatment with or overproduction of steroids (aldosterone or other
mineralocorticoids)
Ingestion or administration of excess bicarbonate or other bases
METABOLIC ALKALOSIS Intravenous bicarbonate
Ingestion of bicarbonate or other bases (e.g., antacids)
Metabolic, or nonrespiratory, alkalosis occurs when there is Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
an excessive loss of fixed acids from the body, or it may occur New York: McGraw-Hill Medical, 2007.
380 SECTION VI Pulmonary Physiology

diuretic therapy, treatment with steroids (or the overpro- mechanism or the breathing apparatus itself. Compensa-
duction of endogenous steroids), and conditions leading to tion for acidosis or alkalosis in these conditions must there-
severe potassium depletion may also cause metabolic fore come from outside the respiratory system. The
alkalosis. respiratory compensatory mechanism can operate very
rapidly (within minutes) to partially correct metabolic aci-
dosis or alkalosis.
COMPENSATORY MECHANISMS
Uncompensated primary acid–base disturbances, such as RENAL COMPENSATORY MECHANISMS
those indicated by points B–D and G in Figure 37–2, seldom
The kidneys can compensate for respiratory acidosis and met-
occur because respiratory and renal compensatory mecha-
abolic acidosis of nonrenal origin by excreting fixed acids and
nisms are called into play to offset these disturbances. The two
by retaining filtered bicarbonate. They can also compensate
main compensatory mechanisms are functions of the respira-
for respiratory alkalosis or metabolic alkalosis of nonrenal ori-
tory and renal systems.
gin by decreasing hydrogen ion excretion and by decreasing
the retention of filtered bicarbonate. These mechanisms are
RESPIRATORY COMPENSATORY discussed in Chapter 47. Renal compensatory mechanisms for
acid–base disturbances operate much more slowly than respi-
MECHANISMS ratory compensatory mechanisms. For example, the renal
compensatory responses to sustained respiratory acidosis or
The respiratory system can compensate for metabolic acidosis
alkalosis may take 3–6 days.
or alkalosis by altering alveolar ventilation. As discussed in
The kidneys help regulate acid–base balance by altering the
Chapter 33, if carbon dioxide production is constant, the alve-
excretion of fixed acids and the retention of the filtered bicar-
olar Pco2 is inversely proportional to the alveolar ventilation. In
bonate; the respiratory system helps regulate body acid–base
metabolic acidosis, the increased blood hydrogen ion concen-
balance by adjusting alveolar ventilation to alter alveolar Pco2. For
tration stimulates chemoreceptors, which, in turn, increase
these reasons, the Henderson–Hasselbalch equation is in effect:
alveolar ventilation, thus decreasing arterial Pco2. This causes
an increase in pHa, returning it toward normal. (The mecha- Kidneys
pH = Constant + ______
Lungs
(4)
nisms by which ventilation is regulated are discussed in detail
in Chapter 38.) These events can be better understood by look-
ing at Figure 37–2. Point G represents uncompensated meta-
bolic acidosis. As the respiratory compensation for the CLINICAL INTERPRETATION
metabolic acidosis occurs, in the form of an increase in venti- OF ARTERIAL BLOOD GASES
lation, the arterial Pco2 decreases. The point representing blood
pHa, Paco2, and bicarbonate concentration would then move a Samples of arterial blood are usually analyzed clinically to
short distance along the lower-than-normal buffer line (from determine the “arterial blood gases”: the arterial Po2, Pco2, and
point G toward point H) until a new lower Paco2 is attained. pH. The plasma bicarbonate can then be calculated from the
This returns the pHa toward normal; complete compensation pH and Pco2 by using the Henderson–Hasselbalch equation.
does not occur. The respiratory compensation for metabolic This can be done directly, or by using a nomogram, or by
acidosis occurs almost simultaneously with the development graphical analysis such as the pH–bicarbonate diagram
of the acidosis. The blood pH, Pco2, and bicarbonate concen- (the “Davenport plot,” after its popularizer), the pH– Pco2 dia-
tration point does not really move first from the normal gram (the “Siggaard-Andersen”), or the composite acid–base
(point A) to point G and then move a short distance along line diagram. Blood gas analyzers perform these calculations
GH; instead, the compensation begins to occur as the acidosis automatically.
develops, so the point takes an intermediate pathway between Table 37–6 summarizes the changes in pHa, Paco2, and
the two lines. plasma bicarbonate concentration that occur in simple, mixed,
The respiratory compensation for metabolic alkalosis is to and partially compensated acid–base disturbances. It contains
decrease alveolar ventilation, thus increasing Paco2. This the same information shown in Figure 37–2, depicted differ-
decreases pHa toward normal, as can be seen in Figure 37–2. ently. A thorough understanding of the patterns shown in
Point D represents uncompensated metabolic alkalosis; respi- Table 37–6 coupled with knowledge of a patient’s Pco2 and
ratory compensation would move the blood pHa, PaCo2, and other clinical findings can reveal a great deal about the under-
bicarbonate concentration point a short distance along the lying pathophysiologic processes in progress.
new higher-than-normal blood–buffer line toward point F. A simple approach to interpreting a blood gas set is to first
Again the compensation occurs as the alkalosis develops, with look at the pH to determine whether the predominant prob-
the point moving along an intermediate course. lem is acidosis or alkalosis. (Note that an acidemia could rep-
Under most circumstances, the cause of respiratory aci- resent more than one cause of acidosis, an acidosis with some
dosis or alkalosis is a dysfunction in the ventilatory control compensation, or even an acidosis and a separate underlying
CHAPTER 37 Acid–Base Regulation and Causes of Hypoxia 381

TABLE 37–6 Acid–base disturbances. istered to a patient. The base excess or base deficit is the num-
ber of milliequivalents of acid or base needed to titrate 1 L of
pH PCO2 HCO3− blood to pH 7.4 at 37°C if the Paco2 were held constant at
Uncompensated respiratory ↓↓ ↑↑ ↑ 40 mm Hg. It is not, therefore, just the difference between the
acidosis plasma bicarbonate concentration of the sample in question
and the normal plasma bicarbonate concentration because
Uncompensated respiratory ↑↑ ↓↓ ↓
alkalosis respiratory adjustments also cause a change in bicarbonate
concentration: the arterial Pco2 must be considered, although
Uncompensated metabolic ↓↓ ↔ ↓↓ in most cases the vertical deviation of the bicarbonate level
acidosis
above or below the blood–buffer line on the Davenport dia-
Uncompensated metabolic ↑↑ ↔ ↑↑ gram at the pH of the sample is a reasonable estimate. Base
alkalosis excess can be determined by actually titrating a sample or by
using a nomogram, diagram, or calculator program. Most
Partially compensated ↓ ↑↑ ↑↑
respiratory acidosis blood gas analyzers calculate the base excess automatically.
The base excess is expressed in milliequivalents per liter above
Partially compensated ↑ ↓↓ ↓↓ or below the normal buffer-base range—it therefore has
respiratory alkalosis
a normal value of 0 ± 2 mEq/L. A base deficit is also called a
Partially compensated ↓ ↓↓ ↓↓ negative base excess.
metabolic acidosis The base deficit can be used to estimate how much sodium
Partially compensated ↑ ↑↑ ↑↑ bicarbonate (in mEq) should be given to a patient by multiply-
metabolic alkalosis ing the base deficit (in mEq/L) times the patient’s estimated
extracellular fluid (ECF) space (in liters), which is the distri-
Respiratory and metabolic ↓↓ ↑↑ ↓
acidosis
bution space for the bicarbonate. The ECF is usually estimated
to be 0.3 times the lean body mass in kilograms.
Respiratory and metabolic ↑↑ ↓↓ ↑
alkalosis
Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed.
ANION GAP
New York: McGraw-Hill Medical, 2007.
Calculation of the anion gap can be helpful in determining the
cause of a patient’s metabolic acidosis. It is determined by sub-
alkalosis. Similarly, an alkalemia could represent more than tracting the sum of a patient’s plasma chloride and bicarbonate
one cause of alkalosis, an alkalosis with some compensation, or concentrations (in mEq/L) from his or her plasma sodium
even an alkalosis and a separate underlying acidosis.) After concentration:
evaluating the pH, look at the arterial Pco2 to see if it explains
Anion gap =[Na+]–([C1–]+[HCO3–]) (5)
the pH. For example, if the pH is low and the Pco2 is increased,
then the primary problem is respiratory acidosis. If the pH is The anion gap is normally 12 ± 4 mEq/L.
low and the Pco2 is near 40 mm Hg, then the primary problem The sum of all of the plasma cations must equal the sum of
is metabolic acidosis with little or no compensation. If both the all of the plasma anions, so the anion gap exists only because
pH and the Pco2 are low, there is metabolic acidosis with respi- all of the plasma cations and anions are not measured when
ratory compensation. Then look at the bicarbonate concentra- standard blood chemistry is done. Sodium, chloride, and
tion to confirm your diagnosis. It should be slightly increased bicarbonate concentrations are almost always reported. The
in uncompensated respiratory acidosis, high in partially com- normal anion gap is a result of the presence of more unmea-
pensated respiratory acidosis, and low in metabolic acidosis. sured anions than unmeasured cations in normal blood:
If the pH is high and the Pco2 is low, then the primary prob-
lem is respiratory alkalosis. If the pH is high and the Pco2 is [Na+]+[Unmeasured cations]= (6)
near 40 mm Hg, then the problem is uncompensated meta- –
[ C1 ]+[HCO3–]+[Unmeasured anions]
bolic alkalosis. If both the pH and the Pco2 are high, then there
is partially compensated metabolic alkalosis. The bicarbonate [Na+]–([ C1–]+[HCO3–])=
(7)
should be slightly decreased in respiratory alkalosis, decreased [Unmeasured anions]–[Unmeasured cations]
in partially compensated respiratory alkalosis, and increased
The anion gap is therefore the difference between the unmea-
in metabolic alkalosis.
sured anions and the unmeasured cations.
The negative charges on the plasma proteins probably make
BASE EXCESS up most of the normal anion gap, because the total charges of
the other plasma cations (K+, Ca2+, Mg2+) are approximately
Calculation of the base excess or base deficit may be very equal to the total charges of the other anions (PO43−, SO42−,
useful in determining the therapeutic measures to be admin- organic anions).
382 SECTION VI Pulmonary Physiology

An increased anion gap usually indicates an increased num- Low Alveolar PO2
ber of unmeasured anions (those other than C1– and HCO3−)
Conditions causing low alveolar Po2 inevitably lead to low arte-
or a decreased number of unmeasured cations (K+, Ca2+, or
rial Po2 and oxygen contents because the alveolar Po2 deter-
Mg2+), or both. This is most likely to happen when the mea-
mines the upper limit of arterial Po2. Hypoventilation leads to
sured anions, [HCO3−] or [Cl–], are lost and replaced by unmea-
both alveolar hypoxia and hypercapnia (high CO2), as dis-
sured anions. For example, the buffering by HCO3− of H+ from
cussed in Chapter 33. Hypoventilation can be caused by
ingested or metabolically produced acids produces an increased
depression or injury of the respiratory centers in the brain
anion gap.
(discussed in Chapter 38), interference with the nerves supply-
Thus, metabolic acidosis with an abnormally great anion
ing the respiratory muscles, as in spinal cord injury, neuro-
gap (i.e., greater than 16 mEq/L) would probably be caused by
muscular junction diseases such as myasthenia gravis, and
lactic acidosis or ketoacidosis; ingestion of organic anions
altered mechanics of the lung or chest wall, as in noncompliant
such as salicylate, methanol, and ethylene glycol; or renal
lungs due to sarcoidosis, reduced chest wall mobility because
retention of anions such as sulfate, phosphate, and urate.
of kyphoscoliosis or obesity, and airway obstruction. Ascent to
high altitude causes alveolar hypoxia because of the reduced
total barometric pressure encountered above sea level. Reduced
THE CAUSES OF HYPOXIA FIo2 (fractional concentration of inspired oxygen) has a similar
effect. Alveolar carbon dioxide is decreased because of the
Thus far, only two of the three variables referred to as the arte- reflex increase in ventilation caused by hypoxic stimulation, as
rial blood gases, the arterial Pco2, and pH have been discussed. will be discussed in Chapter 71. Hypoventilation and ascent to
Many abnormal conditions or diseases can cause a low arterial high altitude lead to decreased venous Po2 and oxygen content
Po2. They are discussed in the following section about the as oxygen is extracted from the already hypoxic arterial blood.
causes of tissue hypoxia in the discussion of hypoxic hypoxia. Administration of increased oxygen concentrations in the
The causes of tissue hypoxia can be classified (in some cases inspired gas can alleviate the alveolar and arterial hypoxia in
rather arbitrarily) into four or five major groups (Table 37–7). hypoventilation and in ascent to high altitude, but it cannot
The underlying physiology of most of these types of hypoxia reverse the hypercapnia of hypoventilation. In fact, adminis-
has already been discussed in this or previous chapters. tration of increased FIo2 to spontaneously breathing patients
hypoventilating because of a depressed central response to car-
bon dioxide (see Chapter 38) can further depress ventilation.
HYPOXIC HYPOXIA
Hypoxic hypoxia refers to conditions in which the arterial Po2 Diffusion Impairment
is abnormally low. Because the amount of oxygen that will Alveolar–capillary diffusion is discussed in greater detail in
combine with hemoglobin is mainly determined by the Po2, Chapter 35. Conditions such as interstitial fibrosis and inter-
such conditions may lead to decreased oxygen delivery to the stitial or alveolar edema can lead to low arterial Po2 and con-
tissues if reflexes or other responses cannot adequately increase tents with normal or elevated alveolar Po2. High FIo2 that
the cardiac output or hemoglobin concentration of the blood. increases the alveolar Po2 to very high levels may increase the

TABLE 37–7 A classification of the causes of hypoxia.


Increased FIO2
Classification PAO2 PaO2 CaO2 P –V O2 C–V O2 Helpful?

Hypoxic hypoxia

Low alveolar PO Low Low Low Low Low Yes


2

Diffusion impairment N Low Low Low Low Yes

Right-to-left shunts N Low Low Low Low No

V̇/ Q̇ mismatch N Low Low Low Low Yes

Anemic hypoxia N N Low Low Low No

CO poisoning N N Low Low Low Possibly

Hypoperfusion hypoxia N N N Low Low No

Histotoxic hypoxia N N N High High No

N, normal.
Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.
CHAPTER 37 Acid–Base Regulation and Causes of Hypoxia 383

arterial Po2 by increasing the partial pressure gradient for oxy- rocytes, or interference with the chemical combination of oxy-
gen diffusion. gen and hemoglobin. Carbon monoxide poisoning, for example,
results from the greater affinity of hemoglobin for carbon mon-
Shunts oxide than for oxygen. Methemoglobinemia is a condition in
which the iron in hemoglobin has been altered from the Fe2+ to
True right-to-left shunts, such as anatomic shunts and abso-
the Fe3+ form, which does not combine with oxygen.
lute intrapulmonary shunts, can cause decreased arterial Po2
Anemic hypoxia results in a decreased oxygen content when
with normal or even increased alveolar Po2. Patients with
both alveolar and arterial Po2 are normal. Standard analysis of
intrapulmonary shunts have low arterial Po2, but may not have
arterial blood gases could therefore give normal values unless
significantly increased Pco2 if they are able to increase their alve-
blood oxygen content is measured independently. Venous Po2
olar ventilation or if they are mechanically ventilated. This is a
and oxygen content are both decreased. Administration of
result of the different shapes of the oxyhemoglobin dissociation
high FIo2 is not effective in greatly increasing the arterial oxy-
curve (see Figure 36–1) and the carbon dioxide dissociation
gen content (except possibly in carbon monoxide poisoning).
curve (see Figure 36–5). The carbon dioxide dissociation curve
is almost linear in the normal range of arterial Pco2, and arterial
Pco2 is very tightly regulated by the respiratory control system HYPOPERFUSION HYPOXIA
(see Chapter 38). Carbon dioxide retained in the shunted blood
Hypoperfusion hypoxia (sometimes called stagnant hypoxia)
stimulates increased alveolar ventilation, and because the car-
results from low blood flow. This can occur either locally, in a
bon dioxide dissociation curve is nearly linear, increased venti-
particular vascular bed, or systemically, in the case of a low car-
lation will allow more carbon dioxide to diffuse from the
diac output. The alveolar Po2 and the arterial Po2 and oxygen
nonshunted blood into well-ventilated alveoli and be exhaled.
content may be normal, but the reduced oxygen delivery to the
On the other hand, increasing alveolar ventilation will not get
tissues may result in tissue hypoxia. Venous Po2 and oxygen con-
any more oxygen into the shunted blood and, because of the
tent are low. Increasing the FIo2 is of little value in hypoperfusion
shape of the oxyhemoglobin dissociation curve, very little more
hypoxia (unless it directly increases the perfusion) because the
into the unshunted blood. This is because the hemoglobin of
blood flowing to the tissues is already oxygenated normally.
well-ventilated and perfused alveoli is nearly saturated with
oxygen, and little more will dissolve in the plasma. Similarly,
arterial hypoxemia caused by true shunts is not relieved by high HISTOTOXIC HYPOXIA
FIo2 because the shunted blood does not come into contact with
the high levels of oxygen. The hemoglobin of the unshunted Histotoxic hypoxia refers to a poisoning of the cellular machin-
blood is nearly completely saturated with oxygen at a normal ery that uses oxygen to produce energy. Cyanide, for example,
FIo2 of 0.21, and the small additional volume of oxygen dis- binds to cytochrome oxidase in the respiratory chain and effec-
solved in the blood at high FIo2 cannot make up for the low tively blocks oxidative phosphorylation. Alveolar Po2 and arte-
hemoglobin saturation of the shunted blood. rial Po2 and oxygen content may be normal (or even increased,
because low doses of cyanide increase ventilation by stimulat-
VENTILATION–PERFUSION MISMATCH ing the arterial chemoreceptors). Venous Po2 and oxygen con-
tent are increased because oxygen is not utilized in the tissues.
Alveolar–capillary units with low ventilation–perfusion (V̇/Q̇)
ratios contribute to arterial hypoxia, as already discussed. Units
with high V̇/Q̇ do not by themselves lead to arterial hypoxia, of THE EFFECTS OF HYPOXIA
course, but large lung areas that are underperfused are usually
associated either with overperfusion of other units or with low Hypoxia can result in reversible tissue injury or even tissue
cardiac outputs (see the section “Hypoperfusion Hypoxia”). death. The outcome of an hypoxic episode depends on whether
Hypoxic pulmonary vasoconstriction (discussed in Chapter the tissue hypoxia is generalized or localized, how severe
34) and local airway responses (discussed in Chapter 32) nor- the hypoxia is, the rate of development of the hypoxia (see
mally help minimize V̇ / Q̇ mismatch. Chapter 71), and the duration of the hypoxia. Different cell
Note that diffusion impairment, shunts, and V̇ / Q̇ mismatch types have different susceptibilities to hypoxia; unfortunately,
increase the alveolar–arterial Po2 difference (see Table 35–1 brain cells and heart cells are the most susceptible.
and the first two columns in Table 37–7).

ANEMIC HYPOXIA CLINICAL CORRELATION


A 15-year-old adolescent entered the emergency depart-
Anemic hypoxia is caused by a decrease in the amount of func- ment with dyspnea (shortness of breath), a feeling of chest
tioning hemoglobin, which can be a result of decreased hemo- tightness, coughing, wheezing, and anxiety. His nail beds
globin or erythrocyte production, the production of abnormal and lips were blue (cyanosis).
hemoglobin or red blood cells, pathologic destruction of eryth-
384 SECTION VI Pulmonary Physiology

He has had frequent episodes of dyspnea and wheezing CHAPTER SUMMARY


for several years, especially in the spring, and has been ■ Hypoventilation causes respiratory acidosis; the compensation
diagnosed with asthma. Pulmonary function tests done at for respiratory acidosis is renal retention of base and excretion
the time of his diagnosis showed lower-than-predicted of hydrogen ions.
forced expiratory volume in the first second (FEV1), forced ■ Hyperventilation causes respiratory alkalosis; the compensa-
vital capacity (FVC), FEV1/FVC, and peak expiratory flow tion for respiratory alkalosis is renal excretion of base and
(PEF). Inhaling a bronchodilator improved all of these. retention of hydrogen ions.
An arterial blood gas was obtained to help determine the ■ Ingestion, infusion, overproduction, or decreased renal
severity of the episode. The arterial Po2 was 55 mm Hg, the excretion of hydrogen ions, or loss of bicarbonate ions, can
arterial Pco2 was 32 mm Hg, the arterial pH was 7.52, and the cause metabolic acidosis; the compensation for metabolic
acidosis is increased alveolar ventilation.
bicarbonate was 25 mEq/L, indicating hypoxemia and uncom-
■ Ingestion, infusion, or excessive renal reabsorption of bases, or
pensated respiratory alkalosis.
loss of hydrogen ions, can cause metabolic alkalosis; the
Asthma is an episodic obstructive disease and it is reason-
compensation for metabolic alkalosis is decreased alveolar
able to assume that it would cause CO2 retention and there- ventilation.
fore respiratory acidosis during attacks. This is true in very
■ Metabolic acidosis with an abnormally elevated anion gap
severe asthma attacks, but most asthma attacks result in indicates an increased plasma concentration of anions other
hypocapnia and respiratory alkalosis. As the asthma attack than chloride and bicarbonate or a decreased plasma concen-
occurs, bronchial smooth muscle spasm and mucus secretion tration of potassium, calcium, or magnesium ions.
obstruct the ventilation to some alveoli. Although some ■ Tissue hypoxia can be a result of low alveolar Po2, diffusion
hypoxic pulmonary vasoconstriction may occur, it is not suf- impairment, right-to-left shunts, or ventilation–perfusion
ficient to divert all of the mixed venous blood flow away from mismatch (hypoxic hypoxia), decreased functional hemoglo-
these poorly ventilated alveoli. This results in a right-to-left bin (anemic hypoxia), low blood flow (hypoperfusion hypoxia),
shunt or shuntlike state (Chapter 35), which would therefore or an inability of the mitochondria to use oxygen (histotoxic
be expected to cause the arterial Po2 to decrease and the arte- hypoxia).
rial Pco2 to increase. However, the Pco2 decreases because the
patient increases alveolar ventilation if he or she is able to.
Irritant receptors in the airways are stimulated by the STUDY QUESTIONS
mucus and by chemical mediators released during the 1–4. Match each of the following sets of blood gas data to one
attack. Hypoxia caused by the shunt stimulates the arterial of the underlying problems listed below. Assume the body
chemoreceptors; the patient also has the feeling of dyspnea temperature to be 37°C, and the hemoglobin concentration
(many asthma attacks have an emotional component). All to be 15 g Hb/100 mL blood. FIo2 is 0.21 (room air).
of these factors cause increased breathing and therefore A) acute methanol ingestion
increased alveolar ventilation. B) diarrhea
Increasing ventilation will get more CO2 out of the blood C) accidental hypoventilation of a patient on a mechanical
perfusing ventilated alveoli (and therefore out of the body) ventilator for 10 minutes
but it will not get much oxygen into alveoli supplied by D) chronic obstructive pulmonary disease
obstructed airways, nor will it get much more oxygen into 1. pHa = 7.25, Paco2 = 50 mm Hg , [HCO3−] = 26 mEq/L, Pao2
the blood of the unobstructed alveoli because of the shape = 70 mm Hg, anion gap = 11 mEq/L.
of the oxyhemoglobin dissociation curve. Remember that 2. pHa = 7.34, Paco2 = 65 mm Hg, [HCO3−] = 40 mEq/L, Pao2
the hemoglobin is already 97.4% saturated with oxygen and = 65 mm Hg, anion gap = 11 mEq/L.
not much more will dissolve in the plasma. Therefore, dur-
3. pHa = 7.25, Paco2 = 30 mm Hg, [HCO3−] = 15 mEq/L, Pao2
ing the attack the patient has hypoxemia, hypocapnia, and
= 95 mm Hg, anion gap = 10 mEq/L.
respiratory alkalosis. It is only when the attack is so severe
that the patient cannot do the additional work of breathing 4. pHa = 7.25, Paco2 = 30 mm Hg, [HCO3−] = 15 mEq/L, Pao2
that hypercapnia and respiratory acidosis occur. = 95 mm Hg, anion gap = 25mEq/L.
Acute treatment of asthma is aimed at dilating the airways
with a bronchodilator, such as a β2-adrenergic agonist, and
relieving the hypoxemia with oxygen. Mechanical ventila-
tion may be used in more severe cases. Chronic treatment
includes bronchodilators such as β2-adrenergic agonists;
anticholinergics, to block parasympathetically mediated
constriction and mucus production; antileukotriene drugs
and inhaled corticosteroids, to prevent inflammation; and
inhibition of mast cells to prevent them from releasing
cytokines.
38
C H A P T E R

Control of Breathing
Michael Levitzky

O B J E C T I V E S

■ Describe the general organization of the respiratory control system.


■ Localize the centers that generate the spontaneous rhythm of breathing.
■ Describe the groups of neurons that effect inspiration and expiration.
■ Describe the other centers in the brainstem that may influence the
spontaneous rhythm of breathing.
■ List the cardiopulmonary and other reflexes that influence the breathing
pattern.
■ State the ability of the brain cortex to override the normal pattern of
inspiration and expiration temporarily.
■ Describe the effects of alterations in body oxygen, carbon dioxide, and
hydrogen ion levels on the control of breathing.
■ Describe the sensors of the respiratory system for oxygen, carbon dioxide,
and hydrogen ion concentration.

ORGANIZATION OF THE ratory system during exercise or may even be temporarily


superseded or suppressed during speech or breath holding.
RESPIRATORY CONTROL SYSTEM The respiratory control centers in the brainstem affect the
automatic rhythmic control of breathing via a final common
Breathing is spontaneously initiated in the central nervous pathway consisting of the spinal cord, the innervation of the
system. A cycle of inspiration and expiration is automatically muscles of respiration such as the phrenic nerves, and the
generated by neurons located in the brainstem. Usually, breath- muscles of respiration themselves. Alveolar ventilation is
ing occurs without a conscious initiation of inspiration and therefore determined by the interval between successive
expiration. groups of discharges of the respiratory neurons and the inner-
This spontaneously generated cycle of inspiration and expi- vation of the muscles of respiration, which determines the
ration can be modified, altered, or even temporarily sup- respiratory rate or breathing frequency; and by the frequency
pressed by a number of mechanisms. As shown in Figure 38–1, of neural discharges transmitted by individual nerve fibers to
these include reflexes arising in the lungs, the airways, and the their motor units, the duration of these discharges, and the
cardiovascular system; information from receptors in contact number of motor units activated during each inspiration or
with the cerebrospinal fluid; and commands from higher cen- expiration, which determine the depth of respiration or the
ters of the brain such as the hypothalamus, the centers of tidal volume. Note that some pathways from the cerebral cor-
speech, or other areas in the cerebral cortex. The centers that tex to the muscles of respiration, such as those involved in vol-
are responsible for the generation of the spontaneous rhythm untary breathing, bypass the medullary respiratory center
of inspiration and expiration are, therefore, able to alter their described below and travel directly to the spinal α motor neu-
activity to meet the increased metabolic demand on the respi- rons. These are represented by the dashed line in Figure 38–1.

385

Ch38_385-396.indd 385 11/26/10 10:11:26 AM


386 SECTION VI Pulmonary Physiology

Influences from
higher centers

Cycle of
inspiration
and
Reflexes from: expiration Reflexes from:
Lungs Arterial
Airways chemoreceptors
Cardiovascular Central
system chemoreceptors
Muscles and
joints
Skin
Muscles of
breathing

FIGURE 38–1 Schematic representation of the organization of the respiratory control system. A cycle of inspiration and expiration
is automatically established in the medullary respiratory center. Its output represents a final common pathway to the respiratory muscles,
except for some voluntary pathways that may go directly from higher centers to the respiratory muscles (dashed line). Reflex responses from
chemoreceptors and other sensors may modify the cycle of inspiration and expiration established by the medullary respiratory center.
(Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

THE GENERATION OF ryngeal) and the 10th cranial nerve (the vagus). These nerves
carry information about the arterial Po2, Pco2, and pH from the
SPONTANEOUS RHYTHMICITY carotid and aortic arterial chemoreceptors (Figure 38–3)
and information concerning the systemic arterial blood pres-
The centers that initiate breathing are located in the reticular sure from the carotid and aortic baroreceptors (see Chapter 29).
formation of the medulla, beneath the floor of the fourth ven- In addition, the vagus carries information from stretch recep-
tricle. This area, known as the medullary respiratory center, tors and other sensors in the lungs that may also exert pro-
consists of inspiratory neurons, that fire during inspiration to found influences on the control of breathing. The effects of
stimulate inspiratory muscles to contract, and expiratory neu- information from these sensors on the control of breathing
rons, that fire during expiration to stimulate expiratory mus- will be discussed later in this chapter. The location of the DRG
cles to contract. Because expiration is passive in normal quiet within the NTS suggests that it may be the site of integration of
breathing, the expiratory neurons may not discharge unless various inputs that can reflexly alter the spontaneous pattern
expiration is active. of inspiration and expiration.
There are two dense bilateral aggregations of respiratory The ventral respiratory groups are located bilaterally in the
neurons in the medullary respiratory center known as the dor- retrofacial nucleus, the nucleus ambiguus, the nucleus
sal respiratory groups (DRG) and the ventral respiratory para-ambigualis, and the nucleus retroambigualis. They
groups (VRG) (Figure 38–2). Inspiratory and expiratory neu- consist of both inspiratory and expiratory neurons. The neu-
rons are anatomically intermingled to a greater or lesser extent rons in the nucleus ambiguus are primarily vagal motor neu-
within these areas. The dorsal respiratory groups are located rons that innervate the ipsilateral laryngeal, pharyngeal, and
bilaterally in the nucleus of the tractus solitarius (NTS). They tongue muscles involved in breathing and in maintaining
consist mainly of inspiratory neurons that project primarily to the patency of the upper airway. They are both inspiratory
the contralateral spinal cord. They serve as the principal initia- and expiratory neurons. Other neurons from the ventral
tors of the activity of the phrenic nerves and maintain the respiratory groups mainly project contralaterally to innervate
activity of the diaphragm. Dorsal respiratory group neurons inspiratory muscles and the expiratory muscles. The retrofa-
send many collateral fibers to those in the ventral respiratory cial nucleus, located most rostrally in the ventral respiratory
group, but the ventral respiratory group sends only a few col- groups, mainly contains expiratory neurons in a group of
lateral fibers to the dorsal respiratory group. The NTS receives cells called the Bötzinger complex. Neurons in the area
visceral afferent fibers of the 9th cranial nerve (the glossopha- called the pre-Bötzinger complex have been identified as the
CHAPTER 38 Control of Breathing 387

Pons

Pneumotaxic center

Apneustic center

Medulla

Ventral respiratory group


(VRG)
Dorsal respiratory group
(DRG) Pre-Bötzinger complex
(rhythm-generating neurons)
Inspiratory neurons
Inspiratory Expiratory
neurons neurons

Inspiratory Expiratory
Spinal motor neurons
FIGURE 38–2 Brainstem respiratory
control centers responsible for respiratory
rhythm generation, activation of inspiratory Inspiratory Expiratory
and expiratory neuron and muscle Muscles
activation, and monitoring lung inflation via
pulmonary stretch receptors and alveolar
ventilation via changes in arterial blood gas
Lung stretch receptors Ventilation
partial pressures. Input from the central Lung
chemoreceptors was omitted for clarity.
(Reproduced with permission from Widmaier EP, Raff H,
Strang KT: Vander’s Human Physiology, 11th ed.
Arterial chemoreceptors Blood gas partial pressures
McGraw-Hill, 2008.)

pacemakers of the respiratory rhythm—the respiratory neurons. Descending axons with inspiratory activity excite
rhythm generator. phrenic and external intercostal motor neurons and also
An area in the in the pons (the part of the brainstem just inhibit internal intercostal motor neurons by exciting spinal
rostral to the medulla) called the apneustic center appears to inhibitory interneurons. They are actively inhibited during
be an integration site for afferent information that can termi- expiratory phases of the respiratory cycle.
nate inspiration. The specific group of neurons that function Ascending pathways in the spinal cord, carrying informa-
as the apneustic center has not been identified. tion from pain, touch, and temperature receptors, as well as
A group of respiratory neurons rostral to the apneustic cen- from proprioceptors, can also influence breathing, as will be
ter known as the pontine respiratory groups (also called the discussed in the next section. Inspiratory and expiratory fibers
pneumotaxic center, as in Figure 38–2) functions to modu- appear to be separated in the spinal cord.
late the activity of the apneustic center. These cells, located in The spontaneous rhythmicity generated in the medullary
the upper pons in the nucleus parabrachialis medialis and respiratory center can be completely overridden (at least tem-
the Kölliker-Fuse nucleus, probably function to “fine-tune” porarily) by influences from higher brain centers. In fact, the
the breathing pattern and smooth the transitions between greatest minute ventilations obtainable from healthy conscious
inspiration and expiration. The pontine respiratory groups human subjects can be attained voluntarily, exceeding those
may also modulate the respiratory control system response to obtained with the stimuli of severe exercise, hypercapnia, or
stimuli such as lung inflation, hypercapnia, and hypoxia. hypoxia. This is the underlying concept of the maximum vol-
In the spinal cord axons projecting from the DRG, the VRG, untary ventilation (MVV) test often used to assess respiratory
the cortex, and other supraspinal sites descend in the spinal function. Conversely, the respiratory rhythm can be com-
white matter to influence the diaphragm and the intercostal pletely suppressed for several minutes by voluntary breath
and abdominal muscles of respiration, as already discussed. holding, until the chemical drive to breath (high Pco2 and low
There is integration of descending influences as well as the Po2 and pH) overrides the voluntary suppression of breathing
presence of local spinal reflexes that can affect these motor at the breakpoint. During speech, singing, or playing a wind
388 SECTION VI Pulmonary Physiology

Inflation of the lungs of anesthetized spontaneously breath-


Sensory nerves ing animals decreases the frequency of the inspiratory effort
Sensory nerves or causes a transient apnea (cessation of breathing). The
stimulus for this reflex is pulmonary inflation. The sensors
Carotid bodies are stretch receptors located within the smooth muscle of
large and small airways. They are sometimes referred to as
slowly adapting pulmonary stretch receptors because their
activity is maintained with sustained stretches. The afferent
pathway consists of large myelinated fibers in the vagus,
which enter the brainstem and project to the DRGs, the
apneustic center, and the pontine respiratory groups. The
Hering–Breuer inflation reflex was originally believed to be
Common carotid artery an important determinant of the rate and depth of ventila-
tion, but recent studies have cast doubt on this conclusion
because the threshold of the reflex is much higher than the
normal tidal volume during eupneic breathing. Tidal vol-
Aortic bodies umes of 800–1,500 mL are generally required to elicit this
reflex in conscious eupneic adults. The Hering–Breuer infla-
Aorta
tion reflex may help minimize the work of breathing by
inhibiting large tidal volumes as well as prevent overdisten-
tion of the alveoli. It may also be important in the control of
breathing in neonates. Neonates have Hering–Breuer infla-
Heart tion reflex thresholds within their normal tidal volume
ranges, and the reflex may be an important influence on their
tidal volumes and respiratory rates.
Deflation of the lungs increases the ventilatory rate. This
could be a result of decreased stretch receptor activity or of
stimulation of other pulmonary receptors, or rapidly adapting
receptors such as the irritant receptors and J receptors, which
FIGURE 38–3 Location of the carotid and aortic bodies.
Note that the carotid bodies are close to the carotid sinuses, the
will be discussed later in this chapter. The afferent pathway is
location of the major arterial baroreceptors. (Reproduced with the vagus, and the effect is increased minute ventilation
permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, (hyperpnea). This reflex may be responsible for the increased
11th ed. McGraw-Hill, 2008.) ventilation elicited when the lungs are deflated abnormally, as
in pneumothorax, or it may play a role in the periodic sponta-
neous deep breaths (sighs) that help prevent atelectasis. These
sighs occur occasionally and irregularly during the course of
instrument, the normal cycle of inspiration and expiration is
normal, quiet, spontaneous breathing. They consist of a slow
automatically modified by higher brain centers. In certain
deep inspiration (larger than a normal tidal volume) followed
emotional states, chronic hyperventilation severe enough to
by a slow deep expiration. This response appears to be very
cause respiratory alkalosis may occur.
important because patients maintained on mechanical ventila-
tors must be given large tidal volumes or periodic deep breaths
or they develop diffuse atelectasis, which may lead to arterial
RESPIRATORY REFLEXES hypoxemia.
The Hering–Breuer deflation reflex may be very important
A large number of sensors located in the lungs, the cardiovas-
in helping to actively maintain functional residual capacities
cular system, the muscles and tendons, and the skin and vis-
(FRCs) in infants. It is very unlikely that infants’ FRCs are
cera can elicit reflexes in the control of breathing. They are
determined passively like those of adults because the inward
summarized in Table 38–1, which lists the stimulus, receptor,
recoil of their lungs is considerably greater than the outward
afferent pathway, and effects for each reflex.
recoil of their very compliant chest walls.
After partly blocking the vagus nerves with cold tempera-
PULMONARY STRETCH RECEPTORS tures, lung inflation causes a further inspiration instead of the
apnea expected when the vagus nerves are completely func-
Three respiratory reflexes can be elicited by the activity of pul- tional. The receptors for this paradoxical reflex are located in
monary stretch receptors: the Hering–Breuer inflation reflex, the lungs, but their precise location is not known. Afferent
the Hering–Breuer deflation reflex, and the “paradoxical” information travels in the vagus; the effect is very deep inspi-
reflex. rations. This reflex may also be involved in the sigh response,
CHAPTER 38 Control of Breathing 389

TABLE 38–1 Respiratory reflexes.


Stimulus Reflex Name Receptor Afferent Pathway Effects

Lung inflation Hering–Breuer Stretch receptors within Vagus Respiratory


inflation reflex smooth muscle of large Cessation of inspiratory effort, apnea,
and small airways or decreased breathing frequency;
bronchodilation
Cardiovascular
Increased heart rate, slight vasoconstriction

Lung deflation Hering–Breuer Possibly J receptors, Vagus Respiratory


deflation reflex irritant receptors in Hyperpnea
lungs, or stretch
receptors in airways
Lung inflation Paradoxical reflex Stretch receptors in Vagus Respiratory
lungs Inspiration

Negative pressure in Pharyngeal dilator Receptors in nose, Trigeminal, laryngeal, Respiratory


the upper airway reflex mouth, upper airways glossopharyngeal Contraction of pharyngeal dilator muscles

Mechanical or Cough Receptors in upper Vagus Respiratory


chemical irritation airways, Cough; bronchoconstriction
of airways tracheobronchial tree

Sneeze Receptors in nasal Trigeminal, olfactory Sneeze; bronchoconstriction


mucosa Cardiovascular
Increased blood pressure

Face immersiona Diving reflex Receptors in nasal Trigeminal Respiratory


mucosa and face Apnea
Cardiovascular
Decreased heart rate; vasoconstriction

Pulmonary J receptors in pulmonary Vagus Respiratory


embolism vessels Apnea or tachypnea

Pulmonary vascular J receptors in pulmonary Vagus Respiratory


congestion vessels Tachypnea, possibly sensation of dyspnea

Specific chemicals in Pulmonary J receptors in pulmonary Vagus Respiratory


the pulmonary chemoreflex vessels Apnea or tachypnea; bronchoconstriction
circulation

Low PaO , high Arterial Carotid bodies, aortic Glossopharyngeal, Respiratory


2
PaCO , low pHa chemoreceptor reflex bodies vagus Hyperpnea; bronchoconstriction, dilation
2
of upper airway
Cardiovascular
Decreased heart rate (direct effect),
vasoconstriction

Increased systemic Arterial baroreceptor Carotid sinus stretch Glossopharyngeal, Respiratory


arterial blood reflex receptors vagus Apnea, bronchodilation
pressure Cardiovascular
Decreased heart rate,
Aortic arch stretch vasodilation etc.
receptors
Stretch of muscles, Muscle spindles, tendon Various spinal Respiratory
tendons, movement organs, proprioreceptors pathways Provide respiratory controller with
of joints feedback about work of breathing,
stimulation of proprioreceptors in
joints causes hyperpnea

Somatic pain Pain receptors Various spinal Respiratory


pathways Hyperpnea
Cardiovascular
Increased heart rate, vasoconstriction, etc.
a
Discussed in Chapter 71.
Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.
390 SECTION VI Pulmonary Physiology

or it may be involved in generating the first breath of the respond to low arterial Po2, high arterial Pco2, and low arterial
newborn baby; very great inspiratory efforts must be gener- pH (as will be discussed later in this chapter), with the carotid
ated to inflate the fluid-filled lungs. bodies generally capable of a greater response than the aortic
bodies. The afferent pathway from the carotid body is Hering’s
nerve, a branch of the glossopharyngeal nerve; the afferent
RECEPTORS IN THE AIRWAYS pathway from the aortic body is the vagus. The reflex effects of
AND THE LUNGS stimulation of the arterial chemoreceptors are hyperpnea,
bronchoconstriction, dilation of the upper airway, and
Negative pressure in the upper airway causes reflex contrac- increased blood pressure. The direct effect of arterial chemore-
tion of the pharyngeal dilator muscles. The receptors for the ceptor stimulation is a decrease in heart rate; however, this is
pharyngeal dilator reflex appear to be located in the nose, usually masked by an increase in heart rate secondary to the
mouth, and upper airways; the afferent pathways appear to be increase in lung inflation. The arterial baroreceptors exert a
in the trigeminal, laryngeal, and glossopharyngeal nerves. very minor influence on the control of ventilation. Low blood
This reflex may be very important in maintaining the patency pressure may stimulate breathing.
of the upper airway during strong inspiratory efforts and dur-
ing sleep.
Mechanical or chemical irritation of the airways (and pos- OTHER RECEPTORS IN MUSCLE,
sibly the alveoli) can elicit a reflex cough or sneeze, or it can TENDONS, SKIN, AND VISCERA
cause hyperpnea, bronchoconstriction, and increased blood
pressure. The receptors are located in the nasal mucosa, upper Stimulation of receptors located in the muscles, the tendons,
airways, tracheobronchial tree, and possibly the alveoli them- and the joints can increase ventilation. Included are receptors
selves. Those in the larger airways of the tracheobronchial in the muscles of respiration (e.g., muscle spindles) and rib
tree, which also respond to stretch, are sometimes referred to cage as well as other skeletal muscles, joints, and tendons.
as rapidly adapting pulmonary stretch receptors because These receptors may play an important role in adjusting the
their activity decreases rapidly during a sustained stimulus. ventilatory effort to elevated workloads and may help mini-
The afferent pathways are the vagus nerves for all but the mize the work of breathing. They may also participate in initi-
receptors located in the nasal mucosa, which send information ating and maintaining the elevated ventilation that occurs
centrally via the trigeminal and olfactory tracts. The cough during exercise, as will be discussed in Chapter 72. Somatic
and the sneeze reflexes were discussed in Chapter 32. pain generally causes hyperpnea; visceral pain generally causes
apnea or decreased ventilation.

PULMONARY VASCULAR
RECEPTORS (J RECEPTORS) THE RESPONSE
Pulmonary embolism causes rapid shallow breathing (tachy- TO CARBON DIOXIDE
pnea) or apnea; pulmonary vascular congestion also causes
The respiratory control system normally reacts very effectively
tachypnea. The receptors responsible for initiating these
to alterations in the internal “chemical” environment of the
responses are located in the walls of the pulmonary capillaries
body. Changes in the Pco2, pH, and Po2 result in alterations in
or in the interstitium; therefore, they are called J (for juxtapul-
alveolar ventilation designed to return these variables to their
monary capillary) receptors. These receptors may also be
normal values. Chemoreceptors alter their activity when their
responsible for the dyspnea (a feeling of difficult or labored
own local chemical environment changes and can therefore
breathing) encountered during the pulmonary vascular con-
supply the central respiratory controller with the afferent
gestion and edema secondary to left ventricular failure or even
information necessary to make the appropriate adjustments in
the dyspnea that healthy people feel at the onset of exercise.
alveolar ventilation to change the whole-body Pco2, pH, and
The afferent pathway of these reflexes is slow-conducting non-
Po2. The respiratory control system therefore functions as a
myelinated vagal fibers. Other receptors that may contribute
negative-feedback system as discussed in Chapter 1.
to the sensation of dyspnea include the arterial chemorecep-
The arterial and cerebrospinal fluid partial pressures of car-
tors, stretch receptors in the heart and blood vessels, and
bon dioxide are probably the most important inputs to the
receptors in the respiratory muscles.
ventilatory control system in establishing the breath-to-breath
levels of tidal volume and ventilatory frequency. (Of course,
OTHER CARDIOVASCULAR RECEPTORS changes in carbon dioxide lead to changes in hydrogen ion
concentration, so the effects of these two stimuli are comple-
The arterial chemoreceptors are located bilaterally in the mentary.) An increase in carbon dioxide is a very powerful
carotid bodies, which are situated near the bifurcations of the stimulus to ventilation: only voluntary hyperventilation and
common carotid arteries, and in the aortic bodies, which are the hyperpnea of exercise can surpass the minute ventilations
located in the arch of the aorta as shown in Figure 38–3. They obtained with hypercapnia. However, the arterial Pco2 is so
CHAPTER 38 Control of Breathing 391

20 drive. Metabolically produced carbon dioxide will then build


PaO  35 mm Hg
2
PaO  50 mm Hg
up and restore breathing.)
2
The ventilatory response of a normal conscious person to
physiologic levels of carbon dioxide is shown in Figure 38–4.
15 Alveolar (and arterial) Pco2 in the range of 38–50 mm Hg increase
Alveolar ventilation (L/min)

alveolar ventilation linearly. The slope of the line is quite steep


and varies from person to person. It decreases with age.
10 The figure also shows that hypoxia potentiates the ventila-
tory response to carbon dioxide. At lower arterial Po2 (e.g., 35
and 50 mm Hg), the response curve is shifted to the left and
the slope is steeper; that is, for any particular arterial Pco2, the
5 ventilatory response is greater at a lower arterial Po2. This may
PaO  100 mm Hg be caused by the effects of hypoxia at the chemoreceptor itself
2

or at higher integrating sites; changes in the central acid–base


0
status secondary to hypoxia may also contribute to the
30 35 40 45 50 enhanced response.
PaCO2 (mm Hg) Other influences on the carbon dioxide response curve are
FIGURE 38–4 Ventilatory carbon dioxide response curves at illustrated in Figure 38–5. Sleep shifts the curve slightly to
three different levels of arterial PO . (Modified with permission from the right. The arterial Pco2 normally increases during slow-
2
Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.) wave sleep, increasing as much as 5–6 mm Hg during deep
sleep. Because of this rightward shift in the CO2 response
curve during non-REM sleep, it is possible that there is a
precisely controlled that it changes little (<1 mm Hg) during “wakefulness” component of respiratory drive. A depressed
exercise severe enough to increase metabolic carbon dioxide response to carbon dioxide during sleep may be involved in
production 10-fold. central sleep apnea, a condition characterized by abnormally
Acutely increasing the level of carbon dioxide in the inspired long periods (1–2 minutes) between breaths during sleep. This
air (the Fico2) increases minute ventilation. The effect is most lack of central respiratory drive is a potentially dangerous con-
pronounced in Fico2 in the range of 0.05–0.10 (5–10% CO2 in dition in both infants and adults. (In obstructive sleep apnea,
inspired gas), which produces alveolar Pco2 between about 40 the central respiratory controller does issue the command to
and 70 mm Hg. Above 10–15% CO2 in inspired air, there is breathe, but the upper airway is obstructed because the pha-
little further increase in alveolar ventilation: very high arterial ryngeal muscles do not contract properly, there is too much fat
Pco2 (>70–80 mm Hg) may directly produce respiratory depres- around the pharynx, or the tongue blocks the airway.) Narcot-
sion. (Very low arterial Pco2 caused by hyperventilation may ics and anesthetics may profoundly depress the ventilatory
temporarily cause apnea because of decreased ventilatory response to carbon dioxide. Indeed, respiratory depression is

15 Metabolic acidosis Awake normal

Sleep
Alveolar ventilation (L/min)

10 Narcotics,
chronic obstruction

5
Deep anesthesia

0
25 35 45 55 65
PaCO (mm Hg)
2

FIGURE 38–5 The effects of sleep, narcotics, chronic obstructive pulmonary disease, deep anesthesia, and metabolic acidosis on
the ventilatory response to carbon dioxide. (Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)
392 SECTION VI Pulmonary Physiology

the most common cause of death in cases of overdose of opiate influence the cerebrospinal fluid; in fact, the cerebrospinal
alkaloids and their derivatives, barbiturates, and most anes- fluid may have changes in hydrogen ion concentration oppo-
thetics. Chronic obstructive pulmonary diseases (COPD) site to those seen in the blood in certain circumstances, as will
depress the ventilatory response to hypercapnia, in part be discussed later in this chapter.
because of depressed ventilatory drive secondary to central The composition of the cerebrospinal fluid is considerably
acid–base changes, and because the work of breathing may be different from that of the blood. The pH of the cerebrospinal
so great that ventilation cannot be increased normally. Meta- fluid is normally about 7.32, compared with the pH of 7.40 of
bolic acidosis displaces the carbon dioxide response curve to arterial blood. The Pco2 of the cerebrospinal fluid is about
the left, indicating that for any particular Paco2, ventilation is 50 mm Hg—about 10 mm Hg higher than the normal arterial
increased during metabolic acidosis because of hydrogen ion Pco2 of 40 mm Hg. The concentration of proteins in the cere-
stimulation of the arterial chemoreceptors. brospinal fluid is only in the range of 0.02-0.05 g/100 mL,
As already discussed, the respiratory control system consti- whereas the concentration of proteins in the plasma normally
tutes a negative-feedback system. This is exemplified by the ranges from 6.6 to 8.6 g/100 mL. This does not even include
response to carbon dioxide. Increased metabolic production of the hemoglobin in the erythrocytes. As a result, bicarbonate is
carbon dioxide increases the carbon dioxide brought to the lung. the main buffer in the cerebrospinal fluid. Arterial hypercap-
If alveolar ventilation stayed constant, the alveolar Pco2 would nia will therefore lead to greater changes in cerebrospinal fluid
increase, as would arterial and cerebrospinal Pco2. This stimu- hydrogen ion concentration than it does in the arterial blood.
lates alveolar ventilation by stimulating the arterial and central The brain produces carbon dioxide as an end product of
chemoreceptors (described later in this chapter). Increased metabolism. Brain carbon dioxide levels are higher than those
alveolar ventilation decreases alveolar and arterial Pco2, as was of the arterial blood, which explains the high Pco2 of the cere-
discussed in Chapter 33, returning the Pco2 to the original value. brospinal fluid.
The Pco2, pH, and Po2 are the principal controlled variables in The central chemoreceptors respond to local increases
the respiratory control system. To act as a negative-feedback in hydrogen ion concentration or Pco2, or both. They do not
system, the respiratory controller must receive information respond to hypoxia.
concerning the levels of the controlled variables from sensors About 80–90% of the normal total steady-state response to
in the system. The sensors are the arterial chemoreceptors increased inspired carbon dioxide concentrations comes
(peripheral chemoreceptors) and the central chemorecep- from the central chemoreceptors; the arterial chemoreceptors
tors located bilaterally near the ventrolateral surface of the contribute only 10–20% of the steady-state response. However,
medulla in the brainstem and other sites. The arterial chemore- the response comes from the arterial chemoreceptors when
ceptors are exposed to arterial blood; the central chemorecep- rapid changes in arterial Pco2 occur, that is, the central chemore-
tors are exposed to cerebrospinal fluid. The central ceptors may be mainly responsible for establishing the resting
chemoreceptors are therefore on the brain side of the blood– ventilatory level but the arterial chemoreceptors are more
brain barrier. Both the peripheral and central chemoreceptors important in short-term transient responses to carbon dioxide.
respond to increases in the partial pressure of carbon dioxide, Both the arterial and central chemoreceptors likely respond to
although the response may be related to the local increase in hydrogen ion concentration, not Pco2. Of course, they are usu-
hydrogen ion concentration that occurs with increased Pco2; ally very closely related in the body, so it is difficult to distin-
that is, the sensors may be responding to the increased carbon guish their effects.
dioxide concentration, the subsequent increase in hydrogen There may be other sensors for carbon dioxide in the body
ion concentration, or both. that may influence the control of ventilation. Chemoreceptors
The arterial chemoreceptors increase their firing rate in within the pulmonary circulation or airways have been pro-
response to increased arterial Pco2, decreased arterial Po2, or posed but have not as yet been substantiated or localized.
decreased arterial pH. The response of the receptors is both
rapid enough and sensitive enough that they can relay infor-
mation concerning breath-to-breath alterations in the compo-
sition of the arterial blood to the medullary respiratory center. THE RESPONSE TO
The response of the arterial chemoreceptors changes nearly HYDROGEN IONS
linearly with the arterial Pco2 over the range of 20–60 mm Hg.
The central chemoreceptors are exposed to the cerebrospi- Ventilation increases nearly linearly with changes in hydrogen
nal fluid and are not in direct contact with the arterial blood. ion concentration over the range of 20–60 nEq/L (nmol/L), as
As shown in Figure 38–6, the cerebrospinal fluid is separated shown in Figure 38–7. A metabolic acidosis of nonbrain origin
from the arterial blood by the blood–brain barrier. Carbon results in hyperpnea coming entirely from the peripheral
dioxide can easily diffuse through the blood–brain barrier, but chemoreceptors. Hydrogen ions cross the blood–brain barrier
hydrogen ions and bicarbonate ions do not. Because of this, too slowly to affect the central chemoreceptors initially. Aci-
alterations in the arterial Pco2 are rapidly transmitted to the dotic stimulation of the peripheral chemoreceptors increases
cerebrospinal fluid in about 60 seconds. Changes in arterial alveolar ventilation, and the arterial Pco2 decreases. Because
pH that are not caused by changes in Pco2 take much longer to the cerebrospinal fluid Pco2 is in a sort of dynamic equilibrium
CHAPTER 38 Control of Breathing 393

BRAIN TISSUE BRAIN TISSUE CSF


Venous blood

CO2 CO2 CO2

barrier

barrier
Central
Blood-brain

Metabolic H Chemoreceptor
H

Blood-brain
CO2
production

CO2 CO2 CO2 CO2

Dilates
HCO3 HCO3
Slow (h)
Smooth muscle

Smooth muscle

Dilates
CO2

Arterial CSF:
blood: pH  7.32
pH  7.40 PCO  50 mm Hg
2
PCO  40 mm Hg Little protein
2
Protein Buffers

FIGURE 38–6 Representation of the central chemoreceptor showing its relationship to carbon dioxide (CO2), hydrogen (H+), and
bicarbonate (HCO3−) ions in the arterial blood and cerebrospinal fluid (CSF). CO2 crosses the blood–brain barrier easily; H+ and HCO3− do not.
(Modified with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

with the arterial Pco2, carbon dioxide diffuses out of the cere-
brospinal fluid and the pH of the cerebrospinal fluid increases,
thus decreasing stimulation of the central chemoreceptor. If
15 the situation lasts a long time (hours to days), the bicarbonate
concentration of the cerebrospinal fluid decreases slowly,
12
returning the pH of the cerebrospinal fluid toward the normal
7.32. The mechanism by which this occurs is not completely
VE (L/min)

agreed on. It may represent the slow diffusion of bicarbonate


9 ions across the blood–brain barrier, active transport of bicar-
bonate ions out of the cerebrospinal fluid, or decreased forma-
6 tion of bicarbonate ions by carbonic anhydrase as the
cerebrospinal fluid is formed.
Similar mechanisms must alter the bicarbonate concentra-
40 41 42 43 44 45 46 tion in the cerebrospinal fluid in the chronic respiratory aci-
 dosis of chronic obstructive lung disease because the pH of
Plasma [H ], nEq/L
the cerebrospinal fluid is nearly normal. In this case, the cere-
FIGURE 38–7 The ventilatory response to increased plasma brospinal fluid concentration of bicarbonate increases nearly
hydrogen ion concentration. (Modified with permission from Levitzky MG: proportionately to its increased concentration of carbon
Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.) dioxide.
394 SECTION VI Pulmonary Physiology

ented. She has vomited twice and says she is thirsty and her
stomach hurts. The symptoms developed gradually over-
night. Her heart rate is 110/min, her blood pressure is
70 95/75 mm Hg, and her respiratory rate is 22/min with
obvious large tidal volumes. Her blood glucose is very
high at 450 mg/dL, her arterial Po2 is slightly increased at
60
105 mm Hg, her arterial Pco2 is 20 mm Hg (normal range
35–45 mm Hg), and her arterial pH is 7.15 (normal range
Alveolar ventilation (L/min)

50 7.35–7.45). Her bicarbonate concentration is 15 mEq/L


(normal range 22–26 mEq/L) and her anion gap is 22
mEq/L (normal range 8–16 mEq/L).
40
The patient has type 1 diabetes mellitus; she is in dia-
PaCO2 50 mm Hg betic ketoacidosis (see Chapter 66). The drug she did not
30 bring with her is insulin; as a result, her blood glucose
PaCO2 45 mm Hg concentration is very high and she is producing ketone
20 bodies (see Chapter 66). Nausea, vomiting, abdominal
pain, and confusion are common symptoms and signs of
PaCO  38 mm Hg diabetes mellitus, as will be discussed in Sections 7 and 9.
2
10 Hydrogen ions from the ketone bodies, which are acids,
have been buffered by bicarbonate and have been exhaled
0 as carbon dioxide, explaining the low bicarbonate concen-
20 40 60 80 100 120 140
PaO (mm Hg) tration and the elevated anion gap (see Chapter 37). The
2
hydrogen ions are stimulating her arterial chemorecep-
FIGURE 38–8 The ventilatory responses to hypoxia at three tors causing her to hyperventilate, as shown by her low
different levels of arterial PCO . (Modified with permission from Levitzky MG:
2
arterial Pco2. Her central chemoreceptors are not contrib-
Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.) uting to the hyperventilation because the hydrogen ions
do not cross her blood–brain barrier and therefore cannot
stimulate them (Figure 38–6); it is likely that her central
THE RESPONSE TO HYPOXIA chemoreceptors have decreased activity because as she
hyperventilates her cerebrospinal fluid Pco2 decreases and
The ventilatory response to hypoxia arises solely from the periph- her cerebrospinal fluid pH increases. Her acid–base disor-
eral chemoreceptors. The carotid bodies are much more impor- der can be described as a primary metabolic acidosis,
tant in this response than are the aortic bodies. In the absence of with increased anion gap, with a secondary respiratory
the peripheral chemoreceptors, the effect of increasing degrees of alkalosis.
hypoxia is a progressive direct depression of the central respira-
tory controller. Therefore, when the peripheral chemoreceptors
are intact, their excitatory influence on the central respiratory
controller must offset the direct depressant effect of hypoxia.
The response of the respiratory system to hypoxia is shown CHAPTER SUMMARY
in Figure 38–8. The figure shows that at a normal arterial Pco2 ■ A cycle of inspiration and expiration is automatically generated
of about 38–40 mm Hg, there is very little increase in ventila- by neurons in the medulla; this cycle can be modified or
tion until the arterial Po2 decreases below about 50–60 mm Hg. temporarily suppressed by reflexes or influences from higher
The response to hypoxia is potentiated at higher arterial Pco2. brain centers.
Experiments have shown that the respiratory response to ■ The respiratory control system functions as a negative-feedback
hypoxia is related to the change in Po2 rather than the change system; arterial Po , Pco , and pH and cerebrospinal fluid Pco
2 2 2

in oxygen content. Hypoxia alone, by stimulating alveolar and pH are the regulated variables.
ventilation, causes a decrease in arterial Pco2, which may lead ■ The increases in alveolar ventilation in response to increases
to respiratory alkalosis. This will be discussed in Chapter 71. in arterial Pco and hydrogen ion concentrations are nearly
2

linear within their normal ranges; the increase in alveolar


ventilation in response to decreases in arterial Po is small
2

near the normal range and very large when the Po falls below
CLINICAL CORRELATION 50–60 mm Hg.
2

■ The arterial chemoreceptors rapidly respond to changes in


A 14-year-old girl forgot her prescription drug on a week-
arterial Po , Pco , and pH; the central chemoreceptors are on the
end sleepover party at her friend’s house. She arrives in the 2 2

brain side of the blood–brain barrier and respond to changes in


emergency department lethargic, confused, and disori-
cerebrospinal fluid Pco and pH.
2
CHAPTER 38 Control of Breathing 395

STUDY QUESTIONS 3. Stimulation of which of the following receptors should result in


decreased ventilation?
1. The ventral respiratory groups A) aortic chemoreceptors
A) are located in the nucleus of the tractus solitarius. B) carotid chemoreceptors
B) include the pacemaker for breathing. C) central chemoreceptors
C) consist solely of inspiratory neurons. D) Hering–Breuer inflation (stretch) receptors
D) consist solely of expiratory neurons. E) all of the above
E) all of the above. 4. Which of the following would be expected to increase the
2. Which of the following conditions would be expected to ventilatory response to carbon dioxide, shifting the CO2
stimulate the central chemoreceptors? response curve to the left?
A) mild anemia A) barbiturates
B) severe exercise B) hypoxia
C) hypoxia due to ascent to high altitude C) slow-wave sleep
D) acute airway obstruction D) deep anesthesia
E) all of the above E) all of the above
This page intentionally left blank
SECTION VII RENAL PHYSIOLOGY

39
C H A P T E R

Renal Functions, Basic


Processes, and Anatomy
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ State the seven major functions of the kidneys.


■ Define the balance concept.
■ Define the gross structures and their interrelationships: renal pelvis,
calyces, renal pyramids, renal medulla (inner and outer zones), renal
cortex, and papilla.
■ Define the components of the nephron–collecting duct system and
their interrelationships: renal corpuscle, glomerulus, tubule, and collecting
duct system.
■ Draw the relationship between glomerulus, Bowman’s capsule, and the
proximal tubule.
■ Define juxtaglomerular apparatus and describe its three cell types; state the
function of the granular cells.
■ List the individual tubular segments in order; state the segments that
comprise the proximal tubule, Henle’s loop, and the collecting duct system;
define principal cells and intercalated cells.
■ Define the basic renal processes: glomerular filtration, tubular reabsorption,
and tubular secretion.
■ Define renal metabolism of a substance and give examples.

FUNCTIONS FUNCTION 1: REGULATION OF WATER


The kidneys perform a number of essential functions that go
AND ELECTROLYTE BALANCE
far beyond their well-known role of eliminating waste. This The balance concept states that our bodies are in balance for
chapter describes these functions and presents an overview of any substance when the inputs and outputs of that substance are
how the kidneys perform them. Ensuing chapters develop matched (see Figure 1–4). The kidneys vary the output of water
more detail of the mechanisms involved. and an array of electrolytes and other substances in close pace
with their input, thereby keeping the body content of those sub-
stances nearly constant, that is, in balance. As an example, our
input of water is enormously variable and is only sometimes

397

Ch39_397-408.indd 397 11/26/10 10:08:52 AM


398 SECTION VII Renal Physiology

driven in response to body needs. We drink water when thirsty, realize the kidneys’ crucial role in controlling blood pressure
but we also drink water because it is a component of beverages (BP). BP ultimately depends on blood volume, and the kidneys’
that we consume for reasons other than hydration. In addition, maintenance of sodium and water balance achieves regulation of
solid food often contains large amounts of water. The kidneys blood volume. Thus, through volume control, the kidneys par-
respond by varying the output of water in the urine, thereby ticipate in BP control. They also participate in direct regulation
maintaining balance for water (i.e., constant total body water of BP via the generation of vasoactive substances that regulate
content). Similarly, electrolytes such as sodium, potassium, and smooth muscle in the peripheral vasculature.
magnesium are components of foods and generally present far
in excess of body needs. As with water, the kidneys excrete elec-
trolytes at a highly variable rate that, in the aggregate, matches FUNCTION 5: REGULATION OF RED
input. One of the amazing feats of the kidneys is their ability to BLOOD CELL PRODUCTION
regulate each of these minerals independently (i.e., we can be
on a high-sodium, low-potassium diet or low-sodium, high- Erythropoietin is a peptide hormone that is involved in the con-
potassium diet, and the kidneys adjust excretion of each of these trol of erythrocyte (red blood cell) production by the bone mar-
substances appropriately). The reader should be aware that row. Its major source is the kidneys, although the liver also secretes
being in balance does not by itself imply a normal state or good small amounts. The renal cells that secrete it are a particular group
health. A person may have an excess or deficit of a substance, yet of cells in the interstitium. The stimulus for its secretion is a
still be in balance so long as output matches input. This is often reduction in the partial pressure of oxygen in the kidneys, as
the case in chronic disorders of renal function or metabolism. occurs, for example, in anemia, arterial hypoxia (see Chapter 71),
and inadequate renal blood flow. Erythropoietin stimulates the
bone marrow to increase its production of erythrocytes. Renal
FUNCTION 2: REGULATION OF disease may result in diminished erythropoietin secretion, and
ACID–BASE BALANCE the ensuing decrease in bone marrow activity is one important
causal factor of the anemia of chronic renal disease.
Acids and bases enter the body fluids via ingestion and from
metabolic processes. The body has to excrete acids and bases
to maintain balance, and it also has to regulate the concentra- FUNCTION 6: REGULATION OF
tion of free hydrogen ions (pH) within a limited range. The VITAMIN D PRODUCTION
kidneys accomplish both tasks by a combination of elimina-
tion and synthesis. These interrelated tasks are among the When we think of vitamin D, we often think of sunlight or
most complicated aspects of renal function and will be explored additives to milk. In vivo vitamin D synthesis involves a
thoroughly in Chapter 47. series of biochemical transformations, the last of which
occurs in the kidneys. The active form of vitamin D
(1,25-dihydroxyvitamin D) is actually made in the kidneys,
FUNCTION 3: EXCRETION OF and its rate of synthesis is regulated by hormones that control
METABOLIC WASTE AND BIOACTIVE calcium and phosphate balance that will be discussed in
detail in Chapter 64.
SUBSTANCES
Our bodies continuously form the end products of metabolic FUNCTION 7: GLUCONEOGENESIS
processes that for the most part serve no function and are
harmful at high concentrations; thus, they must be excreted at Our central nervous system is an obligate user of blood glucose
the same rate they are produced. These include urea (from regardless of whether we have just eaten sugary doughnuts or
protein), uric acid (from nucleic acids), creatinine (from mus- gone without food for a week. Whenever the intake of carbo-
cle creatine), and the end products of hemoglobin breakdown hydrate is stopped for much more than half a day, our body
(which give urine much of its color). In addition, the kidneys begins to synthesize new glucose (the process of gluconeogen-
participate with the liver in removing drugs, hormones, and esis) from noncarbohydrate sources (amino acids from protein
foreign substances. Clinicians have to be mindful of how fast and glycerol from triglycerides). Most gluconeogenesis occurs
drugs are excreted in order to prescribe a dose that achieves in the liver (see Chapters 66 and 69), but a substantial fraction
the appropriate body levels. occurs in the kidneys, particularly during a prolonged fast.

FUNCTION 4: REGULATION OF OVERVIEW OF RENAL PROCESSES


ARTERIAL BLOOD PRESSURE
Most of what the kidneys do to perform these various functions
Although most people appreciate that the kidneys excrete waste is, at least conceptually, fairly straightforward. Of the consider-
substances such as urea (hence the name urine) and salts, few able volume of plasma entering the kidneys each minute, they
CHAPTER 39 Renal Functions, Basic Processes, and Anatomy 399

Cortex

Diaphragm Medulla

Papilla
Kidney
Renal vein
Renal artery
Calyx

Ureter
Pelvis
Ureter

Capsule
Bladder
FIGURE 39–2 Major structural components of the kidney.
(Reproduced with permission from Kibble J, Halsey CR: The Big Picture, Medical
Urethra
Physiology. New York: McGraw-Hill, 2009.)

FIGURE 39–1 Urinary system in a female, indicating the broad bases of the pyramids facing the outside, top, and bot-
location of the kidneys below the diaphragm and well above the tom of the kidney. The pyramids constitute the medulla of the
bladder, which is connected to the kidneys via the ureters. kidney. Overlying the medullary tissue is a cortex, and cover-
(Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human ing the cortical tissue on the very external surface of the kid-
Physiology, 11th ed. McGraw-Hill, 2008.)
ney is a thin connective tissue capsule.
The working tissue mass of both the cortex and medulla is
constructed almost entirely of tubules (nephrons and collect-
transfer (by filtration) about one fifth of it, minus the larger pro- ing tubules) and blood vessels (mostly capillaries and capil-
teins, into the renal tubules and then selectively reabsorb varying larylike vessels). In the cortex, tubules and blood vessels are
fractions of the filtered substances back into the blood, leaving intertwined randomly, something like a plateful of spaghetti.
the unreabsorbed portions to be excreted. In some cases addi- In the medulla, they are arranged in parallel arrays like bun-
tional amounts are added by secretion or synthesis. In essence, dles of sticks. In both cases, blood vessels and tubules are
the renal tubules operate like assembly lines; they accept the fluid always close to each other. Between the tubules and blood ves-
coming into them, perform some segment-specific modification sels lies the interstitium, which comprises less than 10% of the
of the fluid, and send it on to the next segment. renal volume. The interstitium contains a small amount of
fluid and scattered interstitial cells (fibroblasts and others)
that synthesize an extracellular matrix of collagen, proteogly-
ANATOMY OF THE KIDNEYS cans, and glycoproteins.
It is important to realize that the cortex and medulla have
AND URINARY SYSTEM very different properties both structurally and functionally. On
The kidneys lie just under the rib cage on each side of the ver- close examination, we see that (1) the cortex has a highly gran-
tebral column, behind the peritoneal cavity and in front of the ular appearance, absent in the medulla, and (2) each medullary
major back muscles (Figure 39–1). Each of the two kidneys is pyramid is divisible into an outer zone (adjacent to the cortex)
a bean-shaped structure about the size of a fist, with the and an inner zone, which includes the papilla. All these distinc-
rounded, outer convex surface of each kidney facing the side tions reflect the different arrangement of the various tubules
of the body, and the indented surface, called the hilum, facing and blood vessels in the different regions of the kidney.
the spine. Each hilum is penetrated by blood vessels, nerves,
and a ureter, which carries urine out of the kidney to the blad-
der. Each ureter is formed from funnel-like structures called THE NEPHRON
major calyces, which, in turn, are formed from minor calyces
(Figure 39–2). The minor calyces fit over underlying cone- Each kidney contains approximately 1 million nephrons, one
shaped renal tissue called pyramids. The tip of each pyramid of which is shown diagrammatically in Figure 39–3. Each
is called a papilla and projects into a minor calyx. The calyces nephron begins with a spherical filtering component, called
act as collecting cups for the urine formed by the renal tissue the renal corpuscle, followed by a long tubule leading out of it
in the pyramids. The pyramids are arranged radially around that continues until it merges with the tubules of other
the hilum, with the papillae pointing toward the hilum and the nephrons to form collecting ducts, which are themselves long
400 SECTION VII Renal Physiology

Afferent TABLE 39–1 Terminology for the tubular segments.


arteriole
Macula Distal tubule
Bowman’s densa Combination Terms
capsule Sequence of Segments Used in Text
Proximal Proximal convoluted tubule Proximal tubule
tubule Proximal straight tubule
Cortical
Descending thin limb of Henle’s loop Henle’s loop
collecting
duct Ascending thin limb of Henle’s loop
Thick Thick ascending limb of Henle’s loop
ascending (contains macula densa near end)
limb Distal convoluted tubule
Thin
descending Connecting tubule Collecting duct
limb Cortical collecting duct system
Medullary
Thin Outer medullary collecting duct
collecting
ascending Inner medullary collecting duct (last portion
duct
limb is papillary duct)

Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
FIGURE 39–3 Components of the nephron. (Reproduced with 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New York:
McGraw-Hill, 2009.)

THE TUBULE
tubes. Collecting ducts eventually merge with others in the
Throughout its course, the tubule, which begins at and leads out
renal papilla to form a ureter that conveys urine to the bladder
of Bowman’s capsule, is made up of a single layer of epithelial
(Figure 39–3).
cells resting on a basement membrane and connected by tight
junctions that physically link the cells together (like the plastic
THE RENAL CORPUSCLE form that holds a six pack of soft drinks together). Table 39–1
lists the names and sequence of the various tubular segments, as
The renal corpuscle is a hollow sphere (Bowman’s capsule) illustrated in Figure 39–5. Physiologists and anatomists have
filled with a compact tuft of interconnected capillary loops, traditionally grouped two or more contiguous tubular segments
the glomerulus (plural glomeruli) (Figure 39–4a). Blood for purposes of reference, but the terminologies have varied
enters the capillaries inside Bowman’s capsule through an considerably. Table 39–1 also gives the combination terms used
afferent arteriole that penetrates the surface of the capsule at in this text.
one side, called the vascular pole. Blood then leaves the capil- The proximal tubule, which drains Bowman’s capsule, con-
laries through a nearby efferent arteriole on the same side. sists of a coiled segment—the proximal convoluted tubule—
The space within Bowman’s capsule not occupied by the glom- followed by a straight segment—the proximal straight
erulus is called the urinary space or Bowman’s space, and it is tubule—which descends toward the medulla, perpendicular
into this space that fluid filters from the glomerular capillaries to the cortical surface of the kidney.
before flowing into the first portion of the tubule, located The next segment is the descending thin limb of the loop
opposite the vascular pole. of Henle (or simply the descending thin limb). The descend-
The structure and properties of the filtration barrier that ing thin limbs of different nephrons penetrate into the medulla
separates plasma in the glomerular capillaries from fluid in to varying depths, and then abruptly reverse at a hairpin turn
urinary space are crucial for renal function and will be and begin an ascending portion of the loop of Henle parallel to
described thoroughly in the next chapter. For now, we note the descending portion. In long loops (depicted on the left
simply that the functional significance of the filtration barrier side of Figure 39–5) the epithelium of the first portion of the
is that it permits the filtration of large volumes of fluid from ascending limb remains thin, although different functionally
the capillaries into Bowman’s space, but restricts filtration of from that of the descending limb. This segment is called the
large plasma proteins such as albumin. ascending thin limb of Henle’s loop, or simply the ascending
Another cell type—the mesangial cell—is found in close thin limb (see Figure 39–5). Further up the ascending portion
association with the capillary loops of the glomerulus. the epithelium thickens, and this next segment is called the
Glomerular mesangial cells act as phagocytes and remove thick ascending limb of Henle’s loop, or simply the thick
trapped material from the basement membrane. They also ascending limb. In short loops (depicted on the right side of
contain large numbers of myofilaments and can contract in Figure 39–5) there is no ascending thin portion, and the thick
response to a variety of stimuli in a manner similar to vascu- ascending portion begins right at the hairpin loop. The thick
lar smooth muscle cells. The role of such contraction in ascending limb rises back into the cortex to the very same
influencing filtration by the renal corpuscles is discussed in Bowman’s capsule from which the tubule originated. Here it
Chapters 40 and 45. passes directly between the afferent and efferent arterioles at
CHAPTER 39 Renal Functions, Basic Processes, and Anatomy 401

Parietal layer
Bowman's capsule Visceral layer
Renal corpuscle (podocyte)
Glomerular capillary Proximal
(covered by visceral layer) tubule

Afferent arteriole Capillary

Juxtaglomerular
Juxtaglomerular cells a. Blood flows into the glomerulus through
apparatus Macula densa the afferent arterioles and leaves the
glomerulus through the efferent
arterioles. The proximal tubule exits
Distal
Bowman’s capsule.
tubule

Efferent
arteriole
(a)

Cell processes
Podocyte (visceral layer
of Bowman's capsule) Cell body

b. Podocytes of Bowman’s capsule


Filtration surround the capillaries. Filtration slits
slits between the podocytes allow fluid to
pass into Bowman’s capsule. The
glomerulus is composed of capillary
endothelium that is fenestrated.
Glomerular Surrounding the endothelial cells is
capillary (cut) a basement membrane.

(b) Fenestrae

Capsular space

Foot
processes

Filtration
slits c. Substances in the blood are filtered
through capillary fenestrae between
endothelial cells (single layer). The
filtrate then passes across the
basement membrane and through
slit pores between the foot processes
(also called pedicels) and enters the
capsular space. From here, the
Basement membrane filtrate is transported to the lumen
of the proximal convoluted tubule.

Fenestra
Capillary lumen

(c) Endothelium Movement


of filtrate
FIGURE 39–4 The renal corpuscle. a) Cutaway view of the renal corpuscle. b) Close-up of podocytes surrounding capillaries.
c) Transmission EM of the filtration barrier. (Reproduced with permission from Daniel Friend from William Bloom and Don Fawcett, Textbook of Histology, 10th ed.
W.B. Saunders Co. 1975.)
402 SECTION VII Renal Physiology

Peritubular
capillaries

Proximal Distal
convoluted convoluted Efferent arteriole
tubule tubule Afferent arteriole

Artery
Vein

C
o
r
t
Renal corpuscle e
x
Bowman’s
capsule
Glomerulus Cortical
collecting
Macula densa duct

Vein
Loop of Henie Artery

Descending
limb Corticomedullary M
junction e
Thick d
segment of u
ascending l
l
limb a
Medullary
Thin collecting duct
segment of Urine
ascending
limb
Vasa recta
Juxtamedullary nephron Cortical nephron
FIGURE 39–5 Basic structure of nephrons and vascular elements as described in the text. Note the difference between a
juxtamedullary nephron with its renal corpuscle located just above the corticomedullary border (left side of figure) and a cortical nephron with
its renal corpuscle higher in the cortex (right side of figure). Cortical nephrons have efferent arterioles that give rise to peritubular capillaries, and
they have short loops of Henle. In contrast, juxtamedullary nephrons have efferent arterioles that descend into the medulla to form vasa recta,
and they have long loops of Henle. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

the point where they enter and exit the renal corpuscle at its necting tubule, which leads to the cortical collecting tubule,
vascular pole (see Figure 39–4a). The cells in the thick ascend- the first portion of which is called the initial collecting tubule.
ing limb closest to Bowman’s capsule (between the afferent From Bowman’s capsule to the proximal tubule, loop of
and efferent arterioles) are a group of specialized cells known Henle, distal tubule, and initial collecting tubules, each of the
as the macula densa (see Figure 39–6). The macula densa 1 million nephrons in each kidney is completely separate from
marks the end of the thick ascending limb and the beginning the others. However, connecting tubules from several nephrons
of the distal convoluted tubule. This is followed by the con- merge to form cortical collecting tubules, and a number of ini-
CHAPTER 39 Renal Functions, Basic Processes, and Anatomy 403

known more commonly as principal cells. Interspersed among


the segment-specific cells in each of these three segments are
individual cells of the second type, called intercalated cells.
The last portion of the medullary collecting duct contains nei-
ther principal cells nor intercalated cells but is composed
entirely of a distinct cell type called the inner medullary col-
lecting duct cells.

Podocytes
Sympathetic THE JUXTAGLOMERULAR APPARATUS
nerve fiber
Reference was made earlier to the macula densa, a portion of the
late thick ascending limb at the point where this segment comes
Mesangial cells
Juxtaglomerular between the afferent and efferent arterioles at the vascular pole of
Efferent arteriole cells
the renal corpuscle from which the tubule arose. This entire area is
known as the juxtaglomerular apparatus (JG) (see Figure 39–6),
which, as will be described later, plays a very important signaling
Afferent arteriole function. (Do not confuse the term JG with juxtamedullary
Smooth muscle nephron, meaning a nephron with a glomerulus located close to
cells
the cortical–medullary border.) Each JG apparatus is made up of
Macula densa
Distal tubule the following three cell types: (1) granular cells (called juxtaglom-
erular cells in Figure 39–6), which are differentiated smooth mus-
cle cells in the walls of the afferent arterioles; (2) extraglomerular
mesangial cells; and (3) macula densa cells, which are specialized
FIGURE 39–6 Components of the juxtaglomerular (JG)
apparatus. It is made up of (1) juxtaglomerular cells (granular cells), thick ascending limb epithelial cells.
which are specialized smooth muscle cells surrounding the afferent The granular cells are named because they contain secretory
arteriole, (2) extraglomerular mesangial cells, and (3) cells of the macula vesicles that appear granular in light micrographs. These granules
densa, which are part of the tubule. The close proximity of these contain the hormone renin (pronounced REE-nin). As we will
components to each other permits chemical mediators released from describe in Chapter 45, renin is a crucial substance for control of
one cell to easily diffuse to other components. Note that sympathetic renal function and systemic BP. The extraglomerular mesangial
nerve fibers innervate the granular cells. (Reproduced with permission from cells are morphologically similar to and continuous with the
Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.) glomerular mesangial cells, but lie outside Bowman’s capsule. The
macula densa cells are detectors of the composition of the fluid
within the nephron at the very end of the thick ascending limb
tial collecting tubules then join end to end or side to side to and contribute to the control of glomerular filtration rate
form larger cortical collecting ducts. All the cortical collecting (GFR—see below) and to the control of renin secretion.
ducts then run downward to enter the medulla and become
outer medullary collecting ducts, and then inner medullary
collecting ducts. The latter merge to form several hundred large BASIC RENAL PROCESSES
ducts, the last portions of which are called papillary collecting
ducts, each of which empties into a calyx of the renal pelvis. The working structures of the kidney are the nephrons and
Each renal calyx is continuous with the ureter, which empties collecting tubules into which the nephrons drain. Figure 39–7
into the urinary bladder, where urine is temporarily stored and illustrates the meaning of several keywords that we use to
from which it is intermittently eliminated. The urine is not describe how the kidneys function. It is essential that any stu-
altered after it enters a calyx. From this point on, the remainder dent of the kidney grasp their meaning.
of the urinary system serves only to maintain the composition Filtration is the process by which water and solutes in the
of the tubular fluid established by the kidney. blood leave the vascular system through the filtration barrier
Up to the distal convoluted tubule, the epithelial cells form- and enter Bowman’s space (a space that is topologically out-
ing the wall of a nephron in any given segment are homoge- side the body). Secretion is the process of moving substances
neous and distinct for that segment. For example, the thick into the tubular lumen from the cytosol of epithelial cells
ascending limb contains only thick ascending limb cells. How- that form the walls of the nephron. Secreted substances may
ever, beginning in the second half of the distal convoluted originate by synthesis within the epithelial cells or, more
tubule, two cell types are found intermingled in most of the often, by crossing the epithelial layer from the surrounding
remaining segments. One type constitutes the majority of cells renal interstitium. Reabsorption is the process of moving
in the particular segment, is considered specific for that seg- substances from the lumen across the epithelial layer into the
ment, and is named accordingly: distal convoluted tubule cells, surrounding interstitium. In most cases, reabsorbed sub-
connecting tubule cells, and collecting duct cells, the last stances then move into surrounding blood vessels, so that
404 SECTION VII Renal Physiology

Artery organics glucose and urea; amino acids; and peptides such as
Afferent Glomerular Efferent insulin and antidiuretic hormone (ADH).
arteriole capillary arteriole The volume of filtrate formed per unit time is known as the
glomerular filtration rate (GFR). In a healthy young adult
male, the GFR is an incredible 180 L per day (125 mL/min)!
1. Glomerular
filtration
Contrast this value with the net filtration of fluid across all the
2. Tubular other capillaries in the body: approximately 4 L per day. The
1 secretion implications of this huge GFR are extremely important. When
3. Tubular
Bowman’s reabsorption
we recall that the average total volume of plasma in humans is
2
space approximately 3 L, it follows that the entire plasma volume is
Peritubular
filtered by the kidneys some 60 times a day. The opportunity
Tubule capillary to filter such huge volumes of plasma enables the kidneys to
3 excrete large quantities of waste products and to regulate the
Vein constituents of the internal environment very precisely. One of
the general consequences of healthy aging as well as many kid-
Urinary ney diseases is a reduction in the GFR.
excretion
FIGURE 39–7 Fundamental elements of renal function—
glomerular filtration, tubular secretion, and tubular
reabsorption—and the association between the tubule and
TUBULAR REABSORPTION AND
vasculature in the cortex. (Reproduced with permission from Widmaier EP, TUBULAR SECRETION
Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
The volume and composition of the final urine are quite dif-
ferent from those of the glomerular filtrate. Clearly, almost
the term reabsorption implies a two-step process of removal all the filtered volume must be reabsorbed; otherwise, with a
from the lumen followed by movement into the blood. Excre- filtration rate of 180 L per day, we would urinate ourselves
tion means exit of the substance from the body (i.e., the sub- into dehydration very quickly. As the filtrate flows from Bow-
stance is present in the final urine produced by the kidneys). man’s capsule through the various portions of the tubule, its
Synthesis means that a substance is constructed from molec- composition is altered, mostly by removing material (tubular
ular precursors, and catabolism means the substance is bro- reabsorption) but also by adding material (tubular secretion).
ken down into smaller component molecules. The renal As described earlier, the tubule is, at all points, intimately
handling of any substance consists of some combination of associated with the vasculature, a relationship that permits
these processes. rapid transfer of materials between the capillary plasma and
the lumen of the tubule via the interstitial space.
Most of the tubular transport consists of reabsorption rather
GLOMERULAR FILTRATION than tubular secretion. An idea of the magnitude and impor-
tance of tubular reabsorption can be gained from Table 39–2,
Urine formation begins with glomerular filtration, the bulk which summarizes data for a few plasma components that
flow of fluid from the glomerular capillaries into Bowman’s undergo reabsorption. The values in Table 39–2 are typical for
capsule. The glomerular filtrate (i.e., the fluid within Bow- a healthy person on an average diet. There are at least three
man’s capsule) is very much like blood plasma, but contains important generalizations to be drawn from this table:
very little total protein because the large plasma proteins such
1. Because of the huge GFR, the quantities filtered per day
as albumin and the globulins are virtually excluded from mov-
are enormous, generally larger than the amounts of the
ing through the filtration barrier. Smaller proteins, such as
substances in the body. For example, the body contains
many of the peptide hormones, are present in the filtrate, but
about 40 L of water, but the volume of water filtered each
their mass in total is miniscule compared with the mass of
day may be as large as 180 L.
large plasma proteins in the blood. The filtrate contains most
2. Reabsorption of waste products, such as urea, is partial, so
inorganic ions and low-molecular-weight organic solutes in
that large fractions of their filtered amounts are excreted
virtually the same concentrations as in the plasma. Substances
in the urine.
that are present in the filtrate at the same concentration as
3. Reabsorption of most “useful” plasma components (e.g.,
found in the plasma are said to be freely filtered. (Note that
water, electrolytes, and glucose) is either complete
freely filtered does not mean all filtered. It just means that the
(e.g., glucose) or nearly so (e.g., water and most elec-
amount filtered is in exact proportion to the fraction of plasma
trolytes), so that little, if any, of the filtered amounts
volume that is filtered.) Many low-molecular-weight compo-
are excreted in the urine.
nents of blood are freely filtered. Among the most common
substances included in the freely filtered category are the ions For each plasma substance, a particular combination of fil-
sodium, potassium, chloride, and bicarbonate; the uncharged tration, reabsorption, and secretion applies. The relative pro-
CHAPTER 39 Renal Functions, Basic Processes, and Anatomy 405

TABLE 39–2 Average values for several substances handled by filtration and reabsorption.
Substance Amount Filtered Per Day Amount Excreted Reabsorbed (%)

Water (L) 180 1.8 99.0

Sodium (g) 630 3.2 99.5

Glucose (g) 180 0 100

Urea (g) 56 28 50

Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.

portions of these processes then determine the amount excreted. heart. The adrenal cortex secretes the steroid hormones aldos-
A critical point is that the rates of these processes are subject to terone and cortisol, and the adrenal medulla secretes the cat-
physiological control. By triggering changes in the rates of echolamines epinephrine and norepinephrine. All of these
filtration, reabsorption, or secretion when the body content of hormones, but mainly aldosterone, are regulators of sodium
a substance goes above or below normal, these mechanisms and potassium excretion by the kidneys. The posterior pitu-
regulate excretion to keep the body in balance. For example, itary gland secretes the hormone arginine vasopressin (AVP,
consider what happens when a person drinks a large quantity of also called antidiuretic hormone [ADH]). ADH is a major
water: Within 1–2 hours, all the excess water has been excreted regulator of water excretion, and, via its influence on the renal
in the urine, partly as the result of an increase in GFR but vasculature and possibly collecting duct principal cells, prob-
mainly as the result of decreased tubular reabsorption of water. ably sodium excretion as well. The heart secretes hormones—
The body is kept in balance for water by increasing excretion. natriuretic peptides—that increase sodium excretion by the
By keeping the body in balance, the kidneys serve to maintain kidneys. The least understood aspect of regulation lies in the
body water concentration within very narrow limits. realm of intrarenal chemical messengers (i.e., messengers that
originate in one part of the kidney and act in another part). It
is clear that an array of substances (e.g., nitric oxide, puriner-
METABOLISM BY THE TUBULES gic agonists, superoxide, eicosanoids) influence basic renal
processes, but, for the most part, the specific roles of these
Although renal physiologists traditionally list glomerular substances are not well understood.
filtration, tubular reabsorption, and tubular secretion as the
three basic renal processes, we cannot overlook metabolism by
the tubular cells. The tubular cells extract organic nutrients OVERVIEW OF REGIONAL FUNCTION
from the glomerular filtrate or peritubular capillaries and
metabolize them as dictated by the cells’ own nutrient require- We conclude this chapter with a broad overview of the tasks
ments. In so doing, the renal cells are behaving no differently performed by the individual nephron segments. Later, we
from any other cells in the body. In addition, there are other examine renal function substance by substance and see how
metabolic transformations performed by the kidney that are tasks performed in the various regions combine to produce an
directed toward altering the composition of the urine and overall result that is useful for the body.
plasma. The most important of these are gluconeogenesis, The glomerulus is the site of filtration—about 180 L per day
and the synthesis of ammonium from glutamine and the pro- of volume and proportional amounts of solutes that are freely
duction of bicarbonate, both described in Chapter 47. filtered, which is the case for most solutes (large plasma pro-
teins are an exception). The glomerulus is where the greatest
mass of excreted substances enters the nephron. The proximal
REGULATION OF RENAL FUNCTION tubule (convoluted and straight portions) reabsorbs about
two thirds of the filtered water, sodium, and chloride. The
By far the most complex feature of renal physiology is regula- proximal convoluted tubule reabsorbs all of the useful organic
tion of renal processes, details of which will be presented in molecules that the body conserves (e.g., glucose, amino acids).
later chapters. Neural signals, hormonal signals, and intrare- It reabsorbs significant fractions, but by no means all, of many
nal chemical messengers combine to regulate the processes important ions, such as potassium, phosphate, calcium, and
described above in a manner to help the kidneys meet the bicarbonate. It is the site of secretion of a number of organic
needs of the body. Neural signals originate in the sympathetic substances that are either metabolic waste products (e.g., uric
celiac plexus (see Chapter 19). These sympathetic neural sig- acid, creatinine) or drugs (e.g., penicillin) that clinicians must
nals exert major control over renal blood flow, glomerular fil- administer appropriately to make up for renal excretion.
tration, and the release of vasoactive substances that affect The loop of Henle contains different segments that perform
both the kidneys and the peripheral vasculature. Hormonal different functions, but the key functions occur in the thick
signals originate in the adrenal gland, pituitary gland, and ascending limb. As a whole, the loop of Henle reabsorbs about
406 SECTION VII Renal Physiology

20% of the filtered sodium and chloride and 10% of the fil-
tered water. A crucial consequence of these different propor- (osteoporosis). New blood test results reveal elevated levels
tions is that, by reabsorbing relatively more salt than water, the of several waste substances, indicative of a decreased GFR.
luminal fluid becomes diluted relative to normal plasma and The evidence strongly points to chronic renal failure, and
the surrounding interstitium. During periods when the kid- she is referred to a nephrologist for evaluation and treatment.
neys excrete dilute final urine, the role of the loop of Henle in Chronic renal failure that has reached the point of significant
diluting the luminal fluid is crucial. renal dysfunction is called end-stage renal disease (ESRD). It
The end of the loop of Henle contains cells of the macula is the consequence of major loss of functional tissue mass
densa, which sense the sodium and chloride content of the (nephrons and interstitial tissue). One of the common causes
lumen and generate signals that influence other aspects of of ESRD is diabetes mellitus. Chronic hyperglycemia causes
renal function, specifically the renin–angiotensin system the formation of glycosylated proteins that deposit in the
(discussed in Chapter 45). The distal tubule and connecting glomerular filtration apparatus. This interferes with filtration
tubule together reabsorb some additional salt and water, per- function and leads to pathology of glomerular cells. Hyper-
haps 5% of each. The cortical collecting tubule is where sev- tension can be both a cause and an effect of ESRD. The two
eral (6–10) connecting tubules join to form a single tubule. normal kidneys have a considerable reserve capacity. Patients
Cells of the cortical collecting tubule are strongly responsive to can do perfectly well with just one kidney, and ESRD may
and are regulated by the hormones aldosterone and ADH. progress to a considerable degree before symptoms appear.
Aldosterone enhances sodium reabsorption and potassium When enough nephrons are lost, function declines, although
secretion by this segment, and ADH enhances water reabsorp- some functions are preserved better than others; thus, symp-
tion. The degree to which these processes are stimulated or toms do not develop uniformly. Another problem in ESRD is
not stimulated plays a major role in regulating the amount of decreased production of erythropoietin, resulting in decreased
solutes and water present in the final urine. With large amounts red blood cell production and a low hematocrit. The anemia
of ADH present, most of the water remaining in the lumen is and possible accumulations of toxic substances due to the low
reabsorbed, leading to concentrated, low-volume urine. With GFR may account for the fatigue. A more complex problem
little ADH present, most of the water passes on to the final in ESRD involves phosphorus, calcium, and bone. As renal
urine, producing dilute, high-volume urine. function is lost, the ability to excrete phosphate declines and
The medullary collecting tubule continues the functions plasma phosphate rises, in turn leading to excessive loss of
of the cortical collecting tubule in salt and water reabsorp- calcium. The body does not replace the loss, in part because
tion. In addition, it plays a major role in regulating urea reab- of decreased renal production of 1,25-dihydroxyvitamin D.
sorption and in acid–base balance (secretion of protons or Treatment for ESRD includes the possibility of dialysis to
bicarbonate). make up for lost excretory function, renal transplant, and var-
ious dietary controls, including adding phosphate binders to
the diet to prevent accumulation of phosphate in the blood.

CLINICAL CORRELATION
CHAPTER SUMMARY
A 57-year-old woman with type 2 diabetes mellitus has
■ The role of the kidneys in the body includes many functions
managed her condition quite well via dietary control and that go well beyond simple excretion of waste.
has been in good health otherwise. Lately, however, she
■ A major function of the kidneys is to regulate the excretion of
has been feeling increasingly fatigued and so schedules a substances at a rate that, on average, exactly balances their
checkup with her primary care physician (PCP). No input into the body, and thereby maintains appropriate body
remarkable physical signs are noted, except that her BP is content of many substances.
increased at 137/92 mm Hg. Analysis of her blood shows ■ Another major function of the kidneys is to regulate blood
a slightly increased fasting blood glucose of 117 mg/dL volume and vascular resistance, thereby assisting with the
and a low normal hematocrit of 36%. Her PCP reminds maintenance of BP.
her to be extra careful about diet in terms of salt and sugar, ■ The structure of the kidneys reflects the arrangement of tubules
and suggests supplemental iron to maintain her hemoglo- and closely associated blood vessels.
bin. She schedules another checkup in 6 months. ■ Each functional renal unit is composed of a filtering compo-
The fatigue worsens over the next 6 months, and she suf- nent (glomerulus) and a transporting tubular component (the
fers a bone fracture after a seemingly minor fall. At the nephron and collecting duct).
6-month checkup her fasting blood glucose is 121 mg/dL, ■ The tubules are made up of multiple segments with distinct
and hematocrit is decreased to 29%. BP is 135/95 mm Hg. functions.
Her PCP orders additional blood tests and a bone density ■ Basic renal mechanisms consist of filtering a large volume,
scan out of concern for possible loss of bone mineral reabsorbing most of it, and adding substances by secretion,
and, in some cases, synthesis.
CHAPTER 39 Renal Functions, Basic Processes, and Anatomy 407

STUDY QUESTIONS 5. The volume of the ultrafiltrate of plasma entering the tubules by
glomerular filtration in 1 day is typically
1. Renal corpuscles are located A) about three times the renal volume.
A) along the corticomedullary border. B) about the same as the volume filtered by all the capillaries in
B) throughout the cortex. the rest of the body.
C) throughout the cortex and outer medulla. C) about equal to the circulating plasma volume.
D) throughout the whole kidney. D) greater than the total body fluid volume.
2. Relative to the number of glomeruli, how many loops of Henle 6. A substance known to be freely filtered has a certain
and collecting ducts are present? concentration in the afferent arteriole. What can we predict
A) same number of loops of Henle; same number of about its concentration in the efferent arteriole?
collecting ducts A) almost zero
B) fewer loops of Henle; fewer collecting ducts B) close to the value in the afferent arteriole
C) same number of loops of Henle; fewer collecting ducts C) about 20% lower than the value in the afferent arteriole
D) same number of loops of Henle; more collecting ducts D) we cannot predict without knowing what happens in
3. In which of the following lists are all the named substances the tubules
synthesized in the kidneys and released into the blood?
A) insulin, renin, and glucose
B) red blood cells, active vitamin D, and albumin
C) renin, 1,25-dihydroxyvitamin D, and erythropoietin
D) glucose, urea, and erythropoietin
4. The macula densa is a group of cells located in the wall of
A) Bowman’s capsule.
B) the afferent arteriole.
C) the end of the thick ascending limb.
D) the glomerular capillaries.
This page intentionally left blank
40
C H A P T E R

Renal Blood Flow and


Glomerular Filtration
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ Define renal blood flow, renal plasma flow, glomerular filtration rate, and
filtration fraction, and give normal values.
■ State the formula relating flow, pressure, and resistance in an organ.
■ Identify the successive vessels through which blood flows after leaving
the renal artery.
■ Describe the relative resistances of the afferent arterioles and efferent
arterioles.
■ Describe the effects of changes in afferent and efferent arteriolar resistances
on renal blood flow.
■ Describe the three layers of the glomerular filtration barrier, and define
podocyte, foot process, and slit diaphragm.
■ Describe how molecular size and electrical charge determine filterability
of plasma solutes; state how protein binding of a low-molecular-weight
substance influences its filterability.
■ State the formula for the determinants of glomerular filtration rate, and state,
in qualitative terms, why the net filtration pressure is positive.
■ State the reason glomerular filtration rate is so large relative to filtration
across other capillaries in the body.
■ Describe how arterial pressure, afferent arteriolar resistance, and efferent
arteriolar resistance influence glomerular capillary pressure.
■ Describe how changes in renal plasma flow influence average glomerular
capillary oncotic pressure.
■ Define autoregulation of renal blood flow and glomerular filtration rate.

RENAL BLOOD FLOW these, cortical radial arteries project upward toward the kid-
ney surface and give off a series of afferent arterioles, each of
Renal blood flow (RBF) is huge relative to the mass of the which leads to a glomerulus within Bowman’s capsule (see
kidneys—about 1 L/min, or 20% of the resting cardiac output. Figure 39–5). These arteries and glomeruli are found only in
Considering that the volume of each kidney is less than 150 the cortex, never in the medulla. In most organs, capillaries
cm3, this means that each kidney is perfused with over three recombine to form the beginnings of the venous system, but
times its total volume every minute. All of this blood is deliv- the glomerular capillaries instead recombine to form another
ered to the cortex. A small fraction of the cortical blood flow is set of arterioles, the efferent arterioles. The efferent arterioles
then directed to the medulla. Blood enters each kidney at the soon subdivide into a second set of capillaries. These are the
hilum via a renal artery. After several divisions into smaller peritubular capillaries, which are profusely distributed
arteries, blood reaches arcuate arteries that course across the throughout the cortex. The peritubular capillaries then rejoin
tops of the pyramids between the medulla and cortex. From to form the veins by which blood ultimately leaves the kidney.

409

Ch40_409-416.indd 409 11/26/10 10:10:19 AM


410 SECTION VII Renal Physiology

Blood flow to the medulla is far less than cortical blood flow,
perhaps 0.1 L/min, and derives from the efferent arterioles of
glomeruli situated just above the corticomedullary border (jux-
tamedullary glomeruli). These efferent arterioles do not branch Interlobular

CORTEX
into peritubular capillaries, but rather descend downward into artery

the outer medulla, where they divide many times to form bun-
Arcuate
dles of parallel vessels called vasa recta (Latin recta for “straight” Efferent artery
and vasa for “vessels”). These bundles of vasa recta penetrate arteriole

deep into the medulla (see Figure 40–1). Vasa recta on the out-
side of the vascular bundles “peel off ” and give rise to inter-
bundle plexi of capillaries that surround Henle’s loops and the

Outer
stripe
collecting ducts in the outer medulla. Only the center-most vasa Interbundle
plexus

OUTER MEDULLA
Vascular
recta supply capillaries in the inner medulla; thus, little blood bundle
flows into the papilla. The capillaries from the inner medulla
re-form into ascending vasa recta that run in close association
with the descending vasa recta within the vascular bundles. The

stripe
Inner
structural and functional properties of the vasa recta are rather
complex, and will be elucidated further in Chapter 44.
The significance of the quantitative differences between cor-
tical and medullary blood flow is the following: the high blood
flow in the cortical peritubular capillaries maintains the inter-
stitial environment of the cortical renal tubules very close in
composition to that of blood plasma throughout the body. In
contrast, the low blood flow in the medulla permits an intersti-

DVR

AVR
tial environment that is quite different from blood plasma. As
INNER MEDULLA

described in Chapter 44, the interstitial environment in the


medulla plays a crucial role in regulating water excretion.
H2O

NaCl
Urea
FLOW, RESISTANCE, AND BLOOD
PRESSURE IN THE KIDNEYS
Blood flow in the kidneys obeys the basic hemodynamic prin-
ciples described in Chapter 22. The basic equation for blood
flow through any organ is as follows: FIGURE 40–1 The renal microcirculation. Arcuate arteries run
ΔP just above the corticomedullary border, parallel to the surface, and
Q = ___
R
(1) give rise to cortical radial (interlobular) arteries radiating toward the
where Q is organ blood flow, ΔP the mean pressure in the artery surface. Afferent arterioles originate from the cortical radial arteries at
supplying the organ minus mean pressure in the vein draining an angle that varies with cortical location. Blood is supplied to the
peritubular capillaries of the cortex from the efferent flow out of
that organ, and R the total vascular resistance in that organ. The
superficial glomeruli. It is supplied to the medulla from the efferent
high RBF is accounted for by low total renal vascular resistance. flow out of juxtamedullary glomeruli. Efferent arterioles of
The resistance is low because there are so many pathways in juxtamedullary glomeruli give rise to bundles of descending vasa recta
parallel, that is, so many glomeruli and their associated vessels. in the outer stripe of the outer medulla. In the inner stripe of the outer
The resistances of the afferent and efferent arterioles are medulla, descending vasa recta and ascending vasa recta returning
about equal in most circumstances and account for most of the from the inner medulla run side by side in the vascular bundles,
total renal vascular resistance. Resistances in arteries preced- allowing exchange of solutes and water as described in Chapter 44.
ing afferent arterioles (i.e., cortical radial arteries) and in the Descending vasa recta from the bundle periphery supply the
capillaries play some role also, but we concentrate on the arte- interbundle capillary plexus of the inner stripe, whereas those in the
rioles because arteriolar resistances are variable and are the center supply blood to the capillaries of the inner medulla. Contractile
sites of regulation. A change in the afferent arteriole or efferent pericytes in the walls of the descending vasa recta regulate flow. DVR,
descending vasa recta; AVR, ascending vasa recta. (Modified with
arteriole resistance produces the same effect on RBF because
permission from Pallone TL, Zhang Z, Rhinehart K: Physiology of the renal medullary
these vessels are in series. When the two resistances both microcirculation. Am J Physiol Renal Physiol 2003;284(2):F253–F266.)
change in the same direction (the most common state of
affairs), their effects on RBF are additive. When they change in
different directions—one resistance increasing and the other Vascular pressures (i.e., hydrostatic or hydraulic pressures)
decreasing—the changes offset each other. are much higher in the glomerular capillaries than in the
CHAPTER 40 Renal Blood Flow and Glomerular Filtration 411

Sites of largest
vascular resistance
100
Pressure (mm Hg)

75

50

25

0
FIGURE 40–2 Blood pressure decreases as blood flows
le

lary
le

l vein
ry

l vein
capil lar
lary

r terio
r terio
l ar te

eru

capil

through the renal vascular network. The largest drops occur in the
rena

Rena
Glom

ent a
ent a
Rena

sites of largest resistance—the afferent and efferent arterioles. The


bular

Intra

location of the glomerular capillaries, between the sites of high


Effer
Affer

resistance, results in their having a much higher pressure than the


u
Perit

peritubular capillaries. (Reproduced with permission from Kibble J, Halsey CR:


The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)

peritubular capillaries. As blood flows through any vascular capillaries and rest on the basement membrane. The podocytes
resistance, the pressure progressively decreases. Pressure at the have an unusual octopuslike structure. Small “fingers,” called
beginning of a given afferent arteriole is close to mean systemic pedicels (or foot processes), extend from each arm of the podo-
arterial pressure (about 100 mm Hg) and decreases to about 60 cyte and are embedded in the basement membrane (see Fig-
mm Hg at the point where it feeds a glomerulus. Because there ure 39–4c). Pedicels from a given podocyte interdigitate with the
are so many glomerular capillaries in parallel, pressure decreases pedicels from adjacent podocytes. Spaces between adjacent
very little during flow through those capillaries; thus, glomeru- pedicels constitute the path through which the filtrate, once it
lar capillary pressure remains close to 60 mm Hg. Then pres- has passed through the endothelial cells and basement mem-
sure decreases again during flow through an efferent arteriole, brane, travels to enter Bowman’s space. The foot processes are
to about 20 mm Hg at the point where it feeds a peritubular coated by a thick layer of extracellular material, which partially
capillary (see Figure 40–2). The high glomerular pressure of occludes the slits. Extremely thin processes called slit dia-
about 60 mm Hg is necessary to drive glomerular filtration, phragms bridge the slits between the pedicels. Slit diaphragms
whereas the low peritubular capillary pressure of 20 mm Hg is are widened versions of the tight junctions and adhering junc-
equally necessary to permit the reabsorption of fluid. tions that link all contiguous epithelial cells together and are like
miniature ladders. The pedicels form the sides of the ladder, and
the slit diaphragms are the rungs.
GLOMERULAR FILTRATION Both the slit diaphragms and basement membrane are com-
posed of an array of proteins, and while the basement mem-
FORMATION OF GLOMERULAR brane may contribute to selectivity of the filtration barrier,
FILTRATE integrity of the slit diaphragms is essential to prevent excessive
leak of plasma protein (mainly albumin). Some protein-wasting
The glomerular filtrate contains most inorganic ions and low- diseases are associated with abnormal slit diaphragm structure.
molecular-weight organic solutes in virtually the same concen- Selectivity of the barrier to filtered solute is based on both
trations as in the plasma. It also contains small plasma peptides molecular size and electrical charge. Let us look first at size.
and a very limited amount of albumin (see Chapter 43). Filtered The filtration barrier of the renal corpuscle provides no hin-
fluid must pass through a three-layered glomerular filtration drance to the movement of molecules with molecular weights
barrier. The first layer, the endothelial cells of the capillaries, is less than 7,000 d (i.e., solutes this small are all freely filtered).
perforated by many large fenestrae (“windows”), like a slice of This includes all small ions, glucose, urea, amino acids, and
Swiss cheese, which occupy about 10% of the endothelial surface many hormones. The filtration barrier almost totally excludes
area. They are freely permeable to everything in the blood except plasma albumin (molecular weight of approximately 66,000 d).
cells and platelets. The middle layer, the capillary basement (We are, for simplicity, using molecular weight as our reference
membrane, is a gel-like acellular meshwork of glycoproteins and for size; in reality, it is molecular radius and shape that is criti-
proteoglycans, with a structure like a kitchen sponge. The third cal.) The hindrance to plasma albumin is not 100%, however, so
layer consists of epithelial cells (podocytes) that surround the the glomerular filtrate does contain extremely small quantities
412 SECTION VII Renal Physiology

of albumin, on the order of 10 mg/L or less. This is only about released from damaged erythrocytes or myoglobin released
0.02% of the concentration of albumin in plasma and is the rea- from damaged muscles), considerable filtration of these may
son for the use of the phrase “nearly protein-free” earlier. Some occur as well.
small substances are partly or mostly bound to large plasma Electrical charge is the second variable determining filter-
proteins and are thus not free to be filtered, even though the ability of macromolecules. For any given size, negatively
unbound fractions can easily move through the filtration bar- charged macromolecules are filtered to a lesser extent, and
rier. This includes hydrophobic hormones of the steroid and positively charged macromolecules to a greater extent, than
thyroid classes and about 40% of the calcium in the blood. neutral molecules. This is because the surfaces of all the com-
For molecules with a molecular weight ranging from 7,000 ponents of the filtration barrier (the cell coats of the endothe-
to 70,000 d, the amount filtered becomes progressively smaller lium, the basement membrane, and the cell coats of the
as the molecule becomes larger (Figure 40–3). Thus, many podocytes) contain fixed polyanions, which repel negatively
normally occurring small- and medium-sized plasma pep- charged macromolecules during filtration. Because almost all
tides and proteins are actually filtered to a significant degree. plasma proteins bear net negative charges, this electrical
Moreover, when certain small proteins not normally present repulsion plays a very important restrictive role, enhancing
in the plasma appear because of disease (e.g., hemoglobin that of purely size hindrance. In other words, if either albumin
or the filtration barrier were not charged, even albumin would
be filtered to a considerable degree (see Figure 40–3). Certain
diseases that cause glomerular capillaries to become “leaky”
A.
to protein do so by eliminating negative charges in the
1.0 membranes.
Filtrate/plasma concentration ratio

It must be emphasized that the negative charges in the filtra-


tion membranes act as a hindrance only to macromolecules,
Inulin
(~ 5,000) not to mineral anions or low-molecular-weight organic anions.
Thus, chloride and bicarbonate ions, despite their negative
charge, are freely filtered.
0.5

DIRECT DETERMINANTS OF GFR


Variation in glomerular filtration rate (GFR) is a crucial
Hemoglobin (68,000)
Albumin (~ 69,000) determinant of renal function because, everything else being
0 equal, a higher GFR means greater excretion of salt and water.
0 35 70 Regulation of the GFR is straightforward in terms of physical
Molecular weight (thousands)
principles but very complex functionally because there are
many regulated variables. The rate of filtration in all capillaries,
B. including the glomeruli, is determined by the hydraulic perme-
Positively charged = more filtered ability of the capillaries, their surface area, and the net filtra-
tion pressure (NFP) acting across them, given as follows:
Neutral Rate of filtration = (2)
Hydraulic permeability × Surface area × NFP
Because it is difficult to estimate the area of a capillary bed,
Filtration

a parameter called the filtration coefficient (Kf) is used to


denote the product of the hydraulic permeability and the
area.
The NFP is the algebraic sum of the hydrostatic pressures
and the osmotic pressures resulting from protein—the oncotic,
Molecular size or colloid osmotic pressures—on the two sides of the capil-
Negatively charged = less filtered lary wall. There are four pressures to consider: two hydrostatic
pressures and two oncotic pressures. These are the Starling
FIGURE 40–3 A) As molecular weight (and therefore size) forces that were described earlier in Chapter 26. Applying this
increases, filterability declines, so that proteins with a molecular same principle to the glomerular capillaries, we have:
weight above 70,000 d are hardly filtered at all. B) For any given NPF = (PGC – PBC) – (πGC – πBC) (3)
molecular size, negatively charged molecules are restricted far more
than neutral molecules, while positively charged molecules are where PGC is glomerular capillary hydraulic pressure, πBC the
restricted less. (Reproduced with permission from Kibble J, Halsey CR: The Big oncotic pressure of fluid in Bowman’s capsule, PBC the
Picture, Medical Physiology. New York: McGraw-Hill, 2009.) hydraulic pressure in Bowman’s capsule, and πGC the oncotic
CHAPTER 40 Renal Blood Flow and Glomerular Filtration 413

PGC
60
Bowman’s
space NFP
50
PBS
PGC
Glomerular 40

Pressure (mm Hg)


capillary
πGC

30
πGC

20

Forces mmHg
10
Favoring filtration*: PBC

Glomerular capillary blood pressure (PGC) 60

Opposing filtration: 0 50% 100%


Capillary length
Fluid pressure in Bowman’s space (PBS) 15
FIGURE 40–5 Forces affecting glomerular filtration along
Osmotic force due to protein in plasma (π GC) 29 the length of the glomerular capillaries. Note that the oncotic
pressure within the capillaries (πGC) rises due to loss of water and that
Net glomerular filtration pressure = PGC – PBS – πGC 16 the net filtration pressure (shaded region) decreases as a result. (Modified
FIGURE 40–4 Forces involved in glomerular filtration as with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed.
described in the text. (Reproduced with permission from Widmaier EP, Raff H, New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

Table 40–1 presents a summary of these factors. It provides,


pressure in glomerular capillary plasma, shown schemati- in essence, a checklist to review when trying to understand
cally in Figure 40–4, along with typical average values. how diseases or vasoactive chemical messengers and drugs
Because there is normally little total protein in Bowman’s change GFR. It should be noted that the major cause of
capsule, πBC may be taken as zero and not considered in our decreased GFR in renal disease is not a change in these param-
analysis. Accordingly, the overall equation for GFR becomes: eters within individual nephrons, but rather simply a decrease
GFR = Kf (PGC – PBC – πGC) (4) in the number of functioning nephrons, which reduces Kf.
Figure 40–5 shows that the hydraulic pressure changes only
slightly along the glomeruli. This is because there are so many Kf
glomeruli in parallel, and collectively they provide only a small Changes in Kf are caused most often by glomerular disease,
resistance to flow, but the oncotic pressure in the glomerular but also by normal physiological control. The details are still
capillaries does change substantially along the length of the not completely clear, but chemical messengers released within
glomeruli. Water moves out of the vascular space and leaves the kidneys cause contraction of glomerular mesangial cells.
protein behind, thereby increasing protein concentration and, Such contraction may restrict flow through some of the capil-
hence, the oncotic pressure of the unfiltered plasma remaining lary loops, effectively reducing the area available for filtration,
in the glomerular capillaries. Mainly because of this large Kf, and, hence GFR.
increase in oncotic pressure, the NFP decreases from the
beginning of the glomerular capillaries to the end. The NFP
when averaged over the whole length of the glomerulus is PGC
about 16 mm Hg. This average NFP is higher than that found Hydrostatic pressure in the glomerular capillaries (PGC) is
in most nonrenal capillary beds. Taken together with a very influenced by many factors. We can help depict the situation by
high value for Kf, it accounts for the enormous filtration of 180 using the analogy of a leaking garden hose. If pressure in the
L of fluid per day (compared with 3 L per day or so in all other pipes feeding the hose changes, the pressure in the hose and,
capillary beds combined). hence, the rate of leak will be altered. Resistances in the hose
The GFR is not fixed but shows marked fluctuations in differ- also affect the leak. If we kink the hose upstream from the leak,
ing physiological states and in disease. To grasp this situation, it pressure at the region of leak decreases and less water leaks out.
is essential to see how a change in any one factor affects GFR However, if we kink the hose beyond the leak, this increases
under the assumption that all other factors are held constant. pressure at the region of leak and increases the leak rate.
414 SECTION VII Renal Physiology

TABLE 40–1 Summary of direct GFR determinants PBC


and factors that influence them. Changes in this variable are usually of very minor importance.
However, obstruction anywhere along the tubule or in the exter-
Direct Determinants Major Factors that Tend
of GFR: to Increase the Magnitude
nal portions of the urinary system (e.g., the ureter) increases the
GFR = Kf (PGC − PBC − πGC) of the Direct Determinant tubular pressure everywhere proximal to the occlusion, all the
way back to Bowman’s capsule. The result is to decrease GFR.
Kf 1. ↑ Glomerular surface area
(because of relaxation of
glomerular mesangial cells) πGC
Result: ↑ GFR
Oncotic pressure in the plasma at the very beginning of the glom-
PGC 1. ↑ Renal arterial pressure erular capillaries is, of course, simply the oncotic pressure of sys-
temic arterial plasma. Accordingly, a decrease in arterial plasma
2. ↓ Afferent arteriolar resistance
(afferent dilation)
protein concentration, as occurs, for example, in liver disease,
decreases arterial oncotic pressure and tends to increase GFR,
3. ↑ Efferent arteriolar resistance whereas increased arterial oncotic pressure tends to reduce GFR.
(efferent constriction) However, recall that πGC is the same as arterial oncotic pres-
Result: ↑ GFR sure only at the very beginning of the glomerular capillaries;
PBC 1. ↑ Intratubular pressure because πGC then progressively increases along the glomerular capillar-
of obstruction of tubule or ies as protein-free fluid filters out of the capillary, concentrat-
extrarenal urinary system ing the protein left behind. This means that NFP and, hence,
Result: ↓ GFR filtration progressively decrease along the capillary length.
πGC 1. ↑ Systemic plasma oncotic
Accordingly, anything that causes a steeper increase in πGC
pressure (sets πGC at beginning tends to lower average NFP and hence GFR.
of glomerular capillaries) Such a steep increase in oncotic pressure occurs when RBF is
very low. Since blood is composed of cells and plasma, low RBF
2. ↓ Renal plasma flow (causes
increased rise of πGC along means that renal plasma flow (RPF) is also low. When RPF is
glomerular capillaries) low, any given rate of filtration removes a larger fraction of the
Result: ↓ GFR plasma, leaving a smaller volume of plasma behind in the glom-
GFR, glomerular filtration rate; Kf, filtration coefficient; PGC, glomerular capillary
eruli to contain all the plasma protein. This causes the πGC to
hydraulic pressure; PBC, Bowman’s capsule hydraulic pressure; πGC, glomerular reach a final value at the end of the glomerular capillaries that is
capillary oncotic pressure. A reversal of all arrows in the table will cause higher than normal. This increases the average πGC along the
a decrease in the magnitudes of Kf, PGC, PBC, and πGC.
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
capillaries and lowers average NFP and, hence, GFR. Conversely,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.) a high RPF, all other factors remaining constant, causes πGC to
increase less steeply and reach a final value at the end of the cap-
illaries that is less than normal, which will increase the GFR.
These concepts can be expressed as a filtration fraction: the
These same principles apply to PGC and GFR. First, a change in
ratio GFR/RPF, which is normally about 20%. The increase in
renal arterial pressure causes a change in PGC in the same direc-
πGC along the glomerular capillaries is directly proportional to
tion. If resistances remain constant, PGC rises and falls as renal
the filtration fraction (i.e., the greater the percentage of vol-
artery pressure rises and falls. This is a crucial point because a
ume filtered from the plasma, the greater the increase in πGC).
major influence on renal function is arterial blood pressure.
Therefore, if you know that filtration fraction has changed,
Second, changes in the resistance of the afferent and efferent
you can be certain that there has also been a proportional
arterioles have opposite effects on PGC. An increase in afferent
change in πGC and that this has played a role in altering GFR.
arteriolar resistance, which is upstream from the glomerulus,
is like kinking the hose above the leak (it decreases PGC),
whereas an increase in efferent arteriolar resistance is down- FILTERED LOAD
stream from the glomerulus and is like kinking the hose
beyond the leak (it increases PGC). Of course, dilation of the A term we use in other chapters is filtered load. It is the amount
afferent arteriole raises PGC, and hence GFR, while dilation of of substance that is filtered per unit time. For freely filtered sub-
the efferent arteriole lower PGC and GFR. It should also be clear stances, the filtered load is just the product of GFR and plasma
that when the afferent and efferent arteriolar resistances both concentration. Consider sodium. Its normal plasma concentra-
change in the same direction (i.e., they both increase or tion is 140 mEq/L, or 0.14 mEq/mL. A normal GFR in healthy
decrease), they exert opposing effects on PGC. young adult males is 125 mL/min, so the filtered load of sodium
The real significance for this is that the kidney can regu- is 0.14 mEq/mL × 125 mL/min = 17.5 mEq/min. We can do the
late PGC and, hence, GFR independently of RBF. The effects of same calculation for any other substance, being careful in each
changes in arteriolar resistances are summarized in case to be aware of the unit of measure in which concentration
Figure 40–6. is expressed. The filtered load is what is presented to the rest of
CHAPTER 40 Renal Blood Flow and Glomerular Filtration 415

Decreased GFR Increased GFR

Constrict AA Constrict EA

Blood Blood
flow flow
PGC PGC

GFR GFR

(a) (b)

Dilate Dilate
Blood Blood
EA AA
flow flow
PGC PGC

GFR GFR

(c) (d)

FIGURE 40–6 Effect of changes in resistance on GFR. a–d) Constricting the afferent arteriole (AA) or dilating the efferent arteriole (EA)
leads to a decreased GFR, while dilating the AA or constricting the EA leads to increased GFR. (Reproduced with permission from Widmaier EP, Raff H, Strang
KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

the nephron to handle. The filtered load varies with plasma con- RBF varies only modestly when mean arterial pressure changes.
centration and GFR. An increase in GFR, at constant plasma This is partly a result of the myogenic response, which is the
concentration, increases the filtered load, as does an increase in contraction or relaxation of arteriolar smooth muscle in
plasma concentration at constant GFR. response to changes in vascular pressures. Autoregulation is

AUTOREGULATION 1.5 150

Glomerular filtration rate (mL/min)


“Autoregulatory range”
It is extremely important for the kidneys to keep the GFR at a
Renal blood flow (L/min)

level appropriate for the body because the excretion of salt and
water is strongly influenced by the GFR. GFR is strongly influ-
enced by renal arterial pressure. The effect is so strong that
urinary excretion would tend to vary widely with ordinary
daily excursions of arterial pressure. Also, vascular pressure in
the thin-walled glomerular capillaries is higher than in capil-
laries elsewhere in the body, and hypertensive damage ensues
if this pressure is too high.
0 0
To protect the glomerular capillaries from hypertensive 0 80 180
damage and to preserve a healthy GFR at different arterial Renal perfusion pressure (mm Hg)
pressure values, changes in GFR and RBF are minimized by
several mechanisms that we collectively call autoregulation. FIGURE 40–7 Autoregulation of renal blood flow (RBF) and
An increase in renal artery pressure is counteracted by an glomerular filtration rate (GFR). Over the span of renal perfusion
increase in vascular resistance that almost offsets the increase pressure (pressure in renal artery minus pressure in renal vein) from
in pressure. The word “almost” is crucial here. Higher driving 80 to about 170 mm Hg, RBF and GFR rise only modestly as renal
pressures do indeed lead to higher blood flow and GFR, but perfusion pressure increases. Outside of this range, however, the
not proportionally. Consider Figure 40–7. Within the usual changes are much greater. (Reproduced with permission from Kibble J, Halsey
range of mean arterial pressure (renal perfusion pressure), CR: The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)
416 SECTION VII Renal Physiology

also partly the result of rather complicated intrarenal signals CHAPTER SUMMARY
that affect vascular resistance and mesangial cell contraction
■ The kidneys have a very large blood flow relative to their mass
(see Chapter 45). The myogenic response is very fast-acting
because it is regulated for functional reasons rather than
and protects the glomeruli from short-term fluctuations in metabolic demand.
blood pressure. In addition to keeping changes in RBF fairly
■ The cortex has a much higher blood flow and a different
small, autoregulatory processes also keep changes in GFR arrangement of blood vessels than the medulla.
fairly small. Again, GFR does increase slightly with an increase
■ Glomerular filtration proceeds through a three-layered barrier
in arterial pressure. that restricts filtration of large macromolecules.
■ Both molecular charge and size affect the filterability of plasma
solutes.
■ The GFR is determined by the same Starling forces that govern
CLINICAL CORRELATION filtration in blood vessels elsewhere.
A 10-year-old boy developed a severe sore throat and a ■ Control of the resistances of the afferent and efferent arterioles
mild fever. The family pediatrician took a throat swab and permits independent control of glomerular filtration rate
concluded the boy had strep throat and so prescribed a and RBF.
10-day course of antibiotics. The boy recovered and ■ Autoregulation of RBF limits the variation in RBF in the face of
resumed his normal activities. One week later the boy told large changes in arterial pressure.
his mother that his urine was “brownish.” She checked the
bowl after his next urination, and it was indeed a rusty
brown color. As the toilet flushed, she noticed that the STUDY QUESTIONS
water frothed up. Very concerned now, she brought her 1. Blood enters the renal medulla immediately after passing
son back to the pediatrician, who performed a urine dip- through which vessels?
stick test. This revealed increased protein in the urine A) arcuate arteries
(accounting for the frothiness). In addition, his blood B) peritubular capillaries
pressure was somewhat elevated, and the pediatrician C) afferent arterioles
noted some puffiness in the face. Further tests confirmed D) efferent arterioles
that this was a case of poststreptococcal glomerulo- 2. Which cell type is the main determinant of the filterability of
nephritis (inflammation of the glomeruli). plasma solutes?
In glomerulonephritis, mesangial and endothelial cells A) mesangial cells
proliferate within Bowman’s capsule, with deposition of B) podocytes
antigen–antibody complexes between the various cells in C) endothelial cells
the glomeruli. It manifests in several forms; in this case, it D) vascular smooth muscle
was the result of an immune response to the streptococci. 3. Which of the following is not subject to physiological control on a
These events disrupt the barrier function of the podocytes, moment-to-moment basis?
allowing the passage of larger than normal amounts of A) hydrostatic pressure in glomerular capillaries
albumin into the filtrate, which is then excreted in the urine. B) selectivity of the filtration barrier
The rust-brownish color of the urine is due to the presence C) filtration coefficient
D) resistance of efferent arterioles
of intact red blood cells (hematuria) and hemoglobin
released from hemolyzed red blood cells. A paradoxical fea- 4. If autoregulation is effective, we expect to see which one of the
following held almost constant?
ture of some forms of glomerulonephritis is decreased
excretion of sodium, despite the apparent increased leaki- A) pressure in the renal artery.
B) total renal vascular resistance.
ness of the filtration barrier. The accumulation of sodium
C) the filtered load of water and small ions.
and accompanying water can lead to edema (explaining the
D) the contractile state of smooth muscle in the
puffiness in the boy’s face) and elevated blood pressure. afferent arteriole.
When occurring in older adults, glomerulonephritis may
5. In the face of a 20% decrease in arterial pressure, GFR decreases
be accompanied by a greatly reduced RBF and GFR, but by only 2%. What could account for this finding?
this is less likely in children. Some evidence suggests that
A) The resistances of the afferent and efferent arterioles both
increased reabsorption of sodium in the distal nephron is decrease equally.
responsible for the sodium retention in cases where GFR is B) Glomerular mesangial cells contract.
not reduced, demonstrating that the pathology is not lim- C) Efferent arteriolar resistance increases.
ited to Bowman’s capsule. In the case of poststreptococcal D) Afferent arteriolar resistance increases.
glomerulonephritis, no specific treatment is usually neces-
sary, and over time normal renal function returns.
41
C H A P T E R

Clearance
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ Define the terms clearance and metabolic clearance rate, and differentiate
between general clearance and specific renal clearance.
■ List the information required to calculate clearance.
■ State the criteria that must be met for a substance so that its clearance can
be used as a measure of glomerular filtration rate; state which substances are
used to measure glomerular filtration rate and effective renal plasma flow.
■ Given data, calculate the clearance of any excreted substance.
■ Predict whether a substance undergoes net reabsorption or net secretion
by comparing its clearance with that of inulin or by comparing its rate of
filtration with its rate of excretion.
■ Given data, calculate net rate of reabsorption or secretion for any substance.
■ Given data, calculate fractional excretion of any substance.
■ Describe how to estimate glomerular filtration rate from creatinine clearance
and describe the limitations of this estimate.
■ Describe how to use plasma concentrations of urea and creatinine as
indicators of changes in glomerular filtration rate.

Ingested substances and metabolic waste products are con-


stantly being removed from the body (cleared) by a number of
CLEARANCE UNITS
means, including disposal in the urine and feces, biochemical The units of renal clearance are often confusing to the first-
transformation in the liver, and, for volatile substances, exhala- time reader, so let us be sure of the meaning. First, the units are
tion. The rate of removal can be expressed in several ways, for volume per time (not amount of a substance per time). The eas-
example, the plasma half-life. Another way to express removal iest way to think of this is to ask what volume of plasma sup-
rate is clearance, which is the volume of plasma per unit time plies the amount excreted in a given time. For example, if each
from which all of a specific substance is removed. Clearance in a liter of plasma contains 1 mg of substance X, and 0.5 mg of X is
biomedical context has both a general meaning and a specific excreted in 1 hour, then half a liter of plasma supplies the
renal meaning. The general meaning is simply that a substance amount excreted, that is, the clearance is 0.5 L/h. The reader
is removed from the blood by any of the mechanisms men- should appreciate that removing all of a substance from a small
tioned above (see Figure 1–4). Its quantitative measure is called volume of plasma is equivalent to removing some of it from a
the metabolic clearance rate. Renal clearance, on the other larger volume, which is actually the way the kidneys do it. For
hand, means that the substance is removed from the blood and example, if all of substance X is removed from 0.5 L, this is
excreted in the urine. Evaluation of renal clearance is com- equivalent to removing half of it from 1 L, or one quarter of it
monly used clinically as an overall assessment of renal health. from 2 L, etc. The clearance is still 0.5 L/min in all these cases.
Repeated assessments over a period of time can indicate The general meaning and specific renal meaning of clear-
whether renal function is stable or deteriorating. ance can be illustrated by comparing how the body handles

417

Ch41_417-422.indd 417 11/26/10 10:09:24 AM


418 SECTION VII Renal Physiology

two substances with similar-sounding names but very differ- amount now appearing in the urine during this time is the
ent properties: inulin and insulin. Insulin is the familiar pan- product of the urine flow rate (V) and the urine concentration
creatic hormone involved in regulating blood glucose. It is a of X (Ux), that is, amount in urine/time = V × Ux. This equal-
protein with a molecular weight of 5.8 kd and is small enough ity is shown in equations (1) and (2) in Figure 41–1. Finally, by
to be freely filtered by the glomerulus. Once in Bowman’s rearrangement, we solve for clearance (Cx) as shown in equa-
space, it moves along with every other filtered substance into tion (3). Thus, we have equated the amount removed from the
the proximal convoluted tubule, where it is largely taken up by plasma with that appearing in the urine, and by rearrangement
endocytosis and degraded into its constituent amino acids. we end up with clearance in its proper units—volume per time.
Very little insulin escapes this uptake, and very little of the fil- While we are addressing the quantification of clearance, note
tered insulin makes it all the way to the urine. Thus, the kidney that the product of urine flow rate and urine concentration of
takes part in clearing insulin from the blood, but because so X (numerator on the right-hand side of equation (3)) is excre-
little appears in the urine, the specific renal clearance is very tion rate. Therefore, we can also state that the clearance of sub-
low (<1 mL/min). However, the body has additional mecha- stance X is the excretion rate divided by the plasma concentration.
nisms for clearing insulin, and its metabolic clearance rate is The process of clearance and derivation of the clearance for-
quite high (half-life less than 10 minutes). Let us contrast this mula is depicted in Figure 41–1.
with inulin. Inulin is a polysaccharide starch of about 5-kd Let us now examine the clearance of several substances
molecular weight that is not usually found in the body. Like important for the quantification of renal function, starting
insulin, it is freely filtered by the glomerulus, but it is not reab- with inulin. Inulin, as described previously, is a polysaccharide
sorbed or secreted by the nephron. All the inulin that is filtered that is freely filtered and neither reabsorbed nor secreted.
flows through the nephron and appears in the urine. Thus, Thus, once filtered, it must flow through the nephron into the
inulin’s renal clearance is relatively large. Inulin in the blood is urine (Figure 41–2). The volume of plasma cleared of inulin is
not taken up by other tissues, and the kidneys are the only the volume filtered, that is, inulin clearance equals the glom-
excretion route. As we will see, this makes inulin a very special erular filtration rate (GFR). Inulin clearance is indeed the
substance with respect to assessing renal function. hallmark experimental method of measuring the GFR.
Can something have a clearance greater than the GFR?
Indeed, yes. One such substance is para-aminohippurate
QUANTIFICATION OF CLEARANCE (PAH). This is a small (molecular weight of 194 d) water-
soluble organic anion not normally found in the body, but that
Consider again a substance X that is excreted in the urine. is used experimentally. It is freely filtered and also avidly
How do we actually calculate its clearance in the proper units? secreted by the proximal tubule epithelium (via the transcel-
The amount of X removed from a given volume of plasma lular route). The secretion rate is saturable. That is, there is a
equals the amount excreted in the urine. The amount cleared maximum rate of PAH secretion into the tubule. Such a tubu-
from the plasma in a given time is the product of the volume lar maximum, (Tm) is common in transport systems (see
of plasma cleared per unit time (Cx) and the plasma concen- Chapter 42). However, at low plasma concentrations, almost
tration (Px), that is, amount cleared/time = Cx × Px. That same all of the PAH entering the kidney is removed from the plasma

Amount in plasma = Cx × Px

Urine before clearance


Plasma before clearance

Urine during clearance


Plasma during clearance

Amount in urine = V × Px

FIGURE 41–1 Derivation of the basic clearance formula. Urine after clearance
Over time, substance X (dots) is removed from the plasma (large Plasma after clearance
boxes) and put into the urine (small boxes) and excreted. The Amount in plasma = Amount in urine (1)
amount of X removed from the plasma during that time (Cx × Px)
Cx × Px = V × Ux (2)
must equal the amount excreted (V × Ux) in that same time, as
shown in equations (1) and (2). By rearrangement (equation (3)), Cx = V × Ux (3)
we solve for the clearance of X. Px
CHAPTER 41 Clearance 419

Concentration during the period of urine formation and collection or there


of inulin in must be multiple samplings and a complex regression analysis.
plasma = 4 mg/L
For routine assessment of GFR in patients, there is a much
Glomerular easier method: creatinine clearance. Creatinine is an end
capillary
product of creatine metabolism and is exported into our blood
continuously by skeletal muscle. The rate is proportional to
skeletal muscle mass, and to the extent that muscle mass is
Rate of fluid constant in a given individual, the creatinine production is
Bowman’s filtration (GFR) = 7.5 L/h constant. Creatinine is freely filtered and not reabsorbed. A
space Concentration of
inulin in filtrate = 4 mg/L small amount, however, is secreted by the proximal tubule.
Total inulin filtered = 30 mg/h Therefore, the creatinine appearing in the urine represents a
No reabsorption of inulin
filtered component (mostly) and a much smaller secreted
No secretion of inulin component. Because of the secretion, creatinine clearance is
slightly higher than the GFR, normally by about 10–20%. For
Total inulin excreted = 30 mg/h routine assessment of GFR, this degree of error is acceptable.
FIGURE 41–2 Renal handling of inulin. All filtered inulin How does one measure creatinine clearance? Usually, a
is excreted. Since the volume of plasma cleared of inulin is the patient’s urine is collected for 24 hours, and a blood sample is
volume filtered, the inulin clearance equals the GFR. (Modified with taken sometime during the collection period. Blood and urine
permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, are assayed for creatinine concentration, and we apply the
11th ed. McGraw-Hill, 2008.) clearance formula (Figure 41–1, equation (3)) to yield creati-
nine clearance.
For a patient with a very low GFR, the secreted component
and excreted in the urine. About 20% of the excreted PAH is a relatively larger fraction of the total amount excreted;
appears by filtration and the rest by secretion. Its clearance, therefore, the creatinine clearance more severely overestimates
therefore, is nearly as great as the renal plasma flow. In fact, the GFR in patients with a very low GFR than in those with higher
PAH clearance can be used experimentally as a measure of GFR values. Nevertheless, because of low cost and conve-
renal plasma flow, usually called the effective renal plasma nience, creatinine clearance continues to be the most common
flow to indicate that its value is slightly less than the true renal method for routine assessment of patient GFR and the integ-
plasma flow. rity of renal filtration.
What can the clearance of any freely filtered substance tell
us? If we know the GFR (as assessed from inulin clearance)
and the clearance of the substance, then any difference between PLASMA CREATININE AND
clearance and GFR represents net secretion or reabsorption
(or, in a few rare cases, renal synthesis). If the clearance of a UREA CONCENTRATIONS AS
substance exactly equals the GFR, then there has been no net
reabsorption or secretion. If the clearance is greater than the
INDICATORS OF CHANGES IN GFR
GFR, there must have been net secretion. Finally, if the clear- Although creatinine clearance is a valuable clinical determi-
ance is less than the GFR, there must have been net reabsorp- nant of GFR, in routine practice, it is far more common to
tion. The word net in this description is important. As we will measure plasma creatinine alone and to use this as an indica-
see in later chapters, a number of substances are reabsorbed in tor of GFR. If we ignore the small amount secreted, there
certain regions of the nephron and secreted in other regions. should be an excellent inverse correlation between plasma
The net result of these processes is the sum of everything that creatinine concentration and GFR (Figure 41–3).
happens along the nephron. Of course, if a substance is not A healthy person’s plasma creatinine concentration is about
freely filtered, low clearance may simply indicate that little of 1 mg/dL. It remains stable because each day the amount of
the substance entered the tubule. creatinine excreted is equal to the amount of creatinine pro-
duced. Suppose one day, the GFR suddenly decreases by 50%
because of an obstruction in the renal artery. On that day, the
A PRACTICAL METHOD OF person filters only 50% as much creatinine as normal. There-
fore, creatinine excretion is also reduced by 50%. (We are
MEASURING GFR: CREATININE ignoring the small contribution of secreted creatinine.) Assum-
CLEARANCE ing no change in creatinine production, the person transiently
goes into positive creatinine balance, and the plasma creati-
The gold standard for measuring GFR is the inulin clearance, nine increases. However, despite the persistent 50% GFR
and this method is used in research studies. The method is reduction, the plasma creatinine does not continue to rise
cumbersome, however, because either inulin must be infused indefinitely; rather, it stabilizes at 2 mg/dL (i.e., after it has
at a rate sufficient to keep its plasma concentration constant doubled). At this point, the person once again is able to excrete
420 SECTION VII Renal Physiology

surement is a reasonable indicator of GFR. It is not completely


accurate, however, for several reasons: (1) as before, some cre-
8
atinine is secreted; (2) an original creatinine measurement
when GFR was normal may not be available; (3) creatinine
production may not remain completely unchanged. However,
an increasing plasma creatinine is a red flag that there may be
PCr, mg/100 mL

a renal problem.
5 Because urea is also handled by filtration, the same type of
analysis suggests that the measurement of plasma urea con-
4 centration could also serve as an indicator of GFR. However, it
is a much less accurate indicator than plasma creatinine
3
because the range of normal plasma urea concentration varies
2 widely, depending on protein intake and changes in tissue
catabolism, and because urea excretion is under partial hor-
1 monal regulation.

45 90 135 180 225 360


GFR, L/day

FIGURE 41–3 Steady-state relation between plasma CLINICAL CORRELATION


creatinine and GFR for a person with a normal creatinine
production. When GFR is low, plasma creatinine rises to high levels, A 72-year-old, rather thin and frail female is experiencing
making plasma creatinine a convenient indicator of GFR. (Modified with bouts of atrial fibrillation. Her serum electrolytes are all
permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: within normal limits, her creatinine is 1.1 mg/dL (high
Lange Medical Books/McGraw- Hill, Medical Pub. Division, 2009.) normal), and there is no indication of a major renal disor-
der. Her body weight is 47 kg. Her primary care physician
(PCP) prescribes digoxin for its antiarrhythmic effective-
creatinine at the normal rate and so goes back in balance with ness, but is concerned about overdosing and producing
a stable plasma level. The 50% GFR reduction has been just digitalis toxicity. The choice of an appropriate dosing regi-
offset by the doubling of plasma creatinine concentration, men (amount and schedule) is a difficult one, particularly
restoring the filtered load of creatinine to normal. To under- for drugs such as digoxin with significant side effects. The
stand this point, assume an original daily filtration volume of goal is to find a “therapeutic window” in which the body
180 L (1,800 dL). levels of the drug are high enough to be effective, but low
The original normal state is given by: enough not to invoke side effects. For drugs whose major
excretion route is the kidneys, which include digoxin, it is
Filtered creatinine = 1 mg/dL × 1,800 dL per day often necessary to make adjustments based on the patient’s
(4)
= 1,800 mg per day GFR. This is particularly appropriate in patients whose
The new steady state is given by: renal function is impaired due to disease or natural decline
with age. In common medical settings it is not feasible to
Filtered creatinine = 2 mg/dL × 900 dL per day measure GFR via inulin clearance, or even to measure crea-
(5)
= 1,800 mg per day tinine clearance using 24-hour urine collection in nonhos-
pitalized patients. The question is: how does one obtain an
In the new steady state, creatinine excretion is normal, even estimate of GFR?
though the plasma concentration has doubled (the person is in A common method is to estimate creatinine clearance
balance). In other words, creatinine excretion is below normal using a formula, known as the Cockcroft–Gault formula
only transiently until plasma creatinine has increased as much (shown below), that includes plasma creatinine, age, body
proportionally as the GFR has decreased. weight, and gender. The use of this formula, or any of the sev-
What if the GFR then decreased to 300 dL per day? Again, eral others that have been derived over the years, is subject to
creatinine retention would occur until a new steady state had error. However, it is still useful as a guide for drug dosing pur-
been established (i.e., until the person was again filtering 1,800 poses, where a precise clearance value is not really needed:
mg per day). What would the new plasma creatinine be?
creatinine clearance [mL/min] =
1,800 mg per day = Pcr × 300 dL per day (6)
(140–age) × body weight [Kg] × 0.85 [if female]
_________________________________ (8)
Pcr = 6 mg/dL (7) 72 × serum creatinine [mg/dL]
This works out to be 34.3 mL/min in our patient. (For
The increase in plasma creatinine results directly from the comparison, the estimated creatinine clearance using the
decrease in GFR. Therefore, a single plasma creatinine mea-
CHAPTER 41 Clearance 421

2. A drug X has a short plasma half-life and must be administered


Cockcroft–Gault formula for a 21-year-old, 70-kg male frequently to maintain therapeutic levels. The urinary
with a serum creatinine of 1.0 mg/dL is 116 ml/min.) Her concentration of X is much higher than the plasma
GFR, estimated by creatinine clearance, is low relative to concentration. A substantial amount of X also appears in the
that of young adults, but is sufficient to permit adequate feces. What can we say about the renal clearance of X compared
excretory function. However, because the clearance of with the metabolic clearance rate of X?
digoxin will be somewhat slow, her prescription is likely to A) The metabolic clearance rate is higher than the renal
be for lower doses, or for greater intervals between doses, clearance.
than it would be in a much younger patient. B) The renal clearance is higher than the metabolic clearance
rate.
C) The two clearances are the same.
D) There is insufficient information to answer the question.
CHAPTER SUMMARY 3. Inulin clearance is measured twice: the first time at a low inulin
infusion rate, and the second time at a higher infusion rate that
■ Clearance has both a general meaning, describing loss of
results in a higher plasma inulin concentration during the test.
material from the body, and a specific renal meaning involving
Assuming the kidneys behave the same in both cases, which
the kidney’s ability to remove substances from the blood.
measurement will yield a higher inulin clearance?
■ Clearance is always expressed in units of volume per time.
A) the first
■ Renal clearance of any substance is quantified by a general B) the second
clearance formula relating urine flow to urine and plasma C) both measurements are the same
concentrations. D) there is insufficient information to answer the question
■ Inulin clearance can be used to measure GFR because inulin 4. Which of the following indicates correct relative renal
is freely filtered and neither secreted nor reabsorbed. clearances?
■ PAH clearance can be used as an estimate of renal plasma flow. A) Glucose clearance is greater than urea clearance.
■ Creatinine clearance is used as practical estimate of GFR. B) PAH clearance is greater than inulin clearance.
■ Plasma creatinine concentration is used clinically as an C) Urea clearance is greater than PAH clearance.
indicator of the GFR. D) Creatinine clearance is greater than PAH clearance.
5. An acute poisoning episode destroys 80% of a patient’s nephrons.
If the plasma urea concentration prior to the episode was
STUDY QUESTIONS 5 mmol/L, and assuming dietary protein remains the same, what
is the expected value of plasma urea now?
1. We can calculate the renal clearance of any substance if we know
A) 5 mmol/L
which pair of values?
B) 6.25 mmol/L
A) urine flow rate and urine concentration C) 25 mmol/L
B) plasma concentration and urine concentration D) continuously rising
C) GFR and urinary excretion rate
D) plasma concentration and urinary excretion rate
This page intentionally left blank
42
C H A P T E R

Tubular Transport
Mechanisms
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ Identify the major morphological components of an epithelial tissue,


including lumen, interstitium, apical and basolateral membranes, and
tight junctions.
■ State how transport mechanisms combine to achieve active transcellular
reabsorption in epithelial tissues.
■ Define iso-osmotic transport.
■ Define paracellular transport and differentiate between transcellular
and paracellular transport.
■ Describe qualitatively the forces that determine movement of reabsorbed
fluid from the interstitium into peritubular capillaries.
■ Explain why volume reabsorption in the proximal tubule depends on
activity of the Na,K-ATPase.
■ Compare the Starling forces governing glomerular filtration with those
governing peritubular capillary absorption.
■ Compare and contrast the concepts of Tm and gradient-limited transport.
■ Contrast “tight” and “leaky” epithelia.

PROXIMAL TUBULE Most of the solute reabsorbed in the proximal tubule


consists of sodium and the anions (mostly chloride and bicar-
REABSORPTION bonate) that must accompany sodium to maintain electroneu-
trality. These solutes are removed from the tubular lumen
Virtually all of the 180 L of water and several pounds of salt and and move into the interstitium by a combination of processes
other solutes filtered each day into Bowman’s space are reab- that we describe below. The large amount of solute trans-
sorbed, along with large amounts of many other substances. ferred from lumen to interstitium sets up an osmotic gradient
Quantitatively most of this reabsorption occurs in the proxi- that favors the parallel movement of water. The proximal
mal tubule, a process that is very nearly iso-osmotic, meaning tubule epithelium is very permeable to water, which fol-
that water and solutes are reabsorbed in equal proportions. lows the solute across in equal proportions. Thus, both the
Recall that filtration in the glomerulus is also iso-osmotic. fluid removed from the lumen and that remaining behind are
Almost all solutes (except large plasma proteins) are filtered essentially iso-osmotic with the original filtrate. We say
from plasma into Bowman’s space in the same proportion as “essentially” because there must be some difference in osmo-
water; thus, their concentrations in the glomerular filtrate are lality to induce water movement, but for an epithelial barrier
the same as in the plasma. By the end of the proximal tubule, such as the proximal tubule that is very permeable to water, a
about two thirds of the water and two thirds of the solutes have difference of less than 1 mOsm/kg is sufficient to drive reab-
been reabsorbed. In the later portions of the nephron, reab- sorption of water. An osmolality difference of 1 mOsm/kg is
sorption is generally not iso-osmotic, which is crucial for our equal in driving force to 19 mm Hg of hydrostatic pressure.
ability to independently regulate solute and water balance. Once in the interstitium, the solutes and water move from

423

Ch42_423-428.indd 423 11/26/10 10:08:31 AM


424 SECTION VII Renal Physiology

LUMEN INTERSTITIUM

apical Peritubular
membrane Interstital capillary
Channel or space
transporter

Trancellular
reabsorption
FIGURE 42–1 Transcellular and paracellular
reabsorption. Transcellular reabsorption is a two-step Basolateral
membrane
process with separate influx and efflux steps utilizing
transporters or channels. Paracellular reabsorption is always Paracellular
a passive process through the tight junctions. (Modified reabsorption
with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. tight junction Basement
Division, 2009.) membrane

interstitium into the peritubular capillaries and are returned Substances cross the membranes of epithelial cells by an
to the systemic circulation. array of mechanisms. These mechanisms are no different from
Reabsorption across the tubular epithelium can be either those used elsewhere in the body to transport substances across
through the cells (the transcellular route) or around the cell membranes as described in Chapter 3. We can view these
cells (the paracellular route), that is, through the matrix of mechanisms as a physiological toolbox. Renal cells use which-
the tight junctions that link each epithelial cell to its neigh- ever set of tools is most suitable for the task. These mechanisms
bor. The transcellular route is a two-step process: into the include simple diffusion through the bilayer, movement
cells across the apical membrane facing the tubular lumen through channels, and movement through transporters of
and out of the cells across the basolateral membrane facing various types. This variety is depicted in Figure 42–2. Except
the interstitium. These structures and pathways are depicted for simple diffusion, all of these processes are regulated by sig-
in Figure 42–1. naling pathways. It is through regulation of transporters and

Diffusion
Substance A 1
Me
mb

Uniporter
ran

Substance B 2
e

Symporter
Substance C
Substance D 3

Antiporter
Substance E 4
Substance F

Substance G 5 Ion channel

6
Primary active
Substance H
transport
FIGURE 42–2 Mechanisms of transmembrane
solute transport. With the exception of simple diffusion
through the lipid bilayer, all transport involves channels
and transporters that are regulated by signaling ADP + Pi
pathways. (Modified with permission from Eaton DC, Pooler JP:
Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical ATP
Books/McGraw-Hill, Medical Pub. Division, 2009.)
CHAPTER 42 Tubular Transport Mechanisms 425

Lumen Interstitium brane and sites of inactivity or degradation, with lifetimes in


the membrane generally being only a few hours.
While some substances cross the epithelial barrier by the par-
1. Insert or retrieve acellular route, it is transcellular transport that controls the
from storage sites overall process, that is, transcellular processes set up the condi-
tions favoring paracellular transport. All of this requires polar-
2. Transcribe and ization of the epithelial cells, meaning that the transport proteins
synthesize,
present in the apical membrane are different from those in the
or degrade
basolateral membrane. In the proximal tubule, the key process
achieved by this polarized distribution of transport proteins is
3. Attach ligand
L the movement of sodium through the cells from lumen to inter-
stitium. The transport of virtually every other substance depends
on the movement of sodium. Figure 42–4 shows the morphol-
ogy of a generalized proximal tubule epithelium in which salt
FIGURE 42–3 Common mechanisms for regulating channel and water transport can be viewed as a multistep process.
and transporter activity. 1) Transport proteins are shuttled back and Step 1 is the active extrusion of sodium from epithelial cell
forth between the surface membrane, where they function normally, to interstitium across the basolateral membrane. Step 2 is the
and sites of sequestration at the base of microvilli or in intracellular passive entrance of sodium from the tubular lumen across the
vesicles. 2) Transport protein is synthesized and inserted in the apical membrane into the cell to replace the sodium removed
membrane or removed and degraded. 3) Transport proteins are in step 1. Step 3 is the parallel movement of anions that must
activated or inhibited by attaching ligands, either covalently (e.g., accompany the sodium to preserve electroneutrality. Step 4 is
phosphorylation) or reversibly (e.g., ATP). the osmotic flow of water from tubular lumen to interstitium.
Finally, step 5 is the bulk flow of water and solute from inter-
stitium into the peritubular capillary. Let us examine these
channels that the kidneys control the excretion of various sub- steps more closely.
stances. Figure 42–3 depicts some of the ways flux is regulated. The active extrusion of sodium in step 1 is via the Na,K-
This includes trafficking of channels and transporters between ATPase, which is the major energy consumer in the cell. The
the surface membrane and inactive sites within the cell, genetic action of the Na,K-ATPase has several consequences, the key
expression of new protein, degradation of existing protein, and one being that it keeps the concentration of sodium within the
the attachment of ligands that either directly alter protein func- cell low enough to favor the passive entrance of sodium from
tion or target the protein for trafficking. Transporter and chan- lumen to cell in all the processes of step 2.
nel proteins are not permanent fixtures in the membrane; The entrance of sodium into the cell in step 2 is via multiple
rather, they are constantly being shuttled between the mem- pathways. Quantitatively most of sodium enters via the

1. Sodium is actively extruded into the interstitium


2. Sodium enters passively from the tubular lumen
3. Anions follow the sodium
4. Water follows the solute
5. Water and solutes move by bulk flow into the peritubular capillary

Lumen Interstitium

ATP Na+ Peritubular


2 Na+ 1 +
capillary
K

3 Anions

4 Water

Water, anions

FIGURE 42–4 Epithelial salt and water reabsorption. See text for explanation of each individual step.
426 SECTION VII Renal Physiology

sodium–proton antiporter (NHE3 isoform). As we will see tubular lumen to interstitium. Ultimately, this can lead to
later, regulation of this transporter is a key player in regulating increased excretion of water and electrolytes from the body.
sodium excretion. The events just described have consequences for any filtered
Step 3, the movement of anions, is the most complex, as it solute that is not specifically removed from the lumen in step
involves two ions (chloride and bicarbonate) and a variety of 2 or 3. As water follows sodium and its anions across the epi-
transcellular and paracellular processes. We will examine the thelium, the luminal volume decreases, thereby concentrating
details in Chapters 44 and 47, but for now we emphasize that the remaining solutes. If two thirds of the water is removed,
the movement of sodium, which is a cation, must be matched any solute not previously removed will increase in concentra-
quantitatively by the equal movement of anions. tion by a factor of 3. As its luminal concentration rises, this
Step 4 is the osmotic movement of water. The tubular cells generates a concentration gradient across the tight junctions
possess a complement of aquaporins (water channels) in both between the lumen and the interstitium. (The interstitial con-
the apical and basolateral membranes, and the tight junctions centration of transported substances is essentially clamped to
are also permeable to water. Therefore, as steps 1–3 lower the the plasma value because of the high peritubular blood flow
local luminal osmotic concentration by even a few milliosmoles and high permeability of the fenestrated capillaries.) If the
per liter, water flows osmotically from lumen to interstitium. tight junctions are permeable to the substance in question
The movement of water into the interstitium in step (“leaky”), the substance diffuses from the lumen to the inter-
4 promotes step 5. This is the bulk flow of fluid from intersti- stitium, and then into the peritubular capillaries along with
tium to peritubular capillary driven by Starling forces (hydro- sodium and water. This is precisely what happens to many sol-
static and oncotic pressure gradients). The capillary hydraulic utes (e.g., urea, potassium, chloride, calcium, and magnesium)
pressure opposes the uptake of interstitial fluids, but its value in the proximal tubule. The exact fractions that are reabsorbed
of 15–20 mm Hg is much lower than the 60 mm Hg in the depend on the permeability of the tight junctions, but are gen-
glomerular capillaries, where there is net filtration. Meanwhile, erally in the range of one half to two thirds. (One substance
the plasma oncotic pressure has increased to more than that does not move by the paracellular route is glucose, which
30 mm Hg because loss of water by filtration in the glomerular is impermeable in the tight junctions. We will describe the fate
capillaries concentrates the large plasma proteins. There is also of filtered glucose in Chapter 43.)
a small but significant interstitial pressure (Table 42–1). The In summary, active sodium transport by the Na,K-ATPase is
sum of these Starling forces is a net absorptive pressure, and it the crucial process, necessary not only for the reabsorption of
drives fluid movement into the peritubular capillaries. The sodium, but also for creating the conditions that drive the
reader can appreciate the fact that if cortical Starling forces are reabsorption of water and every other solute.
abnormal (e.g., low plasma oncotic pressure as when liver dis-
ease prevents normal production of serum albumin), absorp-
tion of fluid from the cortical interstitium can be slowed, LIMITS ON RATE OF TRANSPORT:
causing a backup of fluid that inhibits fluid movement from
Tm AND GRADIENT-LIMITED
SYSTEMS
TABLE 42–1 Estimated forces involved in movement
Although the transport capacity of the renal cortex is huge, it
of fluid from interstitium into peritubular capillaries.a
is not infinite. There are upper limits to the rate at which
Forces mm Hg sodium or any other solute can be reabsorbed or secreted. In
many situations, these limits are reached, with the conse-
1 Favoring uptake
quence that, in the case of reabsorption, larger than normal
(a) Interstitial hydraulic pressure, PInt 3 amounts of the filtered load are not reabsorbed. In general,
(b) Oncotic pressure in peritubular 33 transport mechanisms can be classified by the properties of
capillaries, πPC these upper limits as either (1) gradient-limited systems or
(2) tubular maximum–limited (Tm) systems. These proper-
2 Opposing uptake
ties are significant both for normal function and, as explained
(a) Hydraulic pressure in peritubular 20 in subsequent chapters, in pathological situations. The classifi-
capillaries, PPC
cation is based on the leakiness of the tight junctions. Con-
(b) Interstitial oncotic pressure, πInt 6 sider first gradient-limited systems. When the tight junctions
3 Net pressure for uptake (1 – 2) 10 are very leaky to a given substance, for example, sodium, it is
a
impossible for the removal of the substance from the lumen to
The values for peritubular capillary hydraulic and oncotic pressures are for the early
portions of the capillary. The oncotic pressure, of course, decreases as protein-free reduce its luminal concentration very much below that in the
fluid enters it (i.e., as absorption occurs) but would not go below 25 mm Hg (the cortical interstitium, because as the luminal concentration
value of arterial plasma) even if all fluid originally filtered at the glomerulus were
absorbed. decreases, the gradient between these two media is increased,
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, causing the substance to leak back as fast as it is removed.
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009. Thus, for sodium and all other substances whose reabsorption
CHAPTER 42 Tubular Transport Mechanisms 427

is characterized by a gradient-limited system, the luminal con-


comprehensive analysis. The results reveal several abnor-
centration remains close to the interstitial concentration.
malities. His urine contains more than trace amounts of glu-
However, the existence of a limit does not stop reabsorption in
cose and amino acids, high amounts of phosphate and
normal circumstances because water is being reabsorbed
potassium, and a low pH (5.5). His blood analysis indicates
simultaneously, so that the luminal concentration does not
low bicarbonate (17 mEq/L), low potassium (3.1 mEq/L),
decrease very much, permitting large amounts of the sub-
and low phosphate (1.7 mg/dL).
stance to be removed. In contrast, if unusual osmotic condi-
The diagnosis is heavy metal–induced damage to the
tions retard water reabsorption, then removal of the substance
proximal tubule cells, producing a constellation of renal
occurs without a corresponding amount of water. Conse-
defects called Fanconi syndrome. The key pathology in
quently, its concentration does decrease and the limiting gra-
Fanconi syndrome is mitochondrial damage that reduces
dient is indeed reached, resulting in unusually high amounts
Na,K-ATPase activity in the proximal tubule. This decreases
of the substance remaining in the large volume of unreab-
reabsorption of sodium and of many substances that are
sorbed water.
directly or indirectly tied to the reabsorption of sodium.
Now consider Tm-limited systems in which the tight junctions
Chief among these are bicarbonate, phosphate, potassium,
are impermeable to the solutes in question. There is no back leak
and, of course, water. Glucose and amino acids in the urine
and no limit on the size of the difference in concentration
provide diagnostic clues, but the losses are not serious. The
between lumen and interstitium. The limit on transport rate
patient’s low bicarbonate is consistent with renal tubular
instead is placed on the capacity of the transporters to remove
acidosis, which is a primary component of Fanconi syn-
the substance (the inherent kinetic properties of the transport
drome. His respiratory system attempts to compensate for
proteins and their density in the membrane). As the filtered
plasma acidity by increasing the rate of ventilation.
load rises, increasing amounts of the filtered substance are reab-
(His breathing discomfort could also be related to the result
sorbed, up to the point of saturating the transporters. Any fur-
of his smoking as well as heavy metal toxicity.) His low
ther increase in filtered load above the saturation point does not
plasma potassium is the result of excessive potassium secre-
increase the rate at which the substance is transported out of the
tion in the distal nephron stimulated by the large amount
lumen; thus, more and more is left behind. In most cases, the
of non-reabsorbed tubular sodium, and accounts for his
amount that escapes reabsorption is excreted.
feeling of muscle weakness. His high excretion of phos-
For many substances governed by a Tm system, their filtered
phate and acidosis lead to complicated effects in bone,
loads are usually well below their Tm, and the tubule has no trou-
including loss of bone mineral that may lead to spontane-
ble reabsorbing virtually all that is filtered. This is the case for
ous fractures. Treatment in this case consists of supple-
glucose and many other organic substances, which, under nor-
mental electrolytes, vitamin D to promote bone health, and
mal circumstances, are completely reabsorbed by the end of the
any measures that will result in a reduction or elimination
proximal tubule. But solutes handled by gradient-limited sys-
of the cadmium exposure.
tems are never reabsorbed completely, because a finite interstitial
concentration ensures there will be a finite tubular concentra-
tion, and therefore a substantial amount passed on to the next
nephron segment. This holds true for sodium. The consequences CHAPTER SUMMARY
of these differences will be discussed in Chapters 43 and 45.
■ Reabsorption in the proximal tubule is iso-osmotic.
■ Flux from lumen to interstitium can be transcellular, using
separate transport steps in the apical and basolateral mem-
CLINICAL CORRELATION branes; or paracellular, around the cells through tight junctions.
■ The kidneys regulate excretion by regulating channels and
A 42-year-old man has worked at a metal recycling plant for transporters in epithelial cell membranes.
the past 5 years, where he has been exposed to toxic heavy ■ The reabsorption of water and almost all solutes is linked,
metals, including cadmium. He has a history of smoking, directly or indirectly, to the activity of Na,K-ATPases in the
but for the most part has been in good health. Over the past basolateral membranes.
year, however, he has noticed gradually increasing cough ■ High water permeability in the proximal tubule epithelium
and lung irritation, and some feeling of difficulty breathing. ensures that water reabsorption is tightly coupled to solute
He also reports vague muscle weakness and pain in his legs. reabsorption.
In addition, he has experienced increased thirst and urinary ■ Volume reabsorption is a multistep process involving transport
frequency, and has developed a craving for salty foods such across epithelial membranes from lumen to interstitium, and
as pickles and potato chips. His physical exam is unremark- bulk flow from interstitium to peritubular capillaries driven by
able except for a somewhat high respiratory rate (tachyp- Starling forces.
nea) of 21 breaths/min. A urine dipstick test reveals a mild ■ All reabsorptive processes have a limit on how fast they can
proteinuria (increased protein in the urine), and samples of occur, either because the transporters saturate (Tm systems) or
because the substance leaks back into the lumen (gradient-
his blood and urine are sent to the clinical laboratory for a
limited systems).
428 SECTION VII Renal Physiology

STUDY QUESTIONS 4. In the proximal tubule, water can move through


A) apical membranes of proximal tubule cells.
1. A healthy patient has a normal plasma osmolality (close B) basolateral membranes of proximal tubule cells.
to 300 mOsm/kg). If 100 mmol of solutes is reabsorbed C) tight junctions.
iso-osmotically from the proximal tubule, approximately D) all of the above.
how much water is reabsorbed with the solute? (Remember: 1
5. A drug X is secreted into the proximal tubule by a Tm-limited
mmol of solute ≅ 1 mOsm and 1 g H2O ≅ 1 mL.)
system. This implies that
A) 100 mL
A) X cannot easily diffuse by the paracellular route.
B) 300 mL
B) all the X that enters the renal vasculature will be secreted.
C) 333 mL
C) the rate of X secretion is independent of the plasma
D) 1,000 mL
concentration.
2. Quantitatively, most sodium gains entrance to proximal tubule D) X is not filtered at the glomerulus.
cells by
A) paracellular diffusion.
B) transcellular diffusion.
C) the Na,K-ATPase.
D) antiport with hydrogen ions.
3. The tight junctions linking proximal tubule cells permit passive
diffusion of
A) glucose.
B) sodium.
C) all filtered solutes.
D) no filtered solutes.
43
C H A P T E R

Renal Handling of
Organic Substances
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ State the physiological utility of either excreting or reabsorbing organic


solutes.
■ State the general characteristics of the proximal tubular systems for active
reabsorption or secretion of organic nutrients.
■ Describe the renal handling of glucose, and state the conditions under which
glucosuria is likely to occur.
■ Describe the renal handling of proteins and small peptides.
■ Describe the secretion of para-aminohippurate.
■ Outline the handling of urate.
■ Describe the secretion of organic cations.
■ Describe how tubular pH affects the excretion and reabsorption of weak
acids and bases.
■ Describe the renal handling of urea, including the medullary recycling of urea
from the collecting duct to the loop of Henle.

OVERVIEW this chapter and again in the following chapter in the discus-
sion of renal handling of water.
As pointed out in Chapter 39, a major function of the kidneys
is the excretion of organic waste products, and foreign chemi-
cals and their metabolites. Furthermore, the kidneys filter PROXIMAL REABSORPTION OF
large amounts of organic substances that they do not excrete; ORGANIC NUTRIENTS: GLUCOSE
therefore, reabsorptive processes must exist to prevent inap-
propriate loss of useful organic nutrients. Because the blood AND AMINO ACIDS
contains many small, filterable molecular species, the kidney
Most of the major cellular nutrients in the plasma that the kid-
has to handle all of them. An analysis of the renal handling of
neys must not lose in the urine are freely filterable and must be
every one of these organic substances would be prohibitive, so
almost completely reabsorbed. These include glucose, amino
we will discuss a few key solutes and establish generalities
acids, acetate, Krebs cycle intermediates, some water-soluble
about the others. In essence, the kidneys perform a kind of tri-
vitamins, lactate, acetoacetate, β-hydroxybutyrate, and many
age. They (1) reabsorb organic metabolites that should not be
others. The proximal tubule is the major site for reabsorption
lost, (2) eliminate waste products and unwanted foreign
of the large quantities of these organic nutrients filtered each
organic substances by not reabsorbing them or actually secret-
day by the renal corpuscles. We can make the following
ing them, and (3) partially reabsorb others.
generalities:
One organic substance, urea, is unique in this regard. It is a
waste product that must be excreted to prevent accumulation. 1. They are actively transported (i.e., are reabsorbed up
However, it also plays a key role in renal regulation of water [against] their respective electrochemical gradients) by
balance. The renal handling of urea is briefly discussed later in transport proteins that are specific for one or only a few

429

Ch43_429-436.indd 429 11/26/10 11:21:33 AM


430 SECTION VII Renal Physiology

solute species. There is not a separate transporter for ev- 900


ery solute in the body. Two or more closely related sub-
800
stances may use the same transporter. For example, amino Filtered

Glucose filtered load, reabsorption


acid transporters are distinct from those for glucose, but load
700
there are not 20 separate amino acid transporters. Rather

or excretion (mg/min)
there is one for arginine, lysine, and ornithine; another for 600
glutamate and aspartate; and so on. In most cases, reab- Transport
Excretion
500
sorption is very close to complete. maximum
2. The “uphill” step is across the apical membrane, usually 400
via a symporter with sodium.
3. Most are characterized as Tm systems (have an upper limit 300
Normal Reabsorption
to the rate at which they can transport). These limits are
200
usually well above the amounts normally filtered. Accord-
ingly, the kidneys have no trouble returning all that is fil- 100
Threshold
tered back to the plasma. However, because none is ex-
creted, the kidneys do not help regulate their levels in the 0
100 200 300 400 500 600 700 800
body; the kidneys are merely reclaiming them from the Plasma glucose concentration
tubular fluid. It is also true, however, that under abnormal (mg/100 ml)
conditions, the plasma concentration of these substances FIGURE 43–1 Glucose handling by the kidney. The filtered
may increase so much that the filtered load exceeds the load, amount reabsorbed, and amount excreted are plotted as a
reabsorptive Tm. In this case, large quantities are excreted function of plasma glucose concentration. At a given GFR, the filtered
in the urine. Examples are glucose, acetoacetate, and load is always exactly proportional to the plasma concentration. At
β-hydroxybutyrate in patients with severe uncontrolled normal levels of plasma glucose the filtered load is well below the Tm,
diabetes mellitus. and therefore all the filtered glucose is reabsorbed and none is
4. The transporters can be inhibited by a variety of drugs, excreted. However, as plasma glucose rises into the hyperglycemic
and several monogenetic diseases are associated with loss range, the Tm is reached, and any glucose filtered in excess of the Tm is
of function in one or more of these proximal reabsorptive excreted. (Reproduced with permission from Widmaier EP, Raff H, Strang KT:
Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
systems. In some cases, the deficit may be highly specific
(e.g., involving only one amino acid), whereas in others,
multiple systems may be involved (e.g., glucose and many the Tm; Figure 43–1). This occurs when plasma glucose
amino acids). These defects also occur when the deficit is increases above roughly 300 mg/dL, a situation often found in
due to an ingested toxin (e.g., heavy metal toxicity) rather untreated diabetes mellitus. In very severe cases, blood glucose
than a genetic abnormality. can exceed 1,000 mg/dL, or over 55 mmol/L, leading to signifi-
cant loss of glucose. Assume that the glucose Tm is 375 mg/min
(a typical value). With a glomerular filtration rate (GFR) of
GLUCOSE 125 mL/min (1.25 dL/min) and a normal plasma glucose of
90 mg/dL, the filtered load is 1.25 dL/min × 90 mg/dL =
Under most circumstances, it would be deleterious to lose glu- 112.5 mg/min, much less than the Tm of 375 mg/min. Thus, the
cose in the urine, particularly in conditions of prolonged fast- kidneys easily reabsorb the entire filtered load. When plasma
ing. Thus, the kidneys normally reabsorb all of the glucose glucose reaches 300 mg/dL, the filtered load is now 1.25 dL/
that is filtered. A typical plasma glucose level is about 90 mg/dL min × 300 mg/dL = 375 mg/min. At this point, the proximal
(5 mmol/L). It increases transiently to well over 100 mg/dL convoluted tubule has reached the upper limit of what it can
during meals and decreases somewhat during fasting. Usually reabsorb, and a little glucose begins to spill into the urine. Fur-
all the filtered glucose is reabsorbed in the proximal tubule. ther increases in plasma glucose above 300 mg/dL lead to pro-
This involves taking up glucose from the tubular lumen along gressively greater renal losses. This leads to an unwanted
with sodium via a sodium-dependent glucose symporter diuresis (high urine volume) that we will discuss later, but one
(SGLUT) across the apical membrane of proximal convoluted can appreciate that any glucose not reabsorbed is an osmole in
tubule epithelial cells, followed by its exit across the basolat- the tubule that has consequences for water reabsorption.
eral membrane into the interstitium via a glucose transporter
(GLUT), a uniporter. Unlike the case for sodium and many
other solutes, the tight junctions are not significantly perme- PROTEINS AND PEPTIDES
able to glucose. Therefore, as glucose is removed from the
lumen and the luminal concentration decreases, there is no Although we sometimes say the glomerular filtrate is protein
backleak, resulting in virtually complete reabsorption. free, it is not truly free of all protein; it just has a total protein
Because glucose reabsorption is a Tm system, abnormally content much lower than plasma. First, peptides and smaller
high filtered loads overwhelm the reabsorptive capacity (exceed proteins (e.g., angiotensin, insulin), although present at low
CHAPTER 43 Renal Handling of Organic Substances 431

concentrations in the blood, are filtered in considerable quan- degradation occurring in renal disease may result in elevated
tities. Second, while the movement of large plasma proteins plasma hormone concentrations.
across the glomerular filtration barrier is extremely limited, a Very small peptides, such as angiotensin II, are catabolized
small amount does make it through into Bowman’s space. For into amino acids or dipeptides and tripeptides within the
albumin, the plasma protein of highest concentration in the proximal tubular lumen by peptidases located on the apical
blood, the concentration in the filtrate is normally about surface of the plasma membrane. These products are then
1 mg/dL, or roughly 0.02% of the plasma albumin concentra- reabsorbed by the same transporters that normally reabsorb
tion (5 g/dL). Because of the huge volume of fluid filtered per filtered amino acids.
day, the total filtered amount of protein is not negligible. Nor- Finally, in certain types of renal damage, proteins released
mally all of these proteins and peptides are reabsorbed com- from damaged tubular cells rather than filtered at the renal
pletely, although not in the conventional way. They are corpuscles may appear in the urine and provide important
enzymatically degraded into their constituent amino acids, diagnostic information.
which are then returned to the blood.
For the larger proteins, the initial step in recovery is endocy-
tosis at the apical membrane. This energy-requiring process is PROXIMAL SECRETION
triggered by the binding of filtered protein molecules to spe-
cific receptors on the apical membrane. The rate of endocyto- OF ORGANIC ANIONS
sis is increased in proportion to the concentration of protein So far we have described reabsorption of useful organic sub-
in the glomerular filtrate until a maximal rate of vesicle forma- stances the body does not normally excrete. There are of course
tion, and thus the Tm for protein uptake, is reached. The many organic anions that it does excrete, both endogenously
pinched-off intracellular vesicles resulting from endocytosis produced and foreign (see Table 43–1 for a partial listing).
merge with lysosomes, whose enzymes degrade the protein to Many of these organic anions are filterable at the renal cor-
low-molecular-weight fragments, mainly individual amino puscles, with proximal secretion adding to the amount filtered.
acids. These end products then exit the cells across the baso- Others, however, are extensively bound to plasma proteins and
lateral membrane into the interstitial fluid, from which they undergo glomerular filtration only to a limited extent; accord-
gain entry to the peritubular capillaries. ingly, proximal tubular secretion constitutes the only signifi-
To understand the potential problem associated with a fail- cant mechanism for their excretion.
ure to take up filtered protein, remember that for a healthy The active secretory pathway for organic anions in the prox-
young adult: imal tubule in some sense is the reverse of reabsorption of
organic solutes: there are active transporters for the anions at
Total filtered protein
the basolateral membrane of tubular epithelial cells that are
= GFR × concentration of protein in filtrate (1)
the rate-limiting step in overall transport. Transport out of the
= 180 L per day × 10 mg/L = 1.8g per day
cell across the apical membrane into the lumen uses a variety
of uniporters or more specific sodium-dependent antiporters.
If this protein was not removed from the lumen, the entire Because the basolateral membrane of proximal convoluted
1.8 g would be lost in the urine. In fact, most of the filtered tubule epithelial cells contains all of these different transport-
protein is endocytosed and degraded so that the excretion of
protein in the urine is normally only 100 mg per day. The
endocytic mechanism by which protein is taken up is easily TABLE 43–1 Some organic anions actively secreted
saturated, so a large increase in filtered protein resulting from by the proximal tubule.
increased glomerular permeability causes the excretion of
large quantities of protein. Endogenous Substances Drugs
Discussions of the renal handling of protein logically tend to Bile salts Acetazolamide
focus on albumin because it is by far the most abundant plasma
protein. There are, of course, many other plasma proteins. Fatty acids Chlorothiazide
Although present in lower levels than albumin, they are smaller Hippurates Ethacrynate
and thus more easily filtered. For example, growth hormone Hydroxybenzoates Furosemide
(molecular weight, 22,000 d) is approximately 60% filterable,
and insulin is 100% filterable. The total mass of these filtered Oxalate Penicillin
hormones is insignificant; however, because even tiny levels in Prostaglandins Probenecid
the plasma have important signaling functions in the body, Urate Saccharin
renal filtration becomes an important influence on concentra-
tions in the blood. Relatively large fractions of these smaller Salicylates
plasma proteins are filtered and then degraded in tubular cells. Sulfonamides
The kidneys are major sites of catabolism of many plasma pro- Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
teins, including polypeptide hormones. Decreased rates of 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
432 SECTION VII Renal Physiology

ers, the proximal tubule has the capacity to secrete all the Although urate reabsorption is greater than secretion, the
organic anions listed in Table 43–1 and many more. Like glu- secretory process is controlled to maintain relative constancy
cose, organic anions are not significantly permeable through of plasma urate. In other words, if plasma urate begins to
tight junctions or lipid membranes, so that their transport is increase because of increased urate production, the active
also characterized by a Tm. If the blood concentration of an proximal secretion of urate is stimulated, thereby increasing
organic anion is too high, it will not be efficiently removed urate excretion.
from the blood by the kidneys. The relatively nondiscriminat- Given these mechanisms of renal urate handling, the reader
ing nature of this collection of transporters accounts for their should be able to deduce the three ways by which altered renal
ability to eliminate so many drugs and other foreign environ- function can lead to decreased urate excretion and hence
mental chemicals from the body. In this regard, the liver’s increased plasma urate, as in gout: (1) decreased filtration of
metabolic transformations are very important. In the liver, urate secondary to decreased GFR, (2) excessive reabsorption
many foreign (and endogenous) substances are conjugated of urate, and (3) diminished secretion of urate.
with either glucuronate or sulfate. The addition of these groups
renders the parent molecule far more water soluble. These two
types of conjugates are actively transported by the organic PROXIMAL SECRETION
anion secretory pathway.
The most intensively studied organic anion secreted by this OF ORGANIC CATIONS
pathway is para-aminohippurate (PAH), the substance used
The proximal tubules possess several closely related transport
for the measurement of effective renal plasma flow (see
systems for organic cations that are analogous to those for
Chapter 41). PAH secretion involves a pair of antiporters, one
organic anions. Because of the large number of different trans-
at each membrane. At the basolateral membrane, PAH is taken
porters, a substantial amount of foreign and endogenous
up in exchange for the anion (base) form of a dicarboxylic
organic cations is transported (Table 43–2). The cations com-
acid. PAH is extruded into the lumen across the apical mem-
pete with one another for transport, and the transporters man-
brane via another antiporter.
ifest Tm limitation. Organic cations enter across the basolateral
As the plasma concentration of an anion secreted by this
membrane via one of the several uniporters, members of the
system increases, so does the rate of secretion (until the Tm for
OCT (organic cation transporter) family, and exit into
that substance is reached). This provides a mechanism for
the lumen via an antiporter, which exchanges a proton for the
regulating the endogenous organic anions handled by the sys-
organic cation.
tem and for speeding the excretion of foreign organic anions.
The proximal secretion of organic cations, as for organic
PAH is typical, in yet another way, of many of the organic
anions, is particularly critical for the excretion of those cations
anions secreted proximally. It undergoes no significant trans-
extensively bound to plasma proteins and not filterable at the
port anywhere else along the nephron. In contrast, some of the
renal corpuscle. However, again similar to organic anions,
other organic anions secreted by the proximal tubule can also
many of the organic cations secreted by the proximal tubules
undergo other forms of transport in both the proximal tubule
and more distal segments. The most important of these is pas-
sive tubular reabsorption or secretion, which is described later.

TABLE 43–2 Some organic cations actively secreted


URATE by the proximal tubule.

Endogenous Substances Drugs


Urate (the base form of uric acid) provides a fascinating
example of the renal handling of organic anions that is partic- Acetylcholine Atropine
ularly important for clinical medicine and is illustrative of Choline Isoproterenol
renal pathology. An increase in the plasma concentration of
urate can cause gout; therefore, its removal from the blood is Creatinine Cimetidine
important. However, it is as though the kidney cannot make Dopamine Meperidine
up its mind what to do with urate. Urate is not protein bound Epinephrine Morphine
and so is freely filterable. Almost all the filtered urate is reab-
sorbed early in the proximal tubule; however, further on in the Guanidine Procaine
proximal tubule, urate undergoes active tubular secretion. Histamine Quinine
Then, in the straight portion, some of the urate is once again Serotonin Tetraethyl ammonium
reabsorbed. The total rate of tubular reabsorption is normally
much greater than the rate of tubular secretion, so the mass of Norepinephrine
urate excreted per unit time is only a small fraction of the mass Thiamine
filtered. Most of this transport involves antiporters that Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
exchange urate for another organic anion. 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
CHAPTER 43 Renal Handling of Organic Substances 433

are not protein bound and, therefore, also undergo glomerular increase passive reabsorption of weak acids (and promote
filtration and tubular secretion; creatinine is a good example. less excretion). For many weak bases, the pH dependence is
Finally, and again analogous to organic anions, some organic just the opposite. At low pH they are protonated cations
cations not only are actively secreted by the proximal tubules, (trapped in the lumen). As the urine becomes acidified, more
but may also undergo other forms of tubular handling, mainly is converted to the impermeable charged form and is trapped
passive reabsorption or secretion. in the lumen. Less is reabsorbed passively, and more is
excreted.
Having said this, what difference does it make? Because so
many medically useful drugs are weak organic acids and bases,
pH DEPENDENCE OF PASSIVE all these factors have important clinical implications. For
REABSORPTION OR SECRETION example, if one wishes to enhance the excretion of a drug that
is a weak acid, one attempts to alkalinize the urine (because
Many substances handled by the kidney are weak acids or this traps the ionic form in the lumen). In contrast, acidifica-
bases. At a given pH, some are in the neutral form and some tion of the urine is desirable if one wishes to prevent excretion
are in the ionized form. The state of ionization affects both of the drug. Of course, exactly the opposite applies to weak
the aqueous solubility and membrane permeability of the organic bases. At any luminal fluid pH, increasing the urine
substance. In most cases, the neutral forms of organic acids flow increases the excretion of both weak acids and bases
and bases are more permeable in lipid membranes than the (Figure 43–2). Finally, excretion can be reduced by adminis-
ionized forms. The neutral forms, being permeable, can tering another drug that interferes with any active proximal
equilibrate with the interstitial levels, while the ionized forms, secretory pathway for the drug.
once in the lumen, are effectively trapped there. Many weak
acids are predominantly neutral at low pH (acid form) and
are dissociated into an anion and a proton at higher pH. The
lower the pH, the greater the amount in the neutral acid form.
UREA
Imagine the case in which the tubular fluid becomes acidified Urea is a very special substance for the kidney. It is an end
relative to the plasma, which it does on a typical Western diet. product of protein metabolism, waste to be excreted, and also
For a weak acid in the tubular fluid, relatively more will be a useful tool for the regulation of water excretion.
converted to the neutral form and, therefore, become more Excess dietary protein not needed for tissue synthesis is
permeable. This favors diffusion out of the lumen (reabsorp- either oxidized or converted to fat and stored for later oxida-
tion). Therefore, highly acidic urine (low pH) tends to tion. During fasting the body breaks down the fat and protein

TUBULAR RENAL TUBULAR PERITUBULAR


LUMEN CELLS PLASMA
(Filtered)
HA A

H H

HA HA A

(Filtered)
BH B FIGURE 43–2 pH dependence of passive reabsorption
or secretion. Acidification of the urine favors reabsorption,
H H andtherefore retention, of weak acids because the neutral,
protonated forms can passively diffuse out of the tubule (top
dashed line). At the same time, acidification favors secretion
B BH (and therefore loss) of weak bases because the protonated
BH (Impermeable)
forms are charged and trapped in the lumen, and the neutral,
 nonprotonated forms can passively diffuse into the tubule
B (bottom dashed line). The processes that acidify the urine are
described in Chapter 47. (Reproduced with permission from Eaton DC,
Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical
Books/McGraw-Hill, Medical Pub. Division, 2009.)
434 SECTION VII Renal Physiology

for fuel, in essence consuming itself. When oxidized for fuel, 110%
protein is first split into its constituent amino acids. These are 5.5
then separated into a nitrogen moiety (ammonium) and a car-
bohydrate moiety. The carbohydrate goes on to further meta-
bolic processing, but the ammonium cannot be further
oxidized and is a waste product. The problem is that ammo-
nium is rather toxic to most tissues (except the medullary
interstitium), and the liver immediately converts most ammo-
nium to urea and a smaller, but crucial amount to glutamine.
(We will take up the fate of this glutamine in Chapter 47.)
100%
Whether a person is well fed or fasting, urea production pro- 1
ceeds continuously. In fact, it constitutes about half of the nor-
mal solute content of urine; thus, it is a crucial player in
osmotic considerations.
The normal level of urea in the blood is quite variable 50%
(3–9 mmol/L), reflecting variations in both protein intake 1.2
and renal handling of urea. (For ease in converting to osmo-
lality, we express urea concentration in units of mmol/L
rather than the clinical unit of blood urea nitrogen [BUN].)
Over days to weeks, renal urea excretion must match hepatic
production; otherwise, plasma levels would increase into
the pathological range, producing a condition called
uremia. On a shorter-term basis (hours to days), urea
excretion rate may not exactly match production rate
because urea excretion is also regulated for purposes other
than keeping a stable plasma level. As discussed in the next
chapter, urea is a key solute involved in regulating excretion
of water.
The gist of the renal handling of urea is the following: it is 50%
25
freely filtered. About half is reabsorbed in the proximal
tubule. Then an amount equal to that reabsorbed is secreted FIGURE 43–3 Urea handling by the kidney. The arrows
back into the loop of Henle. Finally, about half is reabsorbed indicate that urea is reabsorbed in the proximal tubule, secreted in
a second time in the medullary collecting duct. The net the thin portions of the loop of Henle, and reabsorbed again in the
result is that about half the filtered load is excreted medullary collecting ducts. The top halves of boxes indicate the
(Figure 43–3). percentage of the filtered load remaining in the tubule at a given
As a molecule, urea is small (molecular weight, 60 d), water location, and the bottom halves indicate tubular concentration
soluble, and freely filtered. Because of its highly polar nature, relative to plasma. Note that while the amount remaining in the
it does not permeate lipid bilayers, but a set of uniporters collecting duct (and thus excreted) is half the amount filtered,
the concentration is much higher than that in plasma because
(UT family) transports urea in various places in the kidney
most of the water has been reabsorbed. These numbers are highly
and in other sites within the body (particularly red blood
variable, depending on several factors, particularly the hydration
cells). Because urea is freely filtered, the filtrate contains urea status. (Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal
at a concentration identical to that in plasma. Let us assume Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical
a normal plasma level (5 mmol/L). Roughly half the filtered Pub. Division, 2009.)
load is reabsorbed in the proximal tubule. This occurs pri-
marily by the paracellular route. As water is reabsorbed, the
urea concentration increases, driving diffusion through the all the way down to the papilla. For simplicity we consider all
leaky tight junctions. By the time the tubular fluid enters the nephrons together.
loop of Henle, about half the filtered urea has been reab- The interstitium of the medulla has a considerably higher
sorbed, and the urea concentration has increased somewhat urea concentration than does plasma. The concentration
above its level in the filtrate because proportionally more increases from the outer to the inner medulla (part of the so-
water than urea was reabsorbed. At this point, the process called medullary osmotic gradient that we describe in the next
becomes fairly complicated. First, conditions in the medulla chapter). Since the medullary interstitial urea concentration is
depend highly on the individual’s state of hydration. Second, greater than that in the tubular fluid entering the loop of
there is a difference between superficial nephrons, with short Henle, there is a concentration gradient favoring secretion into
loops of Henle that only penetrate the outer medulla, and the lumen. The tight junctions in the loop of Henle are no lon-
juxtamedullary nephrons, with long loops of Henle that reach ger permeable (as they were in the cortex), but the epithelial
CHAPTER 43 Renal Handling of Organic Substances 435

membranes of the thin regions of the Henle loops express urea


through a strainer. Sure enough, she indeed passed a stone
uniporters, members of the UT family. This permits secretion
and brought it to her primary care physician for labora-
of urea into the tubule. In fact, the urea secreted from the
tory analysis. She had never had a stone before. She is
medullary interstitium into the thin regions of the loop of
moderately obese and mildly hypertensive. Previous labo-
Henle replaces the urea previously reabsorbed in the proximal
ratory analysis showed that she has increased plasma
tubule. Thus, when tubular fluid enters the thick ascending
triglycerides. She may be becoming diabetic, and to avoid
limb, the amount of urea in the lumen is at least as large as the
carbohydrate she eats a large amount of seafood and
filtered load (the loop of Henle has essentially reversed what
chicken, but not many vegetables. The stone proved to be
was accomplished in the proximal tubule). However, because
a uric acid stone, which accounts for only about 7% of all
about 80% of the filtered water has now been reabsorbed, the
kidney stones. What might predispose her to develop uric
luminal urea concentration is now several times greater than
acid stones?
that in the plasma. Beginning with the thick ascending limb
Uric acid has a pK of 5.75. In plasma and in all but very
and continuing all the way to the medullary collecting ducts
acidic urine, the vast majority of uric acid exists as the urate
(through the distal tubule and cortical collecting ducts), the
anion, which is the form recognized by renal transporters.
apical membrane urea permeability (and the tight junction
The neutral acid has a lower solubility than the anionic
permeability) is essentially zero. Therefore, an amount of urea
urate. Therefore, increased urinary content of urate, and
roughly equal to the filtered load remains within the tubular
particularly low urinary pH, which converts much of the
lumen and flows from the cortical into the medullary collect-
urate to uric acid, leads to much more urinary uric acid and
ing ducts.
increased likelihood of forming stones. Her diet of fish and
During the transit through the cortical collecting ducts,
chicken has a high content of purines, the precursors of
variable amounts of water are reabsorbed, further concentrat-
uric acid. Therefore, because of the high metabolic produc-
ing the urea. Just how much depends on factors discussed in
tion and low urinary pH, her urine contains high amounts
the next chapter, but the luminal urea concentration can easily
of the rather insoluble uric acid. To complicate the matter,
reach 50 times that of plasma. We indicated earlier that the
her profile fits that of a person with metabolic syndrome, a
urea concentration in the medullary interstitium is greater
cluster of conditions that includes truncal obesity, hyper-
than that in plasma, but the luminal concentration in the
tension, hyperlipidemia, and insulin resistance that will be
medullary collecting ducts is even higher, so in the inner
discussed in Chapter 69. For reasons that are still being
medulla the gradient now favors reabsorption, and urea is
unraveled, patients with metabolic syndrome often have
reabsorbed a second time. It is this reabsorbed urea that leads
decreased urinary ammonium. Because ammonium is a
to the high medullary interstitial concentration that drives
major buffer on which hydrogen ions are excreted (see
urea secretion into the thin regions of the loop of Henle. In
Chapter 47), low ammonium production in the face of nor-
fact, some of the urea recycles, that is, it is reabsorbed from
mal acid loads leads to decreased urinary pH. The urinary
medullary collecting ducts and secreted into thin limbs of the
pH was indeed very low. The stone formation is based pri-
loop of Henle, from where it travels within the tubule to the
marily on her low urinary pH, which, at all pH values less
collecting ducts again to repeat the process. The overall result
than 5.75, converts most of the urate to the less soluble uric
of these events is that half the original amount of filtered urea
acid. The patient was counseled to include more vegetables
passes into the final urine, an amount that, over the long term,
in her diet and consume more water. These measures
must match hepatic production of urea if the body is to remain
should increase her urinary pH and lower the urate con-
in balance for urea. The concentration of urea in the final
centration, and therefore decrease the concentration of uric
urine can be more than 50× that in plasma, depending on how
acid.
much water is reabsorbed. These processes are summarized in
Figure 43–3.

CHAPTER SUMMARY
CLINICAL CORRELATION ■ Important organic metabolites are almost completely reab-
sorbed (saved), whereas waste products are for the most part
A 41-year-old mother of five children presented at a hos- excreted.
pital ER with severe right flank pain. The pain was present ■ Most organic solutes are transported transcellularly by a large
all the time, and was accentuated in waves. A urine speci- number of different saturable multiporters (Tm systems).
men was pink, with a pH of 5.1. The ER personnel sus- ■ Normal filtered loads of glucose are completely reabsorbed,
pected that she had a kidney stone located in the right but in conditions of pathological hyperglycemia the trans-
ureter. Accordingly they gave her large amounts of fluid port saturates, leading to the appearance of glucose in the
and nonsteroidal anti-inflammatories (NSAIDS) to help urine.
ease the pain. After several hours the pain eased, and she ■ Urea is reabsorbed proximally and recycled between the
was sent home with instructions to pass all her urine collecting ducts and loop of Henle in the medulla, resulting
in a net excretion of about half the filtered load.
436 SECTION VII Renal Physiology

STUDY QUESTIONS 4. A high urinary pH favors


A) low excretion of drugs that are weak acids.
1. When plasma glucose reaches such high levels that substantial B) active reabsorption of drugs that are weak bases.
amounts of glucose appear in the urine (glucosuria) C) low excretion of drugs that are weak bases.
A) glucose is leaking back into the tubule through tight D) high passive permeability of drugs that are weak acids.
junctions. 5. The tubular concentration of urea
B) there is not enough luminal sodium to move in symport
A) exceeds the plasma concentration at the hairpin turn of the
with glucose.
loop of Henle.
C) all the glucose transporters are working at their maximum rate.
B) decreases to below the plasma concentration by the end of
D) the glucose transporters are being inhibited by the high
the loop of Henle.
levels of glucose.
C) decreases to below the plasma concentration by the end of
2. Useful small organic metabolites that should not be excreted are the proximal tubule.
A) generally not filtered. D) reaches its highest value in the cortical collecting duct.
B) reabsorbed paracellularly. 6. Urea is secreted into the tubules
C) taken up by endocytosis and degraded.
A) in proximal tubules.
D) reabsorbed transcellularly.
B) in Henle’s loop.
3. Organic anion secretion C) in medullary collecting ducts.
A) involves a step of active influx across the basolateral D) at any of the above sites depending on hydration status.
membrane.
B) is passive and paracellular.
C) is via simple diffusion through the tubular membranes.
D) utilizes the same membrane transporters as organic cation
secretion.
44
C H A P T E R

Basic Renal Processes


for Sodium, Chloride,
and Water
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ List approximate percentages of sodium reabsorbed in major


tubular segments.
■ List approximate percentages of water reabsorbed in major tubular
segments.
■ Describe proximal tubule sodium reabsorption, including the functions of the
apical membrane sodium entry mechanisms and the basolateral Na,K-ATPase.
■ Explain why chloride reabsorption is coupled with sodium reabsorption, and
list the major pathways of proximal tubule chloride reabsorption.
■ State the maximum and minimum values of urine osmolality.
■ Define osmotic diuresis and water diuresis.
■ Explain why there is always an obligatory water loss.
■ Describe the handling of sodium by the descending and ascending limbs,
distal tubule, and collecting duct system.
■ Describe the role of sodium–potassium–2 chloride symporters in the thick
ascending limb.
■ Describe the handling of water by descending and ascending limbs, distal
tubule, and collecting duct system.
■ Describe the process of “separating salt from water,” and discuss why this is
required to excrete either concentrated or dilute urine.
■ Describe how antidiuretic hormone affects water reabsorption.
■ Describe the characteristics of the medullary osmotic gradient.
■ Explain the role of the thick ascending limb, urea recycling, and medullary
blood flow in generating the medullary osmotic gradient.
■ State why the medullary osmotic gradient is partially “washed out” during
a water diuresis.

OVERVIEW The remaining one third of the osmotic content and water is
called the extracellular fluid (ECF). It is mostly accounted for
BODY FLUID COMPARTMENTS by interstitial fluid and blood plasma. Because of the ease with
which water crosses cell membranes (see Chapter 3), the ECF
Body water (about 60% of body weight) is distributed into and ICF are in osmotic equilibrium. Changes in body water
various aqueous spaces in proportion to their osmotic content. affect the volume of one or both compartments, depending on
The collective volume of all the cells in the body is called the how much solute, and what kind, accompanies the water. In
intracellular fluid (ICF). It contains roughly two thirds of the contrast, additions or losses of sodium from the body are
body osmotic content, and therefore two thirds of the water. almost exclusively to or from the ECF only because the actions

437

Ch44_437-448.indd 437 11/26/10 10:29:16 AM


438 SECTION VII Renal Physiology

Plasma
Interstitial
Normal
ECF ICF

Plasma
Add Interstitial
water
ECF ICF

Plasma
Add Interstitial
isotonic
saline ECF ICF

FIGURE 44–1 Distribution of total body water into

Plasma
the intracellular (ICF) and extracellular (ECF) Interstitial
compartments. As explained in the text, addition of water, Add salt
salt, or both alters the volumes of the compartments.
Expanded volumes are indicated by the dashed lines. ECF ICF

of cellular Na,K-ATPases keep intracellular sodium nearly


TABLE 44–1 Normal routes of sodium intake and loss.
constant. The effects of adding sodium and water are depicted
in Figure 44–1. The addition of water alone expands both the Route Amount (g per Day)
ICF and ECF (indicated by the dashed lines). Adding isotonic
Intake
sodium chloride expands only the ECF because the added
osmoles remain in the ECF. Adding sodium chloride without Food 10.5
water does not change the total volume, but causes a shift of Output
water from the ICF to the ECF in order to restore equality of
Sweat 0.25
osmolality between the two compartments.
The transport of water by the renal tubules is straightfor- Feces 0.25
ward—“water follows the osmoles”—so much of the descrip- Urine 10.00
tion of water transport really amounts to describing solute
Total output 10.50
transport, recognizing that in some regions of the kidney, low
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
water permeability limits the amount of water that follows the 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
osmoles. Transport of chloride is more complicated, but
because of the constraints of electroneutrality, it is tied to the
transport of sodium. Sodium transport is clearly the crux of
and chloride are reabsorbed. Table 44–2 summarizes the
the matter because the transport of chloride, water, and many
approximate quantitative contribution of each tubular seg-
other substances is linked to the transport of sodium.
ment to sodium reabsorption. In an individual with an average
The excretory rates of sodium, chloride, and water vary over
salt intake, the proximal tubule reabsorbs 65% of the filtered
an extremely wide range depending on diet. For example, some
sodium, the thin and thick ascending limbs of Henle’s loop
persons may ingest 20–25 g of sodium chloride per day, whereas
25%, and the distal convoluted tubule and collecting duct
a person on a low-salt diet may ingest only 0.05 g. The healthy
system most of the remaining 10%, so that the final urine con-
kidney can readily alter its excretion of salt over this range.
tains less than 1% of the total filtered sodium. As will be
Similarly, urinary water excretion can be varied physiologically
discussed in Chapter 45, reabsorption at several of these tubu-
from approximately 0.4 to 25 L per day, depending on whether
lar sites is under physiological control by neural, hormonal,
one is lost in the desert or maximizing one’s water intake.
and paracrine signals, so that the exact amount of sodium
excreted is regulated. Because so much sodium is filtered, even
SODIUM REABSORPTION a small percentage change in reabsorption results in a rela-
tively large change in excretion.
Table 44–1 is a balance sheet for sodium chloride. The major In all nephron segments, the essential event is the primary
route of salt excretion from the body under normal circum- active transport of sodium from cell to interstitial fluid by the
stances is via the kidneys. The large amount excreted should Na,K-ATPase pumps in the basolateral membrane. These
not obscure the fact that nearly all the filtered loads of sodium pumps keep the intracellular sodium concentration lower than
CHAPTER 44 Basic Renal Processes for Sodium, Chloride, and Water 439

TABLE 44–2 Comparison of sodium and water TABLE 44–3 Normal routes of water gain and loss
reabsorption along the tubule. in adults.

Tubular Segment Filtered Load Reabsorbed (%) Route Amount (mL per Day)

Sodium Water Intake

Proximal tubule 65 65 Beverage 1,200

Descending thin limb of — 10 Food 1,000


Henle’s loop
Metabolically produced 350
Ascending thin limb and 25 —
thick ascending limb of Total 2,550
Henle’s loop Output
Distal convoluted tubule 5 — Insensible loss (skin and lungs) 900
Collecting duct system 4–5 5 (during water Sweat 50
loading)
>24 (during In feces 100
dehydration)
Urine 1,500
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009. Total 2,550
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
that in the surrounding media. Because the inside of the cell is
negatively charged with respect to the lumen, luminal sodium
ions enter the cell passively, down their electrochemical gradi- WATER REABSORPTION
ent. The entry paths are far more numerous than the exit paths.
We will examine these entry paths more closely as we go A balance sheet for total body water is given in Table 44–3.
through the tubule segment by segment. These are average values, which are subject to considerable
variation. The two sources of body water are metabolically
produced water, resulting largely from the oxidation of carbo-
CHLORIDE REABSORPTION hydrates, and ingested water, obtained mostly from liquids,
but also from so-called solid food (e.g., a rare steak is approxi-
Because chloride reabsorption is dependent on sodium reab- mately 70% water). There are several sites from which water is
sorption, the tubular locations that reabsorb chloride and the always lost to the external environment: skin, lungs, gastroin-
percentages of filtered chloride reabsorbed by these segments testinal tract, and kidneys. Menstrual flow and, in lactating
are similar to those for sodium (see Table 44–1). When exam- women, breast milk constitute two other potential sources of
ining chloride reabsorption, it is helpful to keep in mind the water loss in women.
absolute constraint of electroneutrality: any finite volume of The loss of water by evaporation from the cells of the skin
fluid reabsorbed must contain equal amounts of anion and and the lining of respiratory passageways is a continuous pro-
cation equivalents. Let us do a “guesstimate” calculation. One cess, often referred to as insensible loss because people are
liter of normal filtrate contains 140 mEq of sodium, and thus unaware of its occurrence. Additional water evaporates from
must contain about 140 mEq of anions, mainly chloride (110 the skin during production of sweat. Fecal water loss is nor-
mEq) and bicarbonate (24 mEq). (We say “about” because mally quite small but can be severe in diarrhea. Gastrointesti-
there are other cations [e.g., potassium and calcium] and nal loss can also be large during severe bouts of vomiting.
anions [e.g., sulfate and phosphate] that must factor into the Under conditions of normal hydration, the kidneys are, of
calculation to achieve an exact balance, but their contributions course, the main route of water loss.
are much smaller than sodium, chloride, and bicarbonate.) If With a large water load, the renal response is to produce a
65% of the filtered sodium is reabsorbed in the proximal large volume of very dilute urine (osmolality much lower than
tubule, then 0.65 × 140 = 91 mEq of sodium in each liter of that in blood plasma). In contrast, during a state of dehydration,
filtrate is reabsorbed. Therefore, 91 mEq of some combination the urine volume is low and very concentrated (i.e., the urine
of chloride and bicarbonate must also be reabsorbed to accom- osmolality is much greater than that in blood plasma). That the
pany this sodium. As will be described in Chapter 47, about urine osmolality is so variable brings us to a crucial aspect of
90% of the filtered bicarbonate is reabsorbed in the proximal renal function. Terrestrial animals must be able to indepen-
tubule (0.9 × 24 ≈ 22). This leaves 91 – 22 = 69 mEq of chloride dently control excretion of salt and water, because their inges-
that must be reabsorbed in the proximal tubule. This is more tion and loss is not always linked (see Tables 44–1 and 44–3). To
than 60% of the 110 mEq of filtered chloride and almost as excrete water in excess of salt and vice versa (i.e., produce a
much as the fractional reabsorption of sodium and water. range of urine osmolalities), the kidneys must be able to sepa-
Later segments reabsorb almost all of the remaining 40%. rate the reabsorption of solute from the reabsorption of water,
440 SECTION VII Renal Physiology

that is, to “separate salt from water.” How do they do this? LUMEN INTERSTITIUM
Regardless of hydration state, the proximal tubule reabsorbs
water and solute in equal proportions (isosmotic reabsorption),
but the loop of Henle reabsorbs proportionally more solute than
Na+
water. This is a key step in the separation process—reabsorption ATP
HCO3 H+ H+ Na+
of solute, leaving water in the tubule. By the time tubular fluid
Hbase K+
leaves the loop of Henle and enters the distal tubule, the loss of CO2 + H2O
solute has typically decreased the osmolality to about 110 base– base– HCO3
mOsm/kg H2O. If an individual is overhydrated and, therefore, Cl– Na+
requires maximum water excretion, most of this dilute fluid H2O H2O
simply passes through the collecting duct system to appear in Glucose, Cl–
the urine, with only limited further water reabsorption. In con- Na+
phosphate,
trast, when an individual is dehydrated, the vast majority of this amino acids,
dilute water is reabsorbed in the collecting ducts, leaving a low organics etc
H2O, Cl– H2O, Cl–
volume of concentrated final urine.
The ability of the kidneys to produce low-volume hyperos-
motic urine is a major determinant of one’s ability to survive FIGURE 44–2 Major pathways for reabsorption of sodium,
chloride, and water in the proximal tubule. The entire proximal
without water, which for most people is at least several days,
tubule is the major site for reabsorption of salt and water. Sodium entry
and longer if conditions are right. The human kidney can pro- is coupled to the secretion or uptake of a variety of substances, the
duce a maximal urinary concentration of 1,400 mOsm/kg in major one being hydrogen ions, which are secreted in exchange for
extreme dehydration. This is almost five times the osmolality sodium via the NHE3 antiporter. These hydrogen ions, once in the
of plasma. The sum of the urea, sulfate, phosphate, other waste lumen, combine with filtered bicarbonate and secreted organic base
products, and a small number of nonwaste ions excreted each (see text and Chapter 47 for further explanation). Additional sodium
day normally averages approximately 600 mOsm per day. enters in symport with glucose, amino acids, and phosphate. Sodium is
These solutes continue to be excreted even in severe dehydra- transported to the interstitium mostly via the basolateral Na,K-ATPase,
tion; therefore, the minimal volume of water in which this but also in symport with bicarbonate. (The coupling between sodium
mass of solute can be dissolved is roughly 600 mOsm/1,400 and bicarbonate is described fully in Chapter 47.) Chloride that enters
in antiport with organic base leaves mostly via channels. In addition, a
mOsm/L = 0.43 L per day.
substantial amount of chloride is reabsorbed paracellularly. Water
This volume of urine is known as the obligatory water loss.
moves both paracellularly and intracellularly via aquaporins. (Modified
It is not a strictly fixed volume but changes with different phys- with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York,
iological states. For example, increased tissue catabolism, as NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
during fasting or trauma, releases excess solute and so increases
obligatory water loss. The obligatory water loss contributes to
dehydration when a person is deprived of water intake. The
water, cause the secondary active reabsorption of filtered bicar-
obligatory solute loss also explains why a thirsty sailor cannot
bonate. Therefore, in the early proximal tubule, bicarbonate is a
drink seawater for hydration. To excrete all the salt in the sea-
major anion reabsorbed with sodium and the luminal bicarbonate
water plus the obligatory solute would require more urinary
concentration decreases markedly (Figure 44–3). Organic nutri-
water than was contained in the seawater consumed.
ents and phosphate are also absorbed with sodium, and their lumi-
nal concentrations also decrease rapidly.
A major percentage of chloride reabsorption in the proxi-
INDIVIDUAL TUBULAR SEGMENTS mal tubule occurs via paracellular diffusion. The concentra-
tion of chloride in Bowman’s capsule is, of course, essentially
The important principles regarding individual tubular seg-
the same as it is in plasma (about 110 mEq/L). Along the early
ments are how the reabsorption of sodium, chloride, and water
proximal tubule, the reabsorption of water causes the chloride
is related to one another and how the amount of reabsorption
concentration in the tubular lumen to increase somewhat
varies quantitatively from one segment to another.
above that in the peritubular capillaries (see Figure 44–3).
Then, as the fluid flows through the middle and late proximal
PROXIMAL TUBULE tubules, this concentration gradient, maintained by continued
water reabsorption, provides the driving force for paracellular
As shown in Figure 44–2, several apical membrane entry pathways chloride reabsorption by diffusion.
are involved in the active transcellular reabsorption of sodium in There is also an important component of active chloride
the proximal tubule. In the early portion, a large fraction of the transport from lumen to cell in the later proximal tubule. As
filtered sodium enters the cell across the apical membrane via anti- illustrated in Figure 44–2, it uses parallel Na–H and Cl–base
port with protons. As will be described in Chapter 47, these pro- antiporters. Chloride is actively transported into the cell in
tons, which are released when carbon dioxide combines with exchange for the downhill antiport (secretion) of small organic
CHAPTER 44 Basic Renal Processes for Sodium, Chloride, and Water 441

1.2 Cl−

Na+
1.0
osmolarity

FIGURE 44–3 Changes in tubular fluid composition along the


0.8 proximal convoluted tubule, expressed as the ratio of concentration in
TF/P

the tubule relative to the plasma value. Values above 1.0 indicate that
Pi
0.6 relatively less of the substance than water has been reabsorbed, and
therefore its concentration increases. Values below 1.0 indicate that
relatively more of the substance than water has been reabsorbed. Inorganic
0.4 phosphate, bicarbonate, glucose, and lactate are preferentially reabsorbed
with sodium early in the proximal tubule, and their concentrations rapidly
− fall. In contrast, the concentration of chloride increases somewhat because
0.2 HCO3
Amino acids, chloride reabsorption lags behind sodium and, hence, water reabsorption
glucose, lactate in the early proximal tubule. The concentration of sodium and the total
concentration of all solutes (osmolarity) remains nearly the same as in
2 4 6 plasma. (Modified with permission from Rector FC. Sodium, bicarbonate, and chloride
Distance from Bowman's space (mm) absorption by the proximal tubule. Am J Physiol. 1983;244(5):F461–F471.)

base anions. These include formate and oxalate, which are incompletely or not at all by the proximal tubule (e.g., infused
continuously generated in the cell by dissociation of their mannitol, a monosaccharide that is not transported). As water
respective uncharged acids into a proton and the base. Simul- is reabsorbed in the proximal tubule, the concentration of any
taneously, the protons released within the cell by the dissocia- unusual unreabsorbed solute rises, and its osmotic presence
tion of those acids are actively transported into the lumen by retards the further reabsorption of water (here and downstream
the Na–H antiporters. In the lumen, the protons and organic as well). The failure of water to follow the sodium being
bases recombine to form the neutral acids, which then diffuse removed from the lumen means that the sodium concentration
across the apical membrane back into the cell, where the in the proximal tubular lumen decreases slightly below that in
entire process is repeated. Notice that both the protons and the interstitial fluid. This concentration difference, even though
the organic bases endlessly recycle, moving into the cells while small, drives a passive diffusion of sodium across the epithe-
paired as a neutral molecule and then move out via separate lium (mostly the tight junctions) back into the lumen, that is,
transporters after the proton dissociates. The overall achieve- sodium transport reaches the gradient limit we described in
ment of the parallel Na–H and Cl–base antiporters is the Chapter 42. The result is that more than the usual amount of
same as though the Cl and Na were simply cotransported into
the cell together. Importantly, the recycling of protons and
bases means that most of the protons are not acidifying the
lumen but are simply combining with base and moving back
TABLE 44–4 Summary of mechanisms by which
into the cells. It should also be recognized that everything reabsorption of sodium drives reabsorption of other
ultimately depends on the basolateral membrane Na,K- substances in the proximal tubule.
ATPases to establish the gradient for sodium that powers the
Reabsorption of sodium
apical Na–H antiporter.
Regarding water reabsorption, the proximal tubule, as men- 1. Creates transtubular osmolality difference, which favors
reabsorption of water by osmosis; in turn, water reabsorption
tioned, has a very high permeability to water, allowing very
concentrates many luminal solutes (e.g., chloride and urea), thereby
small differences in osmolality (less than 1 mOsm/L) to drive favoring their reabsorption by diffusion
the reabsorption of very large quantities of water, normally
2. Achieves reabsorption of many organic nutrients, phosphate, and
about 65% of the filtered water. This osmolality difference is sulfate by cotransport across the luminal membrane
created by the reabsorption of sodium and the various solutes
linked directly or indirectly with sodium (Table 44–4). 3. Achieves secretion of hydrogen ion by countertransport across the
luminal membrane; these hydrogen ions are required for
If the tight coupling between proximal sodium and water reabsorption of bicarbonate (as described in Chapter 47)
reabsorption is disrupted, we have a phenomenon known as
4. Achieves reabsorption of chloride by indirect cotransport across the
osmotic diuresis. The term diuresis simply means increased luminal membrane (the parallel Na/H and Cl/base
urine flow, and osmotic diuresis denotes the situation in which countertransporters)
the increased urine flow is due to an abnormally high amount Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
of any substance in the glomerular filtrate that is reabsorbed 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
442 SECTION VII Renal Physiology

LUMEN
FIGURE 44–4 Major transport pathways for sodium and INTERSTITIUM
chloride in thick ascending limb. The major transporter in the thick +
Na+ Na Na+
ascending limb is the Na–K–2Cl symporter (NKCC), which is the target
for inhibition by loop diuretics such as furosemide and bumetanide.
2CI– ATP
In addition to NKCC, the cells contain potassium channels that recycle Na+
potassium from the cell interior to the lumen and to the interstitium K+ K+
(see Chapter 46). Besides transcellular routes, some sodium also K+
moves paracellularly in response to the lumen positive potential. The K+
apical membranes and tight junctions have a very low water
permeability, and water is not reabsorbed in this segment. Because
the cells reabsorb salt, but not water, the thick ascending limb is the Cl–
point in the nephron at which salt is separated from water. This K+
ultimately allows water excretion and salt excretion to be controlled Cl–
H2O
independently. (Modified with permission from Eaton DC, Pooler JP: Vander’s
Renal Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical
Pub. Division, 2009.)

sodium is not reabsorbed as well as the unusual solute, and What are the mechanisms of sodium and chloride reabsorp-
both are passed on to the loop of Henle. Thus, osmotic diuret- tion by the ascending limbs? These are mainly passive in the
ics inhibit the reabsorption of both water and sodium (as well thin ascending limb and active in the thick ascending limb.
as other ions). Osmotic diuresis can occur in persons with Water reabsorption in the descending limb concentrates lumi-
uncontrolled diabetes mellitus; the filtered load of glucose nal sodium somewhat. Then when tubular fluid, now contain-
exceeds the tubular maximum (Tm) for this substance, and the ing an increased sodium concentration, reaches the epithelium
unreabsorbed glucose then acts as an osmotic diuretic. of the thin ascending limb, this gradient drives reabsorption,
probably by the paracellular route. As tubular fluid then enters
the thick ascending limb, the transport properties of the epithe-
HENLE’S LOOP lium change again, and active processes become dominant. As
shown in Figure 44–4, the major apical entry step for the
Henle’s loop reabsorbs both salt and water, but, taken as a sodium and chloride in this segment is via the Na–K–2Cl sym-
whole, more salt (about 25% of the filtered loads) than water porter (NKCC2 isoform). (The Na–K–2Cl symporter requires
(10% of the filtered water). See Table 44–2. This is a key differ- that equal amounts of potassium and sodium be transported, a
ence from the proximal tubule, which reabsorbs water and topic we will address in Chapter 46.) The Na–K–2Cl symporter
sodium in essentially equal proportions. is the target for a major class of diuretics collectively known as
Also as shown in Table 44–2, the reabsorption of salt and the loop diuretics, which include the drugs furosemide (Lasix)
reabsorption of water occur in different parts of the loop. The and bumetanide. The apical membrane of this segment also
descending limb reabsorbs water but not sodium or chloride. has a Na–H antiporter isoform, which, like the isoform in the
In contrast, the ascending limbs (both thin and thick) reabsorb proximal tubule, provides another mechanism for sodium
sodium and chloride but little water. As a whole, the loop reab- movement into the cell.
sorbs some water and more salt, leaving a dilute fluid in the In addition to the active transcellular reabsorption of
lumen. The differences between the two limbs reveal that the sodium, a large percentage (approaching 50%) of total sodium
cells lining the descending and ascending regions have differ- reabsorption in this segment occurs by paracellular diffusion.
ent permeability properties. The basolateral membranes of all There is a high paracellular conductance for sodium in the
renal cells are quite permeable to water due to the presence of thick ascending limb, and the luminal potential in this seg-
aquaporins. As a result, the cytosolic osmolality is always ment is positive, which is a significant driving force for cat-
close to that of the surrounding interstitium. But the apical ions. (We will see in later chapters that this paracellular
membranes do not always contain aquaporins. The descend- pathway also allows substantial reabsorption of potassium
ing limbs contain aquaporins, so water is reabsorbed there, and calcium.) However, none of this would work without the
driven by the increasing osmolality of the medullary intersti- continuous operation of the Na,K-ATPase in the basolateral
tium. The ascending limbs do not express aquaporins in the membrane.
apical membranes, and the tight junctions are not permeable To summarize the most important features of the loop of
to water. Therefore, even though an osmotic gradient exists Henle, the descending limb reabsorbs water but not sodium
between the lumen (dilute) and interstitium (concentrated), chloride, whereas the ascending limb reabsorbs sodium chlo-
water does not move down the gradient, and water flowing ride but not water. The crucial events occur in the thick ascend-
into the ascending limb remains there and is passed on to the ing limb. By reabsorbing salt but not water, this assures that
distal tubule. the net for the loop as a whole is reabsorption of more salt than
CHAPTER 44 Basic Renal Processes for Sodium, Chloride, and Water 443

LUMEN INTERSTITIUM LUMEN INTERSTITIUM

ATP ATP
Na+ Na+
Na+
K+ K+
Na+
Na+ K+ K+
Cl–
Cl– H2O H2O

ADH stimulates
H2O

Cl– Cl–

FIGURE 44–5 Major transport pathways for sodium and FIGURE 44–6 Major transport pathways for sodium,
chloride in the distal convoluted tubule. The apical membrane chloride, and water in principal cells of the cortical collecting
contains the Na–Cl symporter (NCC), which is the target for inhibition duct. Sodium reabsorption is via apical sodium channels. Activity of
by thiazide diuretics. There is also some sodium reabsorption via these channels is controlled by the hormone aldosterone (see
apical sodium channels. The apical membranes and tight junctions Chapter 45). Chloride reabsorption is passive via the paracellular
have a very low water permeability. (Modified with permission from pathway. Water reabsorption is via aquaporins, the activity of which
Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical is controlled by the antidiuretic hormone (ADH). (Modified with
Books/McGraw-Hill, Medical Pub. Division, 2009.) permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY:
Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)

water. The ascending limb is called a diluting segment (it


dilutes the tubular fluid), and fluid leaving the loop to enter step is enormously important in controlling sodium excretion,
the distal convoluted tubule is hypo-osmotic (more dilute) and we will expand on this topic in Chapter 45.
compared with plasma. Some sodium chloride reabsorption continues in the med-
ullary collecting ducts, probably via some form of epithelial
sodium channels. Although with modern diets containing
DISTAL CONVOLUTED TUBULE excess sodium there is usually a substantial amount of sodium
in the final urine, it is possible to reabsorb virtually all of the
The distal tubule continues to reabsorb sodium and chlo-
remaining sodium if dietary access to sodium is limited.
ride, the major luminal entry step being via the Na–Cl sym-
Principal cells in the collecting ducts are also the crucial
porter (Figure 44–5). This transporter differs significantly
players in reabsorbing water. As indicated earlier, there is
from the Na–K–2Cl symporter in the thick ascending limb
always a substantial amount of dilute fluid entering the collect-
and is sensitive to different drugs. In particular, the Na–Cl
ing duct system, which reabsorbs variable amounts of the water.
symporter is blocked by the thiazide diuretics. Sodium
The water permeability of the principal cells in the collecting
channels also permit sodium entry in the distal convoluted
duct system—both the cortical and medullary portions—is
tubule. Like the ascending limb of the loop of Henle, the
subject to physiological control by circulating antidiuretic
distal tubule is not permeable to water, so that it further
hormone (ADH; see Figure 44–6). The inner medullary col-
dilutes the already somewhat dilute fluid entering it from
lecting duct has at least a finite water permeability even in the
the thick ascending limb.
absence of ADH, but the outer medullary and cortical regions
have a vanishingly low water permeability without ADH.
COLLECTING DUCT SYSTEM Depending on levels of ADH, therefore, water permeability
for most of the collecting duct system can vary from very low
In the collecting ducts, there is a division of labor among sev- to very high. When water permeability is very low (absence of
eral different cell types. Reabsorption of sodium and water is ADH), the hypo-osmotic fluid entering the collecting duct
associated with principal cells (so called because they make system from the distal convoluted tubule remains hypo-
up approximately 70% of the cells; Figure 44–6). Reabsorption osmotic as it flows along the ducts. When this fluid reaches the
of chloride occurs partially via paracellular pathways, but medullary portion of the collecting ducts, there is now a huge
active reabsorption is also associated with another class of col- osmotic gradient favoring reabsorption, which occurs to some
lecting duct cells, the intercalated cells (see Figure 47–3). extent. That is, although there is little cortical water reabsorp-
The principal cells reabsorb sodium, the luminal entry step tion without ADH, there is still a finite medullary absorption
being via epithelial sodium channels. Regulation of this entry because of the enormous osmotic gradient. However, because
444 SECTION VII Renal Physiology

so much water is not reabsorbed in the cortex, most of the TABLE 44–5 Composition of medullary interstitial
water entering the medullary collecting duct flows on to the fluid and urine during the formation of a concentrated
ureter. The result is the excretion of a large volume of very urine or a dilute urine.
hypo-osmotic (dilute) urine, or water diuresis.
What happens when the collecting duct system’s water perme- Interstitial Fluid at Tip of Urine (mOsm/L)
ability is very high (plentiful ADH)? As the hypo-osmotic fluid Medulla (mOsm/L)
entering the collecting duct system from the distal convoluted Concentrated urine
tubule flows through the cortical collecting ducts, most of the
Urea = 650 Urea = 700
water is rapidly reabsorbed. This is because of the large differ-
ence in osmolality between the hypo-osmotic luminal fluid and +
Na + Cl − = 750 a
Nonurea solutes = 700 (Na+, Cl−, K+,
the isosmotic (285 mOsm/kg) interstitial fluid of the cortex. In urate, creatinine, etc.)
essence, the cortical collecting duct is reabsorbing the large vol- Dilute urine
ume of water that did not accompany solute reabsorption in the
Urea = 300 Urea = 30–60
ascending limbs of Henle’s loop and distal convoluted tubule. In
other words, the cortical collecting duct reverses the dilution car- Na+ + Cl− = 350a Nonurea solutes = 10–40 (Na+, Cl−,
K+, urate, creatinine, etc.)b
ried out by the diluting segments. Once the osmolality of the
a
Some other ions (e.g., K+) contribute to a small degree to this osmolarity.
luminal fluid approaches that of the cortical interstitial fluid, the b
Depending on the sodium balance state, sodium in the urine can vary between
cortical collecting duct then behaves analogously to the proxi- undetectable and the majority of the osmolytes.
mal tubule, reabsorbing approximately equal proportions of sol- Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
ute (mainly sodium chloride) and water. The result is that the 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.
tubular fluid, which leaves the cortical collecting duct to enter
the medullary collecting duct, is isosmotic with cortical plasma,
but its volume is greatly reduced compared with the amount straightforward in that reabsorption of water from the lumen
entering from the distal tubule. into a hyperosmotic interstitium concentrates that luminal
In the medullary collecting duct, solute reabsorption con- fluid, leaving concentrated urine to be excreted. The question
tinues, but in the presence of ADH water reabsorption is pro- is: how do the kidneys generate a hyperosmotic medullary
portionally even greater. This is because the ADH has signaled interstitium? Not only is the medullary interstitium hyperos-
much of the medullary collecting duct epithelium to have high motic, but there is also a gradient of osmolality, increasing
water permeability, and the medullary interstitium is hyperos- from a nearly isosmotic value at the corticomedullary border
motic relative to normal plasma (for reasons discussed later). to a maximum of greater than 1,200 mOsm/kg at the papilla.
Therefore, the tubular fluid becomes more and more hyperos- This peak value is not constant and changes depending on
motic, and reduced in volume. conditions. It is highest during periods of water deprivation
How does ADH convert epithelial water permeability from and dehydration, when urinary excretion is lowest, and is
very low to very high? An alternative name for ADH is argin- “washed out” to only about half of that during excess hydration
ine vasopressin (because the hormone can constrict arterioles and when urinary excretion is high (see Table 44–5).
and thus increase arterial blood pressure), but the major renal The main components of the system that develops the med-
effect of ADH is antidiuresis, that is, “against a high urine vol- ullary osmotic gradient are (1) the addition of sodium to the
ume” (see Chapter 61). ADH acts in the collecting ducts on the medullary interstitium by the thick ascending limb; (2) a vas-
principal cells, the same cells that reabsorb sodium, to recruit culature that minimizes removal of that sodium, due both to
vesicles containing aquaporins into the luminal membrane. In the unusual arrangement of descending components in close
the absence of ADH, the aquaporins are withdrawn from the apposition to ascending components and to a low rate of blood
apical membrane by endocytosis. (As stated earlier, the water flow, and (3) the recycling of urea between the medullary col-
permeability of the basolateral membranes of renal epithelial lecting ducts and the deep portions of the loops of Henle (see
cells is always high because of the constitutive presence of Figure 43–3). Some aspects of the process are still uncertain.
other aquaporin isoforms; thus, the permeability of the lumi- However, the essential points are clear, and it is these essential
nal membrane is rate limiting.) points on which we now focus.
Let us begin with a condition in which there is no gradient
and follow its establishment. Assume that both the plasma enter-
URINARY CONCENTRATION: THE ing the medulla and the medullary interstitium have a normal
MEDULLARY OSMOTIC GRADIENT sodium concentration (140 mEq/L) and that the medullary
interstitium is isosmotic with normal plasma. The reabsorption
The production of hypo-osmotic urine is, we hope by now, an of sodium and chloride by the thick ascending limb in the outer
understandable process: the tubules (particularly the thick medulla is the first step. As sodium is deposited in the intersti-
ascending limb of Henle’s loop) reabsorb relatively more solute tium, the interstitial sodium concentration begins to increase
than water, and the dilute fluid that remains in the lumen is above 140 mEq/L. This drives the diffusion of sodium into
excreted. The production of hyperosmotic urine is also neighboring blood vessels. For those portions of the thick
CHAPTER 44 Basic Renal Processes for Sodium, Chloride, and Water 445

ascending limb in the cortex, the reabsorbed sodium simply What happens to water in the medulla during this time?
mixes with material reabsorbed by the nearby proximal convo- Although solute can accumulate without a major effect on
luted tubules. Because the cortex contains abundant peritubular renal volume, the amount of water in the medullary intersti-
capillaries and a high blood flow, the reabsorbed material imme- tium must remain relatively constant; otherwise, the medulla
diately moves into the vasculature and returns to the general would undergo significant swelling or shrinking. The endothe-
circulation. However, in the medulla, the vascular anatomy and lial cells of descending vasa recta, although not as leaky as the
blood flow are quite different, and reabsorbed sodium that is fenestrated endothelium of ascending vasa recta, contain
deposited in the outer medullary interstitium is not immediately aquaporins, allowing water to be drawn from the plasma into
removed, that is, it accumulates. The degree of accumulation is a the increasingly hyperosmotic medullary interstitium in a
function of the arrangement of the vasa recta, their permeability manner similar to water being drawn out of tubular elements.
properties, and the volume of blood flowing within them. This loss of water from descending vasa recta decreases the
plasma volume of blood penetrating deeper into the medulla
and raises its osmolality, thereby reducing the tendency to
VASA RECTA dilute the inner medullary interstitium. Water leaving descend-
ing vessels moves by bulk flow into nearby ascending vasa
As described in Chapter 40, blood enters and leaves the outer recta and is removed from the medulla; thus, there is no accu-
medulla through parallel bundles of descending and ascend- mulation of water. Just as there is countercurrent exchange of
ing vasa recta. These vessels are permeable to sodium, and solute between descending and ascending vessels, there is
they initially take up most of the sodium that is being trans- countercurrent exchange of water. In descending vessels water
ported by the thick ascending limbs into the interstitium of the leaves and solute enters, while in ascending vessels water enters
outer medulla. The ascending vessels return that sodium to and solute leaves (see Figures 44–7 and 40–1). Water and salt
the general circulation, but the descending vessels carry it reabsorption in the thin regions of Henle’s loops also influ-
down into the inner medulla, where it diffuses out across the ences the osmotic gradient. While their quantitative contribu-
endothelia of the vasa recta and the interbundle capillaries tion is not known, it is surely less than that of the thick
that they feed, thereby increasing the sodium concentration ascending limbs and vasa recta.
(and osmolality) throughout the medulla. It is here that the The magnitude of blood flow in the vasa recta is a crucial vari-
anatomy of the vasculature becomes particularly important. If able. If blood flow is relatively high, water from the isosmotic
inner medullary blood with its somewhat elevated sodium plasma entering the medulla in descending vasa recta dilutes the
concentration simply flowed into a venous drainage system, hyperosmotic interstitium (“washes it out”), which occurs to
very little additional increase in interstitial sodium concentra- some extent during a water diuresis. But medullary blood flow is
tion would occur. However, the interbundle capillaries drain lowest in conditions where medullary osmolality is highest.
into ascending vasa recta that lie right next to descending vasa Therefore, the diluting effect of water diffusing out of descending
recta. The walls of the ascending vasa recta are fenestrated, vasa recta during periods of minimal blood flow is not great.
allowing rapid and thorough equilibration of water and small
solutes between plasma and interstitium. As the total sodium
content of the medulla increases, blood in the ascending ves- UREA
sels acquires an increasingly higher sodium concentration.
Meanwhile, blood entering the medulla has a normal sodium There is one additional major player involved in the develop-
concentration of about 140 mEq/L. Thus, there is a gradient of ment of the medullary osmotic gradient—urea. As indicated
concentration between ascending and nearby descending ves- above, the peak osmolality in the renal papilla reaches over
sels. Accordingly, some of the medullary sodium begins to 1,200 mOsm/kg. About half of this is accounted for by sodium
recirculate, diffusing out of ascending vessels (containing an and chloride, and most of the rest (500–600 mOsm/kg) is
elevated sodium concentration) and reentering nearby accounted for by urea. To develop such a high concentration
descending vessels (containing a normal sodium concentra- of urea (remember that the normal plasma concentration is
tion). The process of crossing between ascending and descend- only about 5 mmol/L), there must be a process of recycling.
ing vessels is called countercurrent exchange. Over time, the This involves the tubules as well as the vasa recta.
content of sodium in the ascending vessels and medullary We described this recycling process in Chapter 43 and review
interstitium increases until a steady state is reached in which the key points here. Urea is freely filtered, and about half is
the amount of new sodium pumped into the outer medulla reabsorbed in the proximal tubule. Urea is secreted in the loop
from thick ascending limbs matches the amount of extra of Henle (thin regions), driven by the high urea concentration
sodium leaving in ascending vasa recta and returning to the in the medullary interstitium, thus restoring the amount of
general circulation. At its peak, the concentration of sodium in tubular urea back to the filtered load. From the end of the thin
the inner medulla may reach 300 mEq/L, which is more than limbs to the inner medullary collecting ducts, little urea trans-
double its value in the general circulation. Since sodium is port occurs. Because the vast majority of water is reabsorbed
accompanied by an anion, mostly chloride, the contribution of prior to the inner medullary collecting ducts (by the cortical
salt to the medullary osmolality is about 600 mOsm/kg. and outer medullary collecting ducts), the luminal urea
446 SECTION VII Renal Physiology

FIGURE 44–7 Renal water handling in states A


of maximum antidiuresis and maximum diuresis. 110
Numbers to the right indicate interstitial osmolality;
numbers in the tubules indicate luminal osmolality.
The dashed line indicates the corticomedullary CORTEX
border. Arrows indicate sites of water movement. In 300 300
both antidiuresis and diuresis, most (65%) of the MEDULLA
filtered water is reabsorbed in the proximal tubule
and another 10% in the descending loop of Henle.
500
The greater relative reabsorption of solute versus Vasa
water by the loop as a whole and distal tubule recta
results in luminal fluid that is quite dilute (110
mOsm) as it enters the collecting ducts. During 900
antidiuresis (A), most of the remaining water is
reabsorbed in the cortical collecting ducts, 1200 1200
stimulated by the actions of ADH, with some
additional reabsorption in the medullary collecting
ducts. The equilibration of tubular fluid with the
high medullary osmolality results in final fluid that is Maximum antidiuresis
very hyperosmotic (1,200 mOsm). During diuresis
(B), no water reabsorption occurs in the cortical B
110
collecting duct, but some occurs in the inner
medullary collecting duct. Despite the medullary
water reabsorption, continued medullary solute
reabsorption reduces solute content relatively more CORTEX
110 300
than water content, and the final urine is very dilute
(70 mOsm). In the parallel vasa recta, there is MEDULLA
considerable exchange of both solute and water, so
that plasma osmolality and solute concentrations 350
Vasa
equilibrate with the surrounding interstitium. The
recta
ascending vasa recta ultimately remove all the
solute and water reabsorbed in the medulla.
400
Because there is always some net volume
reabsorption in the medulla, the vasa recta plasma
flow out of the medulla always exceeds the plasma 70 600
flow in. (Modified with permission from Eaton DC,
Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange
Medical Books/McGraw-Hill, Medical Pub. Division, 2009.) Maximum diuresis

concentration increases up to 50 times its plasma value (i.e., descending vasa recta minimizes removal. Adding to the osmo-
500 mmol/L or more). In the inner medullary collecting ducts, lality of the medulla is urea, which recycles from the inner med-
this drives urea reabsorption via specialized ADH-sensitive ullary collecting ducts to the thin limbs of the loop of Henle.
urea uniporters, increasing the medullary interstitial concen- Urea also participates in countercurrent exchange between
tration close to the same high value as in the tubular lumen. ascending and descending vasa recta for the same reasons that
The concentrated urea remaining in the collecting ducts, typi- salt does. The magnitude of the osmotic gradient is variable,
cally about half the filtered load, is excreted. The combination being greatest during states of dehydration and lowest during a
of a high urea concentration, along with the high sodium and water diuresis (see Figure 44–8). Key controlling factors are levels
chloride, brings the medullary osmolality to a value exceeding of ADH and the magnitude of medullary blood flow.
1,200 mOsm/kg H2O. The importance of urea in contributing Let us conclude by addressing a common student question:
to the medullary osmotic gradient is emphasized in the case of under conditions of high ADH, why does not the water being
low protein intake, which results in a greatly reduced meta- reabsorbed from the medullary collecting ducts dilute the med-
bolic production of urea. In this condition, the ability of the ullary interstitium and destroy the osmotic gradient? The
kidneys to produce highly concentrated urine is reduced. answer is 2-fold. First, with high ADH most of the total water
To summarize the generation of the renal osmotic gradient: reabsorbed from the tubule occurs in the cortex, and there sim-
salt (without water) is deposited in the interstitium of the outer ply is not very much water left to be reabsorbed in the medulla.
medulla by the thick ascending limbs. That salt enters descend- Second, the thick ascending limb keeps adding solute to the
ing vasa recta and is distributed to the inner medulla. It accumu- medulla. Proportionally more sodium is added from the thick
lates in the medullary interstitium because a combination of low ascending limb than water is added from the inner medullary
blood flow and countercurrent exchange between ascending and collecting ducts.
CHAPTER 44 Basic Renal Processes for Sodium, Chloride, and Water 447

A
100%

Volume of remaining filtrate


75%

50% No ADH

25%

Maximum ADH

Proximal tubule Loop of Henle Cortical Medullary


collecting duct collecting duct

B
1200
Maximum ADH
Osmolality (mOsmol/kg)

900

600

300

No ADH

Proximal tubule Loop of Henle Cortical Medullary


collecting duct collecting duct
FIGURE 44–8 Volume of remaining filtrate (A) and tubular osmolality (B) at different sites along the tubule in conditions of
maximum ADH and no ADH. Under all conditions the majority of the filtered volume is reabsorbed in the proximal tubule. Additional
reabsorption occurs in the thin limbs of the loop of Henle, the exact amount depending on ADH because the interstitial osmolality varies with
ADH. In the absence of ADH no further reabsorption occurs until the inner medulla, whereas with maximal ADH most of the remaining volume
is reabsorbed in the cortical collecting ducts. The osmolality of the final urine is strongly dependent on ADH, as is the maximum osmolality in
the loop of Henle because the peak medullary interstitial osmolality also varies with ADH.

CLINICAL CORRELATION which he lost over 10 lb of body weight and showed signs of
severe dehydration, but recovered without ill effects. Blood
A 19-year-old male college freshman is convinced by his tests show normal levels of glucose, urea (BUN), creatinine,
roommates to seek evaluation after noting his amazing calcium, and osmolality. A urine sample, in which he was
amounts of water drinking and urination. He said that he able to void a volume greater than 1 L, reveals no glucose
has always consumed lots of water, and thought there was but it does have an unusually low osmolality of 62 mOsm/
nothing wrong other than the inconvenience of urinating a kg. At this point, he was sent to the university hospital for a
lot. He only urinates once per night, and does not feel spe- pyelogram, consisting of administering contrast media
cial urgency, although he does urinate many times during intravenously and taking serial digital radiographs of the
the day and seeks cold water to drink, sometimes in prefer- kidneys and urinary tracts. The contrast medium appears in
ence to food. At the university clinic, a physical exam reveals the renal cortex almost immediately and is freely filtered. In
nothing unusual. He reports no family history of diabetes another 10 minutes, it makes its way to the renal papilla.
mellitus. While in high school, he had a severe bout of vom- Examination of the student’s radiographs revealed normal
iting and diarrhea, diagnosed as viral gastroenteritis, in kidneys, but rather enlarged ureters and bladder. He was
448 SECTION VII Renal Physiology

kept in the hospital and put on water deprivation overnight,


STUDY QUESTIONS
during which time he urinated a considerable volume and 1. Chloride reabsorption parallels sodium reabsorption
lost 5 lb of body weight. His water deprivation was main- mainly because
tained during the morning, and he continued to produce A) chloride is almost always transported via a symporter
urine. His blood pressure was normal while sitting, but it with sodium.
decreased considerably when he was standing (orthostatic B) chloride is the most abundant negatively charged species
hypotension). A urine sample taken at this time yielded an available to balance the reabsorption of the positive charge
on sodium.
osmolality of 279 mOsm/kg, and his plasma osmolality was
C) chloride has a very high passive permeability.
301 mOsm/kg. D) chloride and sodium are both part of the sodium chloride
His physicians strongly suspected this to be a case of molecule and cannot be separated.
diabetes insipidus. The fact that water loss continued well 2. The obligatory water loss in the kidney
into a state of dehydration ruled out nonosmotic causes
A) is another name for insensible loss of water.
(e.g., psychogenic polydipsia) as a cause of the excessive B) occurs because there is always at least some excretion of
urination. Accordingly, he was given an intravenous injec- waste solutes.
tion of aqueous arginine vasopressin (ADH). Serial urine C) occurs because there is an upper limit to how fast
measurements showed that the urine osmolality increased aquaporins can reabsorb water.
to a peak value of over 500 mOsm/kg, demonstrating that D) is the amount of water that accompanies salt excretion.
the kidneys were able to respond well to the ADH. This is 3. Which region of the tubule secretes water?
a case of diabetes insipidus due to a failure of adequate A) the descending thin limb
ADH secretion by the posterior pituitary gland. In this B) the cortical collecting duct
case, the cause of the patient’s decrease in posterior pitu- C) the medullary collecting duct (when ADH is absent)
itary function could not be ascertained from his medical D) no region secretes water
history. For more on diabetes insipidus, see Chapter 61. 4. If the thick ascending limb stopped reabsorbing sodium, then
the final urine would be
A) isosmotic with plasma in all conditions.
B) dilute, depending on ADH.
CHAPTER SUMMARY C) concentrated, depending on ADH.
D) dilute or concentrated, depending on ADH.
■ The reabsorption of most of the filtered water, anions, and
osmotic content is linked to the active reabsorption of sodium. 5. If a healthy young person drinks a large amount of water, which
of the following is unlikely to happen?
■ In all conditions, the vast majority of the filtered volume is
reabsorbed iso-osmotically in the proximal tubule in a manner A) an increase in medullary blood flow
that is entirely dependent on active sodium reabsorption. B) an increase in water permeability in the medullary
collecting ducts
■ The capacity to generate urine with a variable osmolality depends
C) a decrease in interstitial osmolality at the tip of the
on “separating salt from water” in the diluting segments.
renal papilla
■ Reabsorption of water remaining in the lumen beyond the loop D) a decrease in the urea concentration in the final urine.
of Henle depends on hydration status, allowing the kidneys to
6. A healthy young person drinks a large amount of water. Over
excrete either a high-volume dilute urine, a low-volume
the next several hours, most of the water entering the
concentrated urine, or anything in between.
glomerular filtrate is
■ Levels of ADH determine whether the hypo-osmotic fluid
A) excreted.
leaving the diluting segments is excreted largely as is or
B) reabsorbed in the cortical collecting duct.
whether most of this fluid is subsequently reabsorbed.
C) reabsorbed in the proximal tubule.
■ The existence of the medullary osmotic gradient depends on D) reabsorbed in the loop of Henle.
(1) transport of salt without water into the medullary intersti-
tium by the thick ascending limb, (2) recycling of urea, and
(3) low-volume countercurrent blood flow in the vasa recta.
45
C H A P T E R

Regulation of Sodium
and Water Excretion
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ Describe the relationship between renin and angiotensin II.


■ Describe the detectors that regulate renin secretion.
■ Define pressure natriuresis and diuresis.
■ Define tubuloglomerular feedback, and describe the mechanism for
tubuloglomerular feedback and autoregulation of glomerular filtration rate.
■ List the major factors that regulate sodium excretion.
■ State the tissue origin of aldosterone, its renal sites of action, and its effect
on sodium reabsorption.
■ State the origin of atrial natriuretic peptides, the stimulus for their secretion,
and their effect on sodium reabsorption and glomerular filtration rate.
■ Describe the origin of antidiuretic hormone and the two major reflex controls
of its secretion.
■ Distinguish between the reflex changes that occur when an individual has
suffered iso-osmotic fluid loss because of diarrhea as opposed to a pure
water loss (i.e., solute water loss as opposed to pure water loss).
■ Diagram in flow-sheet form the pathways by which sodium and water
excretions are altered in response to sweating, diarrhea, hemorrhage,
high-salt diet, and low-salt diet.
■ Describe the control of thirst.

GOALS (RBF) and glomerular filtration rate (GFR) are major means
of regulating sodium excretion. However, the kidneys cannot
The excretion of sodium and water is regulated by an array of allow blood flow and filtration to reach such extreme levels that
signals, some originating within the kidneys themselves (intra- they compromise the metabolic health of the kidneys or inter-
renal signals) and some from other regions of the body. The fere with the excretion of substances other than sodium. Thus,
regulatory processes are complex, and it is imperative to grasp another goal is to limit the sodium-related changes in RBF and
the goals of regulation so that individual mechanisms fit into a GFR that might otherwise reach deleterious levels.
logical framework. A key concept is that the kidneys work in
partnership with the cardiovascular system. Together they
ensure that (1) there is enough blood volume to fill the vascular
tree, (2) enough pressure to drive blood flow through periph- SODIUM EXCRETION: THE
eral tissues, and (3) the blood, and therefore the cells through- CARDIOVASCULAR CONNECTION
out the body, has the proper osmolality. One way or another, all
the regulatory mechanisms that control sodium and water The kidneys play a direct role in maintaining cardiac output,
excretion exist for the purpose of meeting these goals. There is first by ensuring that there is sufficient blood volume to allow
also an indirect, but related goal. Variations in renal blood flow the heart to fill between beats, and second by contributing to

449

Ch45_449-462.indd 449 11/26/10 10:33:53 AM


450 SECTION VII Renal Physiology

the plasma and tissue interstitial space. Thus, maintenance of


Direct renal blood volume is largely a matter of maintaining ECF volume.
influences It is obvious why water is crucial for ECF volume, but why is
sodium so important? The answer stems from two facts. First
ECF osmolality is tightly regulated, and second the osmotic
content of the ECF depends critically on its sodium content.
We can approximate ECF osmolality as follows:
Blood cell Total peripheral
ECF volume
production resistance ECF solute content
ECF osmolality = ______________ (1)
ECF volume
By rearrangement the above expression becomes:
ECF solute content
Blood volume
ECF volume = ______________ (2)
ECF osmolality
Furthermore, since almost all of the ECF solute is accounted
FIGURE 45–1 Influence of the kidneys on the cardiovascular for by sodium and an equivalent number of anions (mostly
system. The kidneys affect blood volume via their production of chloride and bicarbonate), the amount of ECF solute is approx-
erythropoietin, which stimulates red blood cell production, and via imately twice the sodium content. We can write the previous
their control of salt and water excretion. They also influence total
expression as follows:
peripheral resistance via the actions on angiotensin II (see text for
details). 2×ECF sodium content
ECF volume = ________________ (3)
ECF osmolality
Therefore, in the face of tightly controlled ECF osmolality,
the control of total peripheral resistance (see Figure 45–1). Let ECF volume varies directly with sodium content.
us examine the renal involvement in these cardiovascular This raises the following question: how do the kidneys know
issues, with blood volume first. how much sodium is present in the ECF? There is no mecha-
Blood is composed primarily of red blood cells (about 45%) nism for the body to assay sodium content per se. Instead the
and blood plasma (about 55%). The kidneys are crucial for detection of sodium content is indirect, based mainly on vas-
both parts—they secrete the hormone erythropoietin, which cular pressures. That is, variations in vascular pressures are
stimulates production of red blood cells, and they regulate interpreted by the body as variations in sodium content.
the extracellular fluid (ECF) volume, of which blood plasma Vascular pressures are assessed by baroreceptors—cells
is a significant part. Although not exact, there is an approxi- that deform in response to changes in local intravascular
mate proportionality between blood volume and total ECF pressure. Three sets of baroreceptors are important for con-
volume. Blood volume tends to increase and decrease as ECF trolling sodium excretion (see Figure 45–2). Two have already
volume increases and decreases due to shifts of fluid between been described in Chapter 29. These are (1) the arterial

Total peripheral resistance


Brainstem
Arterial Cardiac performance
vasomotor
baroreceptors Sympathetic drive to kidney
center
Venous compliance

Brainstem Venous compliance


vasomotor Sympathetic drive to kidney
center Total peripheral resistance
Cardiopulmonary
Cardiac performance
baroreceptors
Hypothalamus ADH

Intrarenal Renin-angiotensin system


baroreceptors GFR, salt and water
reabsorption

FIGURE 45–2 Baroreceptors and the major processes they influence. Arterial baroreceptors sense pressures in the aorta and carotid
arteries and send afferent information to the brainstem vasomotor center, which then regulates cardiovascular and renal processes via
autonomic efferents. Cardiopulmonary baroreceptors sense pressure in the cardiac atria and pulmonary arteries, thereby being responsive to the
filling of the vascular tree. They send afferent information in parallel with the arterial baroreceptors. While there is overlap between the
influences of the two sets of baroreceptors, the cardiopulmonary baroreceptors have a particularly important influence on the hypothalamus,
which regulates secretion of ADH. The intrarenal baroreceptors have a major role in the renin–angiotensin system (see text for details). (Modified
with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
CHAPTER 45 Regulation of Sodium and Water Excretion 451

baroreceptors, nerve cells that mediate the classic barorecep- thetic nerve. Thus, the activity of the granular cells is affected
tor reflex, and (2) the cardiopulmonary baroreceptors that are both by direct sensing of pressure in the renal arterioles and by
also nerve cells and work in parallel with the arterial barore- pressures sensed by neural baroreceptors elsewhere in the
ceptors. The third set of baroreceptors consists of intrarenal body
baroreceptors, which are not nerve cells. We will describe In response to changes in pressures sensed by baroreceptors,
their operation shortly, but first we briefly summarize the a number of renal events are set in motion that have powerful
neural baroreceptors as a review. effects on total peripheral resistance, and, as we will see later,
The medullary cardiovascular centers stimulate vascular on sodium excretion. The critical events are signaling path-
tone (vasoconstriction) throughout the body via the sympa- ways known as renin–angiotensin systems (RAS).
thetic nervous system. In the arterioles of the peripheral vas-
culature this sympathetic tone maintains total peripheral
resistance, and in the peripheral venous system it maintains
central venous pressure via its influence on the compliance of
RENIN–ANGIOTENSIN SYSTEMS
large veins. Arterial baroreceptors exert a tonic inhibition of An important hormone in the control of sodium excretion
the medullary cardiovascular centers, resulting in a brake on and blood pressure is angiotensin II. It is a potent vasocon-
sympathetic drive. An increase in arterial pressure causes strictor, and also a mediator of multiple actions in the kidneys
greater firing of the baroreceptors, more inhibition, and there- that affect sodium excretion. Thus, it affects blood pressure
fore even less sympathetic drive to the periphery, while a directly as a vasoconstrictor and indirectly via regulation of
decrease in arterial pressure reduces firing in the barorecep- renal sodium excretion and therefore blood volume.
tors and less inhibition, allowing more sympathetic drive. The There are many local RAS systems in individual tissues,
resulting changes in vascular tone alter total peripheral resis- including the kidneys, brain, and the heart. There is also a
tance and help stabilize arterial pressure. The cardiopulmo- global or systemic RAS. All RAS systems, whether global or
nary baroreceptors act in the same way, but respond not to local, consist of a large protein substrate called angiotensino-
arterial pressure, but rather to pressures in the cardiac atria gen, several enzymes, and several products. The key product is
and pulmonary vessels. The medullary cardiovascular centers angiotensin II. When angiotensin II binds to cell surface recep-
also send both sympathetic stimulatory and parasympathetic tors, it initiates actions that affect blood pressure and excretion
inhibitory signals to the heart in response to variations in of sodium. The first key enzyme in all RAS systems is renin
baroreceptor input. (pronounced REE-nin). It acts upon angiotensinogen to pro-
The kidneys, being part of the peripheral vasculature, duce a small (10-amino acid) product called angiotensin I.
respond to changes in sympathetic drive and contribute to Angiotensin I is acted upon by another enzyme, angiotensin-
changes in total peripheral resistance. However, as we will converting enzyme (ACE), to produce the highly active eight-
describe later, changes in sodium excretion in response to amino acid peptide angiotensin II. In the global RAS system,
sympathetic drive are even more important than the renal the source of angiotensinogen circulating in the blood is the
contribution to total peripheral resistance. liver. The source of circulating renin is the granule cells in the
kidney. Renin is secreted both into the interstitium of the kid-
ney and into the lumen of the afferent arterioles, where it acts
on circulating angiotensinogen to produce circulating angio-
RENAL CONTROL OF tensin I. ACE, which is expressed on the luminal surface of
VASCULAR RESISTANCE endothelial cells in many parts of the vasculature, then con-
verts angiotensin I to angiotensin II (see Figure 45–3).
If deviations in blood pressure are sustained, the kidneys are The major source of angiotensin II that affects the kidneys is
capable of strongly reinforcing the effects of the vasomotor produced by a local RAS system within the kidneys them-
centers on vascular resistance. How does this work? The major selves. Thus, the kidneys are regulated by both the global RAS
detectors involved in the kidney’s ability to regulate vascular system and a local RAS system.
resistance are the previously described neural baroreceptors, The circulating level of angiotensinogen in the global RAS
and the intrarenal baroreceptors. The intrarenal baroreceptors system is normally high and is not rate limiting. Furthermore,
sense renal afferent arteriolar pressure. Anatomically, these ACE activity usually converts most of the angiotensin I into
structures are not nerve cells and do not send signals to the angiotensin II. Therefore, the major controller of angiotensin II
brainstem vasomotor center, but rather are specializations of production is the amount of angiotensin I produced by the
the cells of the afferent arteriole: granular cells (also called action of renin. Consequently, understanding angiotensin II
juxtaglomerular cells) that form part of the juxtaglomerular production requires an understanding of the regulation of renin
apparatus described in Chapter 40. They act entirely within secretion. Two primary regulators of renin secretion have been
the kidney. Although granular cells acting as intrarenal barore- described: the first are the neural baroreceptors, which influ-
ceptors do not send signals centrally, neural signals originat- ence the activity of renal sympathetic nerves that stimulate
ing in the vasomotor centers (generated in response to neural granular cell production of renin and vasoconstriction at the
baroreceptors) reach the granular cells via the renal sympa- same time. This occurs via a β1-adrenergic receptor–protein
452 SECTION VII Renal Physiology

Stimuli to renin

Liver

Kidney
Angiotensinogen
(453 aa)

Renin (enzyme)

Angiotensin I
(10 aa)

Angiotensin I
Angiotensin-converting Angiotensin-converting
enzyme enzyme
(endothelium) (endothelium)
Angiotensin II

Angiotensin II
(8 aa)

Adrenal
Cardiovascular cortex
system Aldosterone

Kidney

Salt and H2O


FIGURE 45–3 Major components of the global RAS. Vasoconstriction
retention
Angiotensin II acts on the vascular system as a vasoconstrictor
and stimulates adrenal production of aldosterone. It also acts
within the kidneys to promote sodium reabsorption (although
the major source of renal angiotensin II is the intrarenal RAS
system). (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Blood pressure
Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

kinase A-dependent process. The second regulators of renin ratus (Figure 45–4) that monitors the amount of tubular
secretion are the intrarenal baroreceptors, that is, the very same sodium chloride directly bathing the macula densa cells. This
granular cells that respond to adrenergic stimulation also amount of sodium chloride depends on both the rate of filtra-
deform in response to changes in afferent arteriolar pressure; tion (GFR) and the rate of sodium reabsorption in all the
when the pressure decreases, renin production increases. Thus, nephron elements preceding the macula densa. When sodium
granular cells act as both detectors (of renal arteriolar pressure) chloride delivery to the apical surface of macula densa cells
and signal generators (releasing renin) in response to changes increases, renin production decreases, that is, high tubular
in pressure and sympathetic activity. The signals from the vaso- sodium loads inhibit renin production. The utility of this load
motor system to the renin-producing granular cells ensure that detector control of renin secretion is that in situations in which
there is tight coordination between the rapid activity of the sys- the body contains excess sodium and is excreting sodium rap-
temic baroreceptor reflex and the slower-acting RAS. However, idly, it is advantageous not to produce very high quantities of
the intrarenal pressure detector can function in the absence of angiotensin II.
renal innervation (e.g., after a renal transplantation). In review, three separate mechanisms regulate renin secre-
There is also a third detector mechanism that regulates renin tion (neural signals, afferent arteriolar pressure, and NaCl at
release. It is another component of the juxtaglomerular appa- the macula densa). The multiple controls reflect the importance
CHAPTER 45 Regulation of Sodium and Water Excretion 453

Mesangial
cells

Glo
1. Renal sympathetic nerve
er

m
activity stimulates renin ulu
secretion from granular cells. s FIGURE 45–4 Control of renin secretion.
Three primary mechanisms regulate renin
secretion. First, renal sympathetic nerve activity
nt activates β1-adrenergic receptors on granular
ere
Aff riole cells of the afferent arteriole to stimulate renin
2. Changes in systemic e
art

Ef rter
secretion. Second, the granular cells also act as

fe iol
a
blood pressure deform
nin

re e
intrarenal baroreceptors, responding to changes

nt
membrances of granular cells Re R
en
to stimulate release of renin. in in pressure within the afferent arteriole, which,
Granular except in cases of renal artery stenosis, is a
cells reflection of changes in arterial blood pressure.
Deformation of the granular cells alters renin
Chemical transmitters secretion: when pressure falls, renin production
increases. Third, macula densa cells in the thick
ascending limb sense the delivery of tubular
sodium chloride, leading to the release of
Macula d
en chemical transmitters that alter renin secretion
sa
from the granular cells: when sodium chloride
3. Osmotic stretch in response to
changes in NaCI delivery deform
delivery increases, renin production decreases.
membranes to stimulate release of (Modified with permission from Eaton DC, Pooler JP: Vander’s
chemical messengers that reduce Renal Physiology, 7th ed. New York, NY: Lange Medical
renin secretion. Books/McGraw-Hill, Medical Pub. Division, 2009.)

of the RAS system and angiotensin II, in particular, in cardio- additional decrease due to increased renal sympathetic nerve
vascular control. A significant action of circulating angiotensin activity that is part of the neural baroreceptor reflex. These
II produced by the global RAS system is general arteriolar actions all decrease sodium excretion. If the low pressure per-
vasoconstriction. This vasoconstriction acts in parallel with sists, large amounts of renin are released and the subsequent
sympathetically mediated neural signals to increase total actions of angiotensin II reinforce these actions. All of this
peripheral resistance, thereby increasing blood pressure. The makes logical sense: in the face of low arterial pressure, the
importance of this system makes the RAS a natural target for kidneys conserve sodium. On the other hand, if the sodium
pharmacological intervention to reduce high blood pressure. content of the body increases significantly and plasma volume
A number of blood pressure–lowering pharmacological agents is increased, or if there is an inappropriate increase in arterial
are aimed at components of the RAS systems, including ACE pressure, sympathetic vasoconstrictive activity is reduced,
inhibitors and angiotensin II receptor blockers (ARBs). RBF and GFR increase, and sodium excretion increases.
The filtered load of sodium is always enormous, and it is
imperative that the kidneys reabsorb the vast majority of it
CONTROL OF SODIUM under all conditions. Because even a small fractional change in
reabsorption results in a large change in the absolute amount
EXCRETION excreted, precise control over reabsorption is an essential
aspect of maintaining sodium balance. While GFR is an
GLOMERULAR FILTRATION RATE important variable, most controls focus on reabsorption. As
with many other aspects of renal function, certain details of
Many factors influence sodium excretion. One simple principle this control system are still not understood. Thus, we empha-
to remember amid the complexities is that the amount of sodium size again that the most important goal of regulating sodium
excreted is the difference between the filtered load and the balance, even if we do not know all of the mechanistic details,
amount reabsorbed. Therefore, a major control over sodium is to regulate ECF volume.
excretion is regulation of the filtered load via regulation of GFR.
As detailed in Chapter 40, GFR is a function of afferent arte-
riolar pressure (reflecting systemic arterial pressure) and the ECF VOLUME
resistances of the afferent and efferent arterioles. Changes in
resistance are produced by changes in renal sympathetic nerve Dietary sodium loads always expand the ECF volume. Under
activity and levels of angiotensin II. In the face of a decrease in most circumstances, sodium loads are accompanied by water
blood pressure there is a decrease in GFR directly, and an loads, as when one eats pizza and beverage, thus accounting
454 SECTION VII Renal Physiology

for the increase in volume. But even if the load is the unusual ANGIOTENSIN II
case of salt without water, the ECF still expands because the
added osmoles in the ECF increase its osmolality and draw As important as angiotensin II is to vascular resistance, it plays
water from the intracellular space (see Figure 44–1). The kid- an even more important role in regulating sodium reabsorp-
neys respond to sodium loads by decreasing sodium reabsorp- tion in the kidneys. It has direct tubular actions and it stimu-
tion, thus allowing more sodium to be excreted. Much of the lates the secretion of the hormone aldosterone from the
decrease in reabsorption occurs as a result of decreased levels adrenal cortex, another major player in the connection
of angiotensin II, via a mechanism described in the next sec- between sodium excretion and the cardiovascular system
tion. At least some of the signaling is via cardiopulmonary (see Figure 45–3 and Chapter 65). Finally, angiotensin II acts
baroreceptors that inhibit the medullary vasomotor centers in a negative feedback manner to inhibit renin production by
and lead to less sympathetic drive, which in turn leads to less acting directly on granular cells.
production of angiotensin II. In response, the kidneys permit Angiotensin II affects blood pressure by altering peripheral
a large amount of sodium to be excreted. A pure water load, vascular resistance, but within the kidneys, angiotensin II
which also expands the ECF volume, does not lead to increased exerts significant sodium-retaining actions that begin by bind-
sodium excretion. This is partly because pure water loads are ing to G protein–coupled receptors and activating intracellu-
excreted much more rapidly than salt loads and partly because lar signaling cascades: (1) it constricts renal arterioles (as it
pure water loads simultaneously reduce plasma osmolality. does arterioles elsewhere), thereby reducing RBF and GFR; (2)
A major decrease in ECF volume, such as occurs with pro- it constricts glomerular mesangial cells, also reducing GFR;
longed vomiting and diarrhea or a major hemorrhage, always and (3) it stimulates sodium reabsorption in the proximal
involves loss of sodium. The low volume sensed by cardiopul- tubule. All of these actions reduce sodium excretion. The
monary baroreceptors leads to strong sympathetic stimulation number of functioning Na–H exchangers in the apical mem-
of the kidneys, lower RBF, lower GFR, activation of the RAS, brane of proximal tubule cells is strongly influenced by angio-
and stimulation of tubular sodium reabsorption. These actions tensin II. These proteins can shuttle back and forth between
reduce sodium excretion to low levels, thereby helping pre- locations where they are functioning and where they are inac-
serve ECF volume. Figure 45–5 summarizes the multiple tive. When angiotensin II is present, the Na–H exchangers
responses to low ECF volume, of which reduction in sodium move to locations in the apical membrane where they trans-
excretion is a significant part. port ions, thereby increasing the reabsorption of sodium.

Controlled
– LOW EFFECTIVE variable
CIRCULATING VOLUME

JUXTAGLOMERULAR LOW PRESSURE HIGH PRESSURE Sensors


APPARATUS BARORECEPTORS BARORECEPTORS

Renin

Mediators
RENIN-ANGIOTENSIN- ADH SYMPATHETIC NERVOUS
ALDOSTERONE AXIS SYSTEM

+ + + + +
Effectors
RENAL SODIUM
+ STARLING + VASOCONSTRICTION CARDIAC EXCITATION
& WATER
FORCES RETENTION
+ +

+ MEAN ARTERIAL
BLOOD PRESSURE

FIGURE 45–5 Responses to low vascular volume. Because adequate vascular volume is essential for the long-term maintenance of
arterial pressure, loss of volume, as occurs in prolonged diarrhea, vomiting, or hemorrhage, invokes multiple corrective responses. Following a
rapidly acting stimulation of the heart and peripheral resistance, the kidneys are stimulated to reduce excretion of sodium and water, thereby
preserving existing volume. (Reproduced with permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)
CHAPTER 45 Regulation of Sodium and Water Excretion 455

Conversely, when the levels of angiotensin II decrease, the to correct the elevated arterial pressure. Another example of
Na–H exchangers are withdrawn to sites where they are inac- override occurs during exercise. Arterial pressure increases,
tive, along with a concomitant reduction in activity of the but sodium excretion decreases.
basolateral Na,K-ATPase, resulting in much less sodium reab- An implicit assumption about all the processes that control
sorption. Thus, variation in levels of angiotensin II is a very sodium excretion is that there are parallel movements of water,
powerful bidirectional response system. When the levels of and therefore volume. This is indeed true, and maintaining
angiotensin II are high, the kidneys filter less sodium and ECF volume is, as we have said, the most important reason for
reabsorb more, thereby greatly reducing the amount excreted. regulating sodium excretion. While most of the time it is
In contrast, when there is little angiotensin II, large amounts of appropriate that water accompany the movement of sodium,
sodium remain in the tubule and are excreted. it is not always true (because there are occasions when the
inputs of salt and water are not in parallel). Therefore, the kid-
neys possess ways of independently controlling water and
PRESSURE NATRIURESIS sodium excretion. Such independent controls are exerted in
AND DIURESIS regions of the nephron beyond both the proximal tubule and
loop of Henle, namely, in the collecting tubules and ducts. As
Just as ECF volume exerts control over sodium excretion, so described in the next section, the chief player with respect to
does arterial pressure. Increased pressure in the renal artery independent control of sodium excretion is the hormone
causes the kidneys to increase their excretion of sodium. This aldosterone.
phenomenon has been given the name pressure natriuresis
(and because natriuresis tends to increase water excretion, it
can be properly called pressure natriuresis and diuresis). It is
an entirely intrarenal phenomenon, that is, it is not activated LONG-TERM CONTROL:
by external signals (although, as described later, it can be ALDOSTERONE REGULATION
severely attenuated, or even prevented by external signals).
Pressure natriuresis and diuresis serves as a backup system that OF SODIUM BALANCE
comes into play if fast-acting neural reflex systems of regulat-
Aldosterone is a major stimulator of sodium reabsorption in
ing blood pressure fail to completely correct large increases.
the distal nephron; that is, regions of the tubule beyond the
The mechanism of pressure natriuresis is a fascinating inter-
proximal tubule and loop of Henle. Aldosterone-stimulated
action between renal hemodynamics and intricate signaling
sodium retention is an effector system that is vital in correct-
cascades. It begins when higher renal artery pressure drives
ing prolonged reductions in body sodium, blood pressure, and
higher blood flow in the medulla. The medulla has relatively
volume. The most important physiological factor controlling
poor autoregulation (compared with the highly effective auto-
secretion of aldosterone is the circulating level of angiotensin
regulation in the cortex); accordingly medullary blood flow
II, which stimulates the adrenal cortex to produce aldosterone.
tends to vary in closer relation to renal artery pressure. The
This targets the distal nephron to increase sodium reabsorp-
higher medullary blood flow when pressure is high leads to
tion and thus increase total body sodium and blood volume to
increased interstitial pressure that is transmitted throughout
produce a long-term correction to total body sodium content
the kidney. In turn, the higher interstitial pressure activates
and mean blood pressure.
intrarenal signals (arachidonic acid metabolites), which com-
Aldosterone stimulates sodium reabsorption mainly in the
mand the proximal tubule cells to reduce their transport capac-
cortical connecting tubule and cortical collecting duct, spe-
ity. They do this by withdrawing Na–H antiporters (similar to
cifically by the principal cells. An action on this late portion of
what happens when angiotensin II falls) and reducing Na,K-
the nephron is what one would expect for fine-tuning the out-
ATPase activity. Together these events reduce sodium reab-
put of sodium, because more than 90% of the filtered sodium
sorption and increase excretion. Higher interstitial pressure
has already been reabsorbed by the time the filtrate reaches the
may also increase backleak from the interstitium into later
collecting duct system.
portions of the tubule, further reducing reabsorption. If the
The percentage of sodium reabsorption dependent on the
increased pressure is maintained, the RAS system is suppressed
influence of aldosterone is approximately 2% of the filtered
and the production of angiotensin II is lowered, which aug-
load. Thus, all other factors remaining constant, in the com-
ments and sustains the withdrawal of Na–H antiporters.
plete absence of aldosterone, a person would excrete 2% of the
Although pressure natriuresis is an intrarenal process, it can
filtered sodium, whereas in the presence of high plasma con-
be overridden by external signals. When the ECF volume is
centrations of aldosterone, virtually no sodium would be
normal or high and pressure in the renal artery increases, pres-
excreted. Two percent of the filtered sodium may seem trivial
sure natriuresis and diuresis are very effective in increasing
but is actually a significant amount because a large quantity of
excretion of sodium and water and reducing blood volume.
sodium is filtered:
On the other hand, if ECF volume is low and renal artery pres-
sure increases, there is much less salt and water loss. In effect, Total filtered Na per day = GFR × PNa =
(4)
it is more important to prevent further loss of volume than it is 180L per day × 145 mmol/L = 26,100 mmol per day
456 SECTION VII Renal Physiology

Thus, aldosterone controls the reabsorption of 0.02 × 26,100 controlled by renin, this emphasizes once again the impor-
mmol per day = 522 mmol per day. In terms of sodium chlo- tance of the global RAS system. Both renin and aldosterone
ride, the form in which most sodium is ingested, this amounts have relatively short plasma half-lives (~15 minutes), while the
to the control of approximately 30 g NaCl per day, an amount half life of angiotensin II is very short (<1 minute). Therefore,
considerably more than the average person consumes. There- prolonged stimulation by aldosterone requires the continuous
fore, by control of the plasma concentration of aldosterone stimulation of renin secretion. Figure 45–7 shows how
between minimal and maximal, the excretion of sodium can decreased plasma volume, via the RAS–aldosterone system,
be finely adjusted to the intake so that total body sodium leads to decreased volume excretion.
remains constant. We should emphasize that aldosterone
exerts control over sodium, but not water. While changes
in water excretion may accompany aldosterone-mediated AUTOREGULATION REVISITED
changes in sodium excretion, this is not always the case.
Aldosterone also stimulates sodium transport by other epi- The control of sodium excretion is mediated in part by changes
thelia in the body, namely, sweat and salivary ducts and the in RBF and GFR. However, if changes in these processes are
intestine. The net effect is the same as that exerted on the kid- too large, there are negative consequences. In essence, the
ney: movement of sodium from lumen to blood. Thus, aldos- body cannot allow controls of sodium excretion to “take over”
terone is an all-purpose stimulator of sodium retention. the kidneys to either the detriment of the metabolic health of
In the kidney, aldosterone acts like many other steroid hor- the kidneys or the excretion of other substances. Substantial
mones to increase the genetic expression of key proteins reductions in RBF severely compromise already oxygen-poor
(see Chapter 65). The effect of these proteins is to increase the regions of the kidney such as the medulla. Substantial increases
activity or number of apical membrane sodium channels and in glomerular capillary pressures are likely to damage the
basolateral membrane Na,K-ATPase pumps to promote glomeruli. In addition, the ability of the kidneys to regulate the
increased reabsorption of sodium (Figure 45–6). excretion of water and many substances other than sodium
Angiotensin II produced by the global RAS system is the depends on keeping tubular flow (i.e., GFR) within a certain
main stimulator of aldosterone secretion, although there are limited range. These goals are met by several processes that
others, including elevated plasma potassium concentration, as collectively result in autoregulation of both RBF and GFR
described in Chapter 46 in the context of the renal handling of (see Figure 40–7). While these processes do not exist, strictly
potassium. The atrial natriuretic factors (discussed later) speaking, for the purpose of regulating sodium excretion, they
inhibit aldosterone secretion. Since levels of angiotensin II are nevertheless do affect it.
Autoregulation involves the myogenic response described
in Chapter 40 and a rather complicated intrarenal signaling
LUMEN INTERSTITIUM
system called tubuloglomerular feedback. This feedback
(from the tubule to the glomerulus) is associated with the
macula densa sodium chloride load detector (see Figure 45–4).
Inactive 11ß-HSD Glucocorticoids Earlier in this chapter, we described the actions of the macula
products densa in the context of control over renin secretion, where we
receptor Aldosterone said that in conditions when GFR is very high and therefore
filtering a large amount of sodium, it inhibits renin release.
transcription
And, as detailed below, the macula densa also reduces GFR
protein synthesis and RBF.
ATP
Na+ The macula densa cells at the end of the thick ascending
Na+ K+ limb have Na–K–2Cl symporters that avidly take up Na, Cl,
and K when GFR, and hence, NaCl delivery is high. Sodium
also enters the macula densa cells via a Na–H antiporter. Since
the action of this antiporter causes the cells to lose a hydrogen
ion for every sodium ion entering, this increases intracellular
FIGURE 45–6 Mechanism of aldosterone action. Aldosterone pH. A combination of cellular volume change, increased intra-
enters principal cells and interacts with cytosolic aldosterone cellular chloride, and higher intracellular pH initiates intracel-
receptors. The aldosterone-bound receptors interact with nuclear lular signaling processes that lead to the release of ATP from
DNA to promote gene expression. The aldosterone-induced gene
the basolateral surface of the cells in close proximity to the
products activate sodium channels in the apical membrane and
glomerular mesangial cells (see Figure 39–6). This ATP stimu-
sodium pumps in the basolateral membrane, causing increased
sodium reabsorption. Glucocorticoids such as cortisol are also capable
lates purinergic P2 receptors on the mesangial cells and affer-
of binding to the aldosterone receptor. However, they are inactivated ent arteriolar smooth muscle cells. P2 receptor stimulation
by 11β-hydroxysteroid dehydrogenase (11β-HSD). (Modified with increases calcium in these cells and promotes contraction.
permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Contraction of mesangial cells decreases the effective filtration
Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.) area, which decreases GFR. Contraction of the afferent
CHAPTER 45 Regulation of Sodium and Water Excretion 457

Plasma volume

Activity of renal Arterial GFR, which causes


sympathetic nerves pressure flow to macula densa

Direct
effect
of less NaCl delivery
stretch to macula densa

Renal juxtaglomerular cells


Renin secretion

Plasma renin

Plasma angiotensin II

Adrenal cortex
Aldosterone secretion

Plasma aldosterone

FIGURE 45–7 Pathways by which low plasma volume Cortical collecting ducts
Sodium and H2O reabsorption
leads to increased aldosterone secretion and subsequent
reduction in salt and water excretion. (Reproduced with
permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology,
Sodium and H2O excretion
11th ed. McGraw-Hill, 2008.)

arteriolar smooth muscle cells increases afferent resistance


and decreases RBF and GFR. In addition, it is the increased
OTHER MECHANISMS FOR
calcium in the afferent arteriolar cells that reduces renin secre- CONTROLLING SODIUM BALANCE:
tion. The ATP may also be metabolized to adenosine, which
can stimulate adenosine receptors that produce the same result
NATRIURETIC PEPTIDES
as the P2 receptors (in contrast to the vasodilatory actions of Although there are several other renal mechanisms for con-
adenosine in most other tissues). In addition to purinergic trolling sodium balance independent of water balance, under
agonists that mediate tubuloglomerular feedback, there are normal physiological circumstances, none are as important as
other intrarenal signaling systems, specifically including nitric aldosterone. Only under certain pathophysiological conditions
oxide and arachidonic acid metabolites, that participate in do these other mechanisms contribute significantly to the reg-
modulating the strength of the vasoconstrictive actions. ulation of sodium balance. Important among these are a family
Because the actions of the sodium chloride load detector of hormones called natriuretic peptides, so named because
and tubuloglomerular feedback are complicated, we summa- they promote excretion of sodium in the urine. The key ones
rize them here. High salt content in the thick ascending limb are atrial natriuretic peptide (ANP) and brain natriuretic
of a given nephron generates signals that reduce glomerular peptide (BNP; named as such because it was first discovered
blood flow and reduce filtration in that nephron, thus blunting in the brain). The main source of both natriuretic peptides is
(but not eliminating) the increase in sodium excretion initi- the heart. The natriuretic peptides have both vascular and
ated by other processes in conditions (e.g., volume expansion) tubular actions. They relax the afferent arteriole, thereby pro-
in which the appropriate overall response is increased sodium moting increased filtration, and act at several sites in the
excretion. The same signals that reduce filtration also reduce tubule. They inhibit release of renin, inhibit the actions of
the secretion of renin. angiotensin II that normally promote reabsorption of sodium,
458 SECTION VII Renal Physiology

and act in the medullary collecting duct to inhibit sodium intrarenal baroreceptors. Elevated arterial blood pressure also
absorption. The major stimulus for increased secretion of the exerts direct effects on the kidneys (pressure natriuresis) via a
natriuretic peptides is distention of the atria, which occurs separate intrarenal signaling system.
during plasma volume expansion. This is probably the stimu- When considering mechanisms of sodium excretion, it is
lus for the increased natriuretic peptides that occurs in persons useful to consider two conceptually different categories of
on a high-salt diet. Although most experts assume that these mechanisms: (1) GFR and proximal tubule mechanisms that
peptides play some physiological role in the regulation of lead to coupled changes in sodium and water excretion and (2)
sodium excretion in this and other situations in which plasma distal nephron effects in which sodium can be reabsorbed
volume is expanded, it is not currently possible to quantify independently of water. The proximal mechanisms are pri-
their contribution precisely, although it is surely less than marily involved in excreting excess ECF volume, whereas the
aldosterone. These peptides are greatly increased in patients distal mechanisms alter sodium excretion when ingestion of
with heart failure and can serve as diagnostic indicators. sodium is not balanced by ingestion of water. Both types of
mechanisms can alter blood pressure because of the intimate
relationship among total body sodium and water, blood vol-
SUMMARY OF THE CONTROL ume, and blood pressure.
OF SODIUM EXCRETION
An array of signals, some originating in baroreceptors outside
the kidneys and some originating within the kidneys, alters
CONTROL OF WATER EXCRETION
vascular resistances and transport proteins in order to control As with sodium excretion, water excretion is regulated in part-
sodium excretion, with the main goals of maintaining ECF nership with the cardiovascular system. A central goal in regu-
volume acutely and arterial pressure in the long term lating both salt and water excretion is to preserve vascular
(see Figure 45–8). In response to sodium loads and losses, volume. It is also crucial to maintain plasma osmolality at a
consequent changes in pressure are detected by neural and level that is healthy for tissue cells. It is not surprising then that
intrarenal baroreceptors that directly or indirectly signal the signals related to osmolality and volume are the main regula-
kidneys to modify sodium excretion, thereby excreting loads tors of water excretion.
or preserving existing sodium. GFR, which determines the fil- The relation between urinary water excretion, urinary sol-
tered load of sodium, is a function of arterial pressure and the ute excretion, and urine osmolality is shown in the following
amount of smooth muscle contraction in renal arterioles. expression (where, as before, we approximate osmolality sim-
Sodium reabsorption is controlled by a combination of signals ply as total moles divided by volume):
that affect transport proteins in the renal tubules. The main
Urine solute excretion
controlling signals come from renal sympathetic nerves and Urine water excretion = ________________ (5)
Urine osmolality
the RAS–aldosterone hormonal system. The latter is activated
both by sympathetic nerves and by low pressure at the At a given urine osmolality, water excretion varies directly
with urinary solute excretion. More solute excreted (due, e.g.,
to a higher GFR or reduced sodium reabsorption) means more
Dietary Arterial RAS water excreted (because “water follows the osmoles”). This is
GFR the basis for most diuretics, which promote sodium excretion,
sodium pressure system
and therefore water excretion. But the kidneys can also vary
+ Angiotensin II the amount of water accompanying the excreted solute. They
+ +
– do this by regulating how much water is reabsorbed in the col-
ECF +
lecting ducts. As we already know, the kidneys first generate
volume + hypo-osmotic tubular fluid in the loop of Henle. Then as the
Sodium – Aldosterone fluid subsequently flows through the collecting duct system,
excretion
± + variable amounts of water are reabsorbed by allowing the
Intrarenal Natriuretic tubular fluid to equilibrate to varying degrees with the sur-
messengers peptides rounding interstitium. The final osmolality, and hence final
volume, depends on the peak medullary osmolality and how
FIGURE 45–8 Important variables that affect sodium closely the tubular osmolality approaches that value. We also
excretion. Increases in dietary sodium, particularly via the ensuing
know that equilibration with the interstitium is a function of
rise in ECF volume, lead to increased sodium excretion. Increased
water permeability in the collecting ducts via aquaporins
arterial pressure, increased GFR, and the actions of natriuretic
peptides also increase sodium excretion. Activation of the renin–
under the control of the hormone ADH. Therefore, the regula-
angiotensin system (RAS) decreases sodium excretion via the actions tion of water excretion that is independent of solute excretion
of both angiotensin II and aldosterone. Intrarenal messengers, focuses on controls over ADH secretion.
depending on the specific messenger, can either increase or decrease Antidiuretic hormone (ADH; arginine vasopressin [AVP]
sodium excretion. in humans) is a small peptide (nine amino acids) produced by
CHAPTER 45 Regulation of Sodium and Water Excretion 459

neurons in the hypothalamus. The cell bodies are located in the Excess H2O ingested
supraoptic and paraventricular nuclei of the hypothalamus,
and their axons extend downward to the posterior pituitary
gland, from which ADH is released into the blood (for more
Body fluid osmolarity
detail, see Chapter 61). There is normally a moderate rate of ( H2O concentration)
ADH secretion, allowing considerable water reabsorption in the
renal collecting ducts and resulting in urine that is more con-
centrated than plasma. ADH secretion can increase or decrease
Firing by hypothalamic
from this level, giving the control system a bidirectional respon- osmoreceptors
siveness. And because the collecting ducts are very sensitive to
ADH, this allows the body to control water excretion rate over a
very wide range. There are many sources of synaptic input to the
Posterior pituitary
ADH-secreting neurons. The most important signals originate Vasopressin secretion
in osmoreceptors and cardiovascular baroreceptors.

OSMORECEPTOR CONTROL Plasma vasopressin

OF ADH SECRETION
Plasma osmolality is one of the most tightly regulated vari- Collecting ducts
Tubular permeability
ables in the body. It is set mainly by the ratio of ECF sodium to H2O
(plus its associated anions) to water. Other solutes (e.g., glu-
cose and potassium) make some contribution, but those other
H2O reabsorption
solutes are regulated for reasons other than their osmolality.
Thus, except under unusual circumstances, variations in
plasma osmolality reflect variations in sodium concentration.
If the body keeps the inputs and outputs of sodium and water H2O excretion
matched in lock step, osmolality remains constant. But inputs
are often not matched. The major effect of gaining or losing FIGURE 45–9 Mechanism for increased water excretion in
water or salt without corresponding changes in the other is a response to a pure water load. Decreased plasma osmolality leads,
change in the osmolality of the body fluids. When osmolality via osmoreceptors, to decreased secretion of ADH (vasopressin), in
turn causing decreased collecting duct water reabsorption and more
deviates from normal, strong reflexes come into play to change
water excretion. (Reproduced with permission from Widmaier EP, Raff H, Strang
ADH secretion, and thus change the excretion of water. Key
KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
receptors that initiate reflexes controlling ADH secretion are
osmoreceptors: neurons responsive to changes in osmolality.
Most osmoreceptors are located in tissues surrounding the The osmoreceptor–ADH system is very sensitive, respond-
third cerebral ventricle. These tissues contain fenestrated cap- ing to an osmolality change of only 1 or 2 mOsm/kg. However,
illaries, which allow rapid adjustment of interstitial composi- common perturbations are often a good deal greater than this.
tion when plasma composition changes. The hypothalamic For example, if a 70-kg person consumes 1 L of pure water,
cells that secrete ADH receive synaptic input from the osmore- ECF osmolality is reduced by about 7 mOsm/kg. And exercise
ceptors. Via these connections, an increase in osmolality for several hours on a warm day can increase ECF osmolality
increases their rate of ADH secretion. In turn, this causes by 10 mOsm/kg or more. Such routine perturbations result in
water permeability of the collecting ducts to increase, water strong ADH responses that remain active until osmolality
reabsorption is maximal, and a very small volume of highly returns to its previous value. ADH has a plasma half-life of
concentrated (hyperosmotic) urine is excreted. By this means, only a few minutes, so prolonged stimulation of water perme-
relatively less filtered water than solute is excreted, which low- ability in the kidneys requires continuous stimulation of the
ers body fluid osmolality toward normal. Conversely, decreased ADH-secreting neurons.
osmolality inhibits ADH secretion. For example, when a per-
son drinks pure water, the excess water lowers the body fluid
osmolality, which inhibits ADH secretion via the hypotha- BARORECEPTOR CONTROL
lamic osmoreceptors. As a result, water permeability of the OF ADH SECRETION
collecting ducts becomes very low, little water is reabsorbed
from these segments, and a large volume of extremely dilute There is a second major influence on ADH secretion. This
(hypo-osmotic) urine is excreted. In this manner, the excess originates in systemic baroreceptors (the same ones that influ-
water is rapidly eliminated and plasma osmolality is increased ence sympathetic drive to the kidneys). A decreased extracel-
(see Figure 45–9). lular volume or major decrease in arterial pressure reflexively
460 SECTION VII Renal Physiology

Plasma volume
peripheral resistance, which helps restore arterial blood pres-
sure independently of the slower restoration of body fluid vol-
umes. Renal arterioles and mesangial cells also participate in
this constrictor response, so a high plasma concentration of
Venous, atrial, and arterial
pressures
ADH, quite apart from its effect on tubular water permeabil-
ity, may promote retention of both sodium and water by low-
Reflexes mediated ering GFR.
by cardiovascular
baroreceptors We have described two different major afferent pathways
controlling the ADH-secreting hypothalamic cells: one from
Posterior pituitary
Vasopressin secretion baroreceptors and one from osmoreceptors. These hypotha-
lamic cells are, therefore, true integrators, whose activity is
determined by the total synaptic input to them. Thus, a simul-
taneous increase in plasma volume and decrease in body fluid
Plasma vasopressin
osmolality causes strong inhibition of ADH secretion. Con-
versely, a simultaneous decrease in plasma volume and increase
in osmolality produces very marked stimulation of ADH
Collecting ducts secretion. However, what happens when baroreceptor and
Tubular permeability
to H2O osmoreceptor inputs oppose each other (e.g., if plasma volume
and osmolality are both decreased)? In general, because of the
high sensitivity of the osmoreceptors, the osmoreceptor influ-
H2O reabsorption
ence predominates over that of the baroreceptors when
changes in osmolality and plasma volume are small to moder-
ate. However, a dangerous reduction in plasma volume will
H2O excretion take precedence over decreased body fluid osmolality in influ-
encing ADH secretion; under such conditions, water is
FIGURE 45–10 Decreased water excretion in response to retained in excess of solute even though the body fluids become
decreased plasma volume. Low pressure sensed by neural hypo-osmotic (for the same reason, plasma sodium concen-
baroreceptors reduces inhibition of the hypothalamic cells whose tration decreases). In essence, when blood volume reaches a
axons release ADH (vasopressin) from the posterior pituitary gland.
life-threatening low level, it is more important for the body to
The subsequent increase in ADH increases water reabsorption in the
collecting ducts and helps preserve existing volume. (Reproduced with
preserve vascular volume and thus ensure an adequate cardiac
permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed.
output than it is to preserve normal osmolality.
McGraw-Hill, 2008.) The ADH-secreting cells also receive synaptic input from
many other brain areas. Thus, ADH secretion and, hence,
urine flow can be altered by pain, fear, and a variety of other
activates increased ADH secretion. The response is mediated factors, including drugs such as alcohol, which inhibits ADH
by neural pathways originating in cardiopulmonary barore- release. However, this complexity should not obscure the gen-
ceptors, and if arterial pressure decreases, from arterial barore- eralization that ADH secretion is determined over the long
ceptors (see Figure 45–10). term primarily by the states of body fluid osmolality and
Decreased cardiovascular pressures cause less firing by the plasma volume.
baroreceptors, which relieves inhibition of stimulatory path- Figure 45–11 summarizes the major factors known to con-
ways and results in more ADH secretion. In effect, the low car- trol renal sodium and water excretion in response to severe
diovascular pressures are interpreted as low volume, and the sweating. Sweat is a hypo-osmotic solution containing mainly
response of increased ADH appropriately serves to minimize water, sodium, and chloride. Therefore, sweating causes both a
loss of water (i.e., volume). Conversely, baroreceptors are stim- decrease in ECF volume and an increase in body fluid osmola-
ulated by increased cardiovascular pressures, interpreted as lity. The renal retention of water and sodium helps preserve
excess volume, and this causes inhibition of ADH secretion. existing water and salt that are depleted by sweating.
The decrease in ADH results in decreased reabsorption of
water in the collecting ducts, and more excretion. The adap-
tive value of these baroreceptor reflexes is to help stabilize ECF THIRST AND SALT APPETITE
volume and, hence, blood pressure.
There is a second adaptive value to this reflex: large Large deficits of salt and water can be only partly compensated
decreases in plasma volume, as might occur after a major by renal conservation of these substances, and ingestion is the
hemorrhage, elicit such high concentrations of ADH—much ultimate compensatory mechanism. The centers that mediate
higher than those needed to produce maximal antidiuresis— thirst are located in the hypothalamus (very close to those
that the hormone is able to exert direct vasoconstrictor effects areas that produce ADH). The subjective feeling of thirst,
on arteriolar smooth muscle. The result is increased total which drives one to obtain and ingest water, is stimulated both
CHAPTER 45 Regulation of Sodium and Water Excretion 461

Begin

Severe sweating

Loss of hypoosmotic
salt solution

Plasma volume Plasma osmolarity


( H2O concentration)

GFR Plasma Plasma vasopressin


Reflexes Reflexes
aldosterone

Sodium excretion H2O excretion

FIGURE 45–11 Coordinated response to severe sweating. A combination of decreased ECF volume and increased plasma osmolality
activates reflexes that preserve both salt and water. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed.
McGraw-Hill, 2008.)

by reduced plasma volume and by increased body fluid osmo- salt deficient and (2) their drive to obtain salt is markedly
lality. The adaptive significance of both is self-evident. Note increased in the presence of deficiency.
that these are precisely the same changes that stimulate ADH The significance of these animal studies for humans, how-
production, and the receptors—osmoreceptors and the nerve ever, is unclear. Salt craving does seem to occur in humans
cells that respond to the cardiovascular baroreceptors—that who are severely salt depleted, but the contribution of such
initiate the ADH-controlling reflexes are near those that initi- regulatory salt appetite to everyday sodium homeostasis in
ate thirst. The thirst response, however, is significantly less healthy persons is probably slight. On the other hand, humans
sensitive than the ADH response. do seem to have a strong hedonistic appetite for salt, as mani-
There are also other pathways controlling thirst. For exam- fested by almost universally large intakes of sodium whenever
ple, dryness of the mouth and throat causes profound thirst, it is cheap and readily available. Thus, the average American
which is relieved by merely moistening them. Also, when ani- intake of salt is 10–15 g per day even though humans can sur-
mals such as the camel (and humans, to a lesser extent) become vive quite normally on less than 0.5 g per day. As pointed out
markedly dehydrated, they will rapidly drink just enough previously, a large salt intake may be a contributor to the
water to replace their previous losses and then stop. What is pathogenesis of hypertension in susceptible individuals.
amazing is that when they stop, the water has not yet had time
to be absorbed from the gastrointestinal tract into the blood.
Some kind of metering of the water intake by the gastrointes-
tinal tract has occurred, but its nature remains a mystery. Neu- CLINICAL CORRELATION
ral afferents from the pharynx and upper gastrointestinal tract
A 57-year-old man has a long history of smoking and
are likely to be involved.
hypertension, but in prior years, has refused the use of any
Angiotensin II is yet another factor that stimulates thirst: by
medications for treatment. Six months ago, he suffered a
its direct effect on the brain. This hormone constitutes one of
fall from a tractor and injured the right side of his back. He
the pathways by which thirst is stimulated when ECF volume
recovered well, but 2 weeks ago, his blood pressure was
is decreased.
147/102, and his physician finally convinced him to start
Salt appetite, which is the analog of thirst, is also an
on an ACE inhibitor. At this follow-up visit, his blood pres-
extremely important component of sodium homeostasis in
sure was only mildly increased, but he was feeling very tired
most mammals. It is clear that salt appetite in these species is
and agitated, without any specific symptoms. Laboratory
innate and consists of two components: (1) hedonistic appetite
analysis of his blood revealed azotemia (high levels of crea-
and (2) regulatory appetite. In other words, (1) animals like
tinine and urea), and he was referred to a nephrologist.
salt and eat it whenever they can regardless of whether they are
462 SECTION VII Renal Physiology

■ A major action of angiotensin II is to stimulate sodium reabsorp-


Further tests, after taking him off all medications, showed tion via Na–H antiporter activity in the proximal tubule.
activation of the RAS. When he was put back on the ACE ■ Elevated renal artery pressure leads to increased sodium
inhibitor, his renin levels increased markedly, but not his excretion (pressure natriuresis) without external signals.
aldosterone. At this point, a renal angiogram indicated ■ Long-term regulation of sodium excretion and, therefore,
moderate left renal artery blockage and major right renal blood pressure centers on the actions of aldosterone in the
artery blockage. distal nephron.
His diagnosis is renovascular hypertension, caused by ■ Water excretion tends to parallel solute excretion, but is also
activation of the RAS system. He has generalized athero- regulated independently by controlling water reabsorption in
sclerosis, but the major problem is stenosis (narrowing) of the collecting ducts.
his right renal artery, perhaps exacerbated by trauma dur- ■ ADH secretion is regulated by plasma osmolality via hypotha-
ing his accident 6 months prior. Decreased renal perfusion lamic osmoreceptors, and blood pressure, via the baroreceptor–
pressure activates renin secretion from his right kidney, in vasomotor center system.
turn leading to increased angiotensin II and aldosterone.
This is causing a significant increase in blood pressure. The
high blood pressure and levels of angiotensin II suppress STUDY QUESTIONS
secretion of renin from his left kidney, but this makes no
1. Which of the following cell types are not nerve cells?
difference in the face of the high secretion from the right
A) pituitary cells that secrete ADH
kidney. The increased pressure in the left renal artery main-
B) baroreceptors located in pulmonary vessels
tains total GFR sufficiently high to provide adequate excre-
C) baroreceptors located in the arch of the aorta
tion of nitrogenous waste. However, after an ACE inhibitor D) intrarenal baroreceptors
is given, his levels of angiotensin II and aldosterone
2. In the normal RAS system leading to the production of
decrease. Consequently, his systemic and left renal artery aldosterone, the rate-limiting step is
pressure decreases, resulting in a decrease in GFR and
A) the production of angiotensin I.
excretion of nitrogenous waste, leading to the azotemia. B) the production of angiotensinogen.
Furthermore, when he is given an ACE inhibitor and there C) the activity of ACE.
are decreased levels of angiotensin II, the suppressive effect D) the responsiveness of the adrenal gland to angiotensin II.
that the usually high angiotensin II is having on his left kid- 3. A person eats a large bag of very salty potato chips with no
ney is removed, and renin secretion from the left kidney beverage. Which response is most likely to ensue?
rises markedly. Renal artery stenosis is sometimes treated A) movement of aquaporins into the membrane of cortical
by renal artery angioplasty and placement of a stent to collecting duct principal cells
maintain the patency of the renal artery. B) enhanced activity of Na–H antiporters in the proximal
tubule
C) enhanced activity of Na,K-ATPase pumps in collecting duct
principal cells
CHAPTER SUMMARY D) decreased levels of natriuretic peptides in the blood
■ Sodium and water excretion are regulated primarily to meet 4. In response to a major hemorrhage
the needs of the cardiovascular system via preservation of A) GFR increases.
vascular volume, blood pressure, and plasma osmolality. B) ADH secretion is reduced.
■ Baroreceptors at various sites inform the kidneys of vascular C) granular (juxtaglomerular) cells are stimulated by
pressures and volume status. neural input.
■ Angiotensin II, produced by local and systemic RAS, is a crucial D) neural baroreceptor firing rate increases.
regulator of sodium excretion and blood pressure via its actions 5. Tubuloglomerular feedback contributes to the regulation of
in the kidneys, peripheral vasculature, and adrenal glands. A) GFR.
■ Sodium excretion is regulated by both the rate of filtration B) sympathetic neural activity.
(GFR) and rate of reabsorption. C) ADH secretion.
■ ECF volume status is a major determinant of sodium excretion. D) ACE activity.
46
C H A P T E R

Regulation of
Potassium Balance
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ State the normal balance and distribution of potassium between cells and
extracellular fluid.
■ Describe how potassium moves between cells and the extracellular fluid,
and how, on a short-term basis, the movement protects the extracellular
fluid from large changes in potassium concentration.
■ Describe how plasma levels of potassium do not always reflect the status of
total body potassium.
■ State how insulin and epinephrine influence the cellular uptake of potassium,
and identify the situations in which these hormonal influences are most
important.
■ State the relative amounts of potassium reabsorbed by the proximal tubule and
thick ascending limb of Henle’s loop regardless of the state of potassium intake.
■ Describe how nephron segments beyond the thick ascending limb can
manifest net secretion or reabsorption; describe the role of principal cells
and intercalated cells in these processes.
■ List inputs that control the rate of potassium secretion by the distal nephron.
■ Describe the actions of renal outer medulla (ROMK) and BK potassium
channels in conditions of low, normal, and high potassium excretions.
■ Describe how changes in plasma potassium influence aldosterone secretion.
■ State the effects of most diuretic drugs and osmotic diuretics on potassium
excretion.
■ Describe the association between perturbations in acid–base status and the
plasma potassium level.

REGULATION OF POTASSIUM centration of potassium in the ECF is a closely regulated quan-


tity. Major increases and decreases (called hyperkalemia and
BETWEEN THE INTRACELLULAR hypokalemia) from the normal plasma values of 3.5–5 mEq/L
are cause for medical intervention. The importance of main-
AND EXTRACELLULAR taining this concentration relatively constant stems primarily
COMPARTMENTS from the role of potassium in the excitability of nerve and
muscle, especially the heart (review Chapters 4 and 23).
Potassium, like all other important ions, is distributed between Given that the vast majority of body potassium is within
the intracellular fluid and extracellular fluid (ECF) of the body. cells, the extracellular potassium concentration is crucially
The vast majority of potassium is intracellular, and only about dependent on (1) the total amount of potassium in the body
2% of total body potassium is in the ECF. This small fraction, and (2) the distribution of this potassium between the extra-
however, is absolutely crucial for body function, and the con- cellular and intracellular fluid compartments. Total body

463

Ch46_463-470.indd 463 11/26/10 10:34:08 AM


464 SECTION VII Renal Physiology

potassium is determined by the balance between potassium into cells rather than allowing it to accumulate in the ECF.
intake and excretion. Healthy individuals remain in potassium This new potassium then slowly comes out of cells between
balance, as they do in sodium balance, by excreting an amount meals to be excreted. Moreover, a large increase in plasma
of urinary potassium equal to the amount of potassium potassium concentration facilitates insulin secretion at any
ingested minus the small amounts eliminated in the feces and time, and the additional insulin induces greater potassium
sweat. Normally, potassium losses via sweat and the gastroin- uptake by the cells, a negative feedback system for opposing
testinal tract are small, but very large quantities can be lost acute elevations in plasma potassium concentration. In the
from the digestive tract during vomiting or diarrhea. The con- natural order of things, insulin also stimulates glucose uptake
trol of renal potassium handling is the major mechanism by and metabolism by cells: a necessary source of energy to drive
which total body potassium is maintained in balance. the insulin-activated Na,K-ATPase responsible for moving
The fact that most body potassium is intracellular follows potassium into cells.
strictly from the size and properties of the intracellular and The effect of epinephrine on cellular potassium uptake is
extracellular compartments. About two thirds of the body probably of greatest physiological importance during exercise
fluids are intracellular (the collective cytosolic volume of all when potassium moves out of muscle cells that are rapidly
the cells in the body), and typical cytosolic potassium con- firing action potentials. In fact, very intense intermittent
centrations are about 140–150 mEq/L. One third of the body exercise such as wind sprints can transiently double plasma
fluids are extracellular, with a potassium concentration of potassium. However, at the same time, exercise increases
about 4 mEq/L. In a clinical setting, only the extracellular adrenal medullary secretion of epinephrine, which stimulates
concentration can be measured (the intracellular potassium potassium uptake by muscle and other cells. Similarly, trauma
is, in a sense, hidden behind the wall of the cell membrane). causes loss of potassium from damaged cells, and epineph-
Furthermore, the extracellular value does not necessarily rine released due to stress stimulates other cells to take up
reflect total body potassium. A patient may, for example, be plasma potassium.
hyperkalemic and yet at the same time be depleted of total Still another influence on the distribution of potassium
body potassium. between the intracellular fluid and ECF is the ECF hydrogen
The high level of potassium within cells is maintained by the ion concentration: an increase in ECF hydrogen ion concen-
collective operation of the Na,K-ATPase plasma membrane tration (acidemia; see Chapter 47) is often associated with net
pumps, which actively transport potassium into cells. Because potassium movement out of cells, whereas a decrease in ECF
the amount of potassium in the extracellular compartment is hydrogen ion concentration (alkalemia) causes net potassium
very small (40–60 mEq total), even very slight shifts of potas- movement into them. It is as though potassium and hydrogen
sium into or out of cells produce large changes in extracellular ions were exchanging across plasma membranes (i.e., hydro-
potassium concentration. Similarly, a meal rich in potassium gen ions moving into the cell during acidemia and out during
(e.g., steak, potato, and spinach) could easily double the extra- alkalemia and potassium doing just the opposite), but the pre-
cellular concentration of potassium if most of that potassium cise mechanism underlying these “exchanges” has not yet been
were not transferred from the blood to the intracellular com- clarified. However, like the effect of insulin, it probably involves
partment. It is crucial, therefore, that dietary loads be taken up an inhibition (acidemia) or activation (alkalemia) of the Na,K-
into the intracellular compartment rapidly to prevent major ATPase.
changes in plasma potassium concentration. The tissue con-
tributing most to the sequestration of potassium is skeletal
muscle, simply because it contains the largest collective intra- RENAL POTASSIUM HANDLING
cellular volume. Muscle effectively buffers extracellular potas-
sium by taking up or releasing it and keeping the plasma OVERVIEW
potassium concentration close to normal. On a moment-to-
moment basis, this is what protects the ECF from large swings Although other tissues play an important role in the moment-
in potassium concentration. Major factors involved in these to-moment control of plasma potassium concentration, in the
homeostatic processes include insulin and epinephrine, both final analysis, the kidney determines total body potassium
of which cause increased potassium uptake by muscle (and content. Therefore, understanding potassium handling by the
certain other cells) through stimulation of plasma membrane kidneys is the key to understanding potassium balance. Potas-
Na,K-ATPases. Another influence is the GI tract, which con- sium is freely filtered into Bowman’s space. Under all condi-
tains an elaborate neural network (the “gut brain”) that sends tions, almost all the filtered load (~90%) is reabsorbed by the
signals to the central nervous system. It also contains a com- proximal tubule and thick ascending limb of the loop of
plement of enteroendocrine cells that releases an array of Henle. Then, if the body is trying to conserve potassium, most
peptide hormones. Together these neural and hormonal sig- of the rest is reabsorbed in the distal nephron and medullary
nals affect many target organs, including the kidneys (see later collecting ducts, leaving almost none in the urine. In contrast,
discussion) in response to dietary input. if the body is ridding itself of potassium, a large amount is
The increase in plasma insulin concentration after a meal is secreted in the distal nephron, resulting in a large excretion.
a crucial factor in moving ingested and absorbed potassium When secretion occurs at high rates, the amount excreted may
CHAPTER 46 Regulation of Potassium Balance 465

TABLE 46–1 Summary of tubular potassium transport. loads are high. It is in these distal segments where most regula-
tion of potassium excretion is exerted. The distal nephron is
Low-potassium Diet composed of a number of segments, including the distal con-
Normal- or High- or Potassium voluted tubule, connecting tubule, initial collecting tubule,
Transport potassium Diet Depletion
and cortical collecting duct; that is, all of the tubule segments
Proximal tubule Reabsorption Reabsorption (55%) between the end of the thick ascending limb and the medul-
(60–80%) lary collecting duct. It is not possible to finely differentiate
Thick ascending limb Reabsorption Reabsorption (30%) between these segments in terms of function, although the
(5–25%) connecting tubule stands out as being particularly important
Distal convoluted Secretion Reabsorption in potassium handling because of its rich complement of
tubule transport elements. It appears that most of the potassium
Principal cells, Substantial Little secretion secretion occurs prior to segments where most of the water is
connecting tubule, and secretion (>15%) absorbed (cortical collecting duct). Finally, the medullary col-
cortical collecting duct lecting ducts reabsorb small amounts of potassium under all
H,K-ATPase-containing Reabsorption Reabsorption (10%) conditions. When the sum of upstream processes has already
intercalated cells, (10%) reabsorbed almost all the potassium, the medullary collecting
cortical collecting duct ducts bring the final urine excretion down to a few percent of
H,K-ATPase-containing Reabsorption Reabsorption (5%) the filtered load, for an excretion of about 10–15 mEq per day.
cells, medullary (5%) On the other hand, if upstream segments are secreting avidly,
collecting duct the modest reabsorption in the medullary collecting ducts
Percentages are in reference to the filtered load of potassium. H, hydrogen; K, does little to prevent an excretion that can reach 1,000 mEq
potassium; ATPase, adenosine triphosphatase.
per day. Figure 46–1A and B depicts the overall renal handling
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009. of potassium in different tubule regions in conditions of high
and low potassium excretion.
A complication in the renal handling of potassium in all
exceed the filtered load. The chief means of regulation lies in regions, specifically including the proximal tubule and thick
control of secretion in parts of the nephron beyond the loop of ascending limb, is that its active transport is always coupled to
Henle. Let us look at potassium handling by various nephron the active transport of another solute. Active influx of potas-
segments and then address the issue of control. sium across the basolateral membrane via the ubiquitous
Since potassium is freely filtered, a normal plasma level of Na,K-ATPase is coupled to efflux of sodium, while influx of
4 mEq/L and GFR of 150 L per day or more results in a daily potassium across apical membranes via H–K antiporters is
filtered load of about 600 mEq per day. The subsequent events accompanied by efflux of protons. Thus, in describing the
in various tubule segments are summarized in Table 46–1. In renal handling of potassium in various segments, we always
the proximal tubule, about 65% of the filtered load is reab- have to keep in mind the fate of these other solutes. In the
sorbed, mostly via the paracellular route. Much of the flux is proximal tubule, the Na,K-ATPase in the basolateral mem-
driven by the concentration gradient set up when water is brane is very active in moving sodium from the cell to the
reabsorbed (thus concentrating all solutes remaining in the interstitium, necessitating that potassium be simultaneously
tubular lumen). Some may also move by entrainment with the taken up from the interstitium. Since we know that potassium
rapidly reabsorbed water (solvent drag). Either way, this is being put into the interstitium surrounding the proximal
accounts for major potassium absorption in an essentially tubule, this pumped potassium must therefore recycle right
unregulated manner. In the loop of Henle, there is additional back to the interstitium by passive flux through channels in
reabsorption. The major events take place in the thick ascend- the basolateral membrane.
ing limb, where the Na–K–2Cl multiporter in the apical In the thick ascending limb, the interaction with sodium is
membrane reabsorbs potassium (see Figure 44–3). Some of even more complicated. As mentioned above, potassium is
this potassium is returned to the lumen across the apical mem- actively transported into the cells across both membranes and
brane via potassium channels, and the rest exits the cells across exits the cells passively across both membranes. It is pumped
the basolateral membrane by a combination of passive flux into the epithelial cells from the tubular lumen with sodium
through channels and through symporters with chloride, via Na–K–2Cl antiporters and from the interstitium via the
resulting in net transcellular reabsorption. Some potassium is Na,K-ATPase. Since there is far less potassium than sodium in
also reabsorbed by the paracellular route in this segment, the lumen, potassium must recycle back to the lumen by pas-
driven by a lumen-positive voltage. Usually about 25% of the sive channel flux to keep a supply of potassium available to run
filtered load is reabsorbed in the thick ascending limb, so that the multiporter with sodium. Otherwise, sodium reabsorption
only about 10% is passed on to the distal nephron. would be limited only to the amount of potassium present in
In the distal nephron, there is continuous reabsorption if the tubular fluid. Quantitatively, the sum of all these transcel-
dietary loads are very small, but a major superimposed secre- lular and paracellular processes is net reabsorption of about
tion that greatly exceeds the amount reabsorbed when dietary 25% of the filtered load.
466 SECTION VII Renal Physiology

1 1
3 3
2 2

5 4 5 4

A B
FIGURE 46–1 Potassium transport at different locations of the tubule in conditions of low or high excretion. In low excretion (A)
the majority of filtered potassium is reabsorbed in the proximal tubule, mainly by the paracellular route (1). In the thick ascending limb (2)
most of the rest is reabsorbed, mostly by the transcellular route. In the cortical (3) and medullary collecting duct (4) there is some additional
reabsorption via intercalated cells. Some of the potassium reabsorbed into the medullary interstitium recycles back into the thin limbs of the
loop of Henle (5). In high excretion (B) the events in most regions of the tubule are the same as when there is little potassium excretion, but
in the distal nephron, particularly in the connecting tubule, there is major secretion (6) that in some cases is greater than the sum of the
reabsorptive processes. (Modified with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill,
Medical Pub. Division, 2009.)

SECRETION IN THE DISTAL distal nephron, two types of channels stand out as being those
NEPHRON AND ITS REGULATION that secrete potassium in a regulated manner: ROMK (stand-
ing for renal outer medulla, because that is where they were
There are two cell types in the epithelium of the distal nephron: first identified) and BK (since each channel has a “big” capac-
principal cells (about 70% of the cells) and intercalated cells. ity to secrete potassium, also called maxi-K). While ROMK
The intercalated cells are further subdivided into type A (more and BK channels both conduct potassium, they play different
numerous) and type B (sparse) intercalated cells. The princi- roles and are regulated by quite different mechanisms. At very
pal cells secrete potassium at highly variable rates, while the low dietary loads of potassium, there is virtually no secretion
type A intercalated cells reabsorb potassium. The principles
governing both secretion and reabsorption are straightfor-
ward. Secretion of potassium by principal cells involves the LUMEN INTERSTITIUM
uptake of potassium from the interstitium via the Na,K-
ATPase and secretion into the tubular lumen through chan-
nels (Figure 46–2). Type A intercalated cells reabsorb amiloride
potassium via the H,K-ATPase in the apical membrane, which aldosterone
actively takes up potassium from the lumen. They then allow ATP
Na+ Na+ Na+ Na+
potassium to enter the interstitium across the basolateral
K+ K+
membrane via potassium channels.
Regulation of potassium excretion involves multiple con-
trols over the secretory processes in the distal nephron, some- K+ K+ K+ K+
thing like a passenger van where everyone in it has an
accelerator and a brake pedal. As is the case with regulation of
sodium excretion, we cannot predict just how these controls
operate in every situation. Fortunately, with potassium as well
as sodium, the healthy kidneys do a remarkable job of increas-
FIGURE 46–2 Generalized pathway for potassium
secretion by principal cells. Potassium secretion is tied to
ing potassium excretion in response to high dietary loads and sodium reabsorption via the Na,K-ATPase. The drug amiloride
reducing excretion in the face of restricted diets. Much of the inhibits sodium entry, and therefore inhibits potassium secretion.
regulation involves controlling the activity of potassium chan- Aldosterone stimulates both sodium and reabsorption and
nels. The kidneys and other body organs express numerous potassium secretion at multiple points. (Modified with permission
potassium channel species; for simplicity we do not usually from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange
differentiate between types. However, in principal cells of the Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
CHAPTER 46 Regulation of Potassium Balance 467

Low K excretion Normal K excretion High K excretion

ROMK BK ROMK BK ROMK BK


(sequestered) (closed) (open) (closed) (open) (open)

K K K
FIGURE 46–3 Activity of ROMK and BK potassium channels in principal cells under different conditions. When the body is
conserving potassium and little is being excreted, ROMK channels are mostly sequestered in intracellular vesicles and BK channels are closed;
thus, there is virtually no secretion. Under modest potassium loads (normal conditions), ROMK channels secrete potassium, while BK channels
remain closed. When potassium excretion is very high, as on a high-potassium diet, ROMK channel activity is maximized and BK channels are
open, allowing substantial secretion. (Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/
McGraw-Hill, Medical Pub. Division, 2009.)

by either kind of channel. ROMK channels are sequestered in Thus, plasma potassium concentration does exert an in-
intracellular vesicles and BK channels are closed. At normal fluence on potassium excretion, but is not the dominant
potassium loads, ROMK channels are moved to the apical factor under normal conditions.
membrane and secrete potassium. BK channels are still closed, 2. Aldosterone. We discussed the role of aldosterone in reg-
held in reserve and ready to respond to appropriate signals ulating sodium excretion in Chapter 45. Here we describe
when needed. At high excretion rates, both types of channel its role in potassium excretion. One stimulator of aldos-
are present in the luminal membrane and avidly secreting terone secretion is an increase in plasma potassium con-
potassium (Figure 46–3). centration. This is a direct action of potassium on the ad-
Figure 46–4 shows factors known to influence the secretion, renal cortex and does not involve the renin–angiotensin
and thus the ultimate excretion of potassium. The following system. Aldosterone, as well as increasing expression of
provides a brief description of how specific factors affect the Na,K-ATPase, also stimulates expression of ROMK
potassium excretion: channels in the distal nephron. Both actions have the ef-
1. Plasma potassium. The role of plasma potassium is the fect of increasing potassium secretion. Greater pumping
most understandable influence. First, the filtered load is by the Na,K-ATPase supplies more potassium from the
directly proportional to plasma concentration. Second, interstitium to the cytosol of the principal cells, and more
the environment of the principal cells, that is, the cortical ROMK channels provide more pathways for secretion.
interstitium, has a potassium concentration that is nearly 3. Delivery of sodium to the distal nephron. Any change in
the same as in plasma. The Na,K-ATPase that takes up sodium handling prior to the distal nephron determines
potassium is highly sensitive to the potassium concentra- how much is sent on from the thick ascending limb, that is,
tion in this space, and varies its pump rate up and down delivered to the distal nephron. Changes in upstream han-
when potassium levels in the plasma vary up and down. dling of sodium include changes in filtered load and reab-

High flow rate


High Na delivery
High potassium in distal nephron
to distal nephron
diet

Aldosterone

Nonchloride anions High plasma


in distal nephron potassium
Potassium
secretion in
distal
nephron

FIGURE 46–4 Factors that increase secretion of potassium by principal cells as described in the text. (Modified with permission from Eaton
DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
468 SECTION VII Renal Physiology

sorption in prior segments. Sodium delivery influences the apical membrane. Another adaptation to prolonged
potassium secretion because more sodium delivered means periods of low potassium ingestion is an increase in H,K-
more sodium taken up by principal cells, and therefore ATPase activity in intercalated cells, resulting in even
more sodium pumped out by the Na,K-ATPase, in turn more efficient reabsorption of filtered potassium.
causing more potassium to be pumped in. The increased
intracellular potassium can just recycle back to the intersti-
tium, but the usual result is more potassium secretion. PERTURBATIONS IN RENAL
4. Distal nephron flow rate. The role of flow rate in regulat- HANDLING OF POTASSIUM
ing potassium secretion is a story by itself. Increased flow
is detected by mechanosensitive elements of the principal A potential problem in the renal handling of potassium that is not
cells. This includes bending of the central cilium that pro- a problem in healthy kidneys is the simultaneous balance of
trudes from the apical surface into the tubule lumen. sodium and potassium. Given that so much of the transport of
Bending of the central cilium initiates intracellular release these ions is by coupled mechanisms, it is remarkable that the
of calcium and activation of BK channels. Under most kidneys can deal with every combination of dietary load: both
conditions, increased delivery of sodium is the chief cause high, both low, one high, etc. This is all the more remarkable
of increased flow, because the sodium is accompanied by when we consider that aldosterone is a regulator of both. If a per-
water. Thus, increased delivery of sodium implies in- son is consuming very little sodium or potassium, we expect, in
creased flow. Increased flow has another effect. By sweep- order to preserve body stores of sodium, for aldosterone levels to
ing away potassium that reaches the tubule by secretion, be high enough to stimulate avid reabsorption of sodium. But
luminal potassium concentration is kept low enough to this should also lead to avid secretion of potassium, which is an
preserve a favorable concentration gradient for secretion. unwanted action since the body is also trying to conserve potas-
5. Concentration of nonchloride anions. In order for princi- sium. The answer is simply that potassium cannot be secreted
pal cells to secrete potassium there must be a route (open unless there are open apical channels. If the actions of intracellu-
channels and/or functioning transporters) and driving forc- lar signaling cascades have caused most of the ROMK channels
es (electrochemical gradients). In addition to the majority to be sequestered in intracellular vesicles, then potassium that is
secretion via potassium channels, a smaller component in- taken up from the interstitium via the Na,K-ATPase recycles back
volves secretion via potassium-chloride symport. Under via basolateral channels to the interstitium and is not secreted.
conditions in which the luminal chloride concentration is
low due to replacement of luminal chloride with anions that
are not usually in high concentration, the effect is to increase Effects of Diuretics
the electrochemical gradient for chloride secretion. In turn Diuretics are agents that increase urine flow and reduce ECF
this increases the normally modest secretion of potassium volume, usually by increasing the renal excretion of sodium.
via potassium-chloride symport. Most diuretics have the unwanted side effect of simultaneously
6. Dietary potassium. The influence of dietary potassium increasing the renal excretion of potassium. Potassium excre-
on renal function is both the most obvious regulator of tion is almost always increased in individuals undergoing
potassium excretion and the least understood. A major osmotic diuresis (high filtration of solute that is not reab-
task of the kidneys is to maintain potassium balance by sorbed) or treatment with diuretics that block sodium reab-
increasing and decreasing potassium excretion in parallel sorption in the proximal tubule, loop of Henle, or distal
with dietary load. The healthy kidneys do this very well. convoluted tubule (i.e., sites that are upstream from the princi-
The problem is in understanding the signaling—how do pal cells). All of these events increase flow rate past the
the kidneys know how much potassium a person has con- potassium-secreting principal cells, which is a major stimula-
sumed? While very large potassium loads can increase tor of potassium secretion. The potassium loss may cause
plasma potassium somewhat, the changes in excretion as- severe potassium depletion (see Figure 46–5).
sociated with diet do not seem to be accounted for on the Let us integrate this information about diuretics with our
basis of changes in either plasma potassium or the other understanding of the action of aldosterone. High aldosterone
identified factors. However, the previously mentioned levels in individuals with heart failure or other diseases of sec-
gastrointestinal signals influence not only the cellular up- ondary hyperaldosteronism generally do not cause potassium
take of potassium absorbed from the GI tract, but also re- hypersecretion because these patients simultaneously have
nal handling of potassium, and seem to be one of the links low fluid delivery to the distal nephron. However, consider
between dietary load and excretion. One of the manifesta- what happens when such persons are treated with diuretics to
tions of changing dietary loads is to regulate the distribu- eliminate their retained sodium and water. The diuretics
tion of ROMK channels between the apical membrane increase fluid delivery to the distal nephron, and now patients
and intracellular storage, that is, high-potassium diets lead have both increased aldosterone and increased flow. This
to insertion of apical channels and therefore higher potas- combination causes marked increases in potassium secretion
sium secretion. In contrast, during periods of prolonged and excretion. To prevent the potassium loss, drugs that block
low potassium ingestion, there are few ROMK channels in the renal actions of aldosterone may be given; such drugs are
CHAPTER 46 Regulation of Potassium Balance 469

Diuretics affecting proximal


tubule, loop of Henle, or
distal convoluted tubule

Inhibition of NaCI reabsorption


Inhibition of potassium
reabsorption
Inhibition of water reabsorption

Rate of fluid delivery to


cortical collecting duct

Potassium secretion

Potassium excretion

Potassium depletion

FIGURE 46–5 Pathway by which diuretic drugs affecting the proximal tubule, loop of Henle, or distal convoluted tubule cause
potassium depletion. The decrease in potassium reabsorption is a less important factor in causing the increased potassium excretion than the
increased secretion by the principal cells of the cortical collecting ducts. (Modified with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)

weak diuretics because they block only the aldosterone- Similarly, low plasma pH (acidemia) is usually associated
stimulated fraction of sodium reabsorption with its small with hyperkalemia. Whether these relations between acid–
amount of associated water reabsorption. However, unlike base and potassium actually occur in a particular patient
most common diuretics, they are “potassium sparing” because depends on many factors, including the cause of the acid–
they simultaneously block aldosterone’s stimulation of potas- base disturbance.
sium secretion. Another class of “potassium-sparing” diuretics There are two known reasons for the effects of acid–base
blocks sodium channels in the principal cells of the cortical status on potassium. First, changes in the extracellular con-
collecting duct, preventing sodium entry from lumen to cell. centration of hydrogen ions lead to a de facto exchange of
Therefore, the basolateral membrane Na,K-ATPase pumps these ions with cellular cations, the most important of which
slow their uptake of potassium in exchange for sodium, and is potassium. During an alkalemia, for example, the low extra-
potassium secretion across the apical membrane also slows. cellular hydrogen ion concentration induces the efflux of
To recapitulate the effects of diuretics: increased delivery of hydrogen ions that are normally bound to intracellular buffers.
sodium resulting from the filtration of osmotic diuretics or The loss of the positively charged hydrogen ions is balanced
from blocking sodium absorption upstream from the distal by the uptake of other cations, in this case potassium. Thus,
nephron increases potassium secretion; however, blocking an alkalemia (with hydrogen ions leaving tissue cells to replen-
sodium reabsorption in the distal nephron does not. ish the loss from the ECF) induces cells to take up potassium,
causing a hypokalemia. Conversely, a low pH with a concom-
itant cellular uptake of hydrogen ions (“cellular buffering”)
Effects of Acid–Base Changes often leads cells to dump potassium, causing a hyperkalemia.
Primary acid–base disturbances are a major cause of second- In addition to these exchanges of potassium for hydrogen ions,
ary potassium imbalances (and, as discussed in Chapter 47, there is an effect of intracellular pH on cellular Na,K-ATPase and
imbalances in body potassium can perturb acid–base status). potassium channel activity. Low intracellular pH inhibits pumps
The existence of an elevated plasma pH (alkalemia) is often everywhere, allowing potassium to escape from cells (particularly
(i.e., frequently, but not always) associated with hypokalemia. muscle cells) and increase plasma potassium. Ordinarily, the
470 SECTION VII Renal Physiology

increase in plasma potassium would stimulate potassium uptake


promotes the excretion of sodium by blocking sodium chan-
by the Na,K-ATPase in principal cells, but low intracellular pH
nels in the distal nephron without simultaneously increasing
also inhibits the pumps there as well as luminal membrane potas-
potassium excretion. Follow-up work finds hypersecretion
sium channels. Therefore, the principal cells respond inappropri-
of aldosterone by the left adrenal gland due to a benign tumor
ately and do not effectively secrete the excess plasma potassium
(adenoma), and the left adrenal gland is removed surgically.
(paradoxical potassium retention). A high intracellular pH
reverses these effects and relieves this inhibition (effectively stim-
ulating the pump and the potassium channels). Alkalemia pro-
motes potassium loss and contributes to the production of a
CHAPTER SUMMARY
hypokalemia. Thus, a patient suffering from alkalemia (induced, ■ Only a small fraction of body potassium is extracellular, and
for example, by excessive base input) will manifest increased uri- the extracellular concentration may not be a good indicator of
nary excretion of potassium solely as a result of the alkalemia and total body potassium status.
will, therefore, become potassium deficient. ■ On a short-term basis, uptake and release of potassium by
Finally, it should be emphasized that although alkalemia is tissue cells prevent large swings in extracellular potassium
concentration.
often associated with hypokalemia, and acidemia is often asso-
■ Overall renal handling is accomplished by reabsorbing nearly all
ciated with hyperkalemia, there are exceptions when this is
filtered potassium and then secreting an amount of potassium
not the case
that maintains balance between ingestion and excretion.
■ It is mainly the principal cells of the connecting tubule and
cortical collecting duct that alter rates of potassium secretion.
CLINICAL CORRELATION ■ Potassium secretion (and thus excretion) is increased by high
sodium delivery to the distal nephron, particularly when this is
A 60-year-old African American woman has a history of caused by diuretics acting upstream.
hypertension going back several decades, but blood pres-
sure medications have kept it under control and she has
generally been in good health. Recently, however, she has STUDY QUESTIONS
been feeling fatigued and has occasional problems with 1. Potassium excretion is controlled mainly by controlling the rate of
constipation. Furthermore, her blood pressure medications A) potassium reabsorption in the proximal tubule.
no longer seem to be effective. Most recently, she has expe- B) potassium reabsorption in the distal nephron.
rienced shortness of breath (dyspnea) on several occasions. C) potassium secretion in the proximal tubule.
On a visit to her physician, her blood pressure is 142/101. D) potassium secretion in the distal nephron.
Laboratory work shows a normal plasma sodium of 144 2. In the thick ascending limb
mEq/L and a low potassium of 2.9 mEq/L (clearly A) the net amounts of potassium and sodium that are
hypokalemic). A disorder of aldosterone secretion is sus- reabsorbed are the same.
pected, and assays for plasma aldosterone and renin are B) the major pathway for moving potassium from lumen to
ordered. In the meantime, the patient is put on an angio- cell is via the Na,K-ATPase.
tensin receptor blocker and the diuretic amiloride, which is C) most of the potassium that is absorbed into the cells leaks
a potassium-sparing diuretic. Laboratory results reveal an back into the lumen via potassium channels.
D) the major pathway for moving potassium from cell to
increased plasma aldosterone and low renin.
interstitium is via the Na–K–2Cl multiporter.
The reasons for the long-standing hypertension are
3. For which substance is it possible to excrete more than is filtered?
unknown, and the new symptoms were still too vague to per-
mit a definite diagnosis. However, given the clearly identified A) sodium
B) potassium
hypokalemia, those symptoms are consistent with muscular
C) chloride
problems that are created by low plasma potassium. This
D) it is not possible to excrete any of the above-mentioned ions
information, combined with the refractory hypertension, in amounts greater than the filtered loads
points to an overabundance of aldosterone. The aldosterone
4. After a potassium-rich meal, the key action of insulin that
drives excessive distal nephron secretion of potassium, prevents a large increase in plasma potassium is to
accounting for the hypokalemia, and the excessive reabsorp-
A) decrease absorption of potassium from the GI tract.
tion of sodium leads to an upward creep in arterial pressure. B) increase uptake of potassium by tissue cells.
The combination of high aldosterone and low renin identi- C) increase the filtered load of potassium.
fies this as a case of primary hyperaldosteronism (high levels D) increase tubular secretion of potassium.
of aldosterone in spite of low plasma renin; see Chapter 65). 5. A key role of “BK” potassium channels in the kidney is to
The initial precautionary treatment with the angiotensin A) reabsorb potassium when the body is depleted of potassium.
receptor blocker did not correct the high aldosterone, indi- B) recycle potassium in the thick ascending limb.
cating that the high secretion is being driven by something C) secrete potassium when distal nephron flow rate is very low.
other than angiotensin II. The amiloride is helpful because it D) help the body excrete potassium in response to very large loads.
47
C H A P T E R

Regulation of
Acid–Base Balance
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ State the Henderson–Hasselbalch equation for the carbon dioxide–


bicarbonate buffer system.
■ State the major sources for the input of fixed acids and bases into the body,
including metabolic processes and activities of the gastrointestinal tract.
■ Describe how the input of fixed acids and bases affects body levels of
bicarbonate.
■ Explain why body levels of carbon dioxide are usually not altered by the input
of fixed acids and bases.
■ Explain why some low pH fluids alkalinize the blood after they are
metabolized.
■ Describe the reabsorption of filtered bicarbonate by the proximal tubule.
■ Describe how bicarbonate is excreted in response to an alkaline load.
■ Describe how excretion of acid and generation of new bicarbonate are linked.
■ Describe how the titration of filtered buffers is a means of excreting acid.
■ Describe how the conversion of glutamine to ammonium and subsequent
excretion of ammonium accomplishes the goal of excreting acid.
■ Describe how the kidneys handle ammonium that has been secreted in the
proximal tubule.
■ State how total acid excretion is related to titratable acidity and ammonium
excretion.
■ Define the four categories of primary acid–base disturbance and the
meaning of compensation.
■ Describe the renal response to respiratory acid–base disorders.
■ Identify nonrenal problems that may cause the kidneys to generate a
metabolic alkalosis.

OVERVIEW It is essential for the body to regulate the concentration of


free protons (hydrogen ions) in the ECF. While most sub-
Regulating acid–base balance is a key task of the body, and stances regulated by renal processes exist at plasma levels in
perturbations of acid–base balance are among of the most the millimolar range or greater, the normal hydrogen ion con-
important problems confronting clinicians in a hospital set- centration is a seemingly miniscule 40 nmol/L (1 nmol is one
ting. Regulating blood levels of acids and bases is a partner- millionth of a millimole). Even though very small, this level is
ship between the kidneys and the respiratory system (see crucial for body function. As functional groups on membrane
Chapter 37). proteins protonate and deprotonate, the resulting change in

471

Ch47_471-484.indd 471 11/26/10 10:34:37 AM


472 SECTION VII Renal Physiology

charge affects the shape, and therefore the behavior, of those We should emphasize that buffers do not eliminate added acid
proteins. The plasma levels of hydrogen ions are constantly or base equivalents, but only limit the effect of the equivalents
being altered by a number of processes, including (1) metabo- on blood pH. In the face of persistent imbalance between input
lism of ingested food, (2) secretions of the gastrointestinal (GI) and output, the acid or base component of the buffer is gradu-
tract, (3) de novo generation of acids and bases from metabo- ally reduced in concentration as it is converted to the other
lism of stored fat and glycogen, and (4) changes in the produc- component. Eventually acid or base equivalents added to the
tion of carbon dioxide. body, even though transiently associated with blood buffers,
The essence of the physiological response to these changes must be excreted by the kidneys to maintain balance.
comes down to two processes: (1) matching the excretion of Buffers exist in the extracellular fluid, the intracellular fluid
acid/base equivalents to their input, that is, maintaining bal- (the cytosol of the various cells in the body), and the matrix of
ance, and (2) regulating the ratio of weak acids to their conju- bone. Although these buffers are in different compartments,
gate bases in buffer systems. Buffer systems limit changes in pH they communicate with each other. Phosphate and albumin
to a small range. The two processes of excreting acids and bases, are important buffers in the ECF. Hemoglobin in red blood
and regulating physiological buffer concentrations are inti- cells is an important intracellular buffer, since changes in
mately related, but they are not identical. It is possible for them plasma pH lead to uptake or release of protons from red blood
to be in balance even though buffer ratios are inappropriate. cells. For several reasons, the most important buffer system in
the body turns out to be the CO2–bicarbonate buffer system.
Fortunately, we can understand acid–base balance by looking
at this single buffer system alone and ignore the others, because
ACID–BASE FUNDAMENTALS all buffer systems must have ratios of weak acid to conjugate
An acid, when dissolved in solution, dissociates into a hydro- base that result in the same pH.
gen ion and the conjugate base of the acid, thereby increasing One property that sets the CO2–bicarbonate buffer system
the concentration of free hydrogen ions (decreasing the pH). apart from other buffer systems is that the concentrations of
A base, when dissolved in solution, associates with existing CO2 and bicarbonate are regulated independently of each
free hydrogen ions and decreases their concentration (increases other. Because the concentrations of both components are
the pH). These processes are shown in equation (1). The addi- regulated, the ratio of their concentrations is regulated. There-
tion of acids drives the reaction to the right; the addition of fore, this regulates pH.
bases drives it to the left. Strong acids such as hydrochloric In the CO2–bicarbonate buffer system, CO2 is not a weak
acid release all their hydrogen ions, whereas weak acids such acid per se, but it acts like a weak acid because it readily com-
as acetic acid keep most of the hydrogen ions bound and bines with water to form carbonic acid. (CO2 is often called a
release only a small fraction. However, weak acids strongly volatile acid because it can evaporate. All other acids, for
affect plasma levels of free hydrogen ions. A weak acid present example, sulfuric and lactic, are called fixed acids.) Carbonic
at a millimolar concentration, even if it released just a few per- acid dissociates like any other weak acid into a proton and its
cent of its hydrogen ions, would completely overwhelm the conjugate base, which is bicarbonate [equation (4a)]. Consid-
existing nanomolar level of free hydrogen ions if buffering sys- ered this way, and given the ubiquitous presence of water in
tems did not intervene: our body, it is clear that carbon dioxide is effectively an acid.
The concentration of carbonic acid in our blood is trivial
→ Conjugate base + H+
Acid ← (1) (about 3 μmol/L), and at first glance it appears that this system
has little effective buffering capacity. However, the supply of
A buffer system consists of a mixture of a weak acid and its CO2 is effectively infinite because it is being produced contin-
conjugate base. It limits the change in pH on addition of other uously (over 10 mol per day). Any carbonic acid consumed in
acids or bases. When another acid is added, most of the hydro- a reaction is immediately replaced by new generation from
gen ions released by that acid combine with the base of the buf- existing CO2:
fer system, greatly restricting the increase in free hydrogen ions.
Similarly, when another base is added, most of the free hydro- → H CO ←
CO2 + H2O ← → HCO – + H+ (4a)
2 3 3
gen ions removed by the base are replaced by hydrogen ions
that dissociate from the acid of the buffer system. In any buffer (carbonic anhydrase)
→ HCO – + H+
CO2 + H2O ← (4b)
system, the ratio of the acid to its conjugate base fixes the free 3

aqueous concentration of hydrogen ion (which is only a trivial The reaction on the left side of equation (4a) to form carbonic
fraction of the concentration of the acid and base), as shown in acid is rather slow, but most tissues express one or several iso-
equation (2), or in the more familiar pH form (the Henderson– forms of the enzyme, carbonic anhydrase, intracellularly,
Hasselbalch equation), as shown in equation (3): extracellularly, or both. This enzyme greatly speeds the reac-
[Acid] tion between CO2 and water to form bicarbonate and a hydro-
[H+] = K _____ (2)
[Base] gen ion. In so doing it actually skips the step of forming
Base carbonic acid, as shown in equation (4b). However, as with all
[Acid]
pH = pK + log ____ (3) enzyme-catalyzed reactions, the enzyme increases the velocity
CHAPTER 47 Regulation of Acid–Base Balance 473

of the reaction but not the equilibrium concentrations of reac- alents, continuous acid input would soon reduce plasma bicar-
tants and products. bonate to zero. Similarly, no more than a few millimoles of
Unlike the other buffer systems in the body, where addition CO2 are dissolved in the urine, and there is far less carbonic
or loss of hydrogen ions changes the concentration of the weak acid. If somehow the kidneys could convert the acid equiva-
acid, in the CO2–bicarbonate system, the concentration of the lents of CO2 to fixed acid and excrete those acid equivalents, it
weak acid (CO2) is held essentially constant. This is because would require excreting over 10,000 mmol of fixed acids per
the partial pressure of arterial CO2 (Paco2) is regulated by our day—clearly an impossibility.
respiratory system to be about 40 mm Hg (see Chapters 37
and 38). This partial pressure corresponds to a CO2 concentra-
tion in blood of 1.2 mmol/L. Any change in Pco2 resulting from SOURCES OF ACIDS AND BASES
the addition or loss of hydrogen ions or change in metabolism
is sensed by the arterial chemoreceptors and chemorecep- METABOLISM OF DIETARY PROTEIN
tors in the brainstem (see Chapter 38), which alter the rate of
ventilation to restore the concentration. There are times when Although the oxidative metabolism of most foodstuff is acid–
the Pco2 does indeed differ from 40 mm Hg, but this reflects base neutral, protein contains some amino acids that contrib-
changes in the activity of the respiratory system, not a change ute acid or base. When sulfur-containing amino acids and
in Pco2 in response to addition or loss of hydrogen ions. those with cationic side chains are metabolized to CO2, water,
Although adding or removing hydrogen ions from a source and urea, the end result is addition of fixed acid. Phosphory-
other than CO2 does not change Pco2, such changes do alter lated proteins also contribute to an acid load. Similarly, the
the concentration of bicarbonate. Adding hydrogen ions oxidative metabolism of amino acids with anionic side chains
drives the reaction in equations (4a) and (4b) to the left and adds base (consumes hydrogen ions). Depending on whether
reduces bicarbonate on a nearly mole-for-mole basis. We say a person’s diet is high in either meat or fruit and vegetables, the
nearly because the other blood buffers also take up some of net input can be acid or base. For typical American diets, the
the load. Removing hydrogen ions drives the reaction to the input is usually acidic.
right and increases bicarbonate in the same way. There are
many ways of adding or removing hydrogen ions, but, regard-
less of the process, the result is to change the concentration of METABOLISM OF DIETARY WEAK ACIDS
bicarbonate.
Fruits and vegetables, particularly citrus fruit, contain many
Be aware that, from the acid–base perspective, any meta-
weak acids and the salts of those acids (i.e., the conjugate base
bolic process or reaction that produces hydrogen ions is iden-
plus a cation, usually potassium). We all know that citrus juice
tical to the one that removes bicarbonate, because in both
is acidic, with some fruit juices having a pH below 4.0. Inter-
cases the end result is loss of bicarbonate. The same logic
estingly, metabolism of these acidic substances alkalinizes the
applies to processes that remove hydrogen ions. A reaction in
blood, sometimes called the fruit juice paradox. The com-
which a hydrogen ion is a reactant is equivalent to one in
plete oxidation of the protonated form of an organic acid
which bicarbonate is a product, that is, in both cases the end
(e.g., citric acid) to CO2 and water is acid–base neutral, no dif-
result is an increase in bicarbonate.
ferent in principle than the oxidation of glucose. However, the
From the foregoing, we conclude that the task of maintain-
complete oxidation of the base form adds bicarbonate to the
ing hydrogen ion balance really becomes one of maintaining
body, that is, organic anions are precursors of bicarbonate.
bicarbonate balance (again assuming that the respiratory sys-
One can think of the metabolic process as taking a hydrogen
tem keeps Pco2 constant). When hydrogen ions are added
ion from the body fluids to protonate the base, converting it to
(or bicarbonate is removed), the body has to generate new
the acid, and then oxidizing the acid. The loss of the hydrogen
bicarbonate to replace that which was lost. Analogously, the
ion, as emphasized above, adds bicarbonate. Acidic fruits and
removal of hydrogen ions (or addition of base) increases bicar-
vegetables contain a mixture of organic acids in the protonated
bonate, and the extra bicarbonate has to be excreted. Excretion
form and base form. Although the pH is low, there is far more
and generation of new bicarbonate is the responsibility of the
base than free hydrogen ions. Before oxidation, the mixture is
kidneys.
acidic, but on complete oxidation to CO2 and water, the result
Before moving on, let us clarify a common misconception.
is addition of base.
Students sometimes get the impression that somehow fixed
acid equivalents can be converted to CO2 and excreted by
exhalation, or that CO2 can be converted to acids that are GI SECRETIONS
excreted in the urine. Neither is true: fixed acid equivalents
can only be excreted by the kidneys, and CO2 can only be The GI tract, from the salivary glands to the colon, is lined with
removed from the body via the lungs. Fixed acids consume an epithelium and glands that can secrete hydrogen ions, bicar-
bicarbonate and generate CO2, but just exhaling the CO2 does bonate, or a combination. In addition, the major exocrine secre-
not restore the bicarbonate that disappeared when the acid tions of the pancreas and liver that flow into the duodenum
was added. Without actual renal excretion of those acid equiv- contain large amounts of bicarbonate. To accomplish these
474 SECTION VII Renal Physiology

tasks, the GI tract (and the kidneys as we discuss later) uses the conditions lead to production of fixed acids. The anaerobic
CO2–bicarbonate system in an ingenious way. When we gener- metabolism of carbohydrate produces a fixed acid (lactic
ate bicarbonate and protons from CO2 and water in a given acid). In conditions of poor tissue perfusion, this can be a
medium, say in the blood or in a cell, the result is always acidifi- major acidifying factor, and the metabolism of triglyceride to
cation, because the concentration of protons rises. However, β-hydroxybutyrate and acetoacetate also adds fixed acid
cells of the GI tract separate the protons from the bicarbonate. (ketone bodies). These processes normally do not add much
They transport protons out of the cell into one medium (e.g., the of an acid load but can add a huge acid load in unusual meta-
lumen of the GI tract), and bicarbonate into another (the inter- bolic conditions (e.g., severe uncontrolled diabetes mellitus).
stitium bathing the basolateral surface). Therefore, the lumen
becomes acidified, and the surroundings (and therefore the
blood leaving the tissue) become alkalinized (see Figure 47–1). INTRAVENOUS SOLUTIONS:
In other regions of the GI tract, the cells reverse the direction of LACTATED RINGER’S
these processes, that is, they transport bicarbonate into the
lumen (alkalinizing it) and protons into the surroundings. Thus, Another way in which acid–base loads can enter the body is via
different regions of the GI acidify and alkalinize the blood. Nor- intravenous solutions. Hospitalized patients receive a variety of
mally, the sum of GI tract secretions is nearly acid–base neutral intravenous solutions, a common one being lactated Ringer’s
(i.e., the secretion of acid in one site, e.g., the stomach, is bal- solution, a mixture of salts that contains lactate at a concentra-
anced by the secretion of bicarbonate elsewhere, e.g., the tion of 28 mEq/L. The pH is about 6.5. However, this is an alka-
pancreas). Typically, there is a small net secretion of bicarbonate linizing solution for the same reason described as the fruit juice
into the lumen of the GI tract, resulting in addition of protons to paradox earlier. Lactate is the conjugate base of lactic acid, and
the blood. However, in conditions of vomiting or diarrhea, one when oxidized to CO2 and water, it takes a hydrogen ion from
kind of secretion may vastly exceed the other, resulting in a the body fluids, thereby producing bicarbonate. Lactated
major loss of acid or base to the outside world complete with a Ringer’s should not be confused with lactic acidosis associated
major retention of base or acid in the blood. with strenuous exercise or certain forms of shock. In these situ-
ations, the body produces equal numbers of hydrogen ions and
lactate, and the result is to acidify the body fluids.
ANAEROBIC METABOLISM
OF CARBOHYDRATE AND FAT
RENAL HANDLING
The normal oxidative metabolism of carbohydrate and fat is OF ACIDS AND BASES
acid–base neutral. Both carbohydrate (glucose) and triglycer-
ides are oxidized to CO2 and water. Although there are inter- A simplified overview of the renal processing of acids and
mediates in the metabolism (e.g., pyruvate) that are acids or bases is as follows: the kidneys reabsorb most of the filtered
bases, the sum of all the reactions is neutral. However, some bicarbonate in the proximal tubule, thus conserving plasma

LUMEN INTERSTITIUM
H2O CO2

Carbonic
anhydrase

H+ H+ HCO3– HCO3–

H-secreting cell

H2O CO2

FIGURE 47–1 Generic model of hydrogen ion secretion


Carbonic
(upper cell) and bicarbonate secretion (lower cell). The source of anhydrase
secreted ions is CO2 and water. Every hydrogen ion moved out
of a cell across one membrane must be accompanied by the
HCO3– HCO3– H+ H+
transport of a bicarbonate ion out of the cell across the other
membrane. (Modified with permission from Eaton DC, Pooler JP: Vander’s Renal HCO3–secreting cell
Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical
Pub. Division, 2009.)
CHAPTER 47 Regulation of Acid–Base Balance 475

TABLE 47–1 Normal contributions of tubular where they combine with filtered bicarbonate to form water
segments to renal hydrogen ion balance. and carbon dioxide; thus, the filtered bicarbonate “disappears.”
At the same time, the cellular bicarbonate is transported across
Proximal tubule the basolateral membrane into the interstitial fluid and then
Reabsorbs most filtered bicarbonate (normally about 80%)a into the peritubular capillary blood. The overall result is that
Produces and secretes ammonium
the bicarbonate filtered from the blood at the renal corpuscle
Thick ascending limb of Henle’s loop is converted to CO2 and water, replaced by bicarbonate that is
Reabsorbs second largest fraction of filtered bicarbonate
(normally about 10–15%)a
generated inside the cell. Thus, no net change in plasma bicar-
bonate concentration occurs. It is also important to note that
Distal convoluted tubule and collecting duct system the hydrogen ion that was secreted into the lumen is not
Reabsorbs virtually all remaining filtered bicarbonate as well
as any secreted bicarbonate (type A intercalated cells)a excreted in the urine. It has been incorporated into water. Any
Produces titratable acid (type A intercalated cells)a secreted hydrogen ion that combines with bicarbonate in the
Secretes bicarbonate (type B intercalated cells) lumen does not contribute to the urinary excretion of hydro-
a
Processes achieved by hydrogen ion secretion. gen ions, but only to the conservation of bicarbonate.
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, Specific transporters are required for these transmembrane
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009. movements of hydrogen ions and bicarbonate. First, particu-
larly prominent in the apical membrane of the proximal tubule
is the Na–H antiporter (NHE3) as described in Chapter 44
bicarbonate. The proximal tubule also secretes limited amounts and shown in Figure 47–2. This transporter is the major means
of organic bases or weak organic acids and acid equivalents. not only of hydrogen ion secretion, but also of sodium uptake
Then, in the distal nephron (mostly the collecting tubules), the from the proximal tubule lumen. The same NHE3 antiporter
kidneys secrete either protons or bicarbonate to balance the also mediates hydrogen ion secretion in the thick ascending
net input into the body (summarized in Table 47–1). limb. Second, a primary active H-ATPase exists in all the
The first task is to reabsorb most of the filtered bicarbonate. hydrogen ion–secreting distal tubular segments. The type A
Bicarbonate is freely filtered at the renal corpuscles. Reabsorp- intercalated cells of the collecting duct system possess this pri-
tion is an active process, but it is not accomplished in the con- mary active H-ATPase as well as a primary active H,K-ATPase,
ventional manner of importing bicarbonate across the apical which simultaneously moves hydrogen ions into the lumen
membrane and exporting it across the basolateral membrane. and potassium into the cell, both actively (Figure 47–3).
Rather, the mechanism involves the tubular secretion of hydro- The basolateral membrane exit step for bicarbonate gener-
gen ions. An enormous amount of hydrogen ion secretion ated when H ions are secreted is via Cl–HCO3 antiporters or
occurs in the proximal tubule, with additional secretion in the Na–HCO3 symporters (Figures 47–2 and 47–3), depending
thick ascending limb of Henle’s loop and collecting duct sys- on the tubular segment. In both cases, the movement of bicar-
tem. The basic pattern is illustrated in the upper part of bonate is down its electrochemical gradient (i.e., the exit step
Figure 47–1 without indicating any specific transporters. is passive). Symport with sodium is the dominant means of
Within the cells, hydrogen ions and bicarbonate are generated extruding bicarbonate in the proximal tubule and is particu-
from CO2 and water, catalyzed by carbonic anhydrase. The larly interesting because the efflux of sodium is up its electro-
hydrogen ions are actively secreted into the tubular lumen, chemical gradient. This is a rare case of sodium active transport

LUMEN INTERSTITIUM

Filtered H2O CO2


HCO3–
Carbonic
anhydrase

– – FIGURE 47–2 Predominant proximal tubule mechanism


H+ H+ 3HCO3 3HCO3
+
for reabsorption of bicarbonate. Hydrogen ions and bicarbonate
Carbonic Na Na+ are produced intracellularly. The hydrogen ions are secreted via a
anhydrase ATP + Na–H antiporter (member of the NHE family), while the bicarbonate
Na
is transported into the interstitium via an Na–3HCO3 symporter
K+
(member of the NBC family). Because more sodium enters via the
H2O CO2 Na–H antiporter than leaves via the Na–3HCO3 symporter,
additional sodium is removed via the Na,K–ATPase. (Modified with
permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York,
NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
476 SECTION VII Renal Physiology

LUMEN INTERSTITIUM

H2O CO2

Carbonic
anhydrase
ATP
+ + – –
H H HCO3 HCO3
ATP Cl–
H+ H+
K+

Cl–
A

LUMEN INTERSTITIUM

FIGURE 47–3 Type A and type B intercalated cells. H2O CO2


A) Predominant collecting tubule mechanisms in type A
intercalated cells for the secretion of hydrogen ions that Carbonic
result in formation of titratable acidity. The apical membrane anhydrase
ATP
contains H-ATPases and H,K-ATPases, which transport
+
hydrogen ions alone or in exchange for potassium. B) The – – H
HCO3 HCO3
type B intercalated cell secretes bicarbonate and Cl– ATP
simultaneously transports hydrogen ions into the H+
interstitium. The difference between this cell type and the K+
type A cell and those in the proximal tubule is that the
location of the transporters for hydrogen ions and Cl–
bicarbonate are switched between apical and basolateral B
membranes. (Modified with permission from Eaton DC, Pooler JP:
Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/
McGraw-Hill, Medical Pub. Division, 2009.)

that does not use ATP as the energy source, but uses the gradi-
ent of another ion. (However, this process can only occur if the
RENAL EXCRETION
Na,K–ATPase sets up the Na gradient that powers the removal OF ACID AND BASE
of hydrogen ions via Na–H exchange in the apical mem-
brane.) Acid or base loads generated from the processes described ear-
Through its secretion of hydrogen ions, the proximal lier result in changes in plasma bicarbonate. In essence, an
tubule reabsorbs 80–90% of the filtered bicarbonate. The acid or base load, regardless of original source, is turned into
thick ascending limb of Henle’s loop reabsorbs another 10%, an excess or deficit of bicarbonate. In response to base loads,
and almost all the remaining bicarbonate is reabsorbed by the process is relatively straightforward: we reabsorb most
the distal convoluted tubule and collecting duct system of the filtered bicarbonate, but excrete just enough bicarbonate
(although this depends on diet and other conditions; see later in the urine to match the input. The kidneys do this in two
discussion). ways: (1) allow some filtered bicarbonate to pass through to
Throughout the tubule, intracellular carbonic anhydrase the urine and (2) secrete bicarbonate via type B intercalated
is involved in the reactions generating hydrogen ion and cells. The type B intercalated cells, which are found only in the
bicarbonate. In the proximal tubule, carbonic anhydrase is cortical collecting duct, do indeed secrete bicarbonate. In
also located in the lumen-facing surface of apical cell mem- essence, the type B intercalated cell is a “flipped-around” type
branes, and this carbonic anhydrase catalyzes the intralu- A intercalated cell (Figure 47–3B). Within the cytosol, hydro-
minal generation of CO2 and water from the large quantities gen ions and bicarbonate are generated via carbonic anhy-
of secreted hydrogen ions combining with filtered bicar- drase. However, the H-ATPase pump is located in the
bonate. basolateral membrane, and the Cl–HCO3 antiporter is in the
CHAPTER 47 Regulation of Acid–Base Balance 477

apical membrane. Accordingly, bicarbonate moves into the There are two sources of tubular nonbicarbonate buffers:
tubular lumen and hydrogen ion is actively transported out of filtration and synthesis. Normally, the most important filtered
the cell across the basolateral membrane and enters the blood, buffer is phosphate, while ammonia is the most important
where it combines with a bicarbonate ion and reduces plasma synthesized buffer. Ammoniagenesis is crucial to renal acid
bicarbonate. Thus, the overall process achieves the disappear- excretion because its rate can be greatly increased in the face of
ance of excess plasma bicarbonate and the excretion of bicar- large acid loads, whereas the availability of filtered buffers,
bonate in the urine. while somewhat variable, is not regulated for purposes of acid
How do the kidneys excrete an acid load, which always gener- excretion. Figure 47–4 illustrates the sequence of events that
ates a bicarbonate deficit? First, they reabsorb all the filtered achieves hydrogen ion excretion on filtered phosphate and the
bicarbonate. Then they secrete additional hydrogen ions that addition of new bicarbonate to the blood. It must be empha-
attach to buffers in the tubular fluid other than bicarbonate. The sized also that neither filtration per se nor excretion of free
now protonated buffer is excreted. Meanwhile, the bicarbonate hydrogen ions makes a significant contribution to hydrogen
generated in the cell is transported into the blood, replacing the ion excretion. First, the filtered load of free hydrogen ions,
bicarbonate lost when the acid load entered the body. It is when the plasma pH is 7.4 (40 nmol/L/H+), is less than 0.1
important to realize that both parts of this process must occur, mmol per day. Second, there is a minimum urinary pH—
that is, new bicarbonate and excretion of hydrogen ions on buf- approximately 4.4—that can be achieved. This corresponds to
fers. If there were no new bicarbonate, plasma levels would not a free hydrogen ion concentration of 0.04 mmol/L. With a
be restored, and if hydrogen ions were not excreted, they would typical daily urine output of 1.5 L, the excretion of free hydro-
react with and remove the bicarbonate just generated. gen ions is only 0.06 mmol per day, a tiny fraction of the nor-
mal 50–100 mmol of hydrogen ion ingested or produced every
day. To excrete these additional amounts of protons, they must
HYDROGEN ION EXCRETION associate with tubular buffers.
ON URINARY BUFFERS
We see that the identical transport process of hydrogen ion PHOSPHATE AND ORGANIC
secretion achieves both reabsorption of bicarbonate (without
new bicarbonate) and acid excretion, with addition of new ACIDS AS URINARY BUFFERS
bicarbonate to the blood. At first glance, this seems like a con-
Free plasma phosphate exists in a mixture of monovalent
tradiction: how can the same process produce two different
(acid) and divalent forms (base). In the following equation,
end results? The answer lies in the fate of the hydrogen ion
monovalent dihydrogen phosphate (on the left) is a weak acid,
once it is in the lumen. For secreted hydrogen ions that com-
and divalent monohydrogen phosphate (on the right) is its
bine with bicarbonate, we are simply replacing bicarbonate that
conjugate base:
would have left the body. In contrast, when secreted hydrogen
ions combine with a nonbicarbonate buffer in the lumen, the → HPO 2– + H+
H2PO4– ← (5)
4
hydrogen ion is excreted, and the bicarbonate produced in the
cell and transported across the basolateral membrane is new We can write the above equation in the form of the
bicarbonate, not a replacement for filtered bicarbonate. Henderson–Hasselbalch equation:

LUMEN INTERSTITIUM

Filtered
HPO42– H2O CO2

Carbonic
anhydrase

H+ H+ 3HCO3– 3HCO3– FIGURE 47–4 Excretion of hydrogen ions on filtered


H2PO4– Na + Na+ phosphate. Divalent phosphate (base form) that has been filtered
and not reabsorbed reaches the collecting tubule, where it
combines with secreted hydrogen ions to form monovalent
phosphate (acid form), which is then excreted in the urine. The
excreted bicarbonate entering the blood is new bicarbonate, not merely a
H2PO4– replacement for filtered bicarbonate. (Modified with permission from
Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange
Medical Books/McGraw-Hill, Medical Pub. Division, 2009.)
478 SECTION VII Renal Physiology

[HPO 2–]
pH = 6.8 + log _______
4
[H PO –]
(6) 2 amino acids (+oxygen) 2NH4+ + 2HCO3–
2 4 (7)
Urea or glutamine (+ CO2 and water)
At the normal plasma pH of 7.4, we find that about 80% of the
plasma (and filtered) phosphate is in the base (divalent) form When the urea (or glutamine) is excreted, the body has com-
and 20% is in the acid (monovalent) form. As the tubular fluid pleted the catabolism of protein in a manner that promotes
is acidified in the collecting ducts, most of the base form takes total body nitrogen balance, and is acid–base neutral.
up secreted hydrogen ions. Depending on final urine pH, most The renal handling of urea is somewhat complicated from
of the base (HPO42−) has been protonated to acid (H2PO4−). the osmotic point of view, as described in earlier chapters, but
The secreted hydrogen ions that combined with the base form is acid–base neutral. Glutamine, however, is different.
are excreted, and the bicarbonate that was generated intracel- Although the production of glutamine by the liver is acid–base
lularly enters the blood. How much phosphate is available for neutral, it is important to recognize that glutamine can be
this process? The amount is somewhat variable, depending on thought to contain the two components from which it was
a number of factors (see Chapter 48), but a typical plasma synthesized: a base component (bicarbonate) and an acid
concentration is about 1 mmol/L, of which about 90% is free component (ammonium). Ammonium is the protonated form
(the rest being loosely bound to plasma proteins). At a GFR of of ammonia. It is an acid because it contains a dissociable pro-
180 L per day, the total filtered load of phosphate is about ton as shown in equation (8). The pK of ammonium is near
160 mmol per day. The fraction reabsorbed is also variable: 9.2, making it an extremely weak acid (i.e., only at high pH will
from 75% to 90%. Thus, unreabsorbed divalent phosphate it release its proton), but it is an acid nevertheless. At physio-
available to take up secreted hydrogen ions amounts to roughly logical pH, over 98% of the total exists as ammonium, and less
40 mmol per day. In other words, the kidneys can excrete acid than 2% exists as ammonia. For renal acid–base purposes, this
loads using the phosphate buffer system at a rate of about 40 is a good thing because virtually all excreted ammonia is in the
mmol per day. However, the availability of phosphate cannot protonated form and takes a hydrogen ion with it:
be easily increased to increase acid excretion.
→ H+ + NH
NH4+ ← (8)
3

Glutamine released from the liver is taken up by proximal


HYDROGEN ION EXCRETION tubule cells, both from the lumen (filtered glutamine) and
ON AMMONIUM from the renal interstitium. The cells of the proximal tubule
then convert the glutamine back to bicarbonate and NH4+, in
Ordinarily, hydrogen ion excretion associated with phosphate essence reversing what the liver has done. The NH4+ is secreted
and other filtered buffers is not sufficient to balance the nor- into the lumen of the proximal tubule, and the bicarbonate
mal hydrogen ion production of 50–100 mmol per day nor can exits into the interstitium and then into the blood
it take care of any unusually high (usually pathological) pro- (Figure 47–5A). This is new bicarbonate, just like the new
duction of acid loads. To excrete the rest of the hydrogen ions bicarbonate generated by titrating nonbicarbonate buffers.
and achieve balance, there is a second means of excreting Further processing of the NH4+ is complicated, but eventually
hydrogen ions that involves ammoniagenesis and excretion of the ammonium is excreted (Figure 47–5C).
hydrogen ions as ammonium. Quantitatively, far more hydro- The ammonium ion has interesting chemical properties in
gen ions can be excreted by means of ammonium than via fil- that it can masquerade as other ions, in some cases as a hydro-
tered buffers. There are many nuances to hydrogen ion gen ion and in other cases as a potassium ion. This is because
excretion via ammonium, but the basic concepts are straight- some transporters and some channels are not completely
forward. selective for the species they usually move compared with
As described in Chapter 43, the catabolism of protein and ammonium. As the concentration of ammonium rises, there is
oxidation of the constituent amino acids by the liver generates an increasing tendency for ammonium to substitute for these
CO2, water, urea, and some glutamine. Although the metabo- other ions and “sneak” its way across membranes.
lism of the side chains of amino acids can lead to addition of Also, whenever ammonium is present in body fluids, a small
acid or base, the processing of the core of an amino acid—the fraction (2% at physiological pH) always exists as ammonia
carboxyl group and amino group—is acid–base neutral. After because the dissociation, although limited in extent, is nearly
many intermediate steps, processing of the carboxyl group of instantaneous. Ammonium, being a small hydrated ion, is
the amino acid produces bicarbonate, and processing of the essentially impermeant in lipid bilayers and must be handled
amino group produces ammonium. Processing does not stop by channels or transporters if it is to move across membranes,
there, however, because ammonium in more than minuscule but the neutral ammonia has a finite permeability. In terms of
levels is quite toxic. Ammonium is further processed by the cellular handling, cells sometimes transport ammonium as
liver to either urea or glutamine. In both cases, each ammo- such and at other times transport ammonia and a proton in
nium consumed also consumes a bicarbonate. Thus, the bicar- parallel, the end result being the same in both cases.
bonate produced from the carboxyl group is just an intermediate, It would “make sense” if the ammonium secreted into the
consumed as fast as it is made, and the process as a whole is proximal tubule simply stayed in the lumen and was excreted,
acid–base neutral. We can write this process as follows: but the kidneys have a more complicated way of doing
CHAPTER 47 Regulation of Acid–Base Balance 479

LUMEN INTERSTITIUM LUMEN INTERSTITIUM

NH4+

Filtered
glutamine
glutamine 2CI– NH3 NH4+
H+
Na+
NH4+ HCO3

HCO3
– Na+
NH4+
Na+ NH4 +
Na+
K+
NH4+ NH4+
Na+
NH4+
(to thick
A ascending B
limb)

LUMEN INTERSTITIUM

ATP
Na+
ATP
K+ NH3 NH4+

H+ H+ H+

+ NH3
NH4 NH3
NH3 NH3

C (excretion)

FIGURE 47–5 Ammoniagenesis and excretion. A) Ammonium production from glutamine. Glutamine is originally synthesized in
the liver from NH4+ and bicarbonate. When glutamine reaches the proximal tubule cells, it is converted via several intermediate steps (not
shown) back to NH4+ and bicarbonate. B) Ammonium reabsorption in the thick ascending limb. Ammonium reaches the thick ascending
limb from two sources. Most comes as a result of secretion in the proximal tubule. Some also enters the thin limbs from the medullary
interstitium in the form of neutral ammonia and is subsequently reprotonated in the lumen (ammonium recycling). Ammonium is
reabsorbed in the thick ascending limb by several mechanisms, the predominant one being entrance via the NKCC multiporter (where
ammonium substitutes for potassium). C) Ammonium secretion in the inner medulla. Several mechanisms are involved. A prominent one
involves uptake and secretion of neutral ammonia via specific transporters in parallel with hydrogen ion secretion, resulting in reformation
of ammonium in the lumen. In the innermost medulla, the high interstitial ammonium concentration allows ammonium to substitute for
potassium on the Na,K–ATPase. (Modified with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, 7th ed. New York, NY: Lange Medical Books/
McGraw-Hill, Medical Pub. Division, 2009.)

things. An array of channels and transporters participates in Most of the ammonium synthesized from glutamine in the
moving ammonium or ammonia into or out of the tubule in proximal tubule is secreted via the NHE3 antiporter in exchange
various segments. So long as all the ammonium produced for sodium (with ammonium substituting for a hydrogen ion);
from glutamine and secreted in the proximal tubule ends up but some may also diffuse into the lumen as ammonia and then
being excreted, the process accomplishes the goal of excret- combine with a secreted hydrogen ion. The next major trans-
ing acid, even if ammonium is transported as such in some port event occurs in the thick ascending limb (Figure 47–5B).
places and moved as H+ and NH3 separately in other places. In this segment about 80% of the tubular ammonium is reab-
But if ammonium is returned to the circulation, it is metab- sorbed, mostly by the Na–K–2Cl multiporter (with ammo-
olized by the liver back to urea, consuming bicarbonate in nium now substituting for potassium). In the medullary
the process, thereby nullifying the renal generation of bicar- portions of the thick ascending limb, this reabsorption results
bonate. in accumulation of ammonium (and therefore some ammonia)
480 SECTION VII Renal Physiology

in the interstitium, with the concentration progressively increas- TABLE 47–2 Renal contribution of new bicarbonate
ing toward the papilla, analogous to the osmotic gradient. The to the blood in different states.
high interstitial concentration surrounding the loops of Henle
leads to some secretion of ammonia into the thin descending Alkalosis Normal State Acidosis
limbs, where the ammonia becomes protonated in the lumen.
Titratable acid 0 20 40
Therefore, there is a certain amount of recycling, with the con- (mmol per day)
sequence that a considerable amount of ammonium is trapped
Plus NH4+ excreted 0 40 160
in the medullary interstitium (similar to the situation with (mmol per day)
urea). Finally, in the medullary collecting ducts, there is secre-
tion, mainly by parallel transport of hydrogen ions and ammo- Minus HCO3− 80 1 0
excreted (mmol
nia. Thus, the ammonium that was reabsorbed in the thick per day)
ascending limb and accumulated in the medullary interstitium
Total (mmol per day) –80 (lost 59 (added 200 (added
is now put back into the tubule and excreted. from body) to body) to body)

Urine pH 8.0 6.0 4.6

QUANTIFICATION OF RENAL Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology,
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009.

ACID–BASE EXCRETION
We can now quantify the excretion of acid/base equivalents by known neural or hormonal signals conveying acid–base infor-
looking at three quantities in the urine: (1) the amount of mation to the kidneys. In effect, the kidneys act as “pH meters”
titratable acidity, (2) the amount of ammonium, and (3) the and Pco2 detectors and adjust their transport of hydrogen ion
amount of bicarbonate, if any. Titratable acidity can be mea- and ammonium excretion accordingly. Hydrogen ion transport
sured by titrating the urine with strong base (NaOH) to a pH is also stimulated independently by aldosterone.
of 7.4. (The amount of NaOH required to increase the pH back An increase in Pco2, as occurs during respiratory acidosis
to 7.4 must equal the amount of hydrogen ion that was secreted due to hypoventilation, produces a decrease in plasma pH
and combined with phosphate and organic buffers.) Urinary and, thereby, signals an increased tubular hydrogen ion secre-
ammonium equals the urinary volume times the urinary tion. A decrease in Pco2, as occurs during respiratory alkalosis
ammonium concentration. (Ammonium does not contribute due to hyperventilation, causes a decrease in secretion.
to titratable acidity because with a pK of 9.2, few hydrogen Because the tubular membranes are quite permeable to CO2,
ions are removed by titration to pH 7.4.) Similarly, urinary an altered arterial Pco2 causes an equivalent change in Pco2
bicarbonate equals the urinary volume times the urinary within the tubular cells. In turn, this causes altered intracel-
bicarbonate concentration. lular hydrogen ion concentration by driving the reactions
Thus, we can write the net acid excretion as: shown in equations (4a) and (4b) to the right or left. This
Net acid excretion = change in intracellular pH, along with signals generated in
(9) response to altered Pco2 at the basolateral surface, adjusts the
Titratable acid excreted + NH4+ excreted – HCO3– excreted
rate of hydrogen ion secretion.
Note that there is no term for free hydrogen ion in the urine We can see that these renal responses are appropriate. If the
because, even at a minimum urine pH of 4.4, the number of Pco2 is high (causing a decrease in plasma pH), the increased
free hydrogen ions is trivial. hydrogen ion secretion increases plasma bicarbonate, thereby
Typical urine data for the amounts of bicarbonate contrib- increasing plasma pH closer to normal (despite the continued
uted to the blood by the kidneys in three potential acid–base high Pco2). Similarly, if the pH is low because of low bicarbon-
states are given in Table 47–2. Note that in response to acido- ate, the new bicarbonate restores the bicarbonate (and, there-
sis, as emphasized previously, increased production and excre- fore, the pH) toward normal.
tion of NH4+ is quantitatively much more important than
increased formation of titratable acid.
CONTROL OF RENAL GLUTAMINE
METABOLISM AND AMMONIUM
REGULATION OF THE RENAL EXCRETION
HANDLING OF ACIDS AND BASES In addition to regulating hydrogen ion secretion per se, there
Renal acid–base processing is regulated in response to different are several homeostatic controls over the production and
body conditions. The key regulatory signals are the concentra- tubular handling of NH4+. First, the generation of glutamine
tions of free hydrogen ions and CO2 in the fluids to which the by the liver is increased by low plasma pH. In this case, the
various transport elements are exposed, that is, the pH and Pco2 liver shifts some of the disposal of ammonium ion from urea
of the interstitium and cytosol within renal cells. There are no to glutamine. Second, the renal metabolism of glutamine is
CHAPTER 47 Regulation of Acid–Base Balance 481

TABLE 47–3 Summary of processes that acidify or assign acid–base disorders to the following four categories: (1)
alkalinize the blood. high Pco2 is a respiratory acidosis, (2) low Pco2 is a respiratory
alkalosis, (3) low bicarbonate is a metabolic acidosis, and (4)
Nonrenal mechanisms of acidifying the blood high bicarbonate is a metabolic alkalosis:
Consumption and metabolism of protein (meat) containing acidic
[Bicarbonate]
or sulfur-containing amino acids pH = 6.1 + log __________
0.03 P (10)
co2
Consumption of acidic drugs
Metabolism of substrate without complete oxidation (fat to ketones It should be clear from the Henderson–Hasselbalch equation
and carbohydrate to lactic acid).
[equation (10)] for the CO2–bicarbonate buffer system that
GI tract secretion of bicarbonate (puts acid in blood)
changing either the Pco2 or the bicarbonate concentration
Nonrenal mechanisms of alkalinizing the blood increased or decreases the pH. If only one of these is changed,
Consumption and metabolism of fruit and vegetables containing it is called a primary uncompensated disorder. In most cases
basic amino acids or the salts of weak acids the situation is more complicated because there is at least
Consumption of antacids some, and usually considerable, compensation. Compensa-
Infusion of lactated Ringer’s solution tion exists when either Pco2 or bicarbonate levels remain altered
GI tract secretion of acid (puts bicarbonate in the blood) for a period of time and the body changes the other variable in
Renal mechanisms of acidifying the blood the same direction. For example, if the Pco2 is abnormally low,
Allow some filtered bicarbonate to pass into the urine renal compensation consists of reducing plasma bicarbonate.
Secrete bicarbonate (type B intercalated cells) Similarly, if bicarbonate is abnormally low, respiratory com-
pensation consists of reducing arterial Pco2. The compensatory
Renal means of alkalinizing the blood
Secrete protons that form urine titratable acidity changes bring the ratio of bicarbonate to Pco2 closer to a nor-
(type A intercalated cells) mal value, and therefore minimize the change in pH. However,
Excrete NH4+ synthesized from glutamine compensation for an acid–base disorder is not correction,
GI, gastrointestinal. because even if the compensation has returned the pH to the
Reproduced with permission from Eaton DC, Pooler JP: Vander’s Renal Physiology, normal range, both Pco2 and bicarbonate values are still abnor-
7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2009. mal. Consider a case where the Pco2 is too high (respiratory
acidosis) due to hypoventilation (review Chapter 37). The
body compensates by increasing bicarbonate. If bicarbonate is
also subject to control by extracellular pH. A decrease in extra- increased high enough, this restores pH to the normal range;
cellular pH stimulates renal glutamine oxidation by the proxi- however, it does not correct the original respiratory problem
mal tubule, whereas an increase does just the opposite. Thus, that resulted in an increased Pco2 . The same logic applies to any
an acidosis that lowers plasma pH, by stimulating renal glu- other acid–base disorder.
tamine oxidation, causes the kidneys to contribute more new The astute reader may recognize a potential problem in the
bicarbonate to the blood, thereby counteracting the acidosis. interpretation of acid–base disorders. When any acid–base
This pH responsiveness increases over the first few days of an disorder is well compensated, that is, the degree of compen-
acidosis and allows the glutamine–NH4+ mechanism for new sation is such that the pH is in the normal range, both the
bicarbonate generation to become the predominant renal pro- Pco2 and bicarbonate are increased or decreased in the same
cess for opposing the acidosis. Conversely, an alkalosis inhibits direction. Suppose both the Pco2 and bicarbonate are high.
glutamine metabolism, resulting in little or no renal contribu- Is this a respiratory acidosis with renal compensation, or is
tion of new bicarbonate via this route. it a metabolic alkalosis with respiratory compensation?
Table 47–3 provides a summary of the processes of adding Fortunately in a clinical setting it would be rare not to have
acids and bases to the body fluids. The unifying and, therefore, additional information. For example, the high Pco2 of chronic
simplifying principle is that all processes of acid or base addi- bronchitis patient is, in all likelihood, a respiratory acidosis
tion boil down to addition or loss of bicarbonate. All processes resulting from impaired ventilation, not a compensation for
that acidify the blood end up removing bicarbonate, and all a metabolic alkalosis. Nevertheless, in real life there are often
processes that alkalinize the blood end up adding bicarbonate. mixed acid–base disorders that indeed present a challenge in
the clinic.

ACID–BASE DISORDERS
AND THEIR COMPENSATION RENAL RESPONSE TO RESPIRATORY
ACIDOSIS AND ALKALOSIS
In this section we briefly address the topic of acid–base disorders
in the context of the kidney. This topic is covered more thor- In a respiratory acidosis, the low alveolar ventilation causes
oughly in the context of the respiratory system in Chapter 37. an increase in Pco2, in turn causing a decrease in pH. The pH
Acid–base disorders develop when either arterial Pco2, bicar- would be restored to normal if the bicarbonate were increased
bonate, or both deviate from their normal range. Clinicians to the same degree as Paco2. The normal kidneys respond to the
482 SECTION VII Renal Physiology

increased Pco2 by contributing new bicarbonate to the blood in are (1) volume contraction, (2) chloride depletion, and (3) the
the manner previously described. combination of aldosterone excess and potassium depletion.
The renal compensation in response to respiratory alkalosis is In any metabolic alkalosis, by definition the plasma bicarbon-
just the opposite. Respiratory alkalosis is the result of hyperven- ate concentration is elevated. This problem is not a defect in the
tilation, as occurs at high altitude (see Chapter 71), in which the ability of the kidneys to excrete bicarbonate; if a person is fed a
person transiently eliminates carbon dioxide faster than it is pro- large load of bicarbonate, the kidneys can excrete the load with-
duced, thereby decreasing Pco2 and increasing pH. The decreased out a major increase in bicarbonate levels. The problem seems to
Pco2 and increase in extracellular pH signal reduced tubular be in regulation of bicarbonate excretion. The key event in all
hydrogen ion secretion and increased bicarbonate secretion. these situations is oversecretion of hydrogen ion (and sometimes
Bicarbonate is lost from the body, and the loss results in decreased of NH4+ as well), either producing a metabolic alkalosis or fail-
plasma bicarbonate and a return of plasma pH toward normal. ing to respond as usual to an existing metabolic alkalosis.
There is no titratable acid in the urine (the urine is alkaline in
these conditions), and there is little or no NH4+ in the urine
because the alkalosis inhibits NH4+ production and excretion. INFLUENCE OF EXTRACELLULAR
VOLUME CONTRACTION
RENAL RESPONSE TO The presence of total body volume contraction because of salt
METABOLIC ACIDOSIS loss stimulates sodium reabsorption, and also hydrogen ion
secretion because the transport of these ions is linked via the
There are many possible causes of metabolic acidosis, includ- Na/H antiporters in the proximal tubule. In addition, the renin–
ing the kidneys themselves. These include (1) increased input angiotensin system (RAS) is usually activated, resulting in
of acid by ingestion, infusion, or production; (2) decreased stimulation of aldosterone secretion. Besides stimulating
renal production of bicarbonate, as in renal failure; or (3) sodium reabsorption, aldosterone stimulates hydrogen ion
direct loss of bicarbonate from the body, as in diarrhea. The secretion by type A intercalated cells. The net result is that all
result is the same regardless of whether there is loss of bicar- the filtered bicarbonate is reabsorbed so that the already ele-
bonate or addition of hydrogen ions: that is, a lower concen- vated plasma bicarbonate associated with the preexisting meta-
tration of bicarbonate and a lower plasma pH. The renal bolic alkalosis is locked in, and the plasma pH remains high.
response (if the kidneys are not the cause) is to produce more The urine, instead of being alkaline, as it should be when the
bicarbonate, thereby returning pH toward normal. (Note that kidneys are normally responding to a metabolic alkalosis, is
this is a response, not compensation, because the primary somewhat acid. The generation or maintenance of a metabolic
problem is not a respiratory change in Pco2.) To do this, the alkalosis in volume contraction may also occur when the vol-
kidneys must reabsorb all the filtered bicarbonate and contrib- ume is normal or high but the body “thinks” volume is low, spe-
ute more new bicarbonate through increased formation and cifically in congestive heart failure and advanced liver cirrhosis.
excretion of NH4+ and titratable acid. This is precisely what
healthy kidneys do in the case of any acid load, but if the acid
load is too great or the problem is in the kidneys themselves, INFLUENCE OF CHLORIDE DEPLETION
the bicarbonate concentration will remain low.
We referred to extracellular volume contraction without dis-
tinguishing between sodium and chloride losses as the cause
FACTORS CAUSING THE KIDNEYS because loss of either of these ions will lead to extracellular vol-
TO GENERATE OR MAINTAIN A ume contraction. However, we emphasize that specific chloride
METABOLIC ALKALOSIS depletion, in a manner independent of and in addition to extra-
cellular volume contraction, helps maintain metabolic alkalo-
The normal kidneys are able to excrete large amounts of bicar- sis by stimulating hydrogen ion secretion. The most common
bonate. However, in some situations, the kidneys fail to do reasons for chloride depletion are chronic vomiting and heavy
this, and thereby either generate a metabolic alkalosis or main- use of diuretics. The result is that bicarbonate excretion remains
tain a metabolic alkalosis that originates from another cause. essentially zero, and the metabolic alkalosis is not corrected.
Recall that secretion of hydrogen ions, after all filtered bicar-
bonate has been reabsorbed, generates new bicarbonate. Also
recall that secretion of hydrogen ions is coupled to the reab- INFLUENCE OF ALDOSTERONE
sorption of sodium in the proximal tubule and thick ascend- EXCESS AND SIMULTANEOUS
ing limb, and that some is coupled to the reabsorption of POTASSIUM DEPLETION
potassium in the distal nephron. Some conditions that signal
strong reabsorption of sodium or potassium ions have the As noted, aldosterone stimulates hydrogen ion secretion.
undesired effect of causing the secretion of too many hydro- Potassium depletion, by itself, also weakly stimulates tubu-
gen ions. The most important situations in which this occurs lar hydrogen ion secretion and NH4+ production. However,
CHAPTER 47 Regulation of Acid–Base Balance 483

Extensive use of
diuretics

Extracellular-
volume contraction Potassium depletion

Renin secretion

Plasma renin
(Helps maintain the alkalosis)

Plasma angiotensin II

Aldosterone secretion

Plasma aldosterone

FIGURE 47–6 Pathway by which overuse


of diuretics leads to a metabolic alkalosis. NH4+
production and excretion are increased by the
Tubular secretion of hydrogen ion presence of a high aldosterone and potassium
depletion. The extracellular volume contraction,
via both aldosterone and as yet unidentified
nonaldosterone mechanisms, helps to maintain
Reabsorption of all filtered bicarbonate and the alkalosis once it has been generated. If the
contribution of new bicarbonate to blood diuretics have also caused chloride depletion, this
too will contribute to the maintenance of the
metabolic alkalosis (not shown). (Modified with
permission from Eaton DC, Pooler JP: Vander’s Renal
Generation of metabolic alkalosis Physiology, 7th ed. New York, NY: Lange Medical Books/
McGraw-Hill, Medical Pub. Division, 2009.)

the combination of potassium depletion of even moderate diuretic use for weight loss) that can generate a metabolic
degree and high levels of aldosterone stimulates tubular alkalosis (Figure 47–6).
hydrogen ion secretion markedly (NH4+ production also
increases significantly). As a result, the renal tubules not
only reabsorb all filtered bicarbonate, but also contribute
inappropriately large amounts of new bicarbonate to the CLINICAL CORRELATION
body, thereby causing metabolic alkalosis. Note that there
A 7-year-old boy is brought by his mother to his pediatri-
may have been nothing wrong with the acid–base balance to
cian. The boy has been vomiting on and off for over a day
start with: the alkalosis is actually generated by the kidneys
and has not been able to eat or drink anything. He seems
themselves. Of course, if alkalosis were already present, this
jittery and complains of tingling around his mouth. His
high aldosterone–potassium depletion combination would
blood pressure is 107/77, and his heart rate is 89. After the
not only prevent the kidneys from responding appropriately,
pediatrician notes signs of dehydration, he directs them to
but would also make the alkalosis worse. This phenomenon
a local hospital ER. At the hospital, blood gas analysis
is important because the combination of a markedly elevated
shows an arterial pH of 7.52 and Pco2 of 45 mm Hg, for a
aldosterone and potassium depletion occurs in a variety of
calculated bicarbonate of 36 mEq/L, which is high. Analy-
clinical situations, the most common of which is the exten-
sis of his urine, which is scant, yields a chloride of 17 mEq/L,
sive use of diuretic drugs (e.g., from extensive, inappropriate
484 SECTION VII Renal Physiology

which is low. He is given intravenous normal saline (0.9% STUDY QUESTIONS


NaCl) and an antiemetic to reduce the vomiting. 1. A patient excretes 2 L of alkaline (pH 7.6) urine having a
The boy has lost a lot of hydrogen ions, chloride, and bicarbonate concentration of 28 mmol/L. What do we know
water, resulting in hypovolemia and a metabolic alkalosis. about titratable acid excretion?
The relatively rapid development of the alkalosis leads to a A) It is 56 mmol.
transient hypocalcemia (see Chapter 48), in turn leading to B) There is negative titratable acidity.
hyperexcitability of peripheral neurons, tending to produce C) Titratable acid plus ammonium sums to 56 mmol.
muscle spasms and perioral tingling. His slightly high Pco2 D) We cannot determine without data for ammonium and
represents a partial compensation for the alkalemia. His phosphate.
body responds to the volume depletion by decreasing the 2. Which of the following is an acid load per se or becomes an acid
excretion of salt and water via decreased GFR and activa- load after metabolism?
tion of the RAS, which stimulates sodium reabsorption. A) eating a large steak
Given the elevated plasma bicarbonate, the kidneys should B) eating unsweetened grapefruit juice
begin excreting bicarbonate, but it is difficult for them to C) eating sweetened grapefruit juice
D) intravenous infusion of sodium lactate
do so in the face of the volume depletion. First, aldosterone
is stimulating the type A intercalated cells to secrete hydro- 3. How does the proximal tubule handle filtered bicarbonate?
gen ions, just the opposite of what is needed. Second, it is A) Bicarbonate is taken up by the tubular cells on a symporter
difficult for the type B intercalated cells to increase their with sodium.
B) Bicarbonate is taken up by the tubular cells via antiport with
secretion of bicarbonate, a process that is accomplished by
small base anions (e.g., formate).
exchanging cellular bicarbonate for luminal chloride via an C) Bicarbonate is taken up by the tubular the cells via antiport
antiporter. Less luminal chloride is available to exchange with chloride.
with bicarbonate due to upstream reabsorption. Since there D) Bicarbonate combines with a proton in the lumen and is
is no renal pathology per se, and the vomiting is transient, converted to carbon dioxide and water.
his case can be treated by replacing fluid loss and restoring 4. In which situation(s) would you expect to see decreased or no
chloride. renal excretion of acid equivalents?
A) during a major metabolic acidosis such as diabetic
ketoacidosis
B) during a time when the pancreas is secreting a high amount
CHAPTER SUMMARY of bicarbonate-rich fluid into the GI tract
■ Plasma pH is regulated by controlling the concentrations of C) in response to consuming a large number of antacid tablets
CO2 (Pco2) and bicarbonate. D) all of the above situations
■ Fixed acid or base loads are turned into excesses or deficits of 5. What is the fate of ammonium secreted in the proximal tubule?
bicarbonate in the body. A) It flows through to the urine.
■ Fixed acids and bases can enter the body via GI processes, B) It is mostly reabsorbed in the collecting ducts.
metabolism, intravenous infusions, and renal processes. C) It is mostly reabsorbed in the thick ascending limb and
secreted again in the collecting ducts.
■ Under all conditions, the kidneys must recover virtually all
D) It is mostly reabsorbed in the thick ascending limb and
filtered bicarbonate.
combined with bicarbonate to form urea.
■ The kidneys excrete acid by means of filtered or synthesized
urinary buffers. 6. A person develops a decreased arterial Pco2 because of
hyperventilation. If this condition persists, what kind of response
■ Phosphate is the most important filtered urinary buffer.
would we expect by the kidneys?
■ The kidneys excrete acid by converting glutamine to bicarbon-
A) more urinary titratable acidity
ate and ammonium, excreting the ammonium, and returning
B) more urinary bicarbonate
the bicarbonate to the blood.
C) more urinary ammonium
■ Primary acid–base disorders that change either Pco2 or D) a decreased urinary pH
bicarbonate can be compensated by changing the other variable
in the same direction, thereby preserving the ratio of bicarbon-
ate to Pco2.
■ Some situations, including volume contraction and aldosterone
excess, can cause the kidneys to excrete too much acid,
generating a metabolic alkalosis.
48
C H A P T E R

Regulation of Calcium
and Phosphate Balance
Douglas C. Eaton and John P. Pooler

O B J E C T I V E S

■ State the normal total plasma calcium concentration and the fraction that is
free.
■ Describe the distribution of calcium between bone and extracellular fluid
and the role of bone in regulating extracellular calcium.
■ Describe and compare the roles of the gastrointestinal tract and kidneys in
calcium balance.
■ Describe and compare bone remodeling and calcium buffering by bone.
■ Describe the role of vitamin D in calcium balance.
■ Describe how the synthesis of the active form of vitamin D (calcitriol)
is regulated.
■ Describe the regulation of parathyroid hormone secretion, and state the
major actions of parathyroid hormone.
■ Describe renal handling of phosphate.
■ Describe how parathyroid hormone changes renal phosphate excretion.

OVERVIEW to complex with phosphate is a key aspect of the normal for-


mation and maintenance of bone structure. Furthermore,
Calcium plays a key role in a multitude of physiological pro- when levels of calcium and/or small anions such as phosphate
cesses. Therefore, it is essential to regulate both the total are increased, they approach the solubility limit of the com-
amount of calcium in the body and its plasma concentration. plexes, leading to pathologies such as the formation of stones
Regulation is accomplished by the cooperative effort of several in the urinary tract and deposition of calcium complexes in
organ systems, including the gastrointestinal (GI) tract (Chap- soft tissue. For these reasons, the body possesses elaborate
ter 52), the endocrine system (Chapter 64), and the kidneys. means to regulate plasma calcium.
The importance of calcium centers on its being (1) a structural While calcium obeys the principles of input–output balance
component of bone, (2) an intermediate in intracellular sig- as do the other substances we have discussed, its regulation is
naling cascades, and (3) a regulator of the conformation of fundamentally different. First, whole body calcium balance is
membrane proteins. These functions derive largely from the regulated predominantly by input rather than output. Control
chemical properties of the calcium cation. Unlike sodium, of input from the GI tract is a crucial determinant of whole
potassium, and magnesium, calcium is “sticky,” meaning that body calcium. Output by the kidneys plays an important, but
it avidly binds to anionic sites on proteins and readily forms secondary, role.
complexes with small anions such as phosphate and citrate. Second, moment-to-moment regulation of plasma calcium
These associations have consequences for physiological func- is achieved by shifting calcium in and out of bone. Bone stores
tion. When calcium binds and unbinds from membrane pro- of calcium are an enormous buffer system that keeps plasma
teins, this alters the net charge, and hence the conformation of calcium nearly constant regardless of whole body balance.
those proteins. In the case of voltage-gated channels, this Thus, ordinary variations in ingestion and excretion have little
affects membrane excitability (see Chapter 2). The propensity effect on plasma levels because of this tight buffering.

485

Ch48_485-490.indd 485 11/26/10 10:30:24 AM


486 SECTION VII Renal Physiology

The long-term regulation of total calcium in bone is, of course, Lumen Interstitium
important for bone growth during childhood and bone integ-
rity in adult life. Here the kidneys play an important but indi-
rect role because they (1) excrete calcium in the urine and (2) ATP
are involved in forming the active form of vitamin D, which is Calbindin Ca
a major controller of GI input.
Normal levels of plasma calcium are about 10 mg/dL Ca Ca
(2.5 mmol/L or 5 mEq/L). This calcium exists in three general Ca
forms. First, almost half is in the free ionized (Ca2+) form. This Ca Ca
is the only form that is biologically active in target organs. Na
Second, about 15% is complexed to anions with relatively low Ca
molecular weights, such as citrate and phosphate. Third, the
remaining 40% is reversibly bound to plasma proteins.

FIGURE 48–1 Generic method for transcellular calcium


EFFECTOR SITES transport in the GI tract and kidney. In all body cells the free
intracellular calcium concentration must be kept to minuscule levels
FOR CALCIUM BALANCE to prevent formation of insoluble complexes and activation of
deleterious signaling pathways, even though the concentrations of
GI TRACT calcium at the two external surfaces of the cells are thousands of
times higher. The epithelial cells accomplish this by using diffusible
Most dietary calcium simply passes through the GI tract to the calbindins. As calcium enters the cells through channels in the
feces. The amount absorbed depends on many factors, includ- luminal surface, the calcium concentration in the microenvironment
ing the quantity of calcium in the diet. Some of the calcium near the channels is slightly raised, thus promoting the binding of
that is absorbed moves by a regulated active transcellular pro- calcium to calbindins. At the basolateral surface active extrusion of
calcium via ATPases and sodium–calcium antiporters lowers the
cess in the duodenum, while the majority is absorbed by
calcium concentration in the local microenvironment, promoting
unregulated paracellular diffusion in the rest of the small
dissociation of calcium from the calbindins. Ion charge symbols (Ca2+
intestine. In the active transport system calcium enters duode- and Na+) omitted for clarity.
nal cells passively through calcium-selective channels, binds
reversibly to mobile cytosolic calcium-binding proteins (called
calbindins), and is then actively transported out the basolat-
eral side via a Ca-ATPase and to some extent by a Na–Ca anti- via calcium-specific channels, diffusion bound to calbindins,
porter. Calbindins contain multiple binding sites for calcium and active exit across the basolateral membrane by a combina-
and are free to diffuse throughout the cytosol. They act as ferry tion of Ca-ATPase and Na–Ca antiport activity (Figure 48–2).
boats for calcium, permitting large amounts of calcium to Endocrine control of renal calcium handling is exerted in the
move from place to place within a cell, in this case from apical distal tubule.
to basolateral membrane, all the while keeping the concentra- The amount of calcium excreted in the urine, when averaged
tion of free calcium at a low level (see Figure 48–1). over time, is equal to the net addition of new calcium to the
body via the GI tract; thus, the kidneys help maintain a stable
balance of total body calcium. However, the change in renal
KIDNEYS excretion in response to changes in dietary input is much less
than the equivalent responses to dietary sodium, water, or
The kidneys handle calcium by filtration and reabsorption. potassium. For example, only about 5% of an increment in
The free component of plasma calcium is freely filterable. dietary calcium appears in the urine, whereas virtually all of an
Most of the filtered calcium is reabsorbed in the proximal increased ingestion of water or sodium soon appears in the
tubule (about 60% of the filtered load) and the remainder in urine. The reason is that most of the dietary increment never
the thick ascending limb of Henle’s loop, distal convoluted gains entry to the blood because it fails to be absorbed from the
tubule, and collecting duct system. Overall reabsorption is GI tract. In contrast, when dietary intake of calcium is reduced
normally 97–99%, leaving only a few percent of the filtered to extremely low levels, there is a slow reduction of urinary
load to be excreted. calcium, but some continues to appear in the urine for weeks.
Calcium reabsorption in the proximal tubule and thick What determines renal calcium excretion? First, calcium is not
ascending limb of Henle’s loop is largely passive and paracel- secreted. Second, while the filtered load is highly variable, being
lular, and the electrochemical forces driving it are dependent the product of free plasma calcium and GFR, the GFR is regu-
directly or indirectly on sodium reabsorption as they are for so lated mainly to meet the needs of sodium balance, not calcium.
many other substances. In contrast, calcium reabsorption in In fact, an increase in urinary calcium excretion can be induced
the more distal segments is active and transcellular. It uses the simply by administering salt. This feature is used clinically as an
same general mechanism as in the GI tract; that is, entrance emergency procedure when calcium levels in the blood get
CHAPTER 48 Regulation of Calcium and Phosphate Balance 487

Lumen set point for plasma calcium concentration maintained by the


Interstitium
rapid movement of calcium in and out of bone is critically
regulated by hormonal control, as discussed later.
Na ATP The slow remodeling process in bone described in Chapter 64
Cl Thiazides Na is important for bone integrity and calcium stores on a much
block K slower time scale, but this action has little effect on plasma cal-
PTH
stimulates cium levels.

Ca Ca
Ca Na
Ca Ca Ca HORMONAL CONTROL
OF EFFECTOR SITES
The regulation of calcium is achieved mostly through the
actions of two hormones: the active form of vitamin D (1,25-
FIGURE 48–2 Mechanism of calcium reabsorption in the (OH)2D; calcitriol) and parathyroid hormone (PTH). The
distal convoluted tubule, which is the major site for regulated
chemistry and production of these hormones is described in
reabsorption. Ca enters via apical Ca channels, under the control of
PTH, and is actively transported across the basolateral membrane via
Chapter 64 along with the actions of calcitonin, which plays
Na–Ca antiport and via a Ca-ATPase. The apical membrane also an extremely minor role, if any. Briefly, calcitriol is synthe-
contains the Na–Cl symporter (NCC), which is the target for inhibition sized through a series of steps beginning with cholesterol and
by thiazide diuretics. Interestingly, inhibition of NCC with thiazide culminating in the kidney, where a crucial hydroxylation step
diuretics promotes calcium reabsorption (probably by enhancing creates the active form. PTH is a small peptide that is synthe-
the basolateral sodium gradient and increasing Na–Ca exchange). sized by parathyroid glands lying behind the thyroid gland.
Thus, thiazides may reduce the calcium loss associated with Simply stated, the active form of vitamin D regulates what
osteoporosis. Ion charge symbols (Ca2+, Cl–, K+, and Na+) omitted for comes into the body and PTH regulates what is in the ECF.
clarity. (Modified with permission from Eaton DC, Pooler JP: Vander’s Renal
Physiology, 7th ed. New York, NY: Lange Medical Books/McGraw-Hill, Medical Pub.
Division, 2009.) VITAMIN D
The major action of calcitriol is to increase active absorption of
alarmingly high, the treatment consisting of administering large calcium and phosphate by the duodenum. A key mechanism is
amounts of saline, with the consequence that large amounts of to stimulate synthesis of the proteins involved in the steps
calcium-containing fluid pass through the kidneys to the urine. described earlier. In addition, vitamin D has some independent
Thus, we are left with changes in the active reabsorption in the actions on bone that are not entirely clear. Vitamin D also stim-
distal nephron as the means of exerting independent control of ulates the renal tubular reabsorption of calcium and phosphate,
calcium excretion. We will describe this control shortly. again by increasing the synthesis of the protein components in
the transport pathway. The influences of vitamin D on bone
and the kidney are far less important than its actions on the GI
BONE tract to stimulate absorption of calcium and phosphate.

Bone serves as the powerful short-term calcium-buffering sys-


tem that prevents large swings in plasma calcium. About 0.5 g PTH
of calcium passes back and forth between bone and the blood
plasma each day. Bone is also the repository of calcium that PTH secretion is acutely regulated by levels of free plasma cal-
keeps blood supplied with calcium during times of negative cium in a reciprocal manner—a decrease in plasma calcium
whole body calcium balance. stimulates secretion, and an increase inhibits secretion. Phos-
The calcium in bone exists in the form of crystalline phate also affects PTH secretion: increased plasma phosphate
hydroxyapatite (see Chapter 64). Calcium can move back and stimulates PTH secretion by stimulating the capacity of the
forth between blood and hydroxyapatite in the inner recesses parathyroid gland to synthesize PTH, so that chronically high
of bone via an interconnected cellular network of osteocytes. levels of phosphate lead to increased PTH.
The equilibrium between crystalline hydroxyapatite and its PTH exerts at least four distinct effects on calcium homeo-
dissolved components is highly labile, dependent on the con- stasis (summarized in Figure 48–3 showing the response to
centrations of calcium, phosphate, hydrogen ions, and specific hypocalcemia and discussed in detail in Chapter 64):
noncollagenous proteins in the immediate environment. The 1. PTH actions on bone normally increase the movement
formation and dissolution of hydroxyapatite protects the blood of calcium from bone into the ECF. They do this by
plasma from short-term swings in calcium concentration. favoring the dissolving of hydroxyapatite into its
This process does not require hormonal signals. However, the components.
488 SECTION VII Renal Physiology

Plasma calcium

PTH

Synthesis of
1,25 (OH)2-vit D

Rapid Rapid Slow Slow

Renal dissolution of Osteoclastic Intestinal


FIGURE 48–3 Physiological responses to Ca reabsorption hydroxyapatite bone resorption Ca absorption
reduced plasma calcium concentration.
Reduced plasma calcium stimulates secretion of
PTH. In response, the bone matrix immediately
releases calcium from hydroxyapatite into the ECF.
PTH also stimulates renal calcium reabsorption.
On a slower time scale PTH stimulates osteoclastic
bone resorption and increased synthesis of
calcitriol (vitamin D) in the kidney, leading to
Restoration of
increased absorption of dietary calcium from the
plasma calcium
GI tract.

2. PTH stimulates the activation of an intermediate form of sate for the lower calcium concentration that originally stimu-
vitamin D to calcitriol. The major control point is the sec- lated PTH secretion. Regarding the fourth effect, when PTH
ond hydroxylation step that occurs in the kidneys. This acts on bone, both calcium and phosphate are released into the
step is stimulated by PTH. If plasma calcium decreases blood. Similarly, calcitriol enhances the intestinal absorption
acutely, the subsequent increase in PTH immediately of both calcium and phosphate, so that the processes that are
stimulates calcium release from bone, thus restoring plas- restoring calcium to its normal level are simultaneously acting
ma calcium levels, and also stimulates calcium uptake (via to increase the plasma phosphate above normal. But this is an
calcitriol from the GI tract, on a slower time scale). This unwanted action because of the tendency to form insoluble
ensures that enough new calcium will enter the body to precipitates of calcium phosphate. Under the influence of
replace that “borrowed” from bone. PTH, plasma phosphate does not actually increase, because of
3. PTH increases renal–tubular calcium reabsorption, PTH’s inhibition of tubular phosphate reabsorption. Indeed,
mainly by an action on the distal convoluted tubule and this effect is so potent that plasma phosphate may actually
connecting tubule. At these locations, it acts rapidly decrease when PTH levels are high.
through activation of kinases that phosphorylate regula- All of the above describes the effects of an increase in PTH
tory proteins on a short-term basis. It also acts, on a slow- induced by a decrease in plasma calcium. An increase in extra-
er time scale, to increase synthesis of all the components cellular calcium concentration reduces PTH secretion, thereby
of the transport pathway. The increased uptake of calcium producing increased urinary and fecal calcium loss and net
from the tubular lumen stimulates basolateral extrusion movement of calcium from the ECF into bone.
(by a combination of Ca-ATPase activity and Na–Ca There are nuances to the actions of PTH on bone that have
antiporter activity) and thus decreases urinary calcium important clinical implications. The response of bone to PTH
excretion. depends on the pattern of its plasma concentration over time.
4. PTH reduces the proximal tubular reabsorption of phos- PTH can either promote resorption of hydroxyapatite (its
phate, thereby increasing urinary phosphate excretion usual action) or, if administered intermittently, promote depo-
and decreasing extracellular phosphate concentration. sition. Primary hyperparathyroidism, resulting from a
primary defect in the parathyroid glands (usually a hormone-
The adaptive values of the first three effects all result in a secreting tumor), generates a continuous excess hormone
higher extracellular calcium concentration and thus compen- level and causes enhanced bone resorption. This leads to bone
CHAPTER 48 Regulation of Calcium and Phosphate Balance 489

thinning and the formation of completely calcium-free areas treatment of hyperphosphatemia associated with chronic renal
or cysts. In this condition, plasma calcium often increases and failure is reduction of phosphate absorption from the GI tract.
plasma phosphate decreases, the latter caused by increased uri- This is accomplished by encouraging the ingestion of high
nary phosphate excretion. A seeming paradox is that urinary doses of calcium. The calcium forms complexes with phos-
calcium excretion is increased despite the fact that tubular cal- phate in the GI tract, reducing the availability of absorbable
cium reabsorption is enhanced by PTH. The reason is that the phosphate. The high levels of PTH in a healthy patient should
increased plasma calcium concentration induced by the effects signal the kidneys to form more calcitriol, but in chronic renal
of PTH causes the filtered load of calcium to increase even failure a further complication is a reduced ability to synthesize
more than it increases the reabsorptive rate. Because the fil- calcitriol in the kidney. Another clinical intervention in this
tered load is so great, there is also an increased amount not case is to provide exogenous calcitriol. This hormone sup-
reabsorbed (i.e., excreted). This result nicely illustrates the presses expression of the PTH gene in the parathyroid gland.
necessity of taking both filtration and reabsorption (and secre- The calcitriol increases GI absorption of phosphate, the very
tion, if relevant) into account when analyzing excretory thing we are trying to inhibit, but its ability to lower the syn-
changes of any substance. And, as mentioned earlier, the high thesis of PTH is the more important action because this
urinary calcium content promotes the formation of stones. reduces the excessive resorption of bone stimulated by PTH.
In contrast to what happens with the continuous presence of Thus, administering vitamin D or its active metabolite calcit-
elevated PTH that accelerates bone resorption and release of riol is a useful clinical tool.
calcium, intermittent increases (produced by injections once
per day) actually increase deposition of calcium in bone. Inter-
mittent injection of PTH is used therapeutically to increase
bone density in osteoporosis patients. CLINICAL CORRELATION
A 53-year-old African American male is hospitalized with
major vomiting, diarrhea, dehydration, and confusion, and
OVERVIEW OF RENAL the physicians are unable to obtain a history. His heart rate is
84, and blood pressure is 122/63. Temperature is 36.7°C.
PHOSPHATE HANDLING His blood work is normal, except for his calcium and phos-
Normally, approximately 75% of the filtered phosphate is phate. Calcium is high at 15 mg/dL, and phosphate is low at
actively reabsorbed, almost entirely in the proximal tubule in 1.6 mg/dL. He is given intravenous saline to replace fluid loss,
symport with sodium. Reabsorption is a tubular maximum– and then additional saline and a loop diuretic to induce
limited (Tm) system, and the normal filtered load is just a little excretion of calcium. After a day, this procedure has brought
higher than the Tm. Thus, while most filtered phosphate is his serum calcium down to 11.5 mg/dL (still somewhat high).
reabsorbed, some always spills into the urine. (Recall that this His physicians suspect primary hyperparathyroidism, which
phosphate is responsible for accepting hydrogen ions in the is usually caused by a parathyroid adenoma producing too
collecting duct and is the primary ion responsible for titratable much PTH and causing his high calcium and low phosphate.
acidity.) Since the reabsorptive capacity is saturated at normal Measurement of plasma PTH however shows a suppressed
filtered loads, any increase in filtered load simply adds to the intact PTH concentration. If the parathyroid glands are nor-
amount excreted. This occurs when plasma phosphate con- mal and the hypercalcemia is due to some other cause, the
centration increases for any reason, such as increased dietary production of intact PTH should be very low, because secre-
phosphate intake or release of phosphate from bone. Systemic tion should be suppressed by the elevated blood calcium. On
acidosis promotes release of calcium and phosphate from the other hand, if he has primary hyperparathyroidism, the
bone. The increase in plasma phosphate and the consequent PTH levels should be much higher than what was measured.
increase in filtered load of phosphate provides more titratable His physicians are unsure of a diagnosis, so they look for
buffer in the collecting tubule to help remove the excess hydro- causes other than primary hyperparathyroidism.
gen ion that promoted the phosphate release. They realize that this patient is showing the presence of
Much of the physiology we have described is illustrated by something that is acting like intact PTH and binding to the
the case of chronic renal failure, in which a low glomerular PTH receptor throughout the body, but has a different
filtration rate limits the ability of the kidneys to excrete a num- enough amino acid sequence so that it is not detected in the
ber of substances, specifically including phosphate. An almost clinical assay for authentic intact PTH produced by the para-
universal complication of chronic renal failure is increased thyroid glands. Further workup eventually reveals a carci-
plasma phosphate (hyperphosphatemia). Another common noma of the right kidney that is the source of this PTH
finding is increased levels of PTH, due in part to the high receptor agonist—it is called PTH-related peptide (PTHrP)
plasma phosphate. The PTH stimulates excessive bone resorp- and is a common cause of humoral hypercalcemia of malig-
tion, leading to osteoporosis. This is an example of secondary nancy, which is what the patient has. The right kidney is
hyperparathyroidism (because the pathology is not in the removed surgically. On the third postsurgical day, the serum
gland itself, but in the signals that drive it). One goal in the calcium and phosphate have returned to the normal range.
490 SECTION VII Renal Physiology

CHAPTER SUMMARY 3. In response to a sudden decrease in plasma calcium, what is the


source for most of the calcium that restores plasma levels?
■ Moment-to-moment regulation of plasma calcium primarily
A) bone
involves calcium flux between bone and plasma.
B) the GI tract
■ Calcium has a strong tendency to associate with small anions C) the renal tubules
and anionic sites on proteins. D) the organelles of tissue cells
■ The most important action of vitamin D is to ensure adequate 4. Compared with other common plasma cations such as
absorption of calcium from the GI tract. magnesium and potassium, calcium is unusual because
■ PTH is essential both to maintain proper calcium flux between it tends to
bone and plasma and to maintain adequate levels of vitamin D. A) form complexes with small anions and negative groups
■ Keeping phosphate levels in the normal range allows normal on proteins.
calcium retrieval from bone. B) form precipitates of elemental metal.
C) substitute for other ions on transporters.
D) diffuse passively through lipid bilayers.
STUDY QUESTIONS 5. In a case of acute hypercalcemia, one can rapidly lower plasma
calcium and increase urinary calcium excretion by
1. The most important action of calcitriol is to stimulate
A) feeding large amounts of phosphate
A) calcium deposition in bone. B) withholding phosphate from the diet
B) calcium resorption from bone. C) injecting PTH
C) calcium absorption from the GI tract. D) giving large amounts of saline
D) calcium reabsorption from the renal tubules.
6. Which condition(s) would directly or indirectly increase urinary
2. Why does prolonged excessive PTH lead to high urinary levels of excretion of phosphate?
calcium?
A) the actions of PTH on bone
A) PTH inhibits calcium renal calcium reabsorption in the B) the actions of osteoclasts in bone
proximal tubule, allowing more to be excreted. C) the actions of PTH on the kidneys
B) PTH stimulates resorption of calcium from bone, which D) all of the above would increase urinary phosphate excretion
raises plasma calcium and increases the filtered load.
C) PTH inhibits renal calcium reabsorption in the distal
nephron, allowing more to be excreted.
D) PTH stimulates renal calcium secretion in the distal
nephron.
SECTION VIII GI PHYSIOLOGY

49
Overview of the GI C H A P T E R

System—Functional
Anatomy and Regulation
Kim E. Barrett

O B J E C T I V E S

■ Understand the basic functions of the gastrointestinal system and the design
features that subserve these.
■ Describe the functional layers of the gastrointestinal tract and the
specializations that contribute to function.
■ Identify the segments of the gastrointestinal tract and the specialized
functions attributed to each.
■ Understand the circulatory features of the intestine and variations that occur
after meals.
■ Understand the integrated response to a meal and the need for mechanisms
that regulate the function of the gastrointestinal tract as a whole.
■ Describe modes of communication in the gastrointestinal tract.
■ Understand principles of endocrine regulation.
■ Understand the design of the enteric nervous system and neurocrine
regulation.
■ Describe immune and paracrine regulatory pathways.

OVERVIEW OF THE intestine serves to physically reduce the meal into a suspension
of small particles mixed with nutrients in solution. These are
GASTROINTESTINAL SYSTEM then chemically altered, resulting in molecules capable of tra-
versing the intestinal lining. These processes are referred to as
AND ITS FUNCTIONS digestion, and involve gastrointestinal motility as well as the
influences of pH changes, biological detergents, and enzymes.
DIGESTION AND ABSORPTION The final stage in the assimilation of a meal involves move-
The gastrointestinal system exists primarily to convey nutri- ment of digested nutrients out of the intestinal contents, across
ents and water into multicellular organisms. Most nutrients in the intestinal lining, and into either the blood supply to the gut
a normal human diet are macromolecules and thus not readily or the lymphatic system, for transfer to more distant sites in the
permeable across cell membranes. Likewise, nutrients are not body. Collectively, this directed movement of nutrients is
usually taken predominantly in the form of solutions, but referred to as absorption. The efficiency of absorption may vary
rather as solid food. Thus, in addition to the food uptake, the widely for different molecules in the diet, as well as those

491

Ch49_491-506.indd 491 11/26/10 10:28:58 AM


492 SECTION VIII GI Physiology

supplied via the oral route with therapeutic intent, such as drugs. Chopper
The barriers to absorption encountered by a given nutrient will
depend heavily on its physicochemical characteristics, and par-
ticularly on whether it is hydrophilic (such as the products of Blender
Acid sterilizer
protein and carbohydrate digestion) or hydrophobic (such as Reservoir
dietary lipids). Generally, the gastrointestinal tract does not rely
solely on diffusion to provide for uptake, but rather has evolved
Reaction vessel
active transport mechanisms that take up specific solutes with
high efficiency. There is also significant excess capacity in the Enzyme supplier
Detergent Neutralizer
systems for both digestion and absorption of a meal, including supplier
an excess of enzymes and other secreted products as well as an
excess in the surface area available for absorption in healthy
Catalytic and
individuals. Thus, assimilation of nutrients is highly efficient, absorptive surface
assuming adequate amounts are presented to the lumen.

EXCRETION Residue combuster


Dessicator and pelleter

The gastrointestinal system also serves as an important organ


for excretion of substances out of the body. This excretory Emission control device
function extends not only to the nonabsorbable residues of the FIGURE 49–1 The gastrointestinal system as a machine that
meal, but also to specific classes of substances that cannot exit conducts digestive, absorptive, immune, and excretory functions.
the body via other routes. Thus, in contrast to the renal sys- (Modified with permission from Barrett KE, Barman SM, Boitano S, Brooks H:
tem, which handles predominantly water-soluble waste prod- Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
ucts, the intestine works together with the biliary system to
excrete hydrophobic molecules, such as cholesterol, steroids,
and drug metabolites.
DESIGN OF HOLLOW ORGANS
HOST DEFENSE The gastrointestinal tract is a long muscular tube stretching
from mouth to anus. Within the lining of this tube, blind-
The inner surface of the intestinal tract exists in continuity with ended glandular structures invaginate into the wall of the gut
the exterior of the body. This, of course, is essential to its func- and empty their secretions into the lumen, defined as the cav-
tion of bringing nutrients from the environment into the body: ity within the gut. At various points along the length of the
however, this also implies that the intestine, like the skin and gastrointestinal tract, more elaborate glandular organs are also
respiratory system, is a potential portal into the body for less attached to the intestine and are connected to the intestinal
desirable substances. Indeed, we exploit this property to deliver lumen via ducts, allowing secretions to drain into the intestine
drugs via the oral route. In addition, the intestine is potentially where they can be mixed with intestinal contents. Examples of
vulnerable to infectious microorganisms that can enter the gut such organs include the pancreas and salivary glands. Glands
with the ingestion of food and water. To protect, itself and the in general can be considered as structures that convert raw
body, the intestine has evolved a sophisticated system of materials from the bloodstream into physiologically useful
immune defenses. The gastrointestinal immune system is secretions, such as acid and enzyme solutions.
characterized by specific functional capabilities, most notably In general, the glands have a common structure. Specialized
by being able to distinguish between “friend” and “foe”: mount- secretory cells form blind-ended structures known as acini
ing immune defenses against pathogens while being tolerant of where a primary secretion is produced. Clusters of such acini,
dietary antigens and beneficial commensal bacteria. which can be likened to a bunch of grapes, then empty into
tubelike ductular structures, with larger ducts collecting the
secretions from a group of smaller ones until a main collecting
ENGINEERING CONSIDERATIONS duct is reached that connects directly to the gut lumen. The
liver, which will be considered in this section as a critical par-
Given the functions of the gastrointestinal system discussed ticipant in gastrointestinal function overall, has a highly spe-
earlier, we will now consider the anatomic features needed to cialized structure that will be discussed in detail in Chapter 55.
support these functions. In this discussion, the gastrointesti- For now, suffice to say that the liver is designed not only to
nal system can be thought of as a machine (Figure 49–1) in secrete substances into the gastrointestinal lumen via the bil-
which distinct portions conduct the various processes needed iary system, but also to receive absorbed substances from the
for assimilation of a meal without uptake of significant quanti- intestine that travel first to the liver via the portal circulation
ties of harmful substances or microorganisms. before being distributed to the body as a whole.
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 493

Lumen

Epithelium
Basement membrane
Mucosa
Lamina propria

Muscularis mucosa

Submucosa

Circular muscle

Muscularis
Myenteric plexus propria

Longitudinal muscle FIGURE 49–2 Organization of the wall


of the intestine into functional layers. (Adapted
with permission from Madara J. and Anderson J.: Textbook
of Gastroenterology, 4th ed., pp. 151–165. Copyright
Mesothelium (Serosa) Lippincott Williams and Wilkins, Philadelphia, 2003.)

CELLULAR SPECIALIZATION before succumbing to programmed cell death (or apoptosis)


and being shed into the lumen or engulfed by their neighbors.
The tube that comprises the gastrointestinal tract is made Epithelial cells arise from stem cells that are anchored perma-
up of functional layers composed of specialized cell types nently in specific positions in the gut lining, located at the base
(Figure 49–2). The first layer encountered by an ingested of crypts in the intestine and in the middle of gastric glands in
nutrient is the epithelium, which forms a continuous lining the fundic region of the stomach. Following several cycles of
of the entire gastrointestinal tract as well as lining the glands
and organs that drain into the tube. The epithelium must
provide for the selective uptake of nutrients, electrolytes,
and water while rejecting harmful solutes. The surface area Small intestine
of the intestinal epithelium is amplified by being arranged LUMEN
into crypt and villus structures (Figure 49–3). The former
are analogous to the glands discussed earlier, whereas villi
are fingerlike projections that protrude into the intestinal
Villus
lumen, which are covered by epithelial cells. In the large
intestine, only crypts are seen, interspersed with surface epi-
thelium between the crypt openings.
The majority of the gastrointestinal epithelium is columnar
in nature, where a single layer of tall, cylindrical cells separates Crypt
the gut lumen from the deeper layers of the wall of the gut. The
structure of the columnar epithelium can be compared to a six
packs of soda cans, with the cans representing the cells and the Colon
plastic holder that links them as a series of intercellular junc- LUMEN Surface
tions that provide a barrier to passive movement of solutes
around the cells. However, in the first part of the intestinal
tube, known as the esophagus, the epithelial lining is a strati-
fied squamous epithelium. In this site, the epithelium forms Crypt
a multilayer reminiscent of the structure of the skin, with cells
migrating toward the lumen from a basal germinal layer.
The epithelium of the gut as a whole is subject to constant FIGURE 49–3 Comparison of the morphology of the
renewal, unlike the majority of tissues in the adult body. Gas- epithelial layers of the small intestine and colon. (Modified with
trointestinal epithelial cells turn over every 3 days or so in permission from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical
humans, undergoing a cycle of division and differentiation Books/McGraw-Hill, Medical Pub. Division, 2006.)
494 SECTION VIII GI Physiology

division, epithelial cells also differentiate into specialized cell


types with specific functions in the digestive process. Esophagus Upper and lower
In the stomach, some epithelial cells migrate downwards esophageal sphincters
deeper into the gland, and become chief or parietal cells
that contribute specific products to the gastric juice, or Stomach
endocrine cells that regulate the function of the latter secre-
tory cell types. The remaining gastric epithelial cells migrate Liver
Pylorus
upwards to become cells capable of secreting mucus and Sphincter of Oddi
Pancreas
bicarbonate ions.
In the intestine, some stem cells give rise to Paneth cells,
which remain in the crypt base and secrete antimicrobial pep- Gallbladder
Colon
tides. The majority of the daughter cells that arise from stem
cell divisions migrate upwards toward the villus (or surface Small intestine
epithelium in the colon), and of these, most are destined to
differentiate into absorptive epithelial cells with the capacity
for the final steps of nutrient digestion and uptake. A few
cells, however, differentiate into goblet cells, which produce
mucus, or enteroendocrine cells that respond to luminal con-
ditions, and regulate the functions of the other epithelial cell Internal and external
Ileocecal valve anal sphincters
types as well as those of more distant organs.
Beneath the epithelium is a basement membrane, overlying FIGURE 49–4 Overall anatomy of the gastrointestinal
a layer of loose connective tissue known as the lamina pro- system and division of the GI tract into functional segments
pria. This contains nerve endings and blood vessels, as well as by sphincters and valves. (Modified with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical
a rich assortment of immune and inflammatory cells. Taken
Pub. Division, 2006.)
together, the epithelium and lamina propria are referred to as
the mucosa. The mucosa also contains a thin layer of smooth
muscle known as the muscularis mucosae. Beneath this layer,
there is a plexus of nerve cell bodies known as the submucous of nutrients to the capacity of the enzymes required for diges-
(or submucosal) plexus, designed to relay information to and tion and the absorptive surface area. Similarly, the ileocecal
away from the mucosa, including the epithelial cells. Then, valve retains the majority of the gastrointestinal flora within
beyond the mucosa are the smooth muscle layers that provide the colon, opening only intermittently to permit the residues
for overall gut motility. These are arranged circumferentially of the digested meal, along with water and cellular debris, to
around the outer side of the intestinal tube. enter the large intestine. Finally, the sphincter of Oddi relaxes
Closest to the mucosa is a layer of circular muscle that in conjunction with a meal to allow the outflow of both biliary
reduces the diameter of the intestinal lumen when it contracts. and pancreatic secretions into the lumen.
On the outer side of the gut, a layer of smooth muscle in which Most gastrointestinal sphincters are under involuntary
the fibers are arranged longitudinally along the axis of the tube control, and perform their normal cycles of relaxation and
provides for intestinal shortening. Working together, these contraction without conscious input. Many may also func-
two outer muscle layers can provide for complex motility pat- tion in a manner that is largely autonomous of the central
terns that subserve specific gut functions, as will be described nervous system (CNS), being controlled instead by the
in more detail later. Sandwiched between the circular and lon- enteric nervous system. On the other hand, the external
gitudinal muscle layers, the myenteric plexus of nerves regu- anal sphincter can be controlled voluntarily, a skill learned
lates their function. during toilet training in infancy, and the esophageal sphinc-
ters are regulated by the CNS.

DIVISION OF INTESTINE INTO


FUNCTIONAL SEGMENTS ORGANS AND SYSTEMS
Movement of the meal along the length of the intestine is a reg- INVOLVED IN THE RESPONSE
ulated process, and involves selective retention in specific sites TO A MEAL
to promote optimal digestion and absorption. This is accom-
plished by specialized smooth muscular structures known as Several intestinal and extraintestinal tissues cooperate to
sphincters, whose function is also coordinated with that of the respond appropriately to the ingestion of a meal. Collectively,
system as a whole (Figure 49–4). For example, the pylorus, these tissues can sense, signal, and respond to meal ingestion
which controls outflow from the stomach, retains the meal in with altered function. Moreover, the tissues and their func-
the gastric lumen and releases it slowly to match the availability tions are interactive and highly efficient, and redundancy
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 495

exists among the majority of GI regulatory mechanisms. In Lower esophageal sphincter—


this section, we will introduce the functions of each segment prevention of reflux
of the gastrointestinal tract and the structural features that Cardia
underlie these. More detailed discussions will be provided in
subsequent chapters. Specific features of the circulatory sys-
tems designed to carry absorbed nutrients away from the gut,
and the neuromuscular system that provides for motility and

ure
regulation, will also be considered. Secretion Fundus and body

at
v
ur

ture
rc Reservoir
s se

urva
ORAL CAVITY AND ESOPHAGUS Le

c
ter
Mixing

ea
The oral cavity is concerned with initial intake of food and Grinding Gr
with shaping and lubricating the bolus of ingested materials
such that it can be swallowed. The teeth reduce large portions
of food into sizes suitable for passage through the esophagus. Antrum
Salivary glands, which drain into the oral cavity, supply an Pylorus—
aqueous environment and also mucus that coats the surface of control of emptying
the bolus and thus aids in swallowing. The aqueous environ- FIGURE 49–5 Functional regions of the stomach. (Modified
ment also permits diffusion of taste molecules to specific with permission from Barrett KE: Gastrointestinal Physiology. New York: Lange
receptors on the tongue that relay information centrally as to Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
whether the meal is palatable. Salivary secretions also reduce
microbial contamination of the oral cavity. Finally, the struc-
tures of the oral cavity are also intimately involved in swallow- these glands differ in the three regions of the stomach; they are
ing. The esophagus transfers the bolus from the mouth to the shallowest in the cardia, intermediate (though with deep pits)
stomach. Its upper third is surrounded by striated muscle in the antrum, and deepest in the fundus.
overlaid by a thick submucous elastic and collagenous net- The fundic (or gastric) glands contain specific secretory cells
work. The muscle then transitions to smooth muscle that that produce the characteristic components of gastric juice—
works in concert with the swallowing reflex to propel the bolus acid and pepsin—that are products of parietal and chief cells,
toward the stomach. respectively. Thus, the fundus is a secretory region. On the other
Toward the lowest portion of the esophagus, the smooth hand, the antrum (also referred to as the pyloric zone) engages
muscle gradually thickens and interacts with neurogenic and in extensive motility patterns, mixing the gastric contents and
hormonal factors, as well as the diaphragm, to serve function- grinding and sieving ingested particles. Eventually, the meal is
ally as a lower esophageal sphincter. The higher pressure in gradually emptied into the small intestine via the pylorus.
this segment prevents excessive backflow, or reflux, of the gas- The stomach also undergoes receptive relaxation. The gas-
tric contents into the esophageal lumen. Failure of this process tric musculature relaxes as it is stretched during filling, ensur-
leads to gastroesophageal reflux disease (GERD), where ing that the pressure in the stomach does not increase
refluxed contents of the stomach can cause damage to the significantly as its volume expands. This response ensures that
esophageal epithelium. GERD is one of the most common the meal is not forced back into the esophagus, and is integral
gastrointestinal disorders. to the reservoir function of the stomach. An individual lacking
a significant portion of his or her stomach cannot tolerate
large meals due to the loss of this reservoir function, making
STOMACH gastric restriction a treatment for obesity.
The stomach is a muscular bag that functions primarily as a res-
ervoir, controlling the rate of delivery of the meal to more distal DUODENAL CLUSTER UNIT
segments of the gastrointestinal tract. Anatomically, it is divided
into three regions, the cardia (which overlaps with the lower The first segment of the small intestine immediately distal to
esophageal sphincter), fundus, and antrum, each with distinctive the pylorus, approximately 12 in in length, is referred to as the
structures that subserve specific functions (Figure 49–5). duodenum. Together with the pancreas and biliary system,
The cardia begins where the squamous epithelium of the the duodenum makes up the duodenal cluster unit. This seg-
esophagus gives way to the columnar epithelium of the remain- ment of the gastrointestinal system acts as a critical regulator
der of the gastrointestinal tract; it functions mostly to secrete of digestion and absorption. Endocrine cells within the wall of
mucus and bicarbonate to protect the luminal surface from the the duodenum, as well as chemosensitive and mechanosensi-
corrosive gastric contents. At the microscopic level, the sur- tive nerve endings, monitor the characteristics of the luminal
face area of the stomach is amplified by pits, which represent contents and emit signals that coordinate the function of more
the entrances to deep gastric glands. The specific structures of distant regions of the gastrointestinal tract to ready them for
496 SECTION VIII GI Physiology

the arrival of the meal, or to retard the flow of contents from Splenic flexure
the stomach. The exocrine pancreas and gallbladder also drain Transverse colon
Hepatic flexure
into the duodenum with egress of secretions controlled by
opening of the sphincter of Oddi.

SMALL INTESTINE Descending


colon
The remainder of the small intestine consists of the jejunum Tenia
Ascending
and ileum. The jejunum serves as the site of the majority of colon
coli
nutrient absorption in the healthy individual, and has a mark-
Ileum
edly amplified surface area due to the presence of surface folds
and tall, slender villi. The surface area of the jejunum is
also amplified considerably by an abundance of microvilli on Haustra
Cecum
the apical surface of villus epithelial cells. More distally, the
ileum has fewer folds and shorter, sparser villi, and is less actively Appendix Sigmoid colon
engaged in nutrient absorption other than for specific solutes
such as conjugated bile acids, which are exclusively salvaged by Internal anal
Rectum
External sphincter
transporters expressed in the terminal ileum. However, if jeju-
nal absorption is impaired, the ileum represents an anatomic
FIGURE 49–6 Anatomy of the large intestine comprised of
reserve that can be called on for absorption. As a result, the the cecum, colon, rectum, and anus. (Reproduced with permission from
small intestine has excess capacity for both digestion and absorp- Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology,
tion, and thus malabsorption is a relatively rare event. 23rd ed. McGraw-Hill Medical, 2009.)

COLON
by contraction of the external anal sphincter and levator ani
The colon, or large intestine, serves as a reservoir for the stor- muscles, which are under voluntary control. Compared with
age of wastes and indigestible materials prior to their elimina- other segments of the gastrointestinal tract, propulsive motil-
tion by defecation. In general, colonic epithelial cells (or ity in the colon is relatively sluggish until a reflex sufficient to
colonocytes) do not express absorptive transporters for con- trigger mass peristalsis and defecation occurs, and contents
ventional nutrients such as monosaccharides, peptides, amino may remain in the colon for days.
acids, and vitamins but may be actively involved in the uptake
of other luminal constituents. As its name implies, the large
intestine is of a considerably larger diameter than the small SPLANCHNIC CIRCULATION
intestine, with a thicker wall and folds known as haustrations. AND LYMPHATICS
The colon is divided into several regions: the ascending, trans-
verse, descending, and sigmoid colon, which are defined ana- Blood supply to the intestines is vitally important in carrying
tomically but may also subserve different functions (Figure 49–6). away absorbed nutrients, particularly those that are water sol-
For example, in the ascending and transverse colon, there is an uble, to sites of usage elsewhere in the body. Likewise, most
emphasis on reclamation of fluid as well as salvage of other lipids enter the lymphatic drainage of the gut initially, because
dietary byproducts, such as absorption of short-chain fatty acids they are packaged in particles (chylomicrons) too large to pass
produced by the bacterial fermentation of dietary fiber. The through the pores between capillary endothelial cells. Lymph
smooth muscle of the colon, under the influence of the enteric fluid containing absorbed lipids is emptied into the blood-
nervous system, produces mixing motility patterns that maxi- stream via the thoracic duct.
mize the time for reabsorption of fluid and other useful solutes. The circulation of the gastrointestinal tract is unusual because
Other luminal solutes, such as bile acids and bilirubin, are also of its anatomy (Figure 49–7). Unlike venous blood draining
modified in the colon by bacterial metabolism. In fact, the from other organs of the body, which returns directly to the
healthy colon contains an abundant ecosystem comprised pri- heart, blood flow from the intestine flows first to the liver via
marily of anaerobic bacteria, and these symbionts are important the portal vein. Conversely, the liver is unusual in receiving a
contributors to whole body nutritional status. considerable portion of its blood supply not as arterial blood,
The descending colon serves primarily as the storage reser- but as blood that has first perfused the intestine. This anatomic
voir for fecal wastes. When these are propelled through the arrangement ensures that substances absorbed from the gut
sigmoid colon into the rectum, stretch receptors initiate a flow first to the hepatocytes where they can be detoxified if
reflex relaxation of the internal anal sphincter and also send necessary. Gastrointestinal blood flow is also notable for the
afferent impulses to the CNS indicating a need to defecate. range of its dynamic regulation. Even in the fasting state, the
Defecation can, however, be postponed to a convenient time splanchnic circulation receives blood flow (25% of cardiac
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 497

Vena cava myenteric plexus. Many of these fibers are contained in


Hepatic vein the vagus nerve, which follows blood vessels to innervate the
stomach, small intestine, cecum, and ascending and transverse
colon. The remainder of the colon receives parasympathetic
innervation via the pelvic nerve.
The most striking aspect of intestinal neurophysiology is the
enteric nervous system contained wholly within the gut wall.
Hepatic
sinusoids This system consists of neurons with their cell bodies in the
Hepatic artery myenteric or submucosal plexuses. The anatomy of the enteric
nervous system and its relationship to other gut structures is
Intestinal artery
(from mesentery)
shown in Figure 49–8.
The enteric nervous system serves as a relay station to con-
duct and interpret information supplied by extrinsic auto-
Portal vein nomic afferents carrying impulses that originate centrally,
Liver and also to pass information from sensory efferents that have
their endings in the epithelium or smooth muscle. Thus, the
enteric nervous system can ultimately cause changes in motil-
Intestinal capillaries
ity and/or secretory behavior in response to centrally medi-
ated signals. The enteric nervous system can also function
autonomously and mediate reflexes that do not involve the
Intestine CNS at all.
FIGURE 49–7 Schematic anatomy of the splanchnic
circulation. (Modified with permission from Barrett KE: Gastrointestinal Physiology.
New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
Myenteric plexus
Circular muscle
Deep muscular plexus
output) that is disproportionate to the mass of the organs per-
Submucosal
fused (5%). Under these circumstances, the liver receives plexus
approximately 65% of its blood flow via the portal system. In
the postprandial period, blood is diverted from the skeletal Longitudinal
muscles and other body systems, and flow through vessels per- muscle
fusing the intestine can increase more than 5-fold. Under these
circumstances, the liver receives more than 85% of its blood
supply via the portal system. These dramatic changes in blood Submucosal
distribution are produced by hormonal and neurogenic stimuli Mucosa
Muscularis artery
occurring secondary to the ingestion of a meal. A mucosae

Paravascular nerve Perivascular nerves


NEUROMUSCULAR SYSTEM
The motility functions of the gastrointestinal tract are essen- Subserous nerve Mesentery
tial to propel ingested nutrients along the length of the alimen- Myenteric plexus
tary canal, and also to control the length of time available for
digestion and absorption. The motility patterns of the intes-
tine are brought about by the integrated control of the contrac-
tion and relaxation of the circular and longitudinal muscle
layers. Extrinsic innervation of the gut occurs via both sympa-
Deep muscular
thetic and (more prominently) parasympathetic pathways. plexus
Sympathetic innervation primarily involves postganglionic Submucosal
plexus Mucosal plexus
adrenergic nerves originating in prevertebral ganglia. These
nerves synapse mainly with others in the enteric nervous sys- B
tem, discussed later, but a few may directly innervate secretory FIGURE 49–8 Plexuses of the enteric nervous system and
cells in various glands (especially the salivary glands) or the their relationship to the other functional layers of the gut wall.
smooth muscle cells of blood vessels, leading to vasoconstric- Panel A shows intact tissue while Panel B is a transverse section.
tion. Parasympathetic innervation, on the other hand, is via (Adapted with permission from Furness J. and Costa M. Neuroscience 1980;5:1–20.
preganglionic nerve fibers that synapse with cell bodies in the Copyright Pergamon Press.)
498 SECTION VIII GI Physiology

REGULATION OF COMMUNICATION
GASTROINTESTINAL FUNCTION IN THE GI TRACT
For nutrient assimilation to occur, specific tissues and GENERAL FEATURES OF
regions of the gastrointestinal system must sense, signal,
and respond to the ingestion of a meal (Figure 49–9). To
NEUROHUMORAL REGULATION
conduct the business of the gastrointestinal system most Communication that depended simply on diffusion of
efficiently, the various segments must also communicate. locally released signals would not be adequate for the timely
Thus, the activities of the gastrointestinal tract and the transfer of information from one intestinal segment to
organs that drain into it are coordinated temporally via the another. Likewise, the gastrointestinal tract also needs to
action of a series of chemical mediators, with the system communicate its status to organs that drain into it, such as
being referred to collectively as neurohumoral regulation, the pancreas and gallbladder. Thus, the system has evolved
implying the combined action of soluble and neuronal path- mechanisms for communication over significant distances,
ways. The integrated regulation of gastrointestinal function although local messengers also play a role in fine-tuning
underlies the efficiency of the system and its ability to pro- information delivery or, in some cases, amplifying or
vide for the effective uptake of nutrients even when they are antagonizing it. Overall, information is carried between the
in short supply. various sites by chemical entities possessing specific physi-
cochemical properties. Another general principle underly-
ing communication is that of functional redundancy. Several
different mediators may often produce the same physiologic
response, and single mediators may alter the function of
Nutrients more than one system.

Special Chemo/mechanosensitive CHARACTERISTICS OF


senses nerve endings CHEMICAL SIGNALS
Neurohumoral regulation is effected by several classes of
Higher chemical messengers including peptides, derivatives of amino
centers
acids, small molecule neurotransmitters, and lipid mediators.
The gastrointestinal messengers that have been assigned defi-
nite physiologic roles are listed in Table 49–1.
Dorsal
Vagus ENS
vagal
nerve
complex
SPECIFIC MODES OF
Stomach
Intestines
COMMUNICATION
Pancreas Four modes of communication are recognized within the
Gallbladder
Sphincters gastrointestinal system—endocrine, neurocrine, paracrine
(of which autocrine is a special case), and juxtacrine regula-
tion, most often ascribed to cells of the immune system. A
Changes in diagrammatic representation of each of these is provided in
secretion Figure 49–10.
and motility

FIGURE 49–9 Overview of neural control of the ENDOCRINE COMMUNICATION


gastrointestinal system. Nutrients activate both special senses
(smell, taste) and specific sensory nerve endings that exist within Because of its ability to regulate multiple sites in an essentially
the wall of the gut. These responses are conveyed via the autonomic
simultaneous fashion, endocrine regulation is critical to the
nervous system and enteric nervous system (ENS) to alter the
integrated function of the gastrointestinal system in response
function of the gastrointestinal tract and organs draining into it,
resulting in changes in secretion and motility. Such functional
to a meal. The intestine is extremely well supplied with cell
changes may additionally feed back on neural control to allow for types containing endocrine mediators (hormones); in fact, if
appropriate homeostasis of the system. (Modified with permission from all of the endocrine cells within the gut were assembled as a
Barrett KE: Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, single structure, they would make up the largest endocrine
Medical Pub. Division, 2006.) organ in the body.
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 499

TABLE 49–1 Major physiologic neurohumoral regulators of gastrointestinal function.


Endocrine Neurocrine Paracrine Immune/Juxtacrine

Gastrin Acetylcholine Histamine Histamine

Cholecystokinin Vasoactive intestinal polypeptide Prostaglandins Cytokines

Motilin Substance P Somatostatin Reactive oxygen species

Secretin Nitric oxide 5-Hydroxytryptamine Adenosine

Glucose-dependent insulinotropic peptide Cholecystokinin

5-Hydroxytryptamine

Somatostatin

Calcitonin gene-related peptide

Endocrine hormones are packaged within secretory gran- or GIP) are stored in G, S, I, and K cells, respectively. Cells
ules and released in response to nervous activity, as well as containing motilin have not been named, and indeed, there is
chemical and mechanical signals coincident with food inges- some debate as to whether this peptide is stored in endocrine
tion. The endocrine cells of the gut have been identified with cells or nerve endings.
letters to describe their hormonal contents; gastrin, secretin, Some endocrine cells may have processes that contact the
cholecystokinin (CCK), and glucose-dependent insulino- luminal contents and are activated to release their mediators in
tropic peptide (also referred to as gastric inhibitory peptide, response to specific features of luminal composition, such as
acidity, osmolarity, or nutrients such as amino acids and free
fatty acids. In other cases, hormone release in response to
changes in luminal composition can also be activated by a reflex
Endocrine Paracrine (autocrine)
arc that first involves activation of a sensory enteric nerve end-
ing, with subsequent release of specific neurotransmitters close
to the surface of the endocrine cell to stimulate exocytosis. Yet
other endocrine cells are designed to respond to conditions
existing in the interstitium. The hormones that are released
from endocrine cells diffuse into the lamina propria and thence
into the portal circulation. From there, they travel to target
organs and modify secretion, motility, and cell growth. All of
the currently known GI hormones are peptides, but not all pep-
tides isolated from the gastrointestinal tract are hormones.
Neurocrine Immune/Juxtacrine The GI hormones are synthesized in various segments of the
gastrointestinal tract (Figure 49–11), but only gastrin appears
to be present in the stomach of healthy individuals. The
remaining hormones are present in greatest amounts in the
duodenum and jejunum, with tapering expression of CCK and
secretin into the ileum. However, under normal conditions,
most of the release of gastrin occurs in the stomach, and of the
other hormones in the duodenum and to some extent the jeju-
num. Ileal expression of some hormones, therefore, represents
another example of the “reserve capacity” of the intestine that
can be called upon to regulate gastrointestinal function if
required. Further, in health, there appears to be little, if any,
expression of gastrointestinal hormones in the colon.

FIGURE 49–10 Modes of communication in the


gastrointestinal system. Information is conveyed by endocrine, NEUROCRINE REGULATION
neurocrine, paracrine, and immune/juxtacrine routes. Autocrine
regulation is a special class of paracrine regulation. (Modified with Neurocrine regulation is mediated by specific nerve endings
permission from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical of both the enteric and central nervous systems. Neurotrans-
Books/McGraw-Hill, Medical Pub. Division, 2006.) mitters stored in these nerve endings are released on receipt
500 SECTION VIII GI Physiology

Gastrin CCK Secretin GIP Motilin

Fundus

Antrum

Duodenum

Jejunum

Ileum

FIGURE 49–11 Sites of production of the


five gastrointestinal hormones along the length
of the gastrointestinal tract. The width of the bars
reflects the relative abundance at each location.
(Modified with permission from Barrett KE, Barman SM, Boitano Colon
S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed.
McGraw-Hill Medical, 2009.)

of an electrical signal, and diffuse across synaptic clefts to IMMUNE COMMUNICATION


alter secretomotor function in the gastrointestinal system.
These neurotransmitters thus provide information exchange A final class of communication in the gastrointestinal system is
that is exquisitely spatially specific, and because of their rela- mediated by the release of substances by cells of the mucosal
tive instability, there is very little spillover of information immune system. These cells are activated by antigenic sub-
conveyed by neurotransmitters even to immediately adjacent stances or products of pathogenic microorganisms, and release
sites. Some endocrine mediators may also convey informa- a variety of chemical mediators including amines (such as his-
tion among the various parts of the gastrointestinal system tamine), prostaglandins, and cytokines. Immune regulation is
by activating nerve endings in addition to their ability to important in changing the function of the secretomotor sys-
circulate to distant sites; the most classic example of this tems of the gastrointestinal tract during times of threat- for
mode of communication is mediated by CCK, for which example, invasion of the mucosa by pathogens. Immune medi-
receptors exist on sensory nerve endings in the small ators may also be responsible for intestinal dysfunction in the
intestinal mucosa. setting of inflammation or conditions such as food allergies,
where inappropriate immune responses to substances that
would normally be innocuous may be deleterious for the host.
PARACRINE COMMUNICATION Finally, immune cell types may be activated by endogenous
substances such as bile acids in the lumen, or by specific pep-
Some substances are designed to act only in the immediate tide neurotransmitters. Immune regulation thus contributes to
area of their release, and yet are released from cell types gastrointestinal regulation not only under pathological circum-
other than nerves. This paracrine communication provides stances, but also in response to normal physiologic events.
an important additional layer of control for gastrointestinal
secretomotor function, particularly in response to changes
in local conditions. Paracrine regulators, like neurotrans-
mitters, are readily metabolized or retaken up to limit PRINCIPLES OF ENDOCRINE
the duration of their activity. A special case of paracrine REGULATION
regulation is labeled autocrine, which involves the release
of a substance that then acts on its cell of origin. Intesti- ESTABLISHED GI HORMONES
nal epithelial cells may engage in autocrine regulation
since they are capable of releasing growth factors that influ- As noted above, five gastrointestinal peptides have fulfilled the
ence their proliferation and/or migration along the crypt– criteria to be named as hormones (Table 49–2). These fall into
villus axis. three groups based on structural and signaling similarities.
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 501

TABLE 49–2 Factors influencing release of gastrointestinal hormones.


Gastrin CCK Secretin GIP Motilin

Proteins/amino acids ↑ ↑ ↔ ↔ ↓a

Fatty acids ↔ ↑ ↑ ↑ ↓a

Glucose ↔ ↔ ↔ ↑ ↓a

Acid ↓ ↔ ↑ ↔ ↔

Neural stimulation ↑ ↑ ↔ ↔ ↑

Stretch ↑ ↔ ↔ ↔ ↔

Peptide-releasing factors ↔ ↑ ↔ ↔ ↔
a
Motilin release is reduced by feeding, but the precise mechanism is unclear.

GASTRIN/CCK FAMILY to distinct receptors on target cells. All of the receptors for these
family members, however, share the common property of signal-
Gastrin and CCK occur in the gastrointestinal system in vari- ing predominantly via associated G proteins of the Gs class, and
ous forms, and are structurally related peptides that also bind thus via increasing intracellular levels of cAMP.
to closely related receptors known as CCK-A and CCK-B. Secretin itself is a 27-amino acid peptide that is synthesized
CCK and gastrin share a common C-terminal pentapeptide, by S cells located predominantly in the duodenal mucosa, and
which is amidated as a final step in processing in I and G cells, is released in response to a low intraluminal pH. This accords
respectively. Amidation is believed to increase the stability of nicely with the major known biological action of secretin,
these hormones by blocking carboxypeptidase activity. which is to stimulate secretion of bicarbonate by the cells lin-
The major biologically active forms of gastrin are 17- and ing the pancreatic and biliary ducts, as well as the duodenal
34-amino-acid peptides, which may or may not be sulfated; epithelial cells themselves. Up to 80% of the bicarbonate secre-
this posttranslational modification is of unknown function. tory response that occurs in the course of digesting and absorb-
CCK also occurs as a family of peptides of decreasing length ing a meal is likely due to the direct influence of secretin.
(CCK-58, CCK-39, CCK-33, and CCK-8), but unlike gastrin, GIP, or glucose-dependent insulinotropic peptide (formerly
all of the released peptides are sulfated. The sulfation of CCK known as gastric inhibitory peptide, which fortuitously has
peptides appears critical for their high-affinity interaction the same initials), is released from intestinal K cells in response
with their receptor (CCK-A). to all of the major components of a meal—carbohydrates, pro-
CCK was named for its ability to contract (-kinin) the gall- tein, and fat. Its primary physiologic actions are to inhibit gas-
bladder (cholecysto-), but it also affects the function of numer- tric acid secretion and to amplify glucose-stimulated release of
ous other tissues and cell types, and can be considered as the insulin from the endocrine pancreas, making it an incretin.
master regulator of the duodenal cluster unit. It has also been The former action represents an example of a feedback regula-
shown to signal the CNS to indicate satiety, or fullness. CCK tory event that contributes to the termination of gastric secre-
also appears to cooperate with a major systemic regulator of tory function once the bulk of the meal has moved into the
food intake—leptin—that is released by adipocytes to signal small intestine. The latter action represents a feedforward sig-
the status of fat stores throughout the body. nal from the gut to the insulin-secreting cells of the endocrine
CCK-A and CCK-B receptors are G protein–coupled recep- pancreas such that the insulin response to absorption of glu-
tors that signal via increases in cytoplasmic calcium. The spec- cose is amplified (see Chapter 66).
ificity of CCK and gastrin for these receptors is defined by
their structures. Gastrin is highly specific for CCK-B, whereas MOTILIN
CCK binds to both CCK-A and (with lower affinity) CCK-B,
giving it a broader activity that may overlap to some extent Human motilin is a 22-amino acid linear peptide that is
with that of gastrin. released cyclically from the gut in the fasting state, and is
responsible for stimulating a specific pattern of gastrointesti-
SECRETIN FAMILY nal motility known as the migrating motor complex that will
be discussed in detail in Chapter 54. A closely related peptide,
The secretin family of gastrointestinal peptides includes not only ghrelin, is predominantly produced in the stomach, and its
the hormones secretin and GIP, but also a systemic hormone, plasma concentrations are increased by fasting and reduced by
glucagon, as well as a neuropeptide, vasoactive intestinal poly- feeding. Ghrelin may be an important mediator of signaling
peptide (VIP). Although there is some homology among the between the intestine and hypothalamus to increase metabolic
amino acid sequences of these peptides, each is believed to bind efficiency at times when nutrient supplies are limited.
502 SECTION VIII GI Physiology

CANDIDATE GI HORMONES an ileal brake, that is, a substance that acts to slow propulsive
motility and reduce luminal fluidity if nutrients remain unab-
As mentioned earlier, the gastrointestinal tract is a rich source sorbed by the time the meal reaches the ileum, thereby maxi-
of stored peptides, and several peptides have received atten- mizing contact time and ability to absorb nutrients.
tion for their potential physiologic roles. The most compelling
evidence exists for three such peptides—enteroglucagon,
pancreatic polypeptide, and peptide YY (tyrosine–tyrosine). PRINCIPLES OF NEUROCRINE
Enteroglucagon is a member of the secretin family, whereas
the other two peptides are related to each other, but not to any REGULATION
of the other hormone families thus far discussed. While none
of these peptides have yet fulfilled all of the criteria needed to “LITTLE BRAIN” MODEL OF
classify them as a hormone, it is possible that they may do so THE ENTERIC NERVOUS SYSTEM
in the future.
Intestinal L cells make peptides that are closely related to The enteric nervous system is often referred to as the “little
pancreatic glucagon, and arise from differential processing of brain” (as opposed to the “big brain” of the CNS) because
the same gene. One of these peptides, glucagonlike peptide-1, many of its responses are autonomous of central input. The
is a 30-amino acid peptide that inhibits gastric secretion and gastrointestinal system is unique in being the only organ sys-
emptying, and also potently augments the secretion of insulin tem outside the CNS with such an extensive system of intrinsic
in response to glucose (making it another incretin). The neural circuits. The various neurons of the enteric nervous
enteroglucagons are released in response to luminal sugars, system can be considered to perform functions in two primary
and thus may contribute to the axis by which circulating glu- areas (Figure 49–12). First, program circuits receive inputs
cose concentrations are regulated during the period of glucose regarding the physiologic status of the intestine, and translate
absorption after a meal, by coordinating the activities of the these into appropriate changes in function. Second, integra-
intestine and endocrine pancreas. As such, these presumed tion circuits relay such information to the CNS, and in turn
enteroglucagons act in concert with GIP. integrate information derived from the CNS with that sup-
Cells of the pancreatic islets synthesize pancreatic polypep- plied from intrinsic circuits to modify functional outcomes.
tide as a 36-amino acid linear peptide and release it in response As discussed earlier, the intrinsic nerves of the gastrointesti-
to ingestion of a meal, although the signals between the gut nal system are arranged into two plexuses—myenteric and
and pancreas have not been defined. Likewise, although the submucosal. Within these plexuses, the neurons can be subdi-
peptide can be shown to inhibit pancreatic enzyme and bicar- vided according to their functions (Table 49–3). In the myen-
bonate secretion, the physiologic significance of this is unclear teric plexus, inhibitory and excitatory nerves control the
because infusion of an antibody to neutralize the actions of function of the circular and longitudinal muscle layers. There
pancreatic polypeptide during meal digestion and absorption are also ascending and descending interneurons that relay
had no effect on the extent of pancreatic secretion. Thus, the information through the myenteric plexus along the length of
precise role of this peptide remains elusive. the gastrointestinal tract. In the submucosal plexus, secreto-
Finally, peptide YY is synthesized and released by enteroen- motor neurons, some of which also innervate blood vessels to
docrine cells in the distal small intestine and colon in response promote vasodilatation, regulate the secretion of fluid and
to the presence of fat in the ileal lumen. Its actions are largely electrolytes and contractions of the muscularis mucosa. The
inhibitory, reducing gastrointestinal motility as well as gastric plexuses also contain cell bodies of primary afferent nerves
acid secretion and secretion of chloride by the intestinal epithe- with projections to the mucosa designed to sense the physio-
lium. Some have proposed that peptide YY can be considered logic environment.

Epithelium

Secretomotor neuron
Sensory
neuron Smooth muscle

FIGURE 49–12 Schematic diagram of the enteric


nervous system (ENS) and its interactions with the central
PC IC
nervous system (CNS). PC, program circuit; IC, integration
circuit. (Reproduced with permission from Barrett KE: Gastrointestinal CNS
Physiology. New York: Lange Medical Books/McGraw-Hill, Medical Pub.
Division, 2006.)
ENS
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 503

TABLE 49–3 Classification of enteric nerves. On the other hand, painful sensations are conveyed via spinal
afferents that pass through the dorsal root ganglia.
Type Primary Neurotransmitters Vagal communication is largely mediated through the
Myenteric neurons enteric nervous system and involves cholinergic transmission.
Parasympathetic vagal input and vagovagal reflexes play a crit-
Stimulatory motor neurons Acetylcholine
ical role in regulating numerous gut functions, particularly
Inhibitory motor neurons Nitric oxide during the early phases of response to a meal. The pelvic nerve
Ascending and descending Acetylcholine, plays an analogous role in the distal colon and rectum. On the
interneurons 5-hydroxytryptamine other hand, sympathetic innervation to the intestine, medi-
ated by norepinephrine, is relatively limited in its extent and
Sensory neurons Substance P
implications under physiologic circumstances. Instead, it
Submucosal neurons seems likely that sympathetic regulation is called upon to over-
Noncholinergic secretomotor Vasoactive intestinal polypeptide ride the normal control of gut function, by slowing motility
neurons and inhibiting secretion, as a defense mechanism during times
Cholinergic secretomotor Acetylcholine of threat to whole body homeostasis.
neurons

Sensory neurons Substance P


PARACRINE AND
IMMUNE REGULATION
ENTERIC NEUROTRANSMITTERS
IMPORTANT MEDIATORS
Most, if not all, enteric neurons store multiple neurotransmit-
ters, but not all of the transmitters in a given nerve may be Paracrine and immune regulation of gastrointestinal function
equally important in terms of information transfer. Some gen- both involve the release of substances from nonexcitable cell
eral patterns are also apparent. Thus, excitatory nerves depend types, including enteroendocrine cells, enterochromaffin
largely on cholinergic neurotransmission. The actions of ace- and enterochromaffinlike (ECL) cells, and immune elements
tylcholine in muscarinic stimulatory pathways for either mus- in the lamina propria, which then act on neighboring cell types
cle contraction or secretory functions may be amplified by in the immediate environment. Important paracrine/immune
coreleased tachykinins such as substance P and neuroki- mediators are summarized, along with their major sources of
nin A. Acetylcholine also serves to deliver information from origin, in Table 49–4.
the parasympathetic branch of the autonomic nervous system, Note that some paracrines are also stored in nerves, and
largely via the vagus nerve, to the enteric neurons, although in thus play a dual role in signaling in the gut. For example,
this case it acts via nicotinic receptors. somatostatin, an important inhibitory peptide in the gut, is
Inhibitory nerves in the myenteric plexus, on the other synthesized by enteroendocrine D cells as well as being stored
hand, exert their effects predominantly via the release of nitric in interneurons of the enteric nervous system. Other para-
oxide, although several other neurotransmitters also play vary- crines may also derive from multiple cell sources. Thus, hista-
ing roles depending on the species and the segment of intes- mine is released from ECL cells in the gastric glands as a classic
tine being considered. These additional inhibitory
neurotransmitters include VIP, ATP, and pituitary adenylate
cyclase activating peptide (PACAP). VIP is also a critical TABLE 49–4 Important paracrine and immune
neurotransmitter for noncholinergic neurons in the submu- mediators in the gastrointestinal tract.
cosal plexus that function to stimulate secretomotor function
as well as vasodilation. Mediator Major Sources Selected Functions
Interneurons in the myenteric plexus utilize various neu- Histamine 1. ECL cells 1. Gastric acid secretion
rotransmitters to deliver information along the vertical axis, 2. Mast cells 2. Intestinal chloride
but one common transmitter in such nerves is serotonin secretion
(5-hydroxytryptamine [5-HT]). Other interneurons contain- 5-Hydroxytryptamine Enterochromaffin Response to luminal
ing acetylcholine and somatostatin have been implicated in cells nutrients
the generation of a motility pattern known as the migrating
Somatostatin D cells Various inhibitory effects
motor complex (see Chapter 54). Finally, the intrinsic primary throughout GI tract
afferents that relay information to the enteric program and
Prostaglandins Subepithelial Intestinal secretion;
integration circuits utilize tachykinins for sensory transmis- myofibroblasts vascular regulation
sion. These neurons ultimately control intestinal movements,
blood flow, and secretion in response to distension, luminal Adenosine Various cell types Intestinal secretion;
vascular regulation
chemistry, and mechanical deformation of the mucosal surface.
504 SECTION VIII GI Physiology

paracrine, but also from mucosal mast cells in response to inhibition controls the rate of delivery of nutrients such that
antigenic stimulation, where it acts as an immune mediator. this is matched with digestive and secretory capacity. Feed-
back mechanisms also terminate gut secretory responses
when they are no longer needed to assimilate a meal, to con-
MECHANISMS OF ACTIVATION serve resources and, in some cases, minimize possible adverse
consequences of overly prolonged exposure to gastrointestinal
Paracrine and immune regulators are primarily responsible
secretions.
for fine-tuning physiologic responses that are set into motion
by hormonal and neural regulation, and as such are usually
released in response to triggers that also act in the immediate CHAPTER SUMMARY
environment. Thus, both the endocrine and immune cells
■ The GI system fulfills the functions of digestion and absorp-
that release these substances can be considered as the gut tion, excretion, and host defense.
equivalent of the taste buds in the tongue that sample various
■ The GI system reflects a complex and cooperative network
components of ingested food and send information about its of various organs.
palatability. More distally, therefore, enteroendocrine cells are
■ Cellular specialization underlies the various functional
triggered in response to specific meal components, or by responses required of the GI system.
potentially injurious solutes in the lumen in the case of
■ The GI system is highly efficient, interactive, and redundant.
immune cells.
■ The circulatory features of the GI tract and liver set them apart
In some cases, the cells responsible for releasing paracrine
from other organs.
and/or immune effectors also receive neural input, and/or
■ Communication between the various segments of the GI tract,
are sensitive to the actions of circulating gastrointestinal
as well as the organs that drain into it, is vital for the integrated
hormones. The gastric ECL cell in the fundic region is an response to a meal.
excellent example of this, releasing histamine in response to
■ Communication is achieved via the endocrine, neurocrine,
both acetylcholine released from enteric nerve endings and paracrine, and immune mediators that act at sites distant from
gastrin traveling through the bloodstream from the gastric the site of stimulation and locally.
antrum. ■ The enteric nervous system serves to regulate the motility and
secretory responses of the gut, and to integrate this regulation
with information from the CNS.
INTEGRATION OF ■ Stimulatory and inhibitory nerves and neurotransmitters
are involved in the communication and regulation of the
REGULATORY SYSTEMS information.
There is considerable cross-talk between the regulatory sys- ■ Paracrine and immune messengers act locally to modulate
tems discussed in this chapter, as well as functional redundancy. endocrine and neurocrine signaling.
Moreover, communication mediated by one mode, for exam-
ple, endocrine, may secondarily activate other modes of com-
munication to amplify the eventual physiologic responses in STUDY QUESTIONS
target organs. An example of this is seen with the GI hormone 1. A patient receiving chemotherapy for a prostate tumor develops
CCK. On release from the gastrointestinal mucosa, CCK not severe abdominal pain and diarrhea. Following the treatment,
only travels through the bloodstream to activate secretory and his or her gastrointestinal symptoms subside. The resolution of
motor responses in other sites, but also binds to receptors on his or her symptoms most likely reflects repair of which of the
primary efferent nerve endings within the intestinal wall that following cell types?
can transmit vagovagal reflexes to propagate additional signal- A) epithelial cells
ing. Conversely, a neurocrine messenger, gastrin-releasing B) smooth muscle cells
peptide, acts on G cells to release a hormone that then can dis- C) lymphocytes
tribute the signal more broadly. D) enteric nerves
E) Paneth cells
Finally, the existence of multiple inputs to many of the cell
types involved in the integrated response to a meal not only 2. A pharmaceutical scientist trying to develop a new drug for
hypertension gives a candidate compound orally to rats. He or
provides functional redundancy, underscoring the importance
she determines that the drug is adequately absorbed from the
of gastrointestinal function for whole body homeostasis, but intestine, but levels in the systemic circulation remain below
also permits synergism, or potentiated responses, at the level the therapeutic range. The drug is most likely metabolized
of the target cell type. Synergism, or a greater than additive by which organ?
physiologic response, can be predicted to occur if the two (or A) small intestine
more) messengers in question activate their target cell by dif- B) kidney
ferent intracellular signaling cascades. C) lung
Integration of intestinal responses also involves the trans- D) liver
mission of negative, or inhibitory, signals. Such feedback E) spleen
CHAPTER 49 Overview of the GI System—Functional Anatomy and Regulation 505

3. A mouse is genetically engineered to lack CCK-B receptors. This 5. In a study of the secretion of gastrointestinal hormones, their
animal would be expected to display increased circulating levels concentrations in the portal vein are measured during luminal
of which of the following hormones? perfusion of the small intestine with solutions of various pH
A) gastrin levels. Which hormone will increase in the portal vein plasma
B) motilin during perfusion with a buffered solution of pH 3?
C) secretin A) CCK
D) CCK B) gastrin
E) insulin C) GIP
4. An experiment was conducted in which a balloon was inflated D) motilin
inside the stomach of a human volunteer and gastric pressures E) secretin
measured. Despite the increase in gastric volume, gastric
pressures remained relatively constant. This remarkable
pressure–volume relationship could be abolished by which of
the following pharmacological agents?
A) adrenergic agonist
B) inhibitor of the enzyme responsible for nitric oxide synthesis
C) cholinergic agonist
D) CCK
E) an antibody to gastrin
This page intentionally left blank
50
C H A P T E R

Gastric Secretion
Kim E. Barrett

O B J E C T I V E S

■ Understand the physiological role of gastric acid secretion, as well as that of


other gastric secretory products.
■ Identify the regions of the stomach and cell types from which the various
gastric secretions originate.
■ Understand how gastric secretion is initiated in response to anticipation of a
meal, and how secretion is amplified once the meal has been ingested.
■ Define cephalic, gastric, and intestinal phases of the secretory response.
■ Describe how secretion is terminated once the meal has left the stomach.
■ Define the cellular basis for acid secretion and the morphological changes
that take place in parietal cells to achieve this.

BASIC PRINCIPLES the digestive process via simple hydrolysis and is also antimi-
crobial. Enzymatic digestion of the meal also occurs as a result
OF GASTRIC SECRETION of gastric secretions. A proteolytic enzyme, pepsin, is secreted
as an inactive precursor, pepsinogen, and autocatalytically
ROLE AND SIGNIFICANCE cleaved at the low pH existing in the stomach lumen. Pepsin is
specialized for its role in mediating protein digestion in the
The stomach is a muscular reservoir into which the meal stomach because it exhibits optimum activity at low pH. The
enters after being swallowed. While limited digestion may gastric juice also contains intrinsic factor, synthesized by pari-
begin in the oral cavity as a result of enzymes contained in etal cells, and lipase, that contributes to the initial digestion of
saliva, the gastric juices represent the first significant source of triglycerides. Intrinsic factor binds to vitamin B12, also known
digestive capacity. However, the digestive functions of the as cobalamin, and is required for the eventual absorption of
stomach are not necessary for assimilation of a mixed meal, this vitamin more distally in the intestine. The stomach also
and indeed, surgical removal of the majority of the stomach secretes products important in protecting the mucosa from the
usually allows adequate nutrition. However, some degree of harsh effects of the luminal mixture of acid and enzymes.
gastric secretory function is required for the absorption of an Throughout the stomach, the surface cells are covered with a
essential vitamin, B12, and gastric acid may also be important layer of mucus. Mucus consists of a mixture of mucin glyco-
in the absorption of dietary nonheme iron. Gastric secretions proteins, surface phospholipids that endow hydrophobic prop-
also serve to sterilize the meal. erties on the surface of the mucus layer, and water. The stability
of this layer is additionally enhanced by the activity of small
GASTRIC SECRETORY PRODUCTS peptides, known as trefoil factors, which interact with the car-
bohydrate side chains of mucin molecules. Bicarbonate ions
The functions outlined in the previous section are subserved are also secreted into the base of this mucus layer and protect
by a number of products secreted by the stomach (Table 50–1). the gastric surface from excessively low and potentially injuri-
The most characteristic secretory product of the stomach is ous pH via simple neutralization. Finally, the stomach secretes
hydrochloric acid. The acidity of the gastric secretions begins a number of products into the mucosa that play critical roles in

507

Ch50_507-516.indd 507 11/26/10 10:28:08 AM


508 SECTION VIII GI Physiology

TABLE 50–1 Important gastric secretory products.


Product Source Functions

Hydrochloric acid Parietal cell Hydrolysis; sterilization of meal

Intrinsic factor Parietal cell Vitamin B12 absorption

Pepsinogen Chief cell Protein digestion

Mucus, bicarbonate Surface mucous cells Gastroprotection

Trefoil factors Surface mucous cells Gastroprotection

Histamine ECL cells Regulation of gastric secretion

Gastrin G cells Regulation of gastric secretion

Gastrin-releasing peptide Nerves Regulation of gastric secretion

Acetylcholine (ACh) Nerves Regulation of gastric secretion

Somatostatin D cells Regulation of gastric secretion

regulating the secretory and motility functions of the stomach,


including gastrin, histamine, and prostaglandins. The roles of Acid, intrinsic factor, pepsinogen
these factors will be discussed in more detail later.

Mucus layer

ANATOMICAL CONSIDERATIONS
FUNCTIONAL REGIONS
Surface mucous cells
OF THE STOMACH (mucus, trefoil peptide,
bicarbonate secretion)
The stomach lies between the esophagus and the duodenum,
and is delimited by the lower esophageal sphincter and the Cell migration Mucous neck cells
pylorus, respectively (Figure 49–5). The wall of the stomach (stem cell compartment)
contains thick vascular folds known as rugae, and the surface
epithelium is invaginated with a series of gastric pits. Each pit
opens to four to five blind-ended gastric glands. The stomach Parietal cells
(acid, intrinsic factor
can also be divided into three major regions by both structure
secretion)
and function. Most proximal is the cardia that represents
approximately 5% of the gastric surface area, and is a transi-
tional zone where the stratified squamous epithelium of the
esophagus gives way to the columnar epithelium that lines the ECL cell
remainder of the stomach and intestinal tract. The fundus or (histamine secretion)
body of the stomach contains approximately 75% of the gastric
glands, and in this region the oxyntic glands consist of spe-
cialized cell types from which arise the characteristic secre-
tions of the stomach (Figure 50–1). Finally, the antrum of the Chief cells
(pepsinogen secretion)
stomach, immediately proximal to the pylorus, is responsible
for the secretion of gastrin, the primary regulator of postpran-
dial gastric secretion. The antrum also fulfills important motil-
ity functions that will be described in Chapter 54.

FIGURE 50–1 Structure of a gastric gland from the fundus


GASTRIC CELL TYPES and body of the stomach. These acid – and pepsinogen-producing
glands are referred to as “oxyntic” glands in some sources. Notice the
The oxyntic, or parietal, glands found in the gastric fundus mitotic figures in several mucous neck cells indicating rapid cell
contain a variety of specific cell types (Figure 50–1). These turnover. (Adapted with permission from Barrett KE: Gastrointestinal Physiology.
include parietal cells, which are specialized to secrete acid and New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
CHAPTER 50 Gastric Secretion 509

A B
Golgi complex Golgi
complex
Intracellular
canaliculus

Tubulovesicular
membrane
Intracellular Mitochondria
canaliculus
Tubulovesicular
membrane
Basal folds Mitochondria
Basal folds Intracellular
Basement lamina
canaliculus
Basement lamina

Parietal cell (Nonsecreting)


Parietal cell (secreting)

FIGURE 50–2 A) Ultrastructural appearance of a resting parietal cell. Note the elaborate system of intracellular membranes and a large
number of mitochondria. B) Ultrastructural appearance of a parietal cell during active secretion. The apical membrane is massively amplified by
fusion of tubulovesicles and the secretory canaliculi. (Modified with permission from Ito S: Functional gastric morphology. In: Physiology of the Gastrointestinal Tract,
2nd ed. Johnson LR (editor). New York: Raven Press, 1987.)

intrinsic factor, and chief cells, which store pepsinogen in api- tionally significant communication with the gastric lumen.
cal granules that can release their contents via exocytosis. The D cells, which secrete somatostatin, are also present.
glands also contain endocrine cells that are responsible for
releasing products that regulate gastric function, particularly
the enterochromaffin-like (ECL) cells that synthesize hista- INNERVATION
mine. Toward the top of the gland where it joins with the gas-
Nerves carried through the parasympathetic vagus nerve,
tric pit, and moving out onto the gastric surface, the gland
with both efferent and afferent pathways, richly innervate the
contains surface mucous cells that secrete mucus. In the isth-
stomach. Vagal afferents convey information from the dorsal
mus and neck region of the gland lie the mucus neck cells,
vagal complex, which is integrated with that coming from
which are the precursors for all of the other differentiated cell
higher centers, such as the hypothalamus, to set the overall
types in the gland. These anchored stem cells give rise to
level of secretory function at any given moment. Visceral
daughter cells, which migrate downward to become parietal,
inputs also contribute to gastric regulation. Notably, the out-
chief, or endocrine cells, or upward to become surface mucous
put of taste receptors travels to a brain region called the
cells. The surface mucous cells turn over every 1–3 days in
nucleus tractus solitarius, where this information is again
adult humans.
translated into signals that regulate secretion and other gastric
The parietal cells are remarkable for their secretory capac-
functions. The enteric nerve plexuses throughout the gastro-
ity and energetic requirements. These cells secrete acid against
intestinal tract also encircle the walls of the stomach. These
a concentration gradient of more than 2.5 million–fold, from
allow for some degree of autonomous function, in addition to
the cytoplasmic pH of 7.2 to a luminal pH of less than 1 when
transmitting effects of central input.
secretion is maximally activated. To sustain such massive rates
The dorsal vagal complex represents an important site
of secretion, the parietal cell is packed with mitochondria,
where the various influences that can alter gastric secretion are
which are estimated to take up about 30–40% of the cell’s vol-
integrated. Thus, the dorsal vagal complex receives central
ume. The resting parietal cell also contains numerous mem-
input from the hypothalamus, as well as visceral input from
branous compartments known as tubulovesicles, as well as a
the nucleus tractus solitarius.
central canaliculus that deeply invaginates the apical mem-
brane (Figure 50–2A). This morphology changes dramatically
on cell stimulation (Figure 50–2B) as will be described more
fully later. REGULATION OF
In the antral mucosa, the glands do not contain parietal or GASTRIC SECRETION
chief cells, but instead are composed of both mucus-secreting
cells and enteroendocrine cells that regulate gastric function. Control of the secretion of the characteristic products of the
Particularly, the glands contain G cells, which synthesize and cell types lining the stomach represents a paradigm for control
release gastrin across their basolateral poles, and have func- of gastrointestinal function as a whole. Thus, the secretory
510 SECTION VIII GI Physiology

capacity of the stomach is closely integrated with signals coin- in the stomach. It participates in the stimulation of parietal,
cident with the ingestion of a meal, and modulated as the meal chief, and ECL cells, as well as the synapses between nerves
moves through the gastrointestinal tract to provide optimum within the enteric nervous system. In addition, a second impor-
digestion. There are many mechanisms, therefore, whereby tant gastric neurotransmitter is gastrin-releasing peptide
the function of the stomach is controlled. (GRP). GRP is released by enteric nerves in the vicinity of gas-
trin-containing G cells in the gastric antrum.

REGULATORY STRATA
Humoral Control
Short and Long Reflexes The gastric secretory response is also regulated by soluble fac-
Neural input provides an important mechanism for regulation tors that originate from endocrine and other regulatory cell
of gastric secretion (Figure 50–3). Reflexes contribute to both types, such as ECL cells (Figure 50–4). The primary endocrine
the stimulation and inhibition of secretion. For example, dis- regulator of gastric secretion is gastrin, which travels through
tension of the stomach wall activates reflexes that stimulate the bloodstream to stimulate parietal and ECL cells via their
acid secretion at the level of the parietal cell. These may be cholecystokinin (CCK)-B receptors.
short reflexes, which involve neural transmission contained Gastric secretion is also modified by paracrine mediators. His-
entirely within the enteric nervous system. In addition, long tamine is released from ECL cells under the combined influence
reflexes involve the activation of primary afferents that travel of gastrin and acetylcholine, and diffuses to neighboring parietal
through the vagus nerve, which in turn are interpreted in the cells to activate acid secretion via histamine H2 receptors. At one
dorsal vagal complex and trigger vagal outflow via efferent time, histamine was thought to be the final common mediator of
nerves that travel back to the stomach and activate parietal cells acid secretion, based in part on the clinical observation that his-
or other components of the secretory machinery. These long tamine H2 receptor antagonists can profoundly inhibit acid
reflexes are also called vagovagal reflexes. Acetylcholine secretion. However, it is now known that parietal cells express
(ACh) is an important mediator of both short and long reflexes receptors for not only histamine, but also acetylcholine (muscar-
inic m3) and gastrin (CCK-B) (Figure 50–5). Because histamine
H2 receptors are linked predominantly to signaling pathways that
involve cAMP, while ACh and gastrin signal through calcium,
Dorsal when the parietal cell is acted upon simultaneously by all three
vagal complex stimuli, a potentiated, or synergistic, effect on acid secretion
results. The physiological implication of this potentiation, or syn-
ergism, is that a greater level of acid secretion can be produced
Vago-vagal with relatively small increases in each of the three stimuli. The
reflex
pharmacological significance is that simply interfering with the
action of any one of them can significantly inhibit acid secretion.
In fact, synergism is a common theme in the control of several
different functions throughout the gastrointestinal system.
Acid secretion is also subject to negative regulation by spe-
cific mediators. Specifically, somatostatin is released from
D cells in the antral mucosa when luminal pH decreases below 3,
Acid Oxyntic and inhibits the release of gastrin from G cells. Elsewhere in the
Pepsin gland
stomach, somatostatin can also exert inhibitory influences on
ECL, parietal, and chief cells.

ENS Luminal Regulators


reflex
Stretch Specific luminal constituents also modulate gastric secretion
G receptor
indirectly. The example of pH is described earlier, but acid
cell
GRP output, at least, is also increased by components of the meal.
Short peptides and amino acids, derived from dietary protein,
Gastrin are capable of activating gastrin release from G cells.

Blood stream
FIGURE 50–3 Neural regulation of gastric secretion in
REGULATION OF SECRETION
response to gastric distension. Stretch of the stomach wall IN THE INTERDIGESTIVE PHASE
increases acid secretion via both intrinsic reflexes and vagovagal
reflexes. (Modified with permission from Barrett KE: Gastrointestinal Physiology. Between meals, the stomach secretes acid and other secretory
New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.) products at a low level, perhaps to aid in maintaining the
CHAPTER 50 Gastric Secretion 511

ANTRUM FUNDUS
Peptides/amino acids

GRP
H+
G cell ACh
H+ −
Parietal cell FIGURE 50–4 Regulation of gastric acid
and pepsin (P) secretion by soluble mediators
D cell
P and neural input. Gastrin is released from
Gastrin G cells in the antrum and travels through the
SST Chief cell circulation to influence the activity of ECL cells
ACh and parietal cells. The specific agonists of chief
? ?
cells are not well understood. Gastrin release is
Histamine
negatively regulated by luminal acidity via the
ACh release of somatostatin from antral D cells. SST;
Circulation ECL cell somatostatin. (Adapted with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical Books/
Nerve ending McGraw-Hill, Medical Pub. Division, 2006.)

sterility of the stomach. However, because no food is present, ing the gastric phase, when the meal is actually present in the
and thus no buffering capacity of the gastric contents, the low stomach. Secretion of other gastric products usually parallels
volume of secretions produced nevertheless has a low pH— that of acid.
usually around 3.0. Basal acid output in the healthy human is
in the range of 0–11 mEq/h, and is believed to reflect the
combined influences of histamine and ACh, released from
Cephalic Phase
ECL cells and nerve endings, respectively. Gastrin secretion Even before the meal is ingested, the stomach is readied to
during the interdigestive period, on the other hand, is mini- receive it by the cephalic (i.e., related to the head) phase of
mal. This is because gastrin release is suppressed by a luminal secretion. In fact, during the cephalic phase, several gastroin-
pH of 3 or below. testinal systems in addition to the stomach become activated,
including the pancreas and gallbladder. Higher brain centers
respond to the sight, smell, taste, and even the thought of food,
REGULATION OF and relay information to the dorsal vagal complex. In turn,
POSTPRANDIAL SECRETION vagal outflow initiates both secretory and motor behavior in
the stomach and more distal segments. Gastric secretion
In conjunction with a meal, gastric acid secretion occurs in occurring during the cephalic phase readies the stomach
three phases—cephalic, gastric, and intestinal—and increases to receive the meal. Vagal outflow activates enteric nerves
to 10–63 mEq/h. The major portion of secretion occurs dur- that in turn release GRP and ACh. Release of GRP in the vicin-

Resting Secreting
Canaliculus

Tubulo- H+, K+ ATPase FIGURE 50–5 Parietal cell receptors


vesicle and schematic representation of the
morphological changes depicted in Figure
50-2. Amplification of the apical surface area
is accompanied by an increased density of
H+,K+-ATPase molecules at this site. Note that
Ca++ Ca++
M3 acetylcholine (ACh) and gastrin signal via
cAMP CCK−B calcium, whereas histamine signals via cAMP.
ACh
M3 CCK−B Gastrin (Adapted with permission from Barrett KE:
H2 H2 Gastrointestinal Physiology. New York: Lange Medical
Histamine Books/McGraw-Hill, Medical Pub. Division, 2006.)
512 SECTION VIII GI Physiology

ity of antral G cells releases gastrin that travels through the gastrin and CCK. The intestinal phase of secretion may serve to
bloodstream to activate parietal and chief cells in an endocrine ready the stomach for its next meal. Of course, there is also over-
fashion. lap between the gastric and intestinal phases of secretion since
the meal moves only gradually into the duodenum.

Gastric Phase
The gastric phase of secretion is quantitatively the most CELLULAR BASIS OF SECRETION
important. In addition to vagal influences continuing from the
cephalic phase, secretion is now amplified further by mechan- ACID SECRETION
ical and chemical stimuli that arise from the presence of the
meal in the lumen. These include the luminal signals discussed The basolateral membrane of the parietal cell contains recep-
earlier, and signals arising from stretch receptors embedded in tors for histamine, gastrin, and ACh (Figure 50–5). The down-
the wall of the stomach. Thus, as the stomach distends to stream targets of the signaling pathways linked to receptor
accommodate the volume of the meal, these receptors initiate occupancy are presumed to include cytoskeletal elements, ion
both short and long reflexes to further enhance secretory channels, and the receptors themselves, the latter representing
responses either directly, via the release of acetylcholine in the a mechanism of negative feedback. Cytoskeletal rearrange-
vicinity of parietal cells, or indirectly, via the activation of ECL ments are implied by the dramatic morphological changes that
or G cells. The gastric phase also involves changes in motility occur as parietal cells transition from rest to secretion. At rest,
and is accompanied by a marked increase in gastric blood flow, the cytoplasm is filled with the tubulovesicles and intracellular
which supplies the metabolic requirements of the actively canaliculi. When the parietal cell is stimulated, the canaliculi
secreting cell types. fuse with the apical plasma membrane (Figure 50–5). The
Due to the combined influence of neurocrine and endocrine intracellular tubulovesicles, in turn, fuse to the canaliculi, mas-
signals, further amplified by histamine release from ECL cells, sively amplifying the surface area of the apical membrane that
secretory cells of the stomach are highly active during the gas- is in contact with the gland lumen by a factor of approximately
tric phase. Moreover, pepsinogen released by chief cells is rap- 5–10-fold. At rest, the tubulovesicles are the site for storage of
idly cleaved to pepsin in an autocatalytic reaction that occurs the majority of a membrane-bound transporter, H+,K+-ATPase,
optimally at pH 2, and this pepsin then acts on ingested pro- or proton pump, where it is therefore sequestered from the
tein to release short peptides and amino acids that further lumen. Following fusion of the tubulovesicles and canaliculi,
enhance gastrin release. Moreover, many dietary substances, the density of proton pumps in the apical pole of the cell is
including proteins, are highly effective buffers. Thus, while massively increased (Figure 50–5). These pumps are the sites of
acid secretory rates remain high, the effective pH in the bulk active pumping of protons into the gastric lumen.
of the lumen may increase to pH 5. This ensures that the rate Protons are generated adjacent to the apical membrane as a
of acid secretion during the gastric phase is not attenuated by result of the activity of the enzyme carbonic anhydrase II
an inhibition of gastrin release due to somatostatin. (Figure 50–6). This enzyme generates protons and bicarbonate
ions from the reaction of water and carbon dioxide. Protons are
then pumped out of the cell across the apical membrane in
Intestinal Phase exchange for potassium ions, with the consumption of cellular
As contents move out of the stomach into the duodenum, the energy. The potassium ions also originate from the cell cytosol,
buffering capacity of the lumen is reduced and the pH begins where they are maintained at levels above their chemical equi-
to decrease. At around pH 3, somatostatin release is triggered librium by the activity of Na+,K+-ATPase. They can therefore
from D cells, and acts to suppress gastrin release. D cells them- readily exit across the apical membrane through potassium
selves may be capable of responding to luminal acidity. There channels that are also localized to the tubulovesicles, and which
is also evidence for a neural pathway, involving the activation are opened when the parietal cell is stimulated. Specialized
of chemoreceptors sensitive to pH, which in turn leads to chloride channels are also present in this site, and serve to allow
release of the neuropeptide calcitonin gene-related peptide the apical exit of chloride ions to match the protons pumped
(CGRP). This peptide may then act on the D cells to induce from the cell. Thus, the final secretory product is actually
release of somatostatin. Other signals also limit the extent of hydrochloric acid. The overall mechanism should also remind
gastric secretion when the meal has moved into the small you of absorption of bicarbonate by the renal tubule, as was
intestine. For example, the presence of fat in the small intes- discussed in Chapter 47.
tine is associated with a reduction in gastric secretion. This A bicarbonate ion is generated for every proton that is
feedback response is believed to involve several endocrine and secreted, and if these were allowed to accumulate in the cyto-
paracrine factors, including GIP. sol, deleterious effects on cellular metabolism would result
Nevertheless, a portion of gastric secretion occurs once the from the resulting increase in pH. Thus, as the protons are
meal is in the intestine. The mediators of this response are largely secreted apically, the parietal cells also discharge bicarbonate
unknown, although CCK may play a role since the CCK-B ions across the basolateral membrane to maintain cytosolic
receptors on parietal cells do not discriminate markedly between pH within narrow limits. At least a portion of this bicarbonate
CHAPTER 50 Gastric Secretion 513

Lumen Blood Stream

Na+, K+ ATPase
+
2K
Potassium
channel
3Na+
H2O + CO2 Na+
NHE-1
H+
C.A.II
K+ HCO3

FIGURE 50–6 Ion transport proteins of


+ +
H , K ATPase
H+ H+ + HCO3 parietal cells. Protons are generated in the
HCO3
cytosol via the action of carbonic anhydrase II
Cl Cl (C.A.II). Bicarbonate ions are exported from
Cl /HCO3 the basolateral pole of the cell either by
ClC
vesicular fusion or via a chloride/bicarbonate
exchanger
Chloride exchanger. NHE-1, sodium–hydrogen
channel exchanger (Adapted with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical
Apical Basolateral Books/McGraw-Hill, Medical Pub. Division, 2006.)

transport occurs in exchange for the chloride ions that are will briefly review how the secretion of these products is con-
needed for apical secretion, via a chloride–bicarbonate trolled at the cellular level, although it should be noted that
exchanger. Some bicarbonate is likely also lost secondary to considerably less information exists on this topic than for
pumping into intracellular vesicles (distinct from the tubulo- gastric acid.
vesicles) that then move to the basolateral membrane and fuse Intrinsic factor is synthesized and released by parietal cells,
with it, discharging their contents. The bicarbonate leaving the presumably via a process of exocytosis, and activated by
cell is then picked up by the bloodstream. The arrangement of the same secretagogues that initiate acid secretion. However,
the microvasculature in the gastric mucosa carries a portion of while intrinsic factor is usually secreted in parallel with acid,
this bicarbonate up to the basolateral pole of surface epithelial these processes are not dependent on each other. Thus, proton
cells, which secrete bicarbonate to defend themselves against pump inhibitors have no inhibitory effect on the secretion of
the potentially injurious effects of acid and pepsin. This move- intrinsic factor. Pepsinogen is secreted by chief cells via a clas-
ment of bicarbonate into the bloodstream during gastric secre- sical process of compound exocytosis, and is thereafter acti-
tion is referred to as the alkaline tide. vated to its catalytic form in the presence of a low pH. The
The transport mechanisms that exist in parietal cells are active enzyme is inactivated if the pH increases above 5 (i.e.,
depicted in Figure 50–6. In addition to those already men- soon after the meal has moved into the duodenum, in healthy
tioned earlier, the basolateral membrane contains a sodium– individuals). As for other cell types that release their products
hydrogen exchanger, NHE-1, which expels protons from the via exocytosis, calcium is a key intracellular mediator effecting
cell in exchange for sodium ions, a process driven secondarily the secretory response, and ACh and GRP, both agents that
by the low intracellular sodium concentration established by elevate intracellular calcium, are known to be important chief
the Na+,K+-ATPase. At first blush, this may seem counterin- cell secretagogues. The precise roles of gastrin and histamine,
tuitive, since basolateral fluxes of protons would be predicted on the other hand, remain controversial. One additional secre-
to oppose the normal secretion of acid across the apical mem- tagogue that may be important, however, is secretin, especially
brane. However, the role of NHE-1 is not to participate in acid during the intestinal phase of gastric secretion. Surface epithe-
secretion, but to fulfill “housekeeping” functions, namely to lial cells throughout the stomach secrete mucus and bicarbon-
allow for the efflux of protons generated in resting cells by ate. The viscosity of mucus may limit diffusion of acid through
ongoing metabolic activities. A basolateral potassium channel the plane of the gel via a mechanism known as viscous finger-
that has also been identified in parietal cells likely also plays a ing. Thus, acid secreted under hydrostatic pressure from the
similar homeostatic role. gastric glands may emerge as a discrete stream through the gel,
restricting access of the acid to the gastric surface. Mucus-
secreting cells also package phospholipids that are secreted
OTHER PRODUCTS concurrently with mucins, in a manner analogous to the secre-
tion of surfactant in the lung. These phospholipids may
The stomach also secretes a number of additional products limit the back-diffusion of apical solutes, such as protons,
that are important in gastrointestinal physiology. Here, we toward the epithelium. Secretion of the components of the
514 SECTION VIII GI Physiology

mucus layer is increased by a variety of secretagogues, and is


even if secreted in normal amounts, due to its role in sus-
presumed to be under the control of both cholinergic and gas-
taining the activation of pepsin, and, in the case of duode-
trin-dependent signaling pathways, as well as local reflexes
nal ulcers, the direct injurious effects of protons on the
that may involve CGRP and tachykinins. Likewise, prostaglan-
epithelial cells in this site. In fact, a clinical adage, “no acid,
dins are potent mucus secretagogues, providing a partial
no ulcer,” also gives clues as to possible treatments. Patients
explanation as to why nonsteroidal anti-inflammatory drugs
with ulcer disease are treated typically with drugs that sup-
(NSAIDs), which prevent prostaglandin synthesis, predispose
press acid secretion, thereby giving the mucosa the oppor-
the gastric mucosa to injury and ulceration.
tunity to heal itself. In the past, this was accomplished
primarily with H2 antihistamines. However, more recently,
profound acid suppression—essentially total in nature—
CLINICAL CORRELATION has been accomplished with proton pump inhibitors. In
addition to acid suppression, ulcer patients who can be
A 55-year-old man is referred to a gastroenterologist for demonstrated to be infected with H. pylori usually receive
evaluation of a persistent burning sensation in his upper antibiotics to eradicate the microorganism, a treatment that
abdomen that has occurred intermittently for several markedly reduces the risk of any relapse.
months. The pain begins 2–3 hours after eating, often
awakens him at night, and is lessened by a meal. His history
reveals that he smokes one pack of cigarettes per day and
ingests half a bottle of red wine every night with dinner. CHAPTER SUMMARY
Initially, over-the-counter antacids reduced his symptoms ■ Gastric secretion plays important roles in digestion, absorption
to some extent, but recently they have provided little, if any, of specific nutrients, and host defense.
relief. The physician refers the patient for an upper endos- ■ Acid secretion occurs in phases that correspond temporally to
copy procedure, which shows several eroded areas of the the ingestion of a meal.
duodenal mucosa. A test for Helicobacter pylori infection ■ Regulation of acid secretion involves neurocrine, paracrine,
is also positive. A diagnosis of peptic ulcer disease is made, and endocrine components.
and the patient is started on a triple regimen of two antibi- ■ The stomach secretes other important products such as
otics and a proton pump inhibitor. The doctor also advises pepsinogen, intrinsic factor, mucus, and bicarbonate and trefoil
him to stop smoking and reduce his intake of alcohol, and peptides.
over the next 2 months, the patient’s symptoms resolve and ■ Various disease states can result from, or are associated with,
do not recur. abnormal gastric secretory function.
Peptic ulcer disease, so called because its pathogenesis is
related to the injurious effects of gastric acid and pepsin,
involves erosions through the epithelial lining of the stom- STUDY QUESTIONS
ach or duodenum that may ultimately lead to bleeding from
1. A 40-year-old man comes to his physician complaining of
mucosal blood vessels. For duodenal ulcer disease, at least,
epigastric pain. An upper endoscopy reveals duodenal erosions,
a failure in mucosal defense mechanisms seems more likely and a test of gastric secretory function reveals markedly elevated
to be the underlying pathogenic defect. There are two major levels of basal acid secretion that are increased only modestly by
exogenous causes of both gastric and duodenal ulcers: gas- intravenous infusion of a gastrin analog. What is the most likely
tric colonization with a gram-negative, spiral-shaped bac- diagnosis?
terium known as H. pylori and ingestion of NSAIDs. In the A) Zollinger–Ellison syndrome (a gastrin-secreting tumor)
absence of NSAID use, the vast majority of all ulcer patients B) H. pylori infection
can be shown to be infected with H. pylori, which is special- C) gastroesophageal reflux disease
ized to colonize the gastric niche because it secretes large D) gastroparesis (impaired gastric motility)
amounts of the enzyme, urease. This product converts urea E) achalasia (failure of the lower esophageal sphincter to relax)
to ammonium ions in the vicinity of the bacteria, thereby 2. In an experiment, rabbits are administered a cholinergic agonist,
protecting them from the deleterious effects of gastric acid- pentagastrin, or histamine intravenously, and gastric acid
ity. In genetically susceptible individuals, infection with secretion measured. Which treatment, when coadministered
H. pylori can have profound effects on both gastric and with each of these agents, would be expected to block gastric acid
duodenal physiology. NSAID-induced ulcers, on the other secretion produced by any of the stimuli?
hand, likely arise because the drugs suppress the synthesis A) histamine H2 antagonist
of prostaglandins that normally protect the mucosa via B) antibodies to gastrin
C) anticholinergic drug
their effects on mucus and bicarbonate secretion as well as
D) histamine H1 antagonist
blood flow. Acid also contributes to ulcer pathogenesis, E) proton pump inhibitor
CHAPTER 50 Gastric Secretion 515

3. A patient suffering from anemia comes to his physician 6. A patient who is being treated for long-standing osteoarthritis
complaining of frequent bouts of gastroenteritis. A blood test with a NSAID also takes a drug that inhibits acid secretion to
reveals circulating antibodies directed against gastric parietal reduce the toxicity of her NSAID treatment. She comes to her
cells. His anemia is ascribable to hyposecretion of which of the physician complaining of recurrent bouts of diarrhea during a
following gastric secretory products? series of business trips to Guatemala. The apparent increase in
A) histamine her sensitivity to infections acquired by the oral route is most
B) gastrin likely due to reduced secretory function of which of the
C) pepsinogen following?
D) hydrochloric acid A) stomach
E) intrinsic factor B) pancreas
4. Two medical students studying for their physiology final decide C) gallbladder
to take a break for a lunchtime hamburger. Before reaching the D) salivary glands
cafeteria, nervous impulses from the dorsal vagal complex will E) lymphocytes
initiate gastric acid secretion by triggering release of which
neurotransmitter from the enteric nervous system?
A) norepinephrine
B) vasoactive intestinal polypeptide
C) substance P
D) GRP
E) nitric oxide
5. Compared to the cephalic phase, the gastric phase of gastric acid
secretion is characterized by which of the following patterns?

Somatostatin
Acid Secretion Gastrin Secretion Secretion

A) Increased Increased Increased

B) Increased Increased Decreased

C) No change Increased No change

D) Decreased Decreased Increased

E) Decreased Decreased Decreased

F) No change Decreased No change


This page intentionally left blank
51
C H A P T E R

Pancreatic and
Salivary Secretion
Kim E. Barrett

O B J E C T I V E S

■ Understand the role played by the pancreas in digestion and absorption of a


mixed meal.
■ Understand the structure of the exocrine pancreas and the cell types that give
rise to proteinaceous and fluid components of the pancreatic juice.
■ Identify the key constituents of the pancreatic juice and the enzymes that are
secreted in inactive forms.
■ Describe the factors that regulate the release of secretin and the role of this
hormone in stimulating pancreatic ductular secretion.
■ Understand the role of CCK and other factors in regulating pancreatic acinar cells.
■ Identify the signaling events activated in pancreatic acinar cells by
secretagogues.
■ Compare and contrast the structure of the salivary glands with that of the
exocrine pancreas.
■ Identify the functions of saliva and the constituents responsible for these.
■ Define the ion transport pathways that modify salivary composition.
■ Define the regulatory pathways for saliva production.

BASIC PRINCIPLES PANCREATIC SECRETORY PRODUCTS


OF PANCREATIC SECRETION The exocrine pancreas is largely the site of synthesis and
secretion of enzymes. These fall into four main groups—
ROLE AND SIGNIFICANCE proteases, amylolytic enzymes, lipases, and nucleases—as
shown in Table 51–1. In addition, other proteins are produced
The exocrine pancreas is the source of the majority of enzymes that modulate the function of pancreatic secretory products,
required for digestion of a mixed meal (i.e., carbohydrate, such as colipase and trypsin inhibitors. Finally, the pancreas
protein, and fat). Pancreatic enzymes are produced in great secretes monitor peptide, which represents an important
excess, underscoring their importance in the digestive process. feedback mechanism linking pancreatic secretory capacity
However, unlike the digestive enzymes produced by the stom- with the requirements of the intestine for digestion. Almost
ach and in the saliva, some level of pancreatic function is nec- 80% (w/w) of the proteins secreted by the exocrine pancreas
essary for adequate digestion and absorption. In general, are proteases. Of the proteases, trypsinogen, the inactive pre-
nutrition is impaired if production of pancreatic enzymes cursor of trypsin, is the most abundant. This likely reflects a
decreases below 10% of normal levels, or if outflow of the pan- central role for trypsin in initiating the digestion of proteins,
creatic juice into the intestine is physically obstructed. which will be discussed further in Chapter 58.

517

Ch51_517-526.indd 517 12/7/10 9:56:58 PM


518 SECTION VIII GI Physiology

TABLE 51–1 Pancreatic acinar cell secretory products.


Amylolytic
Proteases Enzyme Lipases Nucleases Others

Trypsinogena Amylase Lipase Deoxyribonuclease Procolipasea

Chymotrypsinogena Nonspecific esterase Ribonuclease Trypsin inhibitors

Proelastasea Prophospholipase A2a Monitor peptide

Procarboxypeptidase Aa

Procarboxypeptidase Ba
a
Stored and secreted in inactive forms.

Like pepsinogen in the stomach, pancreatic proteases are exocytosis and fuse with each other and the apical membrane,
packaged and stored as inactive precursors. This is also true thereby discharging their contents into the lumen.
for at least one lipolytic enzyme, prophospholipase A2. The
need to store these enzymes in their inactive forms relates to
the toxicity of the active products toward the pancreas. Under DUCTULAR CELLS
normal circumstances, therefore, the pancreas does not
digest itself. The cells lining the intercalated ducts of the pancreas also play
an important role in modifying the composition of the pan-
creatic juice. They are classical columnar epithelial cells, com-
parable to those lining the intestine itself. When stimulated,
ANATOMIC CONSIDERATIONS these cells transport bicarbonate ions into the pancreatic juice,
IN PANCREAS with water following paracellularly. Thus, the effect of the duct
cells is to dilute the pancreatic juice and to render it alkaline.
Endocrine functions of the pancreas are restricted to cells
located in the islets of Langerhans, which are scattered
throughout the pancreatic parenchyma (see Chapter 66). The
Endocrine cells
exocrine functions, on the other hand, are conducted by a of pancreas
series of blind-ended ducts that terminate in structures known Exocrine cells
as acini (Figure 51–1). Many such acini, arranged like clusters (secrete
of grapes, disgorge their products into a branching ductular enzymes)
system that empties into larger and larger collecting ducts,
eventually reaching the main pancreatic duct or Wirsung’s
duct. A minor part of the pancreas is drained by an accessory
collecting duct, known as the duct of Santorini. Pancreatic Duct cells
(secrete
juice mixed with bile from the liver (see Chapter 56) enters bicarbonate)
the duodenum a short distance distal to the pylorus, under
Gallbladder Pancreas
the control of the sphincter of Oddi. Both the acinar and
ductular cells contribute distinct products to the pancreatic
juice, and both are regulated during the course of responding
to a meal.

ACINAR CELLS
Pancreatic duct
Pancreatic acinar cells are the source of the majority of the
proteinaceous components of the pancreatic juice. Their baso-
lateral membrane faces the bloodstream and contains recep-
tors for a variety of neurohumoral agents responsible for
regulating pancreatic secretion. The apical pole of the cell, on Common bile
Duodenum duct from gallbladder
the other hand, is packed at rest with large numbers of zymo-
gen granules that contain the digestive enzymes and other FIGURE 51–1 Structure of the pancreas. (Reproduced with
regulatory factors. When the cell is stimulated by secret- permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology,
agogues, the granules undergo a process of compound 11th ed. McGraw-Hill, 2008.)
CHAPTER 51 Pancreatic and Salivary Secretion 519

REGULATION OF CCK-A receptors localized to the basolateral membranes of aci-


nar cells and via stimulation of vagal afferents evoking vagova-
PANCREATIC SECRETION gal reflexes that stimulate acinar cell secretion through
cholinergic and noncholinergic neurotransmitters. CCK can
PHASES OF SECRETION also modulate the activity of other neurohumoral regulators on
the pancreas in a synergistic fashion. Notably, while CCK is a
Pancreatic secretory activity related to meal ingestion occurs weak agonist of pancreatic ductular secretion of bicarbonate by
in phases. The majority of the secretory response (approxi- itself, it markedly potentiates the effect of secretin on this trans-
mately 60–70%) occurs during the intestinal phase, but there port mechanism. During the integrated response to a meal,
are also significant contributions from the cephalic (20–25%) therefore, it is likely that the ability of secretin to evoke pancre-
and gastric (10%) phases. During the cephalic and gastric atic bicarbonate secretion is amplified by occurring against the
phases, secretions are low in volume with high concentrations background of a CCK “tone.” Nevertheless, CCK predominantly
of digestive enzymes, reflecting stimulation primarily of aci- affects acinar cell secretion. Thus, during the initial response to
nar cells. This stimulation arises from cholinergic vagal input a meal (i.e., the cephalic and gastric phases), pancreatic secre-
during the cephalic phase, and vagovagal reflexes activated by tions are low in volume with a high concentration of enzymes
gastric distension during the gastric phase. During the intesti- and enzyme precursors. The output of pancreatic enzymes, but
nal phase, on the other hand, ductular secretion is strongly not that of bicarbonate, that occurs in response to a meal can
activated, resulting in the production of high volumes of pan- essentially be reproduced by the intravenous administration of
creatic juice with decreased concentrations of protein, although postprandial concentrations of CCK. This situation should be
the total quantity of enzymes secreted during this phase is contrasted with secretory flows occurring in the intestinal
actually also markedly increased. Ductular secretion during phase, where secretin also plays a role, as discussed later.
this phase is driven primarily by the endocrine action of secre-
tin on receptors localized to the basolateral pole of duct epi-
thelial cells. The inputs to the acinar cells during the intestinal Factors Causing CCK Release
phase consist of cholecystokinin (CCK) as well as neurotrans- CCK is synthesized and stored by “I” cells, endocrine cells
mitters including acetylcholine (ACh) and gastrin-releasing located predominantly in the duodenum (Figure 51–3). Con-
peptide (GRP). The large magnitude of the intestinal phase is trol of CCK release is carefully regulated to match needs for
also attributable to amplification by so-called enteropancreatic CCK bioactivity. In part, this is accomplished by the activity of
reflexes transmitted via the enteric nervous system. two luminally active CCK-releasing factors, which are small
peptides. One is derived from the duodenum, and called CCK-
ROLE OF CCK releasing peptide (CCK-RP). It is released in response to fatty
acids and aromatic amino acids. The other luminal peptide that
CCK can be considered a master regulator of the duodenal controls CCK secretion is monitor peptide, which is a product
cluster unit and especially the pancreas (Figure 51–2). It is a of pancreatic acinar cells. Release of monitor peptide can be
potent stimulus of acinar secretion, acting both directly on neurally mediated, including by the release of ACh and GRP
during the cephalic phase, and mediated by subsequent vago-
vagal reflexes during the gastric and intestinal phases of the
CCK response to a meal. Likewise, once CCK release has been stimu-
lated by CCK-RP, it too can cause monitor peptide release.
The significance of factors that regulate CCK release lies in
their ability to match pancreatic secretion of proteolytic
Sphincter enzymes to the need for these enzymes. When meal proteins
Gallbladder Pancreas Stomach and peptides are present in the lumen in large quantities, they
of Oddi
compete for the action of trypsin and other proteolytic
enzymes, meaning that CCK-RP and monitor peptide are
Contraction Acinar Reduced Relaxation
degraded only slowly. Thus, CCK release is sustained, causing
secretion emptying
further secretion of pancreatic juice. On the other hand, once
the meal has been fully digested and absorbed, CCK-RP and
monitor peptide will be degraded by the pancreatic proteases
• Protein, carbohydrate, lipid absorption and digestion
leading to the termination of CCK release, and thus a marked
• Matching of nutrient delivery to digestive and absorptive capacity
reduction in the secretion of pancreatic enzymes.

FIGURE 51–2 Multiple effects of cholecystokinin (CCK) in


the duodenal cluster unit. CCK serves to coordinate nutrient ROLE OF SECRETIN
delivery to match intestinal capacity. (Reproduced with permission from
Barrett KE: Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, The other major regulator of pancreatic secretion is secretin,
Medical Pub. Division, 2006.) released from duodenal S cells. When the meal enters the
520 SECTION VIII GI Physiology

Ach
GRP
as
Protein cre
− Pan
Trypsin
LUMEN
Amino acids
− −
Fatty acids
CCK−RP Monitor peptide

FIGURE 51–3 Mechanisms responsible for controlling I cell


cholecystokinin (CCK) release from duodenal I cells. CCK-RP, EPITHELIUM
CCK-releasing peptide; ACh, acetylcholine; GRP, gastrin-
releasing peptide. Solid arrows represent stimulatory effects
while dashed arrows indicate inhibition. (Reproduced with CCK
permission from Barrett KE: Gastrointestinal Physiology. New York: Lange
BLOODSTREAM
Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)

small intestine, the volume of pancreatic secretions increases The threshold for secretin release appears to be a luminal pH of
rapidly, shifting from a low-volume, protein-rich fluid to a less than 4.5. The mechanism by which the S cells sense the
high-volume secretion. As the secretory rate increases, the pH change in luminal acidity is currently unclear. Nevertheless,
and bicarbonate concentration in the pancreatic juice increases, subjects who are unable to secrete gastric acid also fail to release
with a reciprocal decrease in the concentration of chloride secretin postprandially no matter what type of meal is given.
(Figure 51–4). These latter effects on the composition of the
pancreatic juice are mediated predominantly by the endocrine
mediator, secretin.
CELLULAR BASIS OF
Factors Causing Secretin Release PANCREATIC SECRETION
The S cells can be considered to act functionally as pH meters, ACINAR CELLS
sensing the acidity of the luminal contents (Figure 51–5). As
the pH decreases, due to the entry of gastric acid, secretin is Pancreatic acinar cells synthesize the proteinaceous compo-
released into the blood and binds to receptors on pancreatic nents of pancreatic juice and package them into zymogen gran-
duct cells. These cells, in turn, secrete bicarbonate, thus causing ules that are stored in the apical pole of the cell. The contents of
an increase in pH that will eventually shut off secretin release. these granules are discharged into the lumen of the acinus via a
process of compound exocytosis. The pancreatic enzymes are
then rapidly resynthesized and repackaged into granules, with
pH 7.2 pH 8.0

160
Na+
Concentration (mEq/L)

140 Stomach
HCO3
120
100 −
Acid HCO3−
80
60
Pancreatic
Duodenum
40 ducts
Cl
20
K+
Secretin
0 0.2 0.4 0.6 0.8 1.0 1.2 1.4
Flow rate (mL/min)
FIGURE 51–5 Function of secretin. Secretin is released from
the duodenum in response to reduced pH, and travels through the
FIGURE 51–4 Ionic composition of the pancreatic juice as a bloodstream to evoke bicarbonate secretion from the pancreatic
function of its flow rate. Note that the pancreatic juice becomes ducts (as well as from the biliary ducts and the duodenal mucosa,
alkaline at high rates of secretion. (Reproduced with permission from Barrett not shown), thereby neutralizing gastric acid in the duodenal lumen.
KE: Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical (Modified with permission from Barrett KE: Gastrointestinal Physiology. New York:
Pub. Division, 2006.) Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
CHAPTER 51 Pancreatic and Salivary Secretion 521

Enzymes washed
into duodenum
by ductular secretion

VIP
cAMP
Secretin

Phosphorylation
of structural
and regulatory
proteins FIGURE 51–6 Receptors of the pancreatic acinar
GRP cell and the regulation of secretion. The block arrow
Ca++ indicates that calcium-dependent signaling pathways
ACh Fusion of granules
play the most prominent role in enzyme secretion. VIP,
m3 vasoactive intestinal polypeptide; GRP, gastrin-releasing
with apical membrane
CCK and discharge of contents peptide; ACh, acetylcholine; CCK, cholecystokinin.
CCK−A (Reproduced with permission from Barrett KE: Gastrointestinal
Physiology. New York: Lange Medical Books/McGraw-Hill, Medical
BASOLATERAL APICAL Pub. Division, 2006.)

the process taking less than an hour, leaving the cell ready to tor (CFTR) chloride channel localized to the apical membrane
respond to the next meal. Evidence exists that the synthetic pro- of the cell. This channel allows outflow of chloride ions, which
cess is regulated by CCK and also by other hormones, such as can exchange for bicarbonate across an apical chloride/
insulin. In the long term, moreover, the rate of synthesis of spe- bicarbonate exchanger to provide for movement of bicarbon-
cific classes of enzymes can be regulated in response to changes ate ions into the duct lumen (Figure 51–7). Water and sodium
in the diet. For example, an increase in carbohydrates will even- ions follow paracellularly. CFTR itself may also be permeable
tually result in increased expression of amylase as a proportion to bicarbonate ions under certain circumstances. The bicar-
of the total pancreatic enzymes. Corresponding changes occur bonate required for the transport mechanism derives from
in the hydrolytic enzymes responsible for digestion of each of two sources. Some is generated intracellularly via the activity
the major classes of nutrients (carbohydrates, fats, and proteins) of carbonic anhydrase. Other bicarbonate ions are taken up
in response to increased or decreased ingestion. from the bloodstream via a basolateral sodium–bicarbonate
On their basolateral membranes, acinar cells express recep- cotransporter (NBC). Circulating bicarbonate is derived from
tors for CCK as well as for neural regulators of secretion, includ- the “alkaline tide” that is a byproduct of gastric acid secretion.
ing ACh, GRP, and vasoactive intestinal polypeptide (VIP) The bicarbonate transported by the duct cells, along with the
(Figure 51–6). All of the receptors are G protein–coupled fluid secretion that this transport mechanism drives, is impor-
receptors, and link to various downstream effectors such as tant to wash the proteinaceous components of the gastric juice
phospholipase C and adenylyl cyclase. In general, the phospho- into the intestinal lumen. Moreover, the alkaline nature of this
lipase C-dependent pathway, which is utilized by the receptors secretion is critically important in neutralizing gastric acid. The
for CCK, ACh, and GRP and results in increases in cytoplasmic pancreatic digestive enzymes are optimally active at neutral pH,
calcium, is the most quantitatively significant for acinar secre- as opposed to the acidic pH optimum of gastric pepsin.
tion, with cAMP-dependent signaling playing a subsidiary role.

PANCREATIC PATHOPHYSIOLOGY
DUCTULAR CELLS
The hydrolytic enzymes secreted by the pancreas are produced
In contrast to acinar cells that secrete their characteristic prod- in quantities that are vastly in excess of those needed to digest
ucts via exocytosis, the ductular cells are classical polarized a normal intake of nutrients. It has been calculated that pan-
epithelial cells that conduct vectorial ion transport. As seen creatic enzyme output needs to decrease below 10% of normal
elsewhere in the gastrointestinal tract, while exocytic secretion levels before effects on nutrient absorption are observed. Thus,
predominantly involves calcium-dependent signaling with pancreatic insufficiency is not common. However, under spe-
cAMP playing a modulatory role, the membrane transport cific conditions, it can occur, manifesting as maldigestion and
events that underlie ductular secretion are predominantly malabsorption. Fat absorption is usually the first affected by
driven by cAMP, with calcium playing the subsidiary role. alterations in pancreatic output of enzymes and bicarbonate,
The primary stimulus of duct cell secretion is secretin, which perhaps due to a relatively limited supply of lipase and because
binds to a basolateral receptor that links to adenylyl cyclase. pancreatic lipase is most sensitive to inactivation by low pH.
The primary target is protein kinase A, which phosphorylates Thus, steatorrhea, or fat in the stool, may be an early sign of
the cystic fibrosis transmembrane conductance regula- pancreatic dysfunction.
522 SECTION VIII GI Physiology

Duct lumen Basolateral


CO2 + H2O
C.A H+
− −
NHE-1
HCO3 HCO3 + H+ Na+
− −
Cl /HCO3 −
2HCO3
Exchanger NBC
Na+

3Na+
Na+, K+
FIGURE 51–7 Ion transport pathways
+ ATPase
present in pancreatic duct cells. C.A, carbonic 2K
Cl−
anhydrase; NHE-1, sodium/hydrogen
+ K+ channel
exchanger-1; NBC, sodium–bicarbonate
cotransporter. (Adapted with permission from Barrett cAMP
CFTR
KE: Gastrointestinal Physiology. New York: Lange Medical
Books/McGraw-Hill, Medical Pub. Division, 2006.)

BASIC PRINCIPLES (known as a bolus) that is suitable for swallowing. However, it


also performs additional roles. For example, the ability of
OF SALIVARY SECRETION saliva to solubilize molecules in the meal allows these to dif-
fuse to taste buds on the tongue, affecting appetite and food
We consider salivary secretion here because of analogies intake. Salivary secretion can also begin the digestive process
between this process and that of pancreatic secretion and plays important roles in host defense. It contains a variety
(Figure 51–8). Thus, a primary salivary secretion arises in of antibacterial substances that serve to protect the oral cavity
acini, and is modified as it flows through ducts. Thus, it is from microbial colonization. Saliva is also slightly alkaline.
instructive to compare and contrast these two processes, and This property is important in clearing any refluxed gastric acid
an understanding of one permits understanding of the other. from the esophagus, thus acting to prevent esophageal ero-
sions and injury. Finally, saliva aids in speech.
ROLE AND SIGNIFICANCE
SALIVARY SECRETORY PRODUCTS
Saliva plays a number of roles in gastrointestinal physiology
(Table 51–2). Its primary function is to lubricate ingested food, The protein components of saliva include digestive enzymes.
and to thereby permit formation of a smooth, rounded portion Saliva begins the digestion of carbohydrates via the action of

Smell
Taste
Sound
Sight

Higher
centers

Parotid ACh Otic Pressure


gland ganglion in mouth

Parasympathetics Salivatory
nucleus of
medulla
Submandibular ACh Submandibular −
gland ganglion
Sleep
Fatigue
FIGURE 51–8 Regulation of salivary Increased Fear
secretion by the parasympathetic nervous salivary
system. ACh, acetylcholine. (Adapted with secretion
via effects on
permission from Barrett KE: Gastrointestinal Physiology.
• Acinar secretion
New York: Lange Medical Books/McGraw-Hill, Medical Pub.
Division, 2006.)
• Vasodilation
CHAPTER 51 Pancreatic and Salivary Secretion 523

TABLE 51–2 Constituents of saliva and their salivary glands also receive extensive sympathetic and para-
functions. sympathetic innervation. Sympathetic efferents originate in
the salivatory center adjacent to the dorsal vagal complex,
Constituent Functions whereas parasympathetics come from the salivatory nuclei.
Water Facilitates taste and dissolution of
nutrients; aids in swallowing and
speech ACINAR CELLS
Bicarbonate Neutralizes refluxed gastric acid The salivary glands are heterogenous in their specific struc-
Mucins Lubrication ture and function. The acini of the parotid glands, which
drain into the upper part of the mouth via the parotid duct,
Amylase Starch digestion
consist entirely of serous cells, and thus are responsible for
Lysozyme, lactoferrin, IgA Innate and acquired immune providing the protein constituents of saliva. The sublingual
protection
gland, under the tongue, has predominantly mucous acini
Epidermal and nerve growth Assumed to contribute to mucosal responsible for secreting mucus and water, but also a scatter-
factors growth and protection ing of serous acini as well. The submandibular gland, below
the jaw, has a mixture of serous and mucous acini.

salivary amylase. This latter enzyme is not required for adequate DUCTULAR CELLS
digestion of starch in healthy adults, but may assume greater
importance in neonates, where there is a developmental delay in As the saliva makes its way out of the acini, it passes through a
the expression of pancreatic amylase. Salivary enzymes are ductular system. The intercalated ducts, linked directly to the
“backups” that are only required for digestion if other sources acini, serve predominantly to convey the saliva out of the aci-
are reduced. In patients with pancreatic insufficiency, for exam- nus and to prevent backflow. Cells of the striated intralobular
ple, salivary enzyme synthesis may be modestly increased. ducts, on the other hand, are polarized epithelial cells with
Salivary lysozyme and other antibacterial peptides limit specialized transport functions. The epithelial cells of the
colonization of the oral cavity by microbes. Lactoferrin intralobular ducts, moreover, have well-developed intercellu-
sequesters iron, thereby inhibiting the growth of bacteria that lar tight junctions that significantly limit the permeability of
require this substance. Saliva also contains significant quanti- this segment of the gland relative to the leaky acinus.
ties of secretory IgA, which contribute to immune defense.
In terms of the lubricating and solubilizing functions of
saliva, the most important constituents are mucins and water.
Mucin molecules are large glycoproteins with viscoelastic prop-
REGULATION OF
erties. Water is the main component of saliva and is secreted at SALIVARY SECRETION
very high rates. At maximal rates of secretion, the volumes pro-
duced by salivary glands can exceed 1 mL/min/g of gland tis- NEURAL REGULATION
sue, necessitating high rates of blood flow to supply this fluid.
In an adult, approximately 500 mL of saliva is produced daily. The salivary glands are unusual in the gastrointestinal system
Saliva also contains a variety of inorganic solutes, including in that their regulation appears to be exclusively mediated by
calcium and phosphate, that are important for tooth formation neurocrine pathways and not by gastrointestinal hormones, at
and maintenance. The primary secretion from the salivary acini least in the short term. The salivary glands are also unusual in
has an ionic composition that is comparable to plasma. However, that they are positively regulated by both the parasympathetic
as the secretion moves along the ducts, the composition is mod- and sympathetic branches of the autonomic nervous system.
ified by active transport processes as will be described later. However, quantitatively, the predominant regulation of secre-
tory rate and composition is via parasympathetic pathways
with sympathetic efferents playing only a modifying role.
SALIVARY GLAND ANATOMY
Like the pancreas, the salivary glands are made up of grape-
Parasympathetic and
like clusters of acini that drain into a system of intercalated Sympathetic Regulation
and intralobular (striated) ducts, and eventually into interlob- The parasympathetic nervous system initiates salivary secre-
ular ducts that drain into the oral cavity. The individual acini tion and sustains secretion at high rates. The nerves originate
and associated ducts are also surrounded by a sheath of myo- in the salivatory nucleus of the medulla, and receive input
fibroblasts, which are contractile cells that may be important from higher centers interpret the need for changes in salivary
in providing a hydrostatic force that expels saliva from the secretion under both physiological or pathophysiological cir-
gland, thereby contributing to high rates of secretion. The cumstances Conditioned reflexes, such as smell and taste, as
524 SECTION VIII GI Physiology

well as pressure reflexes transmitted from the oral cavity mark- 160
edly stimulate parasympathetic outflow, whereas fatigue, sleep,
140
fear, and dehydration suppress neurotransmission. Nausea
also strongly stimulates salivation, presumably to protect the

Concentration (mEq/L)
120
oral cavity and esophagus from the injurious effects of vom-
ited gastric acid and other intestinal contents. Parasympathetic 100
Na+
input to the salivary glands is mediated by ACh acting at mus-
80
carinic receptors. In addition to effects on the acinar cells and
HCO3−
ducts of the glands, parasympathetic innervation causes dila- 60
tion of the blood vessels supplying the gland, thereby provid- Cl−
ing both the fluid and metabolic requirements needed to 40
sustain high rates of secretion. 20 K+
Efferents of the sympathetic nervous system passing
through the superior cervical ganglion also terminate at the
0 1 2 3 4
salivary glands. These nerves are not thought to be capable of
Flow of saliva (mL/min)
initiating or sustaining secretion independently, but can
potentiate the effects of parasympathetic regulation via the FIGURE 51–9 Ionic composition of saliva as a function of
release of norepinephrine and beta-adrenergic receptors. its flow rate. Note that saliva becomes less hypotonic as flow rates
increase. (Reproduced with permission from Barrett KE: Gastrointestinal Physiology.
New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)

CELLULAR BASIS OF
SALIVARY SECRETION keep pace with the active reabsorption of sodium and chloride,
and thus saliva becomes hypotonic. Moreover, due to secretion
ACINAR CELLS of bicarbonate into the lumen without an accompanying pro-
ton, the pH of saliva increases progressively to approximately
Acinar cells release their protein and mucus contents via a 8 as the saliva enters the mouth.
process of exocytosis, analogous to enzyme release from the At very high rates of salivary secretion, the concentrations
pancreatic acini. These responses involve mobilization of of sodium and potassium more closely resemble those in
intracellular calcium downstream of the muscarinic receptor plasma. The concentration of chloride also increases as the
for ACh. Acinar cells also actively secrete chloride, bicarbon- flow rate of saliva increases. These changes in composition are
ate, and potassium ions into the primary salivary secretion. due to the fact that the residence time of the saliva in the ducts
Because the acini are relatively leaky, sodium and water follow is too short for the cells to be able to modify salivary composi-
paracellularly via the tight junctions and the initial secretion tion significantly.
has an ionic composition comparable to plasma.

DUCTULAR CELLS CLINICAL CORRELATION


A Caucasian couple brings their two-month old infant to a
As we learned for the pancreas, the duct cells in the salivary
pediatrician for a well-baby check. It is noted that the baby is
glands modify the composition of the saliva as it passes by
underweight, and his mother notes frequent coughing, some
them. The ionic composition of saliva changes as its flow rate
episodes of vomiting after feeding, and a salty taste to the
increases (Figure 51–9). At low rates of secretion, saliva is
child’s skin. After administering the muscarinic agonist,
hypotonic with respect to plasma and has higher concentra-
pilocarpine, to stimulate secretion by the sweat glands, chlo-
tions of potassium than sodium, the opposite of the situation
ride concentrations in the sweat are found to be markedly
in plasma. The chloride concentration is also much lower than
increased. Genetic testing later reveals that both parents are
that found in plasma. These changes in ionic content are
heterozygous for a mutation that results in the deletion of the
brought about by active transport events taking place in the
phenylalanine residue at position 508 of CFTR, and a diag-
duct cells (Figure 51–10). Sodium and chloride are reabsorbed
nosis of cystic fibrosis is made. The child’s condition improves
across the apical membrane, in exchange for protons and bicar-
with supplementation of the diet with an oral preparation of
bonate, respectively. Protons are recycled to transfer potassium
pancreatic enzymes as well as physical therapy designed to
into the duct lumen. At the basolateral membrane, the driving
dislodge thickened respiratory secretions, but recurrent lung
force for sodium uptake is provided by a sodium–potassium
infections often require antibiotic therapy.
ATPase, and a potassium channel supplies potassium for secre-
Respiratory complications due to failure to clear the thick-
tion into the saliva. Because the ductular epithelium has a low
ened mucus from the airways, are usually the most significant
passive permeability, water cannot flow across the tight junc-
cause of morbidity and mortality in cystic fibrosis. Pancreatic
tions fast enough at moderate rates of salivary secretion to
CHAPTER 51 Pancreatic and Salivary Secretion 525

DUCT LUMEN BASOLATERAL

Cl− channel
Cl−/HCO3− HCO3−
Exchanger Cl−

Cl−

3Na+
Na+
NHE
2K+
H+ Na+, K+
ATPase
H+
K+
H+/K+ K+
Exchanger K+ channel
FIGURE 51–10 Ion transport pathways
in salivary duct epithelial cells. NHE, sodium/
H2O hydrogen exchanger (Reproduced with permission
from Barrett KE: Gastrointestinal Physiology. New York: Lange
Tight junctions restrict osmotic reabsorption of water Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)

■ The pancreas has several lines of defense to protect against


function can also be altered in the genetic disorder of cystic autodigestion. When these lines fail, pancreatitis results.
fibrosis, where mutations lead to abnormal function of the ■ Salivary secretion shares several parallels with pancreatic
CFTR chloride channel. Indeed, the disease was named for secretion.
characteristic cystic histological abnormalities observed in the ■ Salivary secretion is predominantly mediated by parasympa-
pancreas in affected patients. Although pancreatic enzyme thetic input arising from higher brain centers. Hormonal
synthesis and secretion are normal in patients with cystic regulation is much less important.
fibrosis, the relative inability of the ducts to secrete bicarbon-
ate and water means that the enzymes cannot be flushed prop-
erly from the organ, and limited quantities reach the intestinal
lumen. Moreover, the enzymes that do reach the lumen are
STUDY QUESTIONS
inactive because of the failure to neutralize gastric acid. These 1. A 4-year-old boy is brought to the pediatrician for an evaluation
findings underscore the role of the duct cells in normal pan- because of failure to thrive and frequent diarrhea characterized
creatic function. In fact, in patients with severe CFTR muta- by pale, bulky, foul-smelling stools. Sweat chloride concentrations
tions causing a marked reduction in channel function, the are measured and found to be elevated. Diminished secretion of
which pancreatic product is most likely to be the primary cause of
exocrine pancreas may be largely destroyed during fetal life,
the patient’s apparent fat malabsorption?
due to the action of retained proteolytic enzymes that become
A) lipase
inappropriately activated and damage the tissue. Such patients
B) procolipase
are said to have pancreatic insufficiency and are treated with
C) monitor peptide
oral supplements of pancreatic enzymes, along with antacids, D) cholecystokinin
to allow for adequate nutrition. Patients with milder muta- E) bicarbonate
tions may retain some degree of pancreatic function, at least 2. In an experiment, recordings are made of electrical activity in
early in life, but are then at greater risk for the development of afferent nerves originating in the small intestinal mucosa during
inflammation of the pancreas (pancreatitis) with aging. sequential luminal perfusion with saline, a solution of hydrolyzed
protein, and a solution of intact protein. Rates of neuronal firing
were shown to increase markedly during the period when intact
protein was infused compared with the other two. Firing in these
CHAPTER SUMMARY nerves was most likely stimulated by an increase in the mucosal
■ Pancreatic secretion provides for digestion of meals. concentration of which of the following?
■ Pancreatic acini supply enzymes, whereas ducts supply fluid; A) gastrin
major regulators of each cell type are CCK and secretin, B) secretin
respectively. C) somatostatin
■ Pancreatic secretion is initiated during the cephalic phase, D) ACh
but is most prominent when the meal is in the duodenum. E) cholecystokinin
526 SECTION VIII GI Physiology

3. A 50-year-old man with a history of alcohol abuse presents at the 5. A 50-year-old female patient who has suffered for several years
emergency room with severe, colicky abdominal pain and a fever. from severe dryness of her eyes due to inadequate tear
A blood test reveals increased levels of serum amylase and an production is referred to a gastroenterologist for evaluation of
endoscopic imaging procedure reveals a narrowed pancreatic chronic heartburn. Endoscopic examination reveals erosions and
duct. Pain in this patient is likely predominantly ascribable to scarring of the distal esophagus just above the lower esophageal
premature activation of pancreatic enzymes capable of digesting sphincter. Reduced production of which of the following salivary
which of the following nutrients? components most likely contributed to the tissue injury?
A) triglyceride A) lactoferrin
B) phospholipids B) mucus
C) protein C) IgA
D) starch D) bicarbonate
E) nucleic acids E) amylase
4. A researcher conducts a study of the regulation of salivary
secretion in a group of normal volunteers under various
conditions. Which of the following conditions was associated
with the lowest rates of secretion?
A) chewing gum
B) undergoing a mock dental exam
C) sleep
D) exposure to a nauseating odor
E) resting control conditions
52
C H A P T E R

Water and Electrolyte


Absorption and Secretion
Kim E. Barrett

O B J E C T I V E S

■ Understand the physiological significance of the regulation of luminal water


content and daily fluid balance.
■ Describe the functional anatomy of the intestinal epithelium that permits it
to function as a regulator of fluid movement.
■ Define the pathways via which electrolytes can be transferred across
epithelial barriers.
■ Describe how a limited collection of membrane transport pathways is
arranged to assemble transepithelial transport mechanisms.
■ Identify the major electrolyte transport pathways of the small and large
intestines and their intracellular mechanisms of regulation.
■ Identify how subepithelial elements and other regulatory systems impact on
epithelial transport function.
■ Understand how transport function is integrated with intestinal motility.
■ Define major pathogenic alterations in intestinal electrolyte transport and
their consequences.

BASIC PRINCIPLES OF this is exceeded that excessive water loss to the stool occurs,
seen clinically as diarrhea.
INTESTINAL FLUID TRANSPORT
ROLE AND SIGNIFICANCE ELECTROLYTES INVOLVED
Control of the amount of fluid in the intestinal lumen is criti- Epithelial cells express several specialized properties that allow
cal for normal intestinal function. This fluid environment per- them to control fluid movement. Most important are the intra-
mits contact of digestive enzymes with food particles, and in cellular tight junctions that restrict the passive flow of solutes
turn the diffusion of digested nutrients to their eventual site of and backflow of these once either secreted or absorbed. Water is
absorption. The fluidity of the intestinal contents also provides transported passively across the intestinal epithelium in response
for their transit along the length of the gastrointestinal tract to osmotic gradients established by the active transport of elec-
without damage to the lining epithelium. trolytes and other solutes. In common with those in other trans-
Large volumes of fluid are handled by the intestine during porting epithelia, such as in the nephron, active electrolyte
the digestion and absorption of meals. Most of this fluid is transport pathways share a number of defining characteristics
supplied by the intestine and the organs that drain into it. The (Table 52–1). These transport pathways move a solute across a
daily fluid load approximates 9 L in normal adults (Figure 52–1). single membrane in a polarized epithelial cell. Transepithelial
In health, this large volume is later reclaimed by the intestine transport mechanisms, in turn, move solutes across the entire
to avoid dehydration. Both the small and large intestines also epithelium. Furthermore, both absorption and secretion can
have a large reserve capacity for absorption, and it is only when occur simultaneously in any given segment of the intestinal

527

Ch52_527-534.indd 527 11/26/10 10:34:53 AM


528 SECTION VIII GI Physiology

Oral intake TABLE 52–2 Small intestinal ion transport


2000 mL
mechanisms.
Bicarbonate secretion
Saliva Sodium-coupled nutrient absorption
1500 mL
Proton-coupled nutrient absorption
Gastric
juice Electroneutral NaCl absorption
2500 mL
Chloride secretion
Bile Pancreatic
juice Sodium-coupled bile acid absorption
500 mL
1500 mL Calcium and iron absorptiona
a
Intestinal Not major determinants of fluid transport.
secretion
1000 mL
ANATOMICAL CONSIDERATIONS
AMPLIFICATION OF
Small INTESTINAL SURFACE AREA
intestinal
absorption The capacity of the intestine for large volumes of water trans-
7000 mL port is related to its massively amplified surface area. In fact, the
(78%) Colonic surface area of the adult small intestine alone exceeds that of a
absorption
1900 mL
doubles tennis court. The intestine is not a simple cylinder, but
(21%) instead is amplified first by folds in the mucosa, then by the
Stool presence of crypts and villi, and finally by the presence of
100 mL (1%) microvilli on the apical poles of individual epithelial cells. Over-
FIGURE 52–1 Daily water balance in the healthy adult human all, the surface area is increased 600-fold by these physical struc-
gastrointestinal tract. The amount of oral intake varies among tures. This amplification of the surface area not only allows for
individuals depending on the types of meals taken. Note that even in handling of the large volumes of fluids required for normal
health there is a significant secretory flux of fluid from the intestine. functioning of the intestine, but also provides the reserve capac-
(Modified with permission from Barrett KE. and Dharmsathaphorn K: Transport of ity for fluid absorption in disease. However, the surface amplifi-
water and electrolytes in the gastrointestinal tract: physiological mechanisms, cation, particularly in the crypts, also carries a liability in that
regulation and methods for study. In: Maxwell and Kleeman’s Clinical Disorders of Fluid there is a corresponding reserve capacity for intestinal fluid
and Electrolyte Metabolism, 5th ed. Narins RG (editor). McGraw-Hill, New York, 1994.) secretion. If such secretion is extensive and prolonged, it can
rapidly lead to serious dehydration if left untreated.
tract. This is primarily because most of the cells of the villi (or
surface cells in the colon) are absorptive, whereas crypt epithe-
lial cells are secretory. In general, absorptive mechanisms for INNERVATION AND
fluid center on the active movement of sodium, whereas secre- REGULATORY CELLS
tory fluxes of fluid in the intestine are driven mostly by the elec-
trogenic movement of chloride ions, although bicarbonate As we learned in earlier chapters, the intestinal epithelium
secretion may assume significance in particular segments. rests on a lamina propria that is a rich source of potential regu-
Moreover, the transport mechanisms that are expressed in the latory factors. In addition to endocrine regulators, epithelial
small intestine and colon differ, due to the relative paucity of electrolyte transport is controlled by paracrine mediators sup-
nutrients in the latter segment (Tables 52–2 and 52–3). plied by local enteroendocrine cells, immune mediators, and

TABLE 52–1 Characteristics of active membrane TABLE 52–3 Colonic ion transport mechanisms.
transport pathways.
Electrogenic sodium absorption
Mediate uphill transport against an electrochemical gradient
Electroneutral NaCl absorption
Effective at low luminal concentrations
Short-chain fatty acid absorption
Demonstrate saturable kinetics
Chloride secretion
Require cellular energy
Potassium absorption/secretiona
Demonstrate high ionic specificity a
Not a major determinant of fluid transport.
CHAPTER 52 Water and Electrolyte Absorption and Secretion 529

neurocrines released from secretomotor efferent nerves origi- the local passage of a food bolus. In turn, this releases
nating predominantly in the submucosal plexus of the enteric 5-hydroxytryptamine from local enterochromaffin cells, fol-
nervous system. The epithelial cells themselves may also pro- lowed by activation of cholinergic efferents that stimulate a cor-
duce autocrine factors that regulate their transport function. responding burst of chloride, and thus fluid, secretion. This
The regulatory systems that mediate changes in epithelial reflex may be important in protecting the epithelium from
transport do not act in isolation. Rather, there is significant physical damage by the passing meal components.
crosstalk between the various modes of communication. For
example, some immunologic mediators may have both direct
effects on epithelial cells and others that are mediated second-
Humoral Control
arily via the activation of enteric nerves. Crosstalk between the Although there appears to be a relatively limited role for clas-
various regulatory systems also provides for coordinated regu- sical endocrine hormones in mediating changes in intestinal
lation of transport and motility functions. transport function, at least in the short term, other soluble
effectors have clear effects and are largely derived from para-
crine or immune sources. For example, local production of
prostaglandins, likely predominantly by subepithelial myofi-
REGULATION OF WATER AND broblasts, plays an important role in stimulating the secretion
of both chloride and bicarbonate. Similarly, histamine,
ELECTROLYTE TRANSPORT released by mast cells residing in the lamina propria, has been
REGULATORY STRATA shown to be an effective chloride secretagogue, although its
effect is transient. Indeed, immune effector cells that release
Much of our knowledge of the control of intestinal fluid move- substances capable of regulating the epithelium can be consid-
ment comes from studies of the factors that regulate fluid secre- ered specialized “sensory” cells that alter transport function in
tion, which is driven primarily by the secretion of chloride ions. response to specific conditions pertaining in the lumen, such
Fluid absorption, particularly in the postprandial period, is more as the presence of food substances to which an individual is
of a passive response that is driven by the presence of nutrients, allergic. These and other putative humoral regulators of intes-
and not highly subject to regulation by intracellular and intercel- tinal secretion and/or absorption are listed in Table 52–4.
lular mechanisms. On the other hand, in the absence of nutri- Humoral regulators of intestinal transport typically bind to
ents, the intestine absorbs fluid to balance secretory pathways via receptors localized to the basolateral pole of intestinal epithe-
the absorption of sodium and chloride ions. These nutrient- lial cells. It should be emphasized, however, that such effectors
independent pathways for fluid absorption are subject to both can alter epithelial function not only via such direct binding,
intracellular and intercellular regulation. In general, regulatory but also via the secondary activation of other subepithelial ele-
pathways that stimulate chloride secretion inhibit sodium chlo- ments. In this way, intestinal secretory and/or absorptive func-
ride absorption, and vice versa. However, this does not apply to tion can be better integrated with other physiological functions
nutrient-coupled absorption, which can continue unopposed of the gut such as motility and blood flow. In turn, agonists
even under circumstances that lead to the stimulation of chloride that alter these latter functions may have indirect effects on
secretion. This last point underlies the efficacy of so-called oral intestinal secretion and absorption. The net rate of movement
rehydration solutions, which are used to treat the dehydration of any substance across the intestinal epithelium will reflect
accompanying severe diarrheal diseases, such as cholera, when not only the “east–west” vector of absorption/secretion, but
intravenous fluids are not available. also the “north–south” vector of movement along the length of
the gastrointestinal tract (Figure 52–2). Thus, if motility is
increased, hastening the transit of substances along the intes-
Short and Long Reflexes tine, there will be less time for absorption to take place (or,
Intestinal epithelial transport is regulated by neurotransmitters conversely, for active secretion to add to luminal fluid loads).
originating from nerve endings of the enteric nervous system. If transit is slowed, absorption can catch up with the presented
The most potent effectors in this regard include acetylcholine
(ACh) and vasoactive intestinal polypeptide (VIP), both of
which can directly stimulate epithelial cells to secrete chloride. TABLE 52–4 Major endogenous regulators of
Some neural input to the control of intestinal transport almost intestinal ion transport.
certainly originates in the central nervous system, and this input
is then interpreted and integrated with local information to Cyclic Nucleotide-dependent Calcium-dependent
impinge ultimately on the activity of secretomotor neurons. In a
Vasoactive intestinal polypeptide (VIP) Acetylcholine (ACh)
similar fashion, vagovagal reflexes likely match intestinal trans-
port function to conditions that result from the physical state of Prostaglandins Histamine
luminal contents, such as via the activation of stretch receptors. Guanylin (cGMP) 5-Hydroxytryptamine
In addition to these “long” reflexes, however, “short” or local
5′AMP/adenosine Bile acids
reflexes can be initiated by stroking the mucosa, which models
530 SECTION VIII GI Physiology

pendent of the presence of a meal, notably as stimulated by


Oral
input from the central nervous system at times of threat or
stress. In either case, while both secretion and absorption
occur simultaneously, in health, the absorptive vector
predominates overall, and most of the fluid used for digestion
and absorption is recycled (Figure 52–3).
Neurotransmitters released from enteric secretomotor neu-
rons, as well as paracrine effectors from local enteroendocrine
cells or other subepithelial elements, alter the functional
EAST-WEST capacity of transporting epithelial cells to conduct transport
VECTOR across their apical and basolateral membranes. Acutely, sec-
Influenced ond messenger pathways evoked by neurohumoral regulators
by surface alter the activation status of the transporters and/or redistrib-
area
ute transporters within the epithelial cells themselves. Deliv-
ery of additional, preformed transporters to the membrane
will increase transport capacity, whereas endocytic retrieval
will reduce it.

NaCl, nutrients
Anal NORTH-SOUTH VECTOR
Influenced by
motility → transit time

FIGURE 52–2 Integration of influences on fluid movement


in the intestine. Overall fluid fluxes depend on the surface area Villus
Health
available for ion transport and residence time in the lumen. (Modified
with permission from Barrett KE: Gastrointestinal Physiology. New York: Lange
Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)

fluid volume. This last principle underlies the efficacy of sev-


eral antidiarrheal medications, and particularly opiate drugs Crypt
such as loperamide.

NaCl
Luminal Regulators Cl−
The epithelium is also positioned to respond to substances
present in the intestinal lumen, and expresses a number of
apical receptors for such agents. Guanylin is a peptide regu-
lator of epithelial chloride secretion that is synthesized by
enteroendocrine cells and released into the lumen. The phys-
iological role of this substance may be to coordinate salt han-
dling by the small and large intestines with that of the Villus
Diarrheal
kidneys. Bile acids, which are synthesized by the liver to aid disease
in fat digestion and absorption, are also apical stimuli of
chloride secretion in the colon. Under normal circumstances,
however, bile acids are reabsorbed in the terminal ileum
when they are no longer needed to solubilize the products of Crypt
Nutrient absorption
fat digestion, and so bile acid–induced diarrhea is only largely normal
observed in the setting of disease.
Cl−
ACUTE REGULATION FIGURE 52–3 Balance between absorption and secretion in
health and in secretory diarrheal disease. Note that small intestinal
Acute regulation of intestinal fluid and electrolyte transport nutrient absorption is usually largely normal in the setting of secretory
occurs to match needs for luminal fluidity on a minute-to- diarrhea. (Modified with permission from Barrett KE: Gastrointestinal Physiology. New
minute basis. Altered intestinal transport can also occur inde- York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
CHAPTER 52 Water and Electrolyte Absorption and Secretion 531

CHRONIC ADAPTATION uptake of sodium and glucose (or galactose) is a coupled pro-
cess that occurs via a cotransporter, SGLT-1. By tying the
The bowel is also capable of altering its ability to conduct water movement of glucose to that of sodium, glucose can be moved
and electrolyte transport in a chronic fashion (over days to against its concentration gradient, even when luminal concen-
weeks), in order to adapt to changes in whole body electrolyte trations of this nutrient are low. Glucose thus absorbed is then
status. utilized by the enterocyte, or is transported to the bloodstream
The hormone, aldosterone, is an important regulator of via a facilitated diffusion pathway (GLUT-2). Anions (largely
sodium transport in the intestine in addition to similar roles in chloride) and water follow passively via the tight junctions.
the renal system (see Chapters 45 and 65). When the diet is low Sodium-coupled transport also allows for the active uptake of
in salt, aldosterone is released and increases the expression of conjugated bile acids, although in this case the transport
transporters required for sodium absorption in the colon. The mechanism is restricted to the terminal ileum. In the colon,
net effect is active sodium retention by the colon. Analogous where luminal glucose is largely absent, a similar mechanism
processes allow for increased or decreased intestinal retention allows for the electrogenic uptake of sodium by replacing
of other important electrolytes. For example, a decrease in SGLT-1 with the epithelial sodium channel (ENaC) (Figure
plasma calcium increases levels of 1,25-dihydroxyvitamin D, 52–5).
which stimulates the expression of proteins needed for calcium Short peptides that are products of digestion of dietary pro-
absorption in the small intestine. Conversely, levels of trans- teins are absorbed via an apical transporter known as PepT1,
porters involved in intestinal iron absorption are decreased in coupled to proton uptake. PepT1 is a remarkable transporter
patients suffering from the disease of hemochromatosis, which in that it can accommodate a wide range of substrates, includ-
is associated with overloading of the body’s stores of iron. ing dipeptides, tripeptides, and perhaps even tetrapeptides
made up of various combinations of the 20 naturally occurring
amino acids. As we will see in Chapter 58, some amino acids,
CELLULAR BASIS OF TRANSPORT including essential ones that cannot be synthesized by the
body, are only efficiently absorbed in peptide form due to a
ABSORPTIVE MECHANISMS relative lack of relevant amino acid transporters.
Between meals, when nutrients are not available in the
Absorptive mechanisms expressed in the small intestine and lumen, fluid absorption can still continue via a mechanism
colon are summarized in Tables 52–2 and 52–3. Throughout that involves the coupled absorption of both sodium and chlo-
the length of the small intestine, sodium is taken up in con- ride (Figure 52–6). Coupled ion exchangers on the apical
junction with a variety of nutrients as exemplified by glucose- membrane carry sodium and chloride into the cell in exchange
coupled sodium absorption (Figure 52–4). This, and related for protons and bicarbonate ions, respectively, and both
transport mechanisms such as those driven by specific amino exchange processes require the activity of the other. Notably,
acids, relies on the low intracellular sodium concentration the NHE3 sodium–hydrogen exchanger isoform that partici-
established by the active, basolateral Na,K-ATPase. Apical pates in this transport mechanism is inhibited by cAMP;
therefore, the overall transport process can likewise be inhib-
ited by this second messenger.

Glucose
GLUT-2
SGLT-1
Glucose
2Na+ 2K+ EN
3Na+ aC

Na+ K+
Na+,K+– 2K+
ATPase 3Na+

Na+,K+ -
ATPase
Cl–

Cl−
FIGURE 52–4 Sodium-coupled nutrient absorption
exemplified by the uptake of glucose from the intestinal lumen.
(Used with permission from Montrose M.H. et al: Secretion and absorption: small FIGURE 52–5 Electrogenic sodium absorption in the colon.
intestine and colon. In: Textbook of Gastroenterology, 4th ed. Yamada T, Alpers DH, Sodium enters the epithelial cells via epithelial sodium channels
Kaplowitz N, Laine L, Owyang C, Powell DW (editors). Philadelphia, PA: Lippincott (ENaC). (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks
Williams and Wilkins; 2003.) H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
532 SECTION VIII GI Physiology

Na+ _
2K+ 3Na+ 2CI
NHE-3?
Na+ TR
H+ NHE-2? Na+,K+ - CF K+ NKCC1
ATPase
Cl− Na+
2K+ 3Na+
+ +
Na , K -
K+
Cl−
KCC1 ATPase
CLD ?
HCO3–
Cl−
K+

FIGURE 52–6 Electroneutral NaCl absorption in the small


intestine and colon. NaCl enters across the apical membrane via the FIGURE 52–7 Chloride secretion in the small intestine and
coupled activity of a sodium/hydrogen exchanger (NHE) and a colon. Chloride uptake occurs via the sodium/potassium/2 chloride
chloride/bicarbonate exchanger (CLD). The route of basolateral cotransporter, NKCC1. Chloride exit is via the cystic fibrosis
chloride exit remains speculative. (Reproduced with permission from Barrett transmembrane conductance regulator (CFTR) chloride channel, as
KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. well as perhaps via additional chloride channels (not shown).
McGraw-Hill Medical, 2009.) (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H:
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

The small intestine also absorbs iron and calcium in their ionic
forms, although the small quantities of these ions that are han- way for chloride exit is the CFTR channel; there is some evi-
dled do not contribute in a major way to fluid handling. dence also to suggest an accessory role played by additional
Calcium absorption is possible along the length of the small chloride channels. The net effect is the electrogenic movement
intestine depending on whole body demands, whereas the major- of chloride from the bloodstream to the lumen; water and
ity of iron absorption occurs in the proximal small intestine due sodium follow passively via the tight junctions to maintain neu-
to specific expression of the membrane transporters required to trality. In response to agonists such as VIP or prostaglandins,
facilitate iron movement. Dietary iron is also handled differently cAMP levels are increased in the crypt cell cytosol, which in
depending on whether it is in the form of heme (derived from turn result in activation of PKA. This enzyme can phosphory-
meat), from which it is released by lysosomal enzymes following late and thereby open the CFTR chloride channel, resulting in
uptake of the intact heme molecule, or in its ionized form. an initial burst of chloride secretion (Figure 52-8). cAMP-
The colon also conducts an additional absorptive transport dependent agonists of this process are additionally notable for
process that reclaims an important byproduct of waste metab- the fact that they evoke sustained secretory responses. On the
olism. Dietary fiber and other complex carbohydrates that other hand, agonists such as ACh, histamine, and likely bile
cannot be digested by mammalian enzymes are degraded in acids evoke chloride secretion by increasing cytosolic calcium
the colon by the resident bacterial flora, and generate short- concentrations. In this case, the primary locus for regulation is
chain fatty acids such as acetate, propionate, and butyrate that a basolateral potassium channel. As potassium leaves the cell,
are taken up by colonic epithelial cells. the driving force for chloride exit increases, allowing chloride to
flow across the apical membrane via the small proportion of
CFTR channels that may be open at any given time. The cal-
SECRETORY MECHANISMS cium-dependent chloride secretory response is smaller and
more transient than that evoked by cAMP elevation. This may
Secretory mechanisms in the gastrointestinal tract center imply a physiological need to be able to call upon both brief and
around the active transport of chloride ions. The mechanism sustained secretory responses under specific circumstances
for secretion of chloride itself is depicted in Figure 52–7. Chlo- during the digestion and absorption of a meal. Moreover, when
ride is taken up across the basolateral membrane of crypt epi- crypt epithelial cells are simultaneously exposed to a combina-
thelial cells via a sodium/potassium/2 chloride cotransporter tion of agonists acting via cyclic nucleotides and calcium, a syn-
called NKCC1. This transporter conducts secondary active ergistic enhancement of secretion results.
uptake of chloride into the cell cytosol by taking advantage of The intestine is also capable of active bicarbonate secretion
the favorable gradient for sodium movement established by the (Figure 52–9). This mechanism is particularly prominent in
basolateral Na,K-ATPase. Potassium that is cotransported is the proximal duodenum, which must defend itself from the
recycled across the basolateral membrane via channels that may potentially injurious effects of the acidic gastric juice, and is
be activated by either cAMP or calcium. Chloride thus accumu- analogous to pancreatic secretion of this ion as discussed
lates in the cytosol, ready to exit the cell across the apical mem- in the previous chapter. Like chloride secretion, the overall
brane when chloride channels are opened in response to second process of bicarbonate secretion can be stimulated by intracel-
messenger pathways. The quantitatively most significant path- lular increases in cAMP, cGMP, or calcium, with prostaglan-
CHAPTER 52 Water and Electrolyte Absorption and Secretion 533

Cl−

APICAL
CFTR 2K+ 3Na+

CFT
R Na+,K+–
Phosphorylation ATPase
P Cl– Cl–
channel
opening
AE1?
CLD? Na+
R NHE-1
C
C HCO3– + H+ H+
HCO3–
Protein CA
CO2 + H2O
kinase A
cAMP 3Na+
2K+
Na+,K+–
Cytoskeleton R? ATPase
CFT
Vesicle
trafficking HCO3–

Gs Na+
NHE-1
A.C. HCO3– + H+ H+
NKCC1 CA
CO2 + H2O
K+ Na+
BASOLATERAL
2Cl
VIP
PGE2
FIGURE 52–9 Bicarbonate secretion in the duodenum. The
FIGURE 52–8 Regulation of chloride secretion by cAMP-
two models depicted differ in the pathway for bicarbonate exit across
dependent agonists such as vasoactive intestinal polypeptide (VIP)
the apical membrane. Both models are likely to be important,
and prostaglandins. These agonists activate adenylyl cyclase (A.C.) via a
although the anion exchanger involved in the upper mechanism has
stimulatory G protein (Gs), leading to an increase in intracellular cAMP.
not been conclusively identified. CA, carbonic anhydrase; AE1/CLD,
This in turn activates the cAMP-dependent protein kinase (protein kinase
anion exchangers; NHE-1, sodium/hydrogen exchanger-1. (Used with
A), causing dissociation of its catalytic (C) subunits from the regulatory
permission from Montrose et al: Secretion and absorption: small intestine and colon.
(R) subunits. The catalytic subunits are thereby freed to phosphorylate
In: Textbook of Gastroenterology, 4th ed. Yamada T, Alpers DH, Kaplowitz N, Laine L,
CFTR leading to channel opening, and to stimulate the insertion of
Owyang C, Powell DW (editors). Philadelphia: Lippincott, Williams and Wilkins; 2003.)
additional NKCC1 cotransporter molecules into the basolateral
membrane. (Reproduced with permission from Barrett KE: Gastrointestinal Physiology.
New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)

dins, guanylin, and ACh representing physiologically important


receive oral solutions of glucose and sodium chloride in
secretagogues that utilize each respective second messenger.
volumes equivalent to their fecal losses recover from their
The primary physiological stimulus for duodenal bicarbon-
acute diarrheal illness and survive. Later microbial analy-
ate secretion appears to be the presence of an acidic pH in
ses reveal that stool samples contain large numbers of a
the lumen.
bacterial pathogen.
Diarrhea can result when intestinal chloride secretion
is stimulated excessively, and the resulting luminal fluid
CLINICAL CORRELATION load exceeds the absorptive capacity of the small and large
intestines (Figure 52–3). The prototypic disease state in
Following a tsunami disaster in which the supply of clean which this occurs is cholera, in which Vibrio cholerae bac-
drinking water is disrupted, hundreds of adults and chil- teria in the intestinal lumen secrete a toxin. The active
dren are brought to emergency medical personnel suffer- subunit of this toxin translocates to the basolateral mem-
ing from profuse, watery diarrhea. The children, in brane of intestinal epithelial cells where it irreversibly
particular, show signs of severe dehydration. Because of activates the stimulatory Gs G-protein, resulting in a mas-
the prevailing conditions and a lack of trained personnel, sive accumulation of cAMP and stimulation of down-
it is not possible to resuscitate patients with intravenous stream signaling pathways. This produces uncontrolled
fluids. Instead, patients are placed on cots that allow fecal and sustained chloride secretion, inhibition of electroneu-
fluid losses to be measured over time. Many patients who tral NaCl absorption, and an outpouring of fluid into the
534 SECTION VIII GI Physiology

lumen. Stool volumes of up to 20 L per day are not uncom-


STUDY QUESTIONS
mon in this disorder, which can rapidly lead to death due 1. Individuals housed in a camp in Southeast Asia following a
to the complications of dehydration if left untreated. natural disaster develop widespread watery diarrhea. Increased
Active intestinal secretion may also underlie diarrhea activity of which of the following transport proteins might be
caused by a number of other enteric pathogens, including exploited therapeutically to reduce fluid losses?
rotavirus and Salmonella. Clostridium difficile is a patho- A) SGLT-1
gen that is often acquired in hospital settings, particularly B) CFTR
C) NHE3
in patients whose normal enteric flora has been disrupted
D) Na,K-ATPase
by the administration of antibiotics. It secretes toxins that E) NKCC1
provoke chloride secretion via calcium-dependent path-
2. A 50-year-old man on a business trip to a developing county
ways in addition to damaging the barrier function of the develops severe diarrhea and begins taking the opiate drug
epithelium. Finally, the endogenous peptide regulator of Imodium (loperamide) in an attempt to lessen his symptoms.
chloride secretion, guanylin, shows homology to a heat- Any relief he obtains can most likely be ascribed to an increase in
stable toxin produced by certain strains of pathogenic which of the following?
E. coli, which is the major cause of traveler’s diarrhea. Secre- A) intestinal transit time
tory diarrhea can also occur in noninfectious settings. In B) mucosal blood flow
particular, when the epithelium is exposed to a barrage of C) chloride secretion
immune and inflammatory mediators, such as in the D) peristalsis
inflammatory bowel diseases of Crohn’s disease and E) epithelial proliferation
ulcerative colitis, there is both a stimulation of chloride 3. A 30-year-old woman with Crohn’s disease undergoes a surgical
secretion and an inhibition of sodium and chloride absorp- resection of her terminal ileum. After recovering from the
tion, seen clinically as diarrhea, which is the most frequent surgery, she develops chronic diarrhea, with a daily stool output
symptom of these conditions. Diarrheal diseases continue 10× normal. Which of the following substances is/are most likely
to represent a major public health problem, particularly in primarily responsible for her symptoms?
developing countries where sanitation is inadequate and A) prostaglandins
represent an important cause of infant mortality in such B) inflammatory cytokines
C) VIP
countries, second only to respiratory infections. Diarrheal
D) bile acids
diseases also have a major impact in developed countries, E) short-chain fatty acids
though more frequently in terms of discomfort, inconve-
4. An infant develops chronic diarrhea and failure to thrive.
nience, and lost productivity than mortality. Nevertheless, Tests reveal glucosuria (increased glucose in the urine).
thousands of deaths from diarrheal diseases occur each Further studies show that urinary excretion of a nondigestible
year even in the United States, and many of these take disaccharide given orally, as well as uptake of oral alanine, is
place even after the patient has reached a health facility comparable to that seen in a normal child, whereas oral galactose
due to an underappreciation of how rapidly diarrhea can absorption is markedly impaired. Diarrhea in this child is most
cause dehydration and metabolic disturbances. likely due to a mutation in which of the following proteins?
A) CFTR
B) ENaC
C) NHE3
CHAPTER SUMMARY D) PepT1
E) SGLT-1
■ The intestine handles large volumes of fluid in fulfilling its
5. In a healthy adult, the volume of fluid presented to the intestine
physiological functions.
on a daily basis is approximately 8 L. Assuming a normal diet,
■ Dysfunction can rapidly impact whole body electrolyte reabsorption of the bulk of this fluid in the small intestine is
homeostasis. driven primarily by which of the following?
■ Water is moved passively in response to active electrolyte A) nutrient-coupled electrogenic sodium absorption
transport. B) electroneutral NaCl absorption
■ Transport mechanisms are heterogeneous along the length of C) nutrient-coupled proton absorption
the intestine and between crypt and villus cells. D) potassium absorption
■ Transport mechanisms consist of the asymmetrical arrange- E) electrogenic sodium absorption via ENaC channels
ment of a limited number of electrolyte transport pathways.
■ Certain pathogens can cause diarrheal disease by hijacking
normal cellular signaling pathways.
■ Diarrheal disease remains a major health problem in both
developed and developing countries.
53
C H A P T E R

Intestinal Mucosal
Immunology and Ecology
Kim E. Barrett

O B J E C T I V E S

■ Understand the role played by the mucosal immune system in protecting


the host from infections acquired via the oral route while not responding
to innocuous antigens.
■ Identify the cell populations that contribute to immunity in the gut and
where they are located
■ Describe the immune responses that occur to antigens encountered in the gut.
■ Understand the origins, makeup, and physiological importance of microbial
populations that exist in the normal intestine.
■ Define the consequences of abnormal immune responses in the intestine.

BASIC PRINCIPLES OF cells known as M cells. M cells overlie organized lymphoid


aggregates known as Peyer’s patches. The lymphocytes in these
MUCOSAL IMMUNOLOGY structures are immunologically naive and represent the affer-
ent arm of the mucosal immune system. After they have been
CONCEPT OF A MUCOSAL stimulated by their cognate antigen, they traffic back to the
IMMUNE SYSTEM lamina propria. During this migration, the lymphocytes mature
and differentiate, and then represent an efferent arm of the sys-
The surface of the gastrointestinal tract represents a vast frontier tem, capable of effector functions in the mucosa. In addition to
that can potentially serve as a portal of entry into the body. T cells, the humoral aspects of mucosal immunity are predom-
Indeed, the intestine is challenged to distinguish between poten- inantly served by secretory immunoglobulin (IgA) molecules,
tially harmful microorganisms, against which it must defend and the intestinal mucosa can also be considered to be con-
itself, and the innocuous antigens that occur in food. This con- stantly in a state of “physiological” inflammation, even in
stant exposure has driven the development of a highly special- health. Presumably this reflects the constant stimulation the
ized branch of the immune system, referred to as the mucosal system receives, and renders the intestine armed and ready to
immune system, which encompasses the mucosa-associated respond rapidly at times of threat by pathogens.
lymphoid tissues, or MALT. In fact, the intestine represents the
largest immunological compartment of the body, and has also
evolved nonimmunological barriers to invasion by pathogens. FUNCTIONAL ANATOMY OF
THE MUCOSAL IMMUNE SYSTEM
SPECIAL FEATURES OF THE IMMUNE
SYSTEM OF THE INTESTINE CELLULAR MEDIATORS
OF INNATE IMMUNITY
The lymphocytes that traffic to mucosal sites encounter anti-
gens in a controlled fashion. This is accomplished by limiting The innate arm of the mucosal immune system is designed to
the uptake of particulate antigens predominantly to specific mount rapid responses to pathogens, via the expression of
sites within the epithelial monolayer, via specialized epithelial pattern recognition receptors that recognize molecules that are

535

Ch53_535-542.indd 535 11/26/10 10:35:05 AM


536 SECTION VIII GI Physiology

important to broad classes of pathogenic microbes. These Follicle-associated epithelium


receptors are prominently expressed on macrophages as well as
on cells not classically considered to be immune effector cells, M cell
such as those in the epithelium. Pattern recognition receptors
include the Toll-like receptors, and proteins that may respond
to pathogen molecules presented intracellularly such as Nod2.
In general, activation of the innate immune response generates
chemotactic molecules that stimulate the influx and activation
T cell area
of more inflammatory cells, including monocytes and neutro-
phils. Collectively, these cells can effect microbial killing.

B cell area
CELLULAR MEDIATORS FIGURE 53–1 Structure of a Peyer’s patch in the small
OF ADAPTIVE IMMUNITY intestinal mucosa. The follicle-associated epithelium contains M, or
microfold, cells that have a subapical pocket in which antigens can be
Adaptive immunity involves the exquisitely specific recogni- presented to immune cells. Lymphocytes are aggregated underneath
tion of literally millions of discrete antigenic sequences via spe- the epithelium with T and B lymphocytes restricted to distinct areas.
cific receptors expressed on T and B cells. T cells recognize Peyer’s patches also contain dendritic cells (not shown) that can
peptides derived from antigenic sequences via a heterodimeric, present antigens to lymphocytes. (Reproduced with permission from Barrett
variable cell surface T-cell receptor. The peptides are presented KE: Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical

bound to major histocompatibility complex (MHC) mole- Pub. Division, 2006.)

cules on antigen-presenting cells. The binding of antigen to a


specific T-cell receptor then drives the expansion of a clone of
A final class of organized lymphoid cells in the intestine
cells expressing that receptor; some of these differentiate into
consists of lymphocytes anchored to the epithelial layer via
effector T cells; others remain as memory cells to jump-start an
specific adhesion molecules. These are called intraepithelial
adaptive immune response if the same antigen is encountered
lymphocytes, and appear predominantly to consist of mem-
again. T cells in the mucosal immune system can be subdivided
ory T cells capable of responding to only a subset of luminal
into those expressing the differentiation marker CD4 and those
antigens. They likely function primarily to secrete cytokines
expressing CD8. The former cell population recognizes extra-
involved in the regulation of the epithelium, and may also par-
cellular antigens displayed on the surface of antigen-presenting
ticipate in immune surveillance for emerging malignancies.
cells, likely including components of pathogenic microorgan-
isms. CD8-positive T cells, on the other hand, recognize abnor-
mal intracellular proteins and provide important protection
against potentially harmful intracellular events, such as viral SECRETORY IgA SYSTEM
infection or malignant transformation. Adaptive immunity is
Seventy to 90% of the B cells in the intestinal lamina propria
also mediated by B cells, which secrete antibodies specific for a
secrete IgA, with the remainder largely making IgM, and a few
given antigen under the influence of T cell–derived cytokines.
cells making IgE. Very few cells in the lamina propria make
IgG. In fact, given the large numbers of lymphocytes that nor-
ORGANIZATION mally reside in the gut, the daily synthesis of IgA exceeds that
of all other immunoglobulins.
OF LYMPHOID TISSUES
It is also important to appreciate how lymphoid tissues are orga- STRUCTURAL ASPECTS OF IgA
nized in the intestine. As mentioned earlier, in the small intes-
tine, the afferent arm of the system (i.e., the arm that responds IgA plasma cells in the lamina propria secrete two IgA mole-
initially to a threat) occurs in Peyer’s patches, as shown dia- cules that are bound together by a short polypeptide sequence
grammatically in Figure 53–1. In the colon, these aggregates of known as the J (or joining) chain. J chain is also a component
lymphocytes are more loosely organized, but analogous in func- of other polymeric immunoglobulins, such as IgM. The other
tion. Naive T and B cells from the bloodstream are targeted to critical component of secretory IgA that is found in the lumen
migrate into Peyer’s patches because they recognize a specific of the intestine derives from intestinal epithelial cells
type of endothelial cell that is found in these lymphoid struc- (Figure 53–2). Thus, dimers of IgA plus J chain are taken up at
tures. The other important components of the Peyer’s patch the basolateral surface of the epithelium by binding to a struc-
include the M cell, and dendritic cells and macrophages, which ture known as the polymeric immunoglobulin receptor
process and present antigens to the T and B cells. Once stimu- (pIgR). The complex of IgA plus pIgR is internalized and
lated, activated T and B cells migrate out of the lymphoid follicle translocated across the epithelial cell. At the apical membrane,
and eventually back to the lamina propria. the IgA dimer is released into the lumen bound to a cleaved
CHAPTER 53 Intestinal Mucosal Immunology and Ecology 537

LUMEN Secretory component encounter vesicles bearing IgA bound to pIgR that are des-
tined for the apical membrane. The IgA in these vesicles can
Secretory
lgA
bind the foreign antigen and traffic it back to the apical mem-
brane, thus resulting in its elimination. Finally, some IgA mol-
ecules may function to sequester antigens that are able to
penetrate to the lamina propria.
IgA has an additional specialization relative to other anti-
body classes that particularly suits it to function in the gut.
Thus, the antibody is not capable of fixing complement via the
Lysosomal
classical pathway, rendering it relatively noninflammatory on
degradation
antigen binding. This is likely an important consideration
given the vast antigenic load that is presented to the intestine,
Epithelial cell representing the combined influences of potentially antigenic
food proteins along with microbial products.

PHYSIOLOGICAL FUNCTIONS
The secretory IgA response protects the body from potentially
injurious substances that might otherwise stimulate a more
generalized immune/inflammatory reaction in the periphery.
plgR Note that the IgA system is not well developed in newborns. In
the breast-fed infant, protection can be obtained via IgA anti-
J chain bodies in the mother’s milk. This is one benefit of the common
mucosal immune system, discussed earlier, where lympho-
cytes activated by antigens encountered in the intestine also
Dimeric lgA traffic to other mucosal sites, including the mammary glands.
lgA plasma cell
The IgA system becomes mature in the child by the age of 5–6
months.

FIGURE 53–2 Secretion of IgA across the intestinal


epithelium. IgA is secreted by plasma cells in the lamina propria as a IMMUNE RESPONSE
dimer, with two IgA molecules linked by a J, or joining, chain. J chain is
recognized by the polymeric immunoglobulin receptor (pIgR) TO ENTERIC ANTIGENS
expressed on the basolateral membrane of epithelial cells, and once
There are three potential outcomes when the adaptive immune
bound, the complex is internalized and trafficked across the cytosol to
the apical membrane. Apical proteases cleave the extracellular portion system encounters an antigen in the intestine. The specific
of pIgR, which remains associated with the IgA dimer as secretory outcome that occurs depends, in part, on the type of antigen
component. The remnant of the pIgR is internalized and degraded. that is encountered and its quantity. A localized response can
(Reproduced with permission from Barrett KE: Gastrointestinal Physiology. New York: occur, such as the stimulation of antigen-specific IgA produc-
Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.) tion. The antigen can also, at least theoretically, drive a sys-
temic immune response with the production of circulating
antibodies plus expansion of antigen-specific T cells. However,
portion of pIgR, known as secretory component. Secretory it is clear that this would be an undesirable outcome under
component further stabilizes the IgA dimer against proteolytic normal circumstances, given the number of antigens encoun-
cleavage by either the digestive juices or bacterial proteases. tered by the intestine on a daily basis and the deleterious con-
sequences of systemic reactions to these. Thus, an additional
response to orally delivered antigens has also evolved in the
MECHANISMS OF mucosal immune system, which limits these adverse conse-
PROTECTIVE EFFECTS quences in the systemic immune system. This specialized
response is referred to as oral tolerance.
Secretory IgA exerts protection in the intestine via several
mechanisms. In the lumen, it can bind microbial antigens,
food antigens, and viruses to prevent their uptake by intestinal ORAL TOLERANCE
epithelial cells. There is also evidence for a second line of
defense that takes place within epithelial cells themselves. Oral tolerance refers to a mucosal immune response where local
Thus, if any antigens are internalized by these cells, they IgA is produced, yet there is no detectable immune response in
538 SECTION VIII GI Physiology

the periphery. Oral tolerance is the most frequent outcome for known as the enteric flora or microbiota. The species that make
antigens that are presumably innocuous, such as from food. The up this flora are referred to as commensal microorganisms.
determination as to whether oral tolerance will occur depends
on the type of antigen, the amount given, the frequency of expo-
sure, and host factors, including the age of the recipient. The DEVELOPMENT OF
lack of response in the periphery apparently requires active sup- INTESTINAL MICROBIOTA
pressive mechanisms, and is highly specific, extending only to
the substance that is fed and not to other antigens. The phenom- At birth, the intestinal tract is sterile. However, by the age of
enon of oral tolerance is being exploited for the experimental 1 month, the infant acquires a rich flora, derived from the
treatment of some systemic autoimmune diseases. environment in an oral-to-anal direction.

IMMUNE RESPONSIVENESS Indigenous Populations


The entire gastrointestinal tract can be shown to be colonized
Under different circumstances, it may be appropriate for the
by bacteria, but the types of bacteria and their numbers vary
intestine to respond to an antigen and additionally for this
along the gastrointestinal tract. The stomach and the majority
response to spread to the periphery. This is particularly the
of the small intestine contain relatively few bacteria, and lack
case for immune responses to pathogens. Generalized immune
anaerobes. On the other hand, beginning in the distal small
responses to otherwise innocuous antigens can also be driven
intestine, the numbers of bacteria increase sharply, and anaer-
by administering these in the presence of an adjuvant. This
obes also appear. The largest numbers of bacteria, by far, occur
latter approach might ultimately prove useful in exploiting the
in the colon, where anaerobes vastly outnumber aerobic bacte-
oral route to vaccinate people against disease-related antigens.
ria (Table 53–1).
Generalized immune responses can also be deleterious to
The flora in the colon is complex, likely containing at least
the host under specific circumstances. For example, if the bar-
400 different bacterial species. However, the precise propor-
rier function of the epithelium is compromised, it is possible
tions of each species tend to differ between individuals, while
to generate both local and systemic immune responses to the
remaining relatively constant in a given individual over time in
normal commensal flora of the gut, which in turn can lead to
the absence of events that alter the flora (see later). This has led
tissue injury.
to the concept that the bacterial flora of a given subject is essen-
tially equivalent to a “fingerprint,” representing the interplay
AUTOIMMUNITY between host and bacterial factors. The major anaerobic spe-
cies include Bacteroides, bifidobacteria, clostridia, Eubacteria,
During the development of immunity, clones of T and B cells and anaerobic streptococci. Likewise, important aerobes
capable of reacting to self-antigens are actively deleted. How- include enterobacteria such as E. coli, streptococci, and staphy-
ever, even in healthy individuals, a few such lymphocytes are lococci.
thought to remain in the mucosal immune system, and may be
needed to respond to microbes that express antigens that
Factors Controlling Microbiota
resemble proteins of the host, in an attempt to evade immunity.
Under normal circumstances, these autoreactive clones are The bacterial colonization of the upper gastrointestinal tract is
likely held in check by active suppressive mechanisms, includ- kept in check by a number of physical and humoral factors.
ing those mediated by regulatory T cells and inhibitory cyto- Many gastrointestinal secretions contain substances that are
kines. Likewise, self-antigens may be presented in the absence toxic to bacteria, including gastric acid, bile acids, small anti-
of appropriate costimulatory molecules, which results in anergy microbial peptides known as defensins, and lysozyme. In the
(the lack of an immune reaction) rather than an immune small intestine, moreover, the overall bacterial burden is kept
response. On the other hand, under pathologic conditions, this
regulation may be lost with the resulting emergence of autore-
activity and tissue inflammation and injury. These mechanisms TABLE 53–1 Enteric bacterial populations.
may be important in contributing to disease in conditions such
Cecum/
as inflammatory bowel diseases (Crohn’s disease and ulcer- Stomach Jejunum Ileum Colon
ative colitis), celiac disease, and atrophic gastritis.
Total bacteria/g 0–103 0–104 104–108 1010–1012

Aerobes and 0–103 0–104 104–105 102–109


INTESTINAL MICROECOLOGY facultative
anaerobes/g
The gastrointestinal tract also represents a unique body com- Anaerobes/g 0 0 103–108 1010–1012
partment with respect to the lifelong, reciprocally beneficial rela-
pH 3 6–7 7.5 6.8–7.3
tionship it establishes with a resident microbial community,
CHAPTER 53 Intestinal Mucosal Immunology and Ecology 539

in check by the combined influences of motility (especially maturation of intestinal lymphoid populations. Second, epi-
peristalsis) and the secretion of fluid and electrolytes that can thelial proliferation and differentiation are slowed.
wash bacteria out of the lumen. Secretion of IgA may also limit Colonic bacteria, in particular, also supply unique metabolic
the growth of some commensals. functions, divided into effects on endogenous substances and
In the colon, on the other hand, the relatively slow motility those on substances that originate outside the body. Colonic
permits the growth of large numbers of bacteria. These are bacteria convert bilirubin, a product of heme metabolism that
largely retained in the large intestine by the action of the ileo- is secreted in the bile, into urobilinogen. This compound
cecal valve. Upwards of 1012 bacteria can be found per gram of undergoes some passive absorption across the wall of the colon
colonic luminal contents, and the majority of the formed mass and appears in the urine. Bacterial dehydroxylases also act on
of the stool, after water, consists of dead bacteria. Indeed, the primary bile acids to generate secondary bile acids that enter
number of colonic bacteria in the average person is greater the enterohepatic circulation, and bacterial enzymes are also
than the total number of cells in the human body. The makeup responsible for deconjugating any conjugated bile acids that
of the colonic flora is relatively insensitive to diet, although a escape active reabsorption in the terminal ileum, to then permit
diet rich in fiber, which constitutes a fuel for anaerobic bacte- their passive reuptake across the colonic mucosa (Table 53–2).
ria, may result in an increase in overall bacterial numbers. Bacterial enzymes also salvage nutrients that cannot be
Conversely, intestinal colonization can be dramatically reduced degraded by pancreatic or other digestive enzymes (Table 53–2).
in patients taking broad-spectrum antibiotics. In this setting, This is particularly important for dietary fiber, a form of carbo-
there is sometimes also overgrowth of harmful bacteria that hydrate that is resistant to breakdown by amylase. Breakdown of
can cause disease, such as Clostridium difficile, commonly fiber occurs via a metabolic process known as fermentation,
acquired in hospitals, because there are fewer commensals to and requires a strict anaerobic environment. Fermentation can
compete with the harmful bacteria for nutrients. also break down any carbohydrates that escape digestion and
absorption in the small intestine. The products of fermentation
are the short-chain fatty acids, which can be absorbed by colonic
PHYSIOLOGICAL FUNCTIONS epithelial cells and used as fuel. Fermentation also yields energy
OF THE MICROBIOTA for the bacteria and the gases hydrogen, carbon dioxide, and
methane. Bacteria can also act on other dietary components to
Experiments in animals reveal that the intestinal microflora is yield byproducts, although these are usually quantitatively less
not essential to life. Thus, animals that are raised in a totally significant than the products of carbohydrate fermentation. A
sterile environment from birth are apparently healthy and final, and likely critical, role of the microflora is to increase the
reproduce normally. Nevertheless, the flora clearly has mea- resistance of the intestinal mucosa to colonization by pathogenic
surable effects on the host. First, in germ-free animals, the microorganisms (Table 53–3). Germ-free animals are exquisitely
mucosal immune system is poorly developed, illustrating the sensitive to enteric pathogens, succumbing to infection with
critical role of luminal stimuli in driving the development and only a few pathogenic bacteria presented orally compared to the

TABLE 53–2 Metabolic effects of enteric bacteria.


Substrate Enzymes Products Disposition

Endogenous substrates

Urea Urease Ammonia Passive absorption or


excretion as NH4+

Bilirubin Reductases Urobilinogen, stercobilins Passive reabsorption


Excreted

Primary bile acids Dehydroxylases Secondary bile acids Passive reabsorption

Conjugated bile acids Deconjugases Unconjugated bile acids Passive reabsorption

Exogenous substrates

Carbohydrates (fiber) Glycosidases SCFAsa Active absorption


H2, CO2, CH4 Expired in breath or excreted
in flatus

Amino acids Decarboxylases and Ammonia, HCO3− Reabsorbed or excreted


deaminases (for ammonia) as NH4+

Cysteine, methionine Sulfatases Hydrogen sulfide Excreted in flatus


a
SCFA, short-chain fatty acid.
540 SECTION VIII GI Physiology

TABLE 53–3 Selected important intestinal pathogens and their pathophysiological mechanisms.
Luminal (Toxigenic) Pathogens Adherent Pathogens Invasive Pathogens Viral Pathogens

V. cholerae Giardia Salmonella spp. Rotavirus

Enterotoxigenic E. coli Enteropathogenic E. coli Shigella Norwalk virus

H. pylori Campylobacter

Listeria

millions needed to cause disease in a normal animal. Likewise,


she is found to be hypotensive, with a red, itchy rash and
the enteric flora protects the host from overgrowth of bacteria
severe laryngeal edema that is starting to impair her breath-
that are innocuous when present in low numbers, but can cause
ing. The physician immediately administers epinephrine as
disease if allowed to dominate the flora.
well as an antihistamine, and the symptoms begin to sub-
side. As she starts to recover, she tells the physician that as
GENERATION OF a child she was unable to eat peanuts but thought she had
GAS IN THE INTESTINE grown out of the problem; one of her friends then points
out that one of the dishes she had tried contained ground
The intestinal flora is also the source of the majority of the gas peanuts in the sauce. The physician discharges her with a
that originates in the gastrointestinal tract. While large vol- prescription for an epinephrine self-injector, and advice to
umes of air may be swallowed with meals, most of this is make an appointment with an allergist for further testing
expelled from the stomach by belching. That which remains is and to carefully avoid ingesting peanuts in the future.
supplemented by carbon dioxide that is generated when gastric In certain individuals, likely as a result of a genetic predis-
acid is neutralized by bicarbonate; the majority of the carbon position, the intestinal immune system inappropriately gen-
dioxide can diffuse across the wall of the bowel and is excreted erates IgE antibodies to food proteins or other components
via the lungs. The gas that remains in the intestine is moved of the diet, such as nucleic acids. These antibodies bind to
back and forth by motility patterns, resulting in bowel sounds— mast cells that reside in the lamina propria, which are then
borborygmi—that can be heard through a stethoscope and
sometimes even by the naked ear as a “rumbling stomach.” The
absence of such bowel sounds is a reliable indicator that bowel
motility has been inhibited, which happens commonly (and Major gases
usually reversibly) after abdominal surgery. More distally,
fermentation and other metabolic pathways promoted by bac-
terial enzymes in the colon result in the formation of large vol-
umes of gas on a daily basis, even in normal individuals. The H2
volume of gas produced by a healthy individual is likely to aver- 20%
age 1 L per day. However, there is considerable variation among
individuals, and the amount of gas also depends on the quan-
tity of fermentable residues that depends, in turn, on the diet. CO2
Most of the gases present in flatus are nonodorous, with the 10%
majority consisting of nitrogen and hydrogen (Figure 53–3). N2
The amount of methane produced varies considerably among 65% CH4
individuals. On the other hand, the gases that are responsible O2 3%
for the odor of flatus are present in very small, or trace, 2%
amounts. These include hydrogen sulfide, indole, and skatole.

CLINICAL CORRELATION Trace gases:


Hydrogen sulfide
A 30-year-old woman is out for dinner with friends when Odorous
Indole
she notices a tingling sensation in her mouth and around Skatole (3-methyl indole)
her lips. Initially, she ignores the symptoms, but when her
face starts to swell and she begins to feel light-headed, her FIGURE 53–3 Composition of normal intestinal flatus.
(Reproduced with permission from Barrett KE: Gastrointestinal Physiology.
friends take her to a nearby emergency room. On arrival,
New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
CHAPTER 53 Intestinal Mucosal Immunology and Ecology 541

said to be “sensitized.” The sensitization process also results STUDY QUESTIONS


in upregulation of pathways that permit transfer of the 1. A scientist interested in gut ecology studies intestinal responses in
offending protein across intestinal epithelial cells. The net germ-free mice. Compared with normally housed animals, what
result is that the next time the person eats the protein in combination of findings would be expected in the colonic lumen?
question, it is transferred rapidly across the epithelium and
cross-links IgE antibodies on the surface of mast cells, caus- Secondary Short-chain
ing cell activation and release of a host of potent chemical sIgA Bile Acids Fatty Acids
mediators. These mediators can increase intestinal chloride A) Increased Increased Decreased
secretion and alter motility, which can be observed clinically
B) Decreased Decreased Decreased
as diarrhea. Stimulation of enteric nerve endings by mast cell
mediators may further amplify these responses. In a seri- C) Increased Decreased Increased
ously allergic subject, the antigen may also gain access to the D) Decreased Increased Increased
circulation from which it can trigger reactions in extraintes-
E) Increased Increased Increased
tinal sites, such as the skin and airways, or cause the general-
ized allergic reaction throughout the body known as systemic
anaphylaxis. Food allergies can be life-threatening. 2. A biotechnology company attempts to develop an oral vaccine
Certain foods are more commonly seen as triggers of aller- for use in developing countries by expressing a viral protein in
transgenic bananas. However, clinical trials reveal that consumption
gic responses, perhaps reflecting the relative stability of com-
of the bananas fails to confer protective immunity against the viral
ponent proteins during the digestive process. Such foods infection. What immune response likely accounts for this failure?
include peanuts, eggs, certain fruits, and shellfish. Unless an
A) IgA secretion
individual is allergic to multiple foods, the best treatment for B) phagocytic uptake
food allergy is to avoid the food in question, especially if very C) oral tolerance
severe allergic reactions occur. However, it may not always be D) T-cell sensitization
a simple matter to avoid a given food, particularly outside the E) Toll-like receptor activation
home, and for this reason, those who are seriously affected by 3. In a diagnostic test conducted in a patient with suspected
food allergies are usually advised to carry a self-injector con- Giardia infection, fecal samples are screened for the presence
taining epinephrine, which can counter serious symptoms. of antibodies reactive with giardial antigens. Assuming such
antibodies are found, what is their likely class?
A) monomeric IgG
B) monomeric IgA
CHAPTER SUMMARY C) dimeric IgA
■ The mucosal immune system is uniquely specialized to D) monomeric IgM
protect the vast interface between the host and environment E) pentameric IgM
represented by the GI tract. 4. A patient with a severe lung infection is treated with a broad-
■ Mucosal immune responses are shared among several mucosal spectrum antibiotic. Shortly after beginning this treatment, she
sites beyond the intestine. develops severe diarrhea. Stool samples are most likely to reveal
■ The intestine can be considered to be “physiologically evidence of overgrowth with which of the following organisms?
inflamed,” even in health, priming it to respond promptly A) Shigella dysenteriae
to invaders. B) Vibrio cholerae
■ Secretory IgA provides important humoral protection against C) Lactobacillus acidophilus
infections in the gut. D) Campylobacter jejuni
E) C. difficile
■ Presentation of antigens via the oral route often leads to a
response known as oral tolerance, where a local immune 5. A scientist working to develop a new diagnostic test administers
response exists in the face of systemic unresponsiveness. a solution of lactulose (a disaccharide that is not digested by host
enzymes, but can be broken down by bacterial enzymes) orally to
■ Oral tolerance may protect us from inappropriate reactions
a group of volunteers as well as to a patient with symptoms of
to food antigens. This response may also be exploited for
malabsorption. She then measures the concentration of
therapeutic benefit in autoimmune diseases.
hydrogen in expired breath from each group. Hydrogen is
■ The intestine maintains a lifelong, reciprocally beneficial present in negligible amounts in both groups prior to lactulose
relationship with a complex microflora that is predominantly administration, and increases in the control group only after a
contained in the colon. 1–2-hour lag period. However, in the patient, breath hydrogen
■ Colonic bacteria supply metabolic functions, especially levels begin to rise almost immediately. What is the most likely
fermentation, and contribute to intestinal gas production. cause of this more rapid appearance?
■ Commensal bacteria may provide important protection against A) celiac disease
colonization by pathogens. B) cystic fibrosis
■ Derangements in intestinal physiology occur when immune C) Crohn’s disease
responses are inappropriately stimulated in the intestine, or D) small bowel bacterial overgrowth
when defenses fail to protect us from infection with pathogens. E) H. pylori infection
This page intentionally left blank
54
C H A P T E R

Intestinal Motility
Kim E. Barrett

O B J E C T I V E S

■ Understand how the motility functions of the esophagus, stomach, and


intestines contribute to the integrated response to a meal.
■ Describe the functional anatomy of the esophagus, stomach, intestines, and
related structures, and their innervation.
■ Understand the roles played by the oral cavity, pharyngeal structures,
esophagus, and esophageal sphincters in transferring food from the mouth
to the stomach during swallowing.
■ Describe receptive relaxation and mixing/grinding patterns of motility and
their regulation.
■ Understand how the stomach is emptied, and how this is coordinated with
the function of downstream segments.
■ Define the motility patterns that characterize movements of the small and
large intestines under fed and fasted conditions and their control
mechanisms.
■ Distinguish between mixing patterns and those that propel contents along
the length of the intestine.
■ Describe reflexes that coordinate the motility functions of the small intestine
and colon with the function of the stomach.
■ Understand the process whereby undigestible residues of the meal are
eliminated from the body.

This chapter reviews the processes that move the meal along
the length of the gastrointestinal tract and provide for its dis-
BASIC PRINCIPLES OF
persion, as well as mixing with digestive secretions. Because ESOPHAGEAL MOTILITY
there are segmental differences in the types and functions of
the motility processes of the alimentary tract, we will consider ROLE AND SIGNIFICANCE
in turn the motility of the esophagus, stomach, small intestine,
and colon. Each segment has a specific role to play in handling The esophagus is a muscular tube that transfers food from the
the meal, but all depend on the properties of the smooth mus- mouth to the stomach. Under normal circumstances, food
cle layers that surround the mucosa. It may therefore be help- resides in the esophagus for only a few seconds. The move-
ful to review the basic properties of smooth muscle as explained ments of the esophagus and related oral and pharyngeal struc-
in Chapter 11. tures must also be carefully regulated to avoid misdirection of

543

Ch54_543-558.indd 543 11/26/10 10:33:41 AM


544 SECTION VIII GI Physiology

Swallowing
Glossopharyngeal
center
+ vagal afferents

Pharynx
(S)
Outflow:
Upper
esophageal • Somatic nerves (S)
sphincter regulate striated
(S) muscle directly
Striated muscle • Autonomic nerves (A)
regulate smooth
muscle via
enteric nervous
system or directly
(A)
Smooth muscle

(A)
FIGURE 54–1 Functional anatomy and innervation of
the esophagus. Note that the mode of innervation differs
Lower
between the portions of the esophagus made up of smooth esophageal
versus striated muscle. (Reproduced with permission from Barrett KE: sphincter
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill,
Medical Pub. Division, 2006.) Stomach

the food into the respiratory tract, or inspired air into the diges- in a circular fashion and the outer oriented longitudinally.
tive system. At rest, the esophagus is a relaxed structure that is However, unlike the exclusive occurrence of smooth muscle in
closed off at both ends by sphincters—the upper esophageal all more distal segments of the gastrointestinal tract, the esoph-
sphincter and lower esophageal sphincter (LES). These agus contains striated (or skeletal) muscle in its upper third,
sphincters not only cooperate in the act of swallowing, or both striated and smooth muscle in its middle third, and exclu-
deglutition, but also prevent backflow of gastric contents into sively smooth muscle in its most distal third. The distinction
the esophageal lumen or oral cavity. However, under specific between muscle types also corresponds approximately to dif-
circumstances, the esophagus does allow for retrograde move- ferent types of neural control, as discussed later.
ment. This occurs normally for air swallowed with the meal, in Other structures associated with the esophagus are impor-
the process of belching, or abnormally during vomiting. Dur- tant in swallowing and normal esophageal function. We have
ing retrograde movement in humans and most mammals, the already mentioned the upper esophageal sphincter and LES,
esophagus itself is a passive conduit, that is, there are no spe- which occlude both ends of the esophagus at rest. The esopha-
cific motility functions that propel vomitus or air along the gus is situated within the low-pressure thorax, and thus the
length of the tube. Swallowing can be initiated voluntarily, but presence of these sphincters is important to prevent the entry
thereafter reflects an automatic reflex that involves, sequentially, of air and gastric contents. The pharynx, which connects the
impulses from the brainstem, processing of this information nose and mouth to both the esophagus and trachea, is also
through vagal centers in the central nervous system, direct critically involved in swallowing. It segregates food and air as
effects of parasympathetic vagal efferents on esophageal muscle they pass through this region.
layers, and relay of information via the enteric nervous system
(Figure 54–1). Movement of materials along the length of the
esophagus is aided by gravity, but predominantly depends on a
INNERVATION
coordinated series of muscle contractions and relaxations that The function of the pharynx is controlled by the central ner-
make up the propulsive motility pattern known as peristalsis. vous system, via outflow from a region known as the central
swallowing center (Figure 54–1). The pharynx thereby per-
mits complex coordination of voluntary swallowing with func-
FUNCTIONAL ANATOMY OF THE tions such as respiration and speech. Central input also
ESOPHAGEAL MUSCULATURE controls the contractile function of the upper one third of the
esophagus. The somatic nerves that innervate these structures
MUSCLE LAYERS have motor end plates that terminate directly on the striated
muscle fibers. They originate in brain regions of the medulla
The esophagus is 18–25-cm long in adult humans. Like the known as the nucleus retrofacialis and the nucleus ambiguus
remainder of the gastrointestinal tract, it is surrounded by two and release acetylcholine (ACh), which acts via nicotinic
muscle layers: the innermost (i.e., closest to the lumen) oriented receptors.
CHAPTER 54 Intestinal Motility 545

The smooth muscle of the esophagus is innervated predom- contractile state of the pharyngeal muscles. These events occur
inantly by the vagus nerve. The vagal efferents synapse with almost simultaneously, which is in contrast to the slower
myenteric neurons via ACh and with the smooth muscle motility changes that occur more distally in the esophagus, as
directly via ACh and substance P. Sensory afferents located in will be discussed later. First, the larynx and soft palate move
the esophagus likewise project via the vagus to the brain region upwards, closing off the airway and nasopharynx, respectively.
of the medulla known as the nucleus tractus solitarius in the Next, contraction of several muscles in the anterior portion of
dorsal vagal complex. Cell bodies in this region also project the pharynx causes forward displacement of the larynx and
to the motor neurons in the nucleus ambiguus, which control pharynx as well as helping to open the upper esophageal
a pattern generator for the oral and pharyngeal components of sphincter. Sphincter opening also depends on relaxation of the
swallowing. This neural circuitry ensures that control of the encircling cricopharyngeal muscle. This is accomplished by a
muscle groups involved in deglutition is linked to the function suppression of impulses normally occurring to this region,
of more distal regions of the esophagus, as well as to the regu- coordinated by the swallowing center via the nucleus ambig-
lation of opening of the LES. uus. Longitudinal contractions of the pharynx also bring the
The esophagus is also richly supplied with enteric neurons. upper esophageal sphincter close to the base of the tongue,
These clearly contribute to both sensing the presence and whereupon a pressure gradient developed by the tongue and
nature of esophageal contents and coordinating local reflexes pharyngeal muscles serves to force the bolus through the
that supplement central control of swallowing and esophageal sphincter. Finally, the posterior wall of the pharynx contracts
peristalsis. This network of enteric neurons can produce sec- in a transverse fashion to clear the area of any remaining food
ondary peristalsis of the smooth muscle portion of the esopha- residues. These transverse contractions are propagated aborally
gus even in the absence of vagal input. (i.e., away from the mouth) and can be considered the harbin-
ger of the peristaltic wave that later will carry the bolus through
the esophagus and down into the stomach. The sequence of
FEATURES OF events involved in normal swallowing is shown diagrammati-
cally in Figure 54–2.
ESOPHAGEAL MOTILITY
The motility functions consist of sequential movement of food
from the mouth into the esophagus itself, propulsion along the PERISTALSIS
length of the esophagus via peristalsis, and relaxation of the
LES to permit entry of the food bolus into the stomach. In Once the bolus has moved through the upper esophageal
health, these components of deglutition are tightly integrated, sphincter into the esophageal lumen, it is moved along the
but for simplicity we will consider each in turn here. length of the tube via peristalsis (Figure 54–3). The sequenc-
ing and thus the direction of this propulsive process appears to
be hardwired, with contraction of more distal segments occur-
SWALLOWING ring at longer latencies following the swallow than in those
close to the pharynx. The striated muscle region contracts
Although the term swallowing can refer to the entire process within 1–2 seconds after the swallow, the middle third of the
required to move food from the mouth to stomach, here we esophagus within 3–5 seconds, and the lower third within
will consider it to include only those motility events that move 5–8 seconds. This means that the ability of the body to transfer
the bolus beyond the upper esophageal sphincter, as well as food from the mouth to stomach is largely independent of
their regulatory controls. As noted earlier, swallowing is initi- body orientation—one can swallow food while hanging upside
ated when we sense that food particles in the mouth have been down. Nevertheless, gravity influences transit rate, particu-
reduced in size sufficiently to permit their passage into the larly for liquids. The peristaltic wave requires up to 10 seconds,
esophagus. While we consider this to be a voluntary response, on average, to sweep solid esophageal contents along its length.
during its course it in fact becomes an involuntary reflex This relatively slow transfer should be contrasted with the
involving significant input from a pattern recognition center rapid events of swallowing itself.
in the brainstem. This recognizes a food bolus as suitable Peristalsis in the esophagus is stimulated by its distension.
for swallowing and generates the required neuromuscular Mechanoreceptors on sensory afferents transmit impulses to
response. However, it is possible to override this recognition the dorsal vagal complex, which in turn activates somatic and
system voluntarily, such as in the case of swallowing a pill or vagal efferents that terminate either directly on the striated
capsule. But in either case, the subsequent events that contrib- muscle in the upper third of the esophagus or onto nerves of
ute to swallowing are entirely involuntary. the enteric nervous system, respectively. The latter activate
First, the tongue shapes and lubricates the bolus and moves enteric nerves capable of releasing ACh (to induce contrac-
it backward in the mouth. Subsequently, a rapid series of pha- tion) above the location of bolus-induced distension, or nitric
ryngeal effects occur, initiated by mucosal mechanoreceptors oxide (to induce relaxation) below the bolus (Figure 54–4).
in the pharynx that activate afferent nerves traveling via the The net effect of the sequential contractions and relaxations is
glossopharyngeal nerve to the swallowing center. In turn, to move the bolus aborally. The primary wave of peristalsis
efferent motor nerves run through the vagi to control the along the length of the esophagus may also be followed by a
546 SECTION VIII GI Physiology

Hard palate Soft palate Pharynx

Food

Tongue
Epiglottis

Glottis Upper
esophageal
Trachea sphincter
Esophagus
(a) (b) (c) (d)

FIGURE 54–2 Movement of food through the pharynx and upper esophagus during swallowing. a) The tongue pushes the food
bolus to the back of the mouth. b) The soft palate elevates to prevent food from entering the nasal passages. c) The epiglottis covers the glottis
to prevent food from entering the trachea, and the upper esophageal sphincter relaxes. d) Food descends into the esophagus. (Reproduced with
permission from Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

secondary wave restricted to the smooth muscle portion into the lower esophagus. The response is triggered by disten-
(Figure 54–5). Secondary peristalsis may clear a bolus that sion, and involves both local reflexes within the enteric ner-
was not wholly expelled from the esophagus during the pri- vous system and vagovagal reflexes (Figure 54–6). Studies
mary wave, or remove any gastric contents that reflux back suggest that the presence of acid alone within the distal esoph-
agus, in the absence of significant distension, may also be suf-
ficient to generate a secondary peristaltic response.
Swallow The esophagus also transmits information about its contents
50 mm Hg back to more proximal segments. Thus, the presence of water
Pharynx 0 or, more potently, acid in the esophagus can be shown to
increase the pressure of the upper esophageal sphincter. This
Upper esophageal 50 mm Hg
response depends, at least in part, on vagovagal signaling. On
sphincter 0

50 mm Hg
Striated muscle 0
esophagus Dorsal vagal complex

50 mm Hg
0 Vagal efferents

Vagal afferent
Smooth muscle Myenteric nerves
esophagus 50 mm Hg
NO
0
ACh

Lower esophageal 50 mm Hg
sphincter 0
5 sec Bolus

Orad contraction
FIGURE 54–3 Primary peristalsis triggered by swallowing in Caudad
the esophagus. Note that the pressure wave that moves down the distension
esophagus is coordinated with opening of the lower esophageal FIGURE 54–4 Control of peristalsis by vagovagal reflexes in
sphincter. (Adapted with permission from Biancani P. et al: Esophageal motor function. the lower esophagus. ACh, acetylcholine; NO, nitric oxide.
In: Textbook of Gastroenterology, 4th ed. Yamada T, Alpers DH, Kaplowitz N, Laine L, (Reproduced with permission from Barrett KE: Gastrointestinal Physiology. New York:
Owyang C, Powell DW (editors). Philadelphia: Lippincott Williams and Wilkins, 2003.) Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
CHAPTER 54 Intestinal Motility 547

Balloon LES RELAXATION


inflated
50 mm Hg The final component of esophageal motility is relaxation of the
Pharynx 0 LES, allowing the bolus to move into the stomach. Under rest-
ing conditions, the LES is tonically contracted. There is evi-
Upper esophageal 50 mm Hg
0
dence to suggest that this tonic contraction is accounted for by
sphincter
a mechanism referred to as myogenic—that is, the contractile
50 mm Hg state of the muscle is independent of neural input, and increases
Striated muscle 0 intrinsically as it is stretched. Moreover, the tone of the sphinc-
esophagus ter can also be increased by neurohumoral agents released in
50 mm Hg concert with ingestion of a meal, including ACh and gastrin.
0 The basal tone of the sphincter is critical to protect the lower
portion of the esophagus from the corrosive effect of the gas-
Smooth muscle 50 mm Hg
tric contents. When a food bolus is swallowed, relaxation of
esophagus 0
the LES is closely coordinated with the preceding motility
50 mm Hg events, such that it occurs just as the peristaltic wave reaches
0 the end of the esophagus (Figures 54–3 and 54–5). The careful
integration of peristalsis and sphincter relaxation is brought
Lower esophageal 50 mm Hg about by the combined activity of the vagus nerve and enteric
sphincter 0 nervous system, and is mediated by the release of nitric oxide
5 sec
from inhibitory nerves whose cell bodies lie in the myenteric
FIGURE 54–5 Secondary peristalsis triggered by distention plexus. Vasoactive intestinal polypeptide (VIP) within these
in the smooth muscle portion of the esophagus. Contraction nerves may also contribute to LES relaxation. In general, the
orad to the bolus is followed by a descending pressure wave that is contractile state of the LES at any given moment can be consid-
coordinated with lower esophageal sphincter opening. (Adapted ered to reflect a summation of both positive and negative
with permission from Biancani P. et al: Esophageal motor function. In: Textbook inputs. Even in healthy individuals, the LES relaxes transiently
of Gastroenterology, 4th ed. Yamada T, Alpers DH, Kaplowitz N, Laine L, Owyang C, from time to time independent of either swallowing or second-
Powell DW (editors). Philadelphia: Lippincott Williams and Wilkins, 2003.) ary peristalsis. Such relaxations may be required to facilitate
belching of swallowed air. However, in individuals with reflux
the other hand, if air is present in the esophagus, it induces disease, these transient relaxations occur more frequently and
opening of the upper esophageal sphincter, which is critical to allow reflux of the gastric contents into the distal esophagus.
belching, or the retrograde movement of air that is swallowed
along with food.
BASIC PRINCIPLES
Interneurons
OF GASTRIC MOTILITY
Sensory ROLE AND SIGNIFICANCE
neuron
As we have learned from previous chapters, the stomach is a
Myenteric plexus segment of the gastrointestinal tract in which important
Excitatory aspects of digestion and secretory function are initiated. How-
motor neuron ever, in addition to these functions, which are largely depen-
ACh NO/VIP dent on gastric secretion, the stomach also plays critical roles
Contraction Relaxation that depend on its motility properties.
of circular of circular First, the stomach can be considered as a homogenizer,
muscle muscle mechanically breaking down ingested food into an emulsion of
Distension
ORAD
low pH
CAUDAD
small particles with a vastly increased total surface area, thereby
amplifying the effects of digestion. Second, the stomach matches
FIGURE 54–6 Control of peristalsis by the enteric nervous food delivery to the digestive and absorptive capacity of more
system. Peristalsis can be triggered when a sensory nerve detects
distension or luminal acidity. Interneurons convey the signal to
distal segments of the gut. Under normal circumstances,
excitatory and inhibitory nerves above and below the site of the stomach allows the delivery of approximately 200 kcal/h into
stimulation, respectively. ACh, acetylcholine; NO, nitric oxide; VIP, the small intestine. To accomplish its reservoir function, the
vasoactive intestinal polypeptide. (Reproduced with permission from stomach exhibits remarkable pressure/volume characteristics,
Barrett P. KE: Gastrointestinal Physiology. New York: Lange Medical Books/ which accommodate the volume of the meal without allowing
McGraw-Hill, Medical Pub. Division, 2006.) significant reflux of the gastric contents back into the esophagus,
548 SECTION VIII GI Physiology

or forcing them prematurely into the duodenum. Distention of arranged circumferentially, and closer to the lumen,
the stomach also delivers important information to downstream and a longitudinal layer that is oriented along the length of the
segments of the gastrointestinal tract, as well as contributing to organ. However, because the stomach is shaped as a sac rather
the signaling of satiety. Finally, the stomach possesses distinct than a simple tube, these different muscle layers may assume
motility functions during the fasted state. The migrating motor greater or lesser importance in the different functional regions
complex (MMC), a “housekeeping” motility pattern, sweeps of the stomach, likely also important for specific motility pat-
undigested materials or ingested foreign objects along the length terns. Thus, circular muscle is prominent throughout the
of the entire gastrointestinal tract, beginning at the stomach. stomach, although it is notable that it is largely electrically iso-
lated from the circular muscle in the small intestine because of
the presence of a connective tissue septum at the level of the
FUNCTIONAL ANATOMY OF pylorus. On the other hand, longitudinal muscle is more
prominent in the distal stomach, and these muscle fibers are
THE GASTRIC MUSCULATURE mostly continuous with those of the duodenum. There is also
a small region of obliquely oriented muscle fibers in the lesser
The stomach is a highly distensible muscular sac with the larg-
curvature of the stomach that is continuous with the gastroe-
est caliber of any intestinal segment. It can be divided into two
sophageal junction, and restricted to the cardia. Finally, the
functional regions for considerations of motility (Figure 54–7).
pylorus represents a specialized region of circular muscle at
The proximal stomach, consisting of the cardia, fundus, and
the point where the caliber of the gastric lumen is sharply
proximal portion of the body (corpus) of the stomach, serves
reduced prior to entry into the duodenum; it serves as a
primarily as a reservoir and to move gastric contents to the
mechanical barrier to food exit that is also enhanced by a
distal stomach. Tonic contractions of the proximal stomach
folded, redundant mucosa.
are additionally important in gastric emptying. The distal
The smooth muscle cells of the different functional regions
stomach, on the other hand, consisting of the distal portion of
of the stomach also display distinctive contractile properties.
the body and the antrum, serves predominantly to grind and
Most important for our discussion is the distinction between
pulverize the meal. Finally, the pylorus acts as a sphincter that
phasic and tonic contractions. Some smooth muscles con-
controls the amount and size of food particles that can exit the
tract and then relax in a matter of seconds, known as phasic
stomach in the fed state. Conversely, full relaxation of the
contractions, which are prominent in the distal stomach. Tonic
pylorus is critical during the housekeeping MMC.
contractions, on the other hand, are sustained contractions
that are prominent in the proximal stomach, and may persist
for many minutes. Each type of contraction is important in
MUSCLE LAYERS mediating the specific motility properties that are needed for
the function of each region of the stomach.
As elsewhere in the gastrointestinal tract, the muscle layers of
the stomach consist of a circular layer of smooth muscle

INNERVATION
Lower esophageal sphincter
The stomach is richly endowed with both intrinsic and extrin-
sic neural inputs. The major extrinsic pathways are parasym-
pathetic and contained within the vagus nerve. Most vagal
Fundus efferents that terminate in the stomach are stimulatory, cho-
linergic nerves although a few nerves with high thresholds for
Reservoir
(Tonic contractions)
activation are inhibitory, releasing VIP and nitric oxide as
their major neurotransmitters. Mechanosensitive and chemo-
Body Pacemaker
region sensitive vagal afferents are also critical for the control of
motility functions. These activate sites in the nucleus tractus
solitarius of the dorsal motor nucleus in the brain. In a more
Antrum limited fashion, sympathetic innervation arrives at the stom-
ach by way of the splanchnic nerve, and these nerves release
noepinephrine as a postganglionic inhibitory neurotransmit-
ter at the level of enteric ganglia. The physiological role of
Antral pump sympathetic innervation to the stomach is relatively unclear,
Pylorus (Phasic contractions) but presumed to be minor compared with vagal influences.
FIGURE 54–7 Regions of the stomach involved in motility On the other hand, sympathetic influences may contribute to
responses, and location of the gastric pacemaker. (Reproduced with a decrease in gastric motility during times of threat.
permission from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical Intrinsic innervation via the enteric nervous system is also
Books/McGraw-Hill, Medical Pub. Division, 2006.) critically important to the full expression of gastric motility
CHAPTER 54 Intestinal Motility 549

responses. Indeed, many of the stereotypical motility responses whereas the duodenum has a BER of 12 cpm. This presumably
of the stomach are largely, if not wholly, independent of central reflects the presence of dominant and separate pacemakers in
input. Myenteric neurons of the stomach also provide for each distinct segment, which then relay electrical information
coordination of gastric motility functions with those of the throughout the segment that they control via the remaining
more distal segments of the gut, particularly during fasting network of interstitial cells of Cajal.
periods. These nerves also communicate with the pacemaker
cells of the intestine, known as interstitial cells of Cajal,
located within the circular muscle layers of the stomach and RECEPTIVE RELAXATION
proximal gut. This communication establishes the maximal
The ability of the stomach to relax as its volume increases is
rate at which contractions of the tissue can occur if an addi-
essential to its reservoir function. The process—receptive
tional excitatory signal is also received, which is known as the
relaxation—results in a drop in gastric pressure immediately
basal electrical rhythm (BER).
after eating that persists until all solids have been emptied
from the stomach. The process involves vagal input coincident
with food intake, vagovagal reflexes, and intrinsic reflexes
FEATURES OF GASTRIC MOTILITY mediated wholly within the wall of the stomach (Figure 54–9).
Vagovagal and intrinsic reflexes are triggered by the activation
BASAL ELECTRICAL RHYTHM of mechanosensitive nerve endings within the stomach wall.
In turn, ACh released by vagal pathways acts presynaptically
The BER refers to waves of rhythmic depolarization of intesti-
to release additional neurotransmitters that actively relax the
nal smooth muscle cells, which originate at a specific point
gastric smooth muscle layers, particularly in the proximal part
and then are propagated along the length of the gastrointesti-
of the stomach. Both VIP and nitric oxide have been impli-
nal tract. The pacemaker potentials originating in this region
cated in this response.
determine the contractile parameters of the stomach as a
Gastric tone may also be affected by feedback signals com-
whole—namely, the maximal frequency of contractions, their
ing from more distal segments of the gastrointestinal tract.
propagation velocity, and the direction in which they propa-
For example, distension of the duodenum or colon, or an
gate. For the stomach, the waves appear to begin at a point in
increase of fat or protein in the duodenum or ileum, results
the body along the greater curvature of the stomach, and then
in a decrease in the tone of the gastric fundus. In this way,
sweep across the stomach toward the pylorus (Figure 54–7). It
gastric emptying is retarded until the duodenum is able to
should also be emphasized that the BER represents only the
process additional nutrients. The response involves reflexes
maximal rate of contraction of the stomach or indeed of any
of the enteric nervous system, as well as CCK by binding to
other segment of the gastrointestinal tract. The waves of depo-
CCK-A receptors expressed on vagal sensory afferents.
larization that occur in response to the pacemaker activity of
Gastric distension, conversely, signals forward to more distal
the network of interstitial cells of Cajal are not of sufficient
segments to ready them for the arrival of the meal. Probably
magnitude to initiate action potentials in the smooth muscle.
the best known of these reflexes is the gastrocolic reflex,
Rather, it is only when the release of stimulatory neurotrans-
which may induce the need to defecate shortly after ingesting
mitters from enteric nerve endings is superimposed on these
a meal.
waves of depolarization that an action potential may occur,
leading in turn to contraction of the smooth muscle
(Figure 54–8). Various patterns of motility can thus be accom- MIXING AND GRINDING
plished depending on whether the stomach is filled with a
meal, or is in the fasted state. The primary motility pattern of the distal portion of the
The BER differs in the various intestinal segments. For exam- stomach during the fed state consists of rapid phasic contrac-
ple, in the stomach the BER is approximately 3 cycles/min (cpm), tions that occur circumferentially, and which can even

Stimulated
0 Resting BER
Membrane
potential
(mV)
FIGURE 54–8 Basal electrical rhythm
established by the gastric pacemaker. Note that
−70 waves of depolarization initiated by the pacemaker
20 s
are insufficient to trigger contractions unless these
5 are superimposed with a contractile stimulus.
Tension (Modified with permission from Barrett KE: Gastrointestinal
(g) Physiology. New York: Lange Medical Books/McGraw-Hill, Medical
0 Pub. Division, 2006.)
550 SECTION VIII GI Physiology

occlude the lumen (Figure 54–10). The contractions proceed


DVC
from the gastric pacemaker region and move toward the
pylorus in a peristaltic pattern. As these waves of contraction
begin, they force the gastric contents toward the outlet of the
stomach. However, as the velocity of the peristaltic wave
increases, it overtakes all but the smallest particles in the gas-
Vago-vagal tric lumen, and thus the majority of the meal is forced back-
reflex wards (retropulsion), mixing the gastric contents with the
VIP NO gastric juice and mechanically reducing the size of the food
NO VIP particles.
Relaxation

GASTRIC EMPTYING AND


THE ROLE OF THE PYLORUS
ACh
When the stomach is filled with a meal, the pylorus remains
closed for prolonged periods with only intermittent openings
h
tc
re

that allow small food particles (less than 1–2 mm) to enter the
St

duodenum. The pylorus is regulated by both inhibitory and


excitatory vagal pathways as well as ascending and descend-
Stretch ing intrinsic reflexes, and its function is clearly regulated
Fat independently of contractions of the gut segments on either
Protein
side. Nitric oxide has been identified as a key mediator of
CCK pyloric relaxation, arising from both vagal and intrinsic path-
ways, whereas opioids released from vagal efferents and CCK
FIGURE 54–9 Intrinsic and vagovagal reflexes involved in have been implicated in contractions. The presence of nutri-
receptive relaxation of the stomach. The figure indicates that ents, hypertonicity, or acid in the duodenum also causes the
signals triggered by nutrients in the duodenal lumen, or duodenal pylorus to close, with ACh and 5-hydroxytryptamine identi-
distension, also result in relaxation of the gastric fundus. ACh, fied as mediators.
acetylcholine; NO, nitric oxide; VIP, vasoactive intestinal polypeptide; Emptying of the stomach involves both tonic contractions
CCK, cholecystokinin; DVC, dorsal vagal complex. (Reproduced with of the proximal region and phasic distal contractions. Extrin-
permission from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical
sic innervation, working together with the enteric nervous
Books/McGraw-Hill, Medical Pub. Division, 2006.)
system, is vital for normal emptying, with 5-hydroxytryptamine

1 2

FIGURE 54–10 Gastric motility patterns Pylorus Gastric


contributing to mixing, grinding, and sieving of Pylorus contents
gastric contents. 1) A circumferential contraction, A, Gastric
contents
sweeps toward the pylorus resulting in anterograde
Contraction B
and retrograde propulsion of material. 2) As
contraction A subsides, a second contraction, B, mixes Contraction A
contents further. 3) Contraction B is sufficient to cause Contraction A
transient and partial opening of the pylorus, allowing
particles smaller than 1 mm to exit the stomach. 3 4
Pylorus
Larger particles are propelled back into the stomach
to be further dispersed by contraction C. 4) Further Pylorus
cycles of contraction against a closed pylorus
continue mixing and grinding until all of the meal is
emptied from the stomach. (Reproduced with permission Contraction C Contraction D
from Barrett KE: Gastrointestinal Physiology. New York: Lange Contraction B
Medical Books/McGraw-Hill, Medical Pub. Division, 2006.) Contraction C
CHAPTER 54 Intestinal Motility 551

being identified as a key mediator. 5HT1 receptors have been Some aspects of neurohumoral control of the MMC are
implicated in delaying gastric emptying while 5HT3 receptors understood. For example, the onset of phase III activity is
increase it. CCK also delays gastric emptying. The rate of gas- independent of either parasympathetic or sympathetic input,
tric emptying also depends on both the physical state of the but rather is correlated with levels of plasma motilin
contents and their chemical characteristics. Inert liquids empty (Figure 54–11). However, the trigger for cyclical release of this
most rapidly. If the liquid contains nutrients, a rapid initial hormone from the duodenal mucosa is not known, other
phase is followed by slowed exit, apparently reflecting feed- than the fact that release is suppressed by feeding. Neverthe-
back from the small intestine. The rate of emptying also less, it enhances propulsive motility via both direct and
depends on the caloric density and osmolarity of the contents. indirect actions, the latter involving the release of ACh,
Emptying of solids from the stomach is slower yet, with a half- 5-hydroxytryptamine, and nitric oxide. Moreover, while phase
time of approximately 1–2 hours. Moreover, emptying of sol- III can occur in the absence of vagal input, impulses from the
ids from the stomach does not begin immediately, but occurs vagus can amplify the response. Similarly, phase II of the MMC
only after a lag phase of up to 1 hour during which retropul- is abolished by vagotomy. The MMC is also reduced during
sion and mixing take place. sleep, and slowed by stress.

GASTRIC MOTILITY DURING FASTING VOMITING


In the period in between meals, the stomach, in common Vomiting primarily results in evacuation of the gastric con-
with more distal segments of the gastrointestinal tract, tents, and reflects the coordinated interaction of neural,
undergoes the stereotypical pattern of motility known as the humoral, and muscular phenomena. Vomiting may be
MMC. In the absence of feeding, cycles of motility lasting prompted by both central and peripheral triggers
approximately 100 minutes, and consisting of three phases, (Figure 54–12), but pivotally requires the involvement of cen-
begin in the stomach and are propagated aborally tral brain regions to coordinate the responses required. A
(Figure 54–11). Phase I is characterized by quiescence. Dur- chemoreceptor trigger zone, located in the area postrema of
ing phase II, contractile activity increases, but with irregular the medulla, receives input from cortical, oral, vestibular, and
contractions that fail to propel luminal contents. Finally, peripheral afferents. In addition, the blood/brain barrier in
phase III involves a 5–10-minute period of intense, luminally this region is relatively leaky. This means that the chemorecep-
occlusive contractions that sweep from the body of the stom- tor trigger zone can sample chemical constituents of both the
ach to the pylorus, and from there move into the duodenum. blood and cerebrospinal fluid. Various stimuli can lead to both
During this time, the pylorus opens fully in normal subjects, nausea (the sensation that vomiting is imminent) and actual
allowing any indigestible residues to be cleared. This house- vomiting, such as endocrine changes in pregnancy, noxious
keeping function is important for intestinal health, since in odors, visual stimuli, somatic pain, or unpleasant tastes.
its absence, large quantities of indigestible materials called Peripheral afferents can also trigger a vomiting response, such
bezoars may accumulate and even obstruct the lumen, par- as when irritants are present in the gastric lumen, or if the
ticularly in the stomach. intestine is obstructed. Some vagal afferents originating in the
Plasma motilin concentration, pg/ml

800

Feeding
600

400

200

FIGURE 54–11 The migrating motor


0 complex as assessed in a dog, followed by
the motility pattern in the fed state. Each
Gut motor pattern

Gastric antrum
antral phase III complex is accompanied by
Upper jejunum an increase in plasma motilin, whereas motilin
Mid-intestine release is suppressed after feeding. (Adapted from
Itoh Z, et al. Changes in plasma motilin concentration and
gastrointestinal contractile activity in conscious dogs.
Time intervals, 1 hr Am J Dig Dis. 1978;23(10):929–935.)
552 SECTION VIII GI Physiology

Programmed Pain
Pharyngeal vomiting Sights
stimulation response Anticipation

Glossopharyngeal Higher
nerve centers

Nucleus Brain stem


tractus solitarius vomiting center

Vagus nerve Area postrema Cerebellum


chemoreceptor
trigger zone

Labyrinth
Gastric
mucosa
Drugs
FIGURE 54–12 Neural pathways eg, opiates, chemotherapy
leading to the initiation of vomiting in Hormones Motion
response to various stimuli. (Reproduced with eg, pregnancy Vertigo
permission from Barrett KE, Barman SM, Boitano S, Ipecac
Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. Cytotoxic drugs
McGraw-Hill Medical, 2009.)
Irritants

stomach, and presumably linked to chemoreceptors, project to entirely dependent on input from extrinsic nerves, coordinated
the area postrema. by the brain centers that also regulate the functions of somatic
A second brain region, the nucleus tractus solitarius, also muscles that support vomiting, as described earlier.
contributes to the emetic cascade, particularly following vagal
activation (Figure 54–12). This region receives inputs from the
area postrema, abdominal vagus, and labyrinths, and in turn BASIC PRINCIPLES OF
coordinates the motor responses required, which are a stereo-
typical program of somatic muscle actions. First, the thoracic, INTESTINAL MOTILITY
diaphragmatic, and abdominal muscles contract concurrently
against a closed glottis, resulting in the phenomenon of retch- ROLE AND SIGNIFICANCE
ing. The high positive intra-abdominal pressure also forces the IN THE SMALL INTESTINE
gastric contents into the esophagus. The brain then coordi-
nates the synchronous contraction of inspiratory and expira- The primary role of the small intestine is to digest the vari-
tory muscles, resulting in a reversal of the thoracic pressure ous components of the meal and to absorb the resulting
gradient. This high positive thoracic pressure acts in turn to nutrients into the bloodstream or lymphatic system. The
drive expulsion of the vomitus. Simultaneously, respiration is motility patterns observed in the small intestine are pro-
suppressed, and movement of laryngeal and pharyngeal struc- foundly altered by eating, with the duration of such changes
tures prevents aspiration and, usually, passage of vomitus into depending on the caloric load. During the fed state, the motil-
the nasal cavity. ity patterns in the small intestine are not designed primar-
Intestinal motility is also regulated during vomiting. The ily to propel the intestinal contents aborally, but rather to
BER is suspended and replaced by bursts of electrical activity mix them with digestive secretions and prolong their expo-
that propagate orally. These result in a motility pattern referred sure to the absorptive epithelium. The speed with which the
to as a retrograde giant contraction, or the retroperistaltic contents are propelled also varies along the length of the
contractile complex, that moves the gastric contents up and out small intestine. Movement is fastest in the duodenum and
of the esophagus. Despite the autonomy of the enteric nervous jejunum, providing for rapid mixing and propulsion of the
system in producing normal patterns of gastric and esophageal contents both orally and aborally. Motility then slows in the
motility, the retrograde propulsion seen during emesis is ileum, providing additional time for the absorption of slowly
CHAPTER 54 Intestinal Motility 553

permeable nutrients, and particularly, lipids. Then, once the junctions, and by the activity of interstitial cells of Cajal.
meal is digested and absorbed, the small intestine converts to These latter pacemaker cells undergo rhythmic cycles of
the MMC to expel undigested residues through the small depolarization, and dictate the BER, or slow waves, that ulti-
intestine and into the colon. mately control the rate and locations of phasic contractions of
the smooth muscle.
ROLE AND SIGNIFICANCE IN THE COLON The large intestine also contains both circular and longitu-
dinal muscle layers that regulate its motility, but the ana-
The functions of the colon are quite distinct from those of the tomic arrangement of these differs somewhat from that seen
small intestine, serving primarily to extract and reclaim water in the small intestine. In the ascending, transverse, and
from the intestinal contents, and process the feces for elimina- descending colon, the circular muscle layer is overlaid by
tion. As a result, even in the fasted state, the motility functions three long nonoverlapping bands of longitudinal muscle ori-
of the colon are considerably more biased toward mixing the ented at 120° to each other, known as the taenia coli. Electri-
contents and retaining them for prolonged periods, and the cal coupling between the circular muscle and taenia coli is
colon does not participate in the MMC. On the other hand, less effective than that between the corresponding muscle
periodically, large propulsive contractions sweep through the layers in the small intestine, which likely contributes to less
colon, transferring its contents to the rectum and ultimately effective propulsive motility in the colon. The circular mus-
promoting the urge to defecate. cle layer is also contracted intermittently to divide the colon
into functional segments known as haustra. Note that the
haustral segments are not permanent structures, however,
FUNCTIONAL ANATOMY and thus they can be smoothed out to permit propulsion of
the colonic contents.
MUSCLE LAYERS In the sigmoid colon and rectum, the intestine becomes
completely enveloped by longitudinal muscle that is impor-
The layer of circular muscle found closest to the mucosa, over- tant to the specialized functions of this region, which include
laid by a longitudinal muscle layer, produces the stereotypical serving as a reservoir and participating in defecation. The
motility patterns of the small intestine. A thin layer of muscle lumen of the rectum is also partially occluded by transverse
sandwiched between the mucosa and submucosa, the muscu- folds, again formed by muscular contraction, which act as
laris mucosa, also confers specific motility functions on shelves to retard the passage of fecal material (Figure 54–13).
mucosal structures, such as the villi. Finally, the most distal portion of the gastrointestinal tract,
The functions of the circular and longitudinal muscle lay- the anal canal, is a specialized region that contains both
ers are closely integrated by electrical coupling through gap smooth and striated muscles in its walls.

Sigmoid colon

Rectosigmoid junction
Rectum
Rectal valves

Muscle layers
making up internal Internal anal sphincter
and external
anal sphincters FIGURE 54–13 Anatomy of the rectum and
External anal sphincter anal canal. (Reproduced with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical Books/
Anal canal McGraw-Hill, Medical Pub. Division, 2006.)
554 SECTION VIII GI Physiology

ENTERIC NERVOUS SYSTEM FEATURES OF


The primary determinant of motility function in both the INTESTINAL MOTILITY
small and large intestines derives from the activity of intrinsic
neural circuits. The number of intrinsic nerves vastly exceeds FED VERSUS FASTED
that of extrinsic inputs, and the role of the latter is normally PATTERNS OF MOTILITY
felt to be restricted largely to modulating motility patterns
established by the “little brain” of the enteric nervous system. For the small intestine, there is a marked distinction between
Enteric nerves are responsible for both contraction and relax- motility observed in the postprandial versus fasting periods;
ation. The major stimulatory neurotransmitters include ACh, however, colonic motility has far less temporal association
neurokinin A, and substance P, whereas the inhibitory nerves with the ingestion of a meal. These differences are mirrored by
express VIP and also produce nitric oxide on activation. the times taken for luminal contents to transit these two
There is also an abundant supply of sensory afferents that intestinal segments. In the small intestine, substances move
respond to the physicochemical characteristics of the luminal from the mouth to the ileocecal valve in a little under 2 hours in
contents. healthy adults, on average, with transit occurring most rapidly
Modulatory influences of extrinsic nerves derive from a proximally. Transit is slowed in proportion to the number of
variety of sources depending on the intestinal segment in calories presented to the intestine. In the colon, on the other
question. The vagus (parasympathetic) and splanchnic hand, transit from the cecum to the rectum may take 1–2 days
(sympathetic) nerves innervate the small intestine, ileoce- on average, with considerable interindividual variability.
cal valve, and proximal colon. The pelvic nerves, on the During fasting, the small intestine exhibits the MMC. When
other hand, are the conduits of extrinsic input to the the meal has emptied from the small intestine, the MMC
remainder of the colon and the internal anal sphincter. resumes with its characteristic three phases (Figure 54–14). As
Finally, the pudendal nerves provide input from the sacral in the stomach, the purpose of the MMC, and of phase III in
region of the spinal cord to the external anal sphincter and particular, appears to be to sweep the intestine clear of any
the muscle layers of the pelvic floor. In fact, unlike the other remaining residues of the meal. The MMC may additionally
gastrointestinal regions discussed earlier, voluntary input play a role in limiting reflux of colonic contents into the ileum
to these latter structures is vital to their function. The abil- since it does not propagate into the colon in humans.
ity to contract the external anal sphincter and pelvic floor After a meal is ingested, motility events in the small intes-
muscles, a behavior learned during toilet training, allows tine become more frequent. The fed pattern of motility rests
the deferral of defecation until a time when it is socially on the BER, generated by the intestinal pacemaker and propa-
convenient. gated to surrounding smooth muscle cells. As in the stomach,
however, the basal rhythm supplies only the nodes at which
contractions can occur at any given moment, because the
SPHINCTERS rhythmic changes in membrane potential that are induced in

The passage of contents along the length of the small intestine


and colon is also regulated by sphincters. The ileocecal valve,
or junction, reflects activity of the circular muscle layer. The D1
primary function of the ileocecal valve is to limit reflux of
colonic contents into the ileum. The elimination of waste
D2
matter from the colon is under the control of the internal and
external anal sphincters. The internal anal sphincter consists
of a thickened band of gastrointestinal circular muscle J1
(Figure 54–13). This sphincter supplies approximately
70–80% of the tone of the anal canal at rest, and its regulation J2
is entirely autonomous. If the rectum is suddenly distended,
the sphincter relaxes and then contributes only 40% of anal J3
tone, with the remainder supplied by the external anal 30 min
sphincter (which is composed of striated muscle). At the
same time, the external anal sphincter pressure is increased. FIGURE 54–14 Migrating motor complexes in the duodenum
and jejunum as recorded from a fasting human subject by
This rectoanal inhibitory reflex, initiated by rectal disten-
manometry. D1, D2, J1, J2, and J3 indicate recording points along the
sion, thus allows for efficient defecation while preventing length of the duodenum and jejunum. Note that the intense
accidental leakage. After a short period of time, however, the contractions occurring rhythmically during phase III propagate
internal anal sphincter accommodates to the new rectal vol- aborally. (Reproduced with permission from Soffer EE et al: Prolonged ambulatory
ume and regains its tone, unless defecation can conveniently duodeno-jejunal manometry in humans: normal values and gender effect. Am J
be completed. Gastroenterol 1998;93:1318–1323. Copyright American College of Gastroenterology.)
CHAPTER 54 Intestinal Motility 555

the muscle cells are insufficient to cause contraction. Rather, it of passage of the food bolus along the wall of the gut or sec-
is only when the effects of neurotransmitters are superimposed ondary to intestinal distension. Rapid stretch of the intestine is
on this rhythm that action potentials can take place. The result most effective in triggering peristalsis.
is a series of intermittent phasic contractions occurring along As in the esophagus, where peristalsis is important in
the length of the small intestine, peaking 10–20 minutes after moving the bolus from mouth to stomach, intestinal peri-
eating. ACh from the enteric nervous system is a critical medi- stalsis involves the influences of both an ascending contrac-
ator of such effects, with any role for hormonal mediators tion and a descending relaxation (Figure 54–15). Activation
much less clear. of stretch and possibly other mechanoreceptors in the
mucosa secondarily induce the release of 5-HT and calci-
tonin gene–related peptide. On the proximal side of the
MIXING AND SEGMENTATION bolus, the circular muscle shortens and the longitudinal
muscle relaxes, pushing the bolus forward. These responses
During the fed state, the primary motility events serve to mix have been attributed to the action of ACh and substance P
the contents, and to propel them slowly, if at all. An isolated from enteric nerve endings. On the distal side, the bolus is
contraction in the absence of others either proximal or distal received by a segment of intestine of increased caliber,
to it will have the effect of mixing the contents of the lumen in brought about by shortening of the longitudinal muscle and
the immediate vicinity of the contraction (Figure 54–15). relaxation of the circular muscle. These responses relate to
Another common pattern is referred to as segmentation. Seg- the activity of VIP and nitric oxide.
menting contractions serve to move intestinal contents back
and forth within a short segment of bowel. The complex pat-
terns of motility in the small intestine after a meal are the COLONIC MOTILITY
result of the almost autonomous effects of the enteric neural
circuitry, and presumably reflect a stereotypical “programmed” The motility patterns of the colon are primarily concerned
response to a given set of physiological conditions. with mixing the contents and retaining them for a sufficient
period to allow optimal salvage of the fluid utilized during the
digestive process. The colon has the capacity to reabsorb even
PERISTALSIS unusually large quantities of fluid, provided adequate contact
time with the mucosa occurs. However, periodically the colon
Peristalsis produces aboral propulsion in both the small intes- also engages in a propulsive motility pattern that essentially
tine and colon. It is a motility response that occurs in response moves the majority of the colonic contents into the rectum. In
to deformation of the mucosa, either via the mechanical effects turn, this induces the need to defecate, which will be dealt
with later.
During mixing, the colon shuttles contents back and forth
Isolated contraction between its haustra, and progressively propels contents from
one haustra to the next in a motility pattern referred to as seg-
mental propulsion. These patterns are accomplished by two
Segmentation
types of contraction that have been characterized in the colon,
short- and long-duration contractions. Short-duration con-
tractions arise in the circular muscle, and are stationary pres-
sure waves lasting approximately 8 seconds on average, which
effect local mixing. Long-duration contractions, on the other
hand, last for 20–60 seconds and may be stationary or may
Peristalsis
propagate for a short distance, and are attributed to contrac-
tion of the longitudinal muscles of the taenia coli. Propagating
contractions account for segmental propulsion.
Contraction High-amplitude propagating contractions are distinct
Relaxation from the motility patterns just described. They propagate
exclusively aborally, and provide for mass movement of the
FIGURE 54–15 Patterns of intestinal mixing and propulsion. feces over long distances. While they precede the urge to def-
An isolated contraction moves contents both orally and aborally.
ecate, they in fact occur about 10 times per day, and are associ-
Segmentation mixes contents over a short length of intestine, as
ated with rising in the morning and with eating. These
indicated by the time sequence from left to right. In the diagram on
the left, the vertical arrows indicate the points at which the next set contractions originate in the cecum and sweep throughout the
of contractions is initiated. Finally, peristalsis, which involves both a colon to the rectum, also resulting in relaxation of the internal
contraction and a relaxation, propels the luminal contents aborally. anal sphincter. The propagation of these contractions is likely
(Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: mediated by both cholinergic and neurokinin-dependent
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) pathways.
556 SECTION VIII GI Physiology

DEFECATION Difficulty in swallowing is referred to as dysphagia, and


can result from abnormalities in any of the components of
Defecation is the process of elimination of solid wastes from
the swallowing reflex or the anatomic structures involved.
the gastrointestinal tract, and it involves several structures in
For example, abnormalities of the tongue can result in dys-
and around the rectum and anus. Filling of the rectum causes
phagia because the bolus cannot be propelled backward
relaxation of the internal anal sphincter via the release of VIP
toward the pharynx with sufficient force. In general, dys-
and nitric oxide from intrinsic nerves in the rectoanal inhibi-
phagia can be considered as arising from either the orophar-
tory reflex, but this response is offset by a simultaneous action
ynx and striated muscle region of the esophagus or the
to increase the tone of the external anal sphincter. Overall, this
smooth muscle portion of the esophagus, corresponding to
reflex can permit efficient defecation while preventing leak-
the different innervation and mechanisms of sensation and
age. The portion of rectal contents that enters the anal canal is
control in these two areas. Likewise, dysphagia can result
also identified as being gaseous, solid, or liquid, thereby initi-
from either structural or functional causes. Dysphagia is a
ating appropriate activity of the external anal sphincter to
common medical problem that is especially frequent in the
retain each of these, or to permit voluntary expulsion.
elderly, and associated with much distress, as well as the risk
When defecation is desired, on the other hand, adopting a
of aspiration, choking, and malnutrition. It is estimated that
sitting or squatting position changes the relative orientation of
up to 13% of hospitalized patients and as many as 60% of
the intestine and surrounding muscular structures to straighten
nursing home residents have feeding problems, most of
the pathway for fecal exit. This is also assisted by relaxation
which are the result of oropharyngeal dysphagia. All
of the puborectalis muscle, which results in a less acute rec-
patients with dysphagia will experience problems with solid
toanal angle. Rectal contraction then produces the propulsive
food, and may have varying degrees of difficulty swallowing
force to move the feces out of the body. Evacuation is enhanced
liquids as well, depending on the severity of the underlying
by simultaneous contraction of the rectus abdominus, dia-
cause. Structural causes of dysphagia extend to diverticula,
phragm, and other levator ani muscles, which increases
or outpouchings of the pharyngeal or esophageal wall in
intra-abdominal pressure. All of these events occur whether
which food can become trapped, or to various forms of
solid (in health) or liquid (in disease) feces are expelled,
obstruction. The latter include mucosal or muscular rings
although less force is obviously needed to evacuate liquid feces.
that circumferentially occlude a portion of the esophageal
On the other hand, the voluntary expulsion of flatus involves
lumen. These can occur in response to long-standing tissue
the contractile functions listed, but the puborectalis muscle
injury secondary to gastroesophageal reflux disease
does not relax and there is no change in the rectoanal angle.
(GERD); the inflammation eventually leads to scarring and
This allows the flatus gas to be forced past the acutely angled
fibrosis that may occlude the lumen. Esophageal tumors
anorectum without the simultaneous loss of feces.
can also impede passage of esophageal contents. Functional
causes of dysphagia relate to either neurological control of
the oropharyngeal phase of swallowing, peristalsis, and
esophageal sphincter relaxation or defects in the muscle
CLINICAL CORRELATION layers themselves. Treatment of dysphagia depends on the
underlying cause. When there are structural abnormalities,
A 90-year-old, bedridden male resident in a nursing surgery to repair diverticula, cut overly tight muscles, or
home is given his daily oral medications (pills). He later remove an obstructing tumor can often bring some relief.
regurgitates a bedtime snack and complains of a feeling Mechanical dilation of a stricture (abnormal narrowing) is
that the pill “did not go all the way down,” so is taken to also attempted, with varying degrees of success. In the case
the emergency room. On physical examination, he is of functional disorders, on the other hand, effective therapy
found to be somewhat underweight, agitated, and clearly usually depends on whether treatment is available for the
distressed. He is also coughing and retching. The aide underlying disorder, and surgery is much less helpful.
that accompanied him reports that this situation has
occurred previously. A gastroenterologist is called and
performs an upper endoscopy, whereupon the pill can
be seen lodged in the mid-esophagus. It is removed via CHAPTER SUMMARY
the esophagus and the patient’s symptoms slowly subside. ■ Regulation of swallowing involves somatic neurotransmission
The gastroenterologist advises the nursing home aide in the upper third of the esophagus, composed of striated
that the patient should be given only semisolid food in muscle, and autonomic regulation via the vagus and enteric
future, and that any needed pills should be crushed and nervous system in the lower two thirds, composed of smooth
mixed with apple sauce to administer them to the patient. muscle. Swallowing is initiated voluntarily, but thereafter
Despite these measures, the patient’s problems worsen, reflects a complex integration of regulatory influences
and he is eventually admitted to the hospital suffering coordinated by the swallowing center in the brain.
from pneumonia. ■ Two sphincters, normally closed, regulate the movement of the
bolus into and out of the esophagus. The upper esophageal
CHAPTER 54 Intestinal Motility 557

sphincter is opened in concert with pharyngeal motility. The 2. A 50-year-old man who is markedly overweight comes to his
LES opens to allow the bolus to enter the stomach, coordinated primary care physician complaining that he suffers nightly from
with esophageal motility. a burning sensation in his chest after retiring, which is made
■ The stomach serves to receive the meal from the esophagus, worse if he has had a snack close to bedtime. Which of the
and it displays motility functions that both initiate the process following would be the most appropriate treatment for this
of digestion and control the delivery of nutrients to more distal patient if his symptoms are not resolved by weight loss and
segments. eliminating nighttime meals?
■ Receptive relaxation of the proximal stomach allows the A) cholinergic agonist
stomach to function as a reservoir and ensures that the B) smooth muscle relaxant
pressure within the stomach changes little as its volume C) nitric oxide donor
expands to receive the meal. D) nicotinic agonist
■ The distal stomach uses phasic contractions to grind the E) proton pump inhibitor
meal, moving only the smallest particles to the pylorus. 3. In an experiment, a balloon is inserted into the stomach of a
■ Emptying of the stomach involves tonic contractions of the human volunteer and gradually inflated while intraluminal
proximal portions, and depends on both the physical and pressures are monitored. Although the volume of the balloon
chemical characteristics of the meal. Liquids empty most increases considerably, pressures remain relatively constant.
rapidly; solids empty only after a lag phase. Nutrients and the This remarkable pressure–volume relationship is thought to
osmolarity of the meal feed back to retard gastric emptying involve release of which of the following patterns of
once they reach the small intestine via both neural and neurotransmitters?
humoral mechanisms.
Vasoactive Intestinal
■ Phase III of the MMC results in large contractions that
Acetylcholine Polypeptide Nitric Oxide
propagate aborally, while the pylorus relaxes maximally,
allowing the exit of even large particles. This phase of the A) Yes Yes Yes
MMC is related to release of the GI hormone motilin.
B) Yes Yes No
■ Motility patterns in the small and large intestines serve not
only to propel intestinal contents, but also to mix them with C) No Yes Yes
enzymes and other digestive juices, and to retain them in a D) No Yes No
given segment long enough for optimal absorption to occur.
E) Yes No Yes
■ The colon serves predominantly salvage and reservoir
functions, with slow transit of contents along its length and
marked dehydration of luminal contents. 4. A mother brings her 2-year-old child to the emergency room,
■ Movement of colonic contents out of the body is controlled by distressed because he has swallowed a quarter while the family
the internal and external anal sphincters, under involuntary was eating dinner at a restaurant. The physician reassures her
and voluntary control, respectively. that the quarter, which can be plainly seen in the stomach by
fluoroscopy, will eventually pass in the stool. What physiological
■ Periodically, large propulsive contractions sweep through the
condition or response will be required to permit exit of the
colon and precede the urge to defecate.
quarter from the stomach?
A) receptive relaxation
B) fasting
STUDY QUESTIONS C) eating another meal
1. In a study of the control of esophageal motility, a scientist instills D) mixing and grinding by the stomach
a small amount of dilute hydrochloric acid into the upper third of E) relaxation of the LES
the esophagus of a human volunteer, using an endoscope. This
treatment is most likely to produce which of the following
responses?
A) peristalsis
B) retroperistalsis
C) esophageal spasm
D) relaxation of the upper esophageal sphincter
E) no response
558 SECTION VIII GI Physiology

5. Four medical students studying for their physiology final develop 6. Which of the following substances is not involved in mediating
headaches and take either regular or enteric-coated aspirin with the fed pattern of intestinal motility?
either milk or water (enteric-coated pills will not dissolve until A) ACh
the pH is neutral). Assuming headache relief is proportional to B) VIP
blood aspirin concentrations, place the following conditions in C) 5-hydroxytryptamine (serotonin)
order of headache relief (from fastest to slowest): D) nitric oxide
1. regular aspirin with water; E) motilin
2. enteric-coated aspirin with water;
3. regular aspirin with milk;
4. enteric-coated aspirin with milk.
A) 1 > 2 > 3 > 4
B) 4 > 3 > 2 > 1
C) 1 > 3 > 2 > 4
D) 2 > 4 > 1 > 3
E) 2 > 4 > 3 > 1
55
C H A P T E R

Functional Anatomy
of the Liver and Biliary
System
Kim E. Barrett

O B J E C T I V E S

■ Understand the role of the liver in whole body homeostasis and the structural
features that subserve its functions.
■ Understand the functions of bile secretion and the anatomy of the biliary
system.
■ Describe the unusual circulatory features of the liver and the relationship
of blood flow to bile flow.
■ Identify the parenchymal and nonparenchymal cell types of the liver, their
anatomic relationships, and their respective functions.

OVERVIEW OF THE LIVER AND many important biochemical compounds from the intermedi-
ate products of carbohydrate metabolism. Many of the sub-
BILIARY SYSTEMS AND THEIR strates for these reactions derive from the products of
carbohydrate digestion and absorption that travel directly to the
FUNCTIONS liver from the gut, as will be described in more detail in Chapter
The liver is the largest organ in the body, and conducts a myr- 58. As a consequence, the liver plays a major role in maintaining
iad of vital metabolic and excretory functions. In addition, by blood glucose concentrations within normal limits, particularly
virtue of its circulatory relationship to the absorptive surface in the postprandial period (see Chapter 69). The liver removes
of the gastrointestinal tract, the liver is the initial site where excess glucose from the blood and returns it as needed, in a pro-
most ingested nutrients, and other substances entering via cess referred to as the glucose buffer function of the liver. The
the gastrointestinal tract, are processed by the body. Thus, the liver also contributes in a major way to fat metabolism. While
liver is a gatekeeper that can process useful substances while many aspects of lipid biochemistry are common to all cells of
detoxifying orally absorbed substances that are potentially the body, others are concentrated in the liver. Specifically, the
harmful. liver supports an especially high rate of oxidation of fatty acids
to supply energy for other body functions. Likewise, the liver
converts amino acids and two-carbon fragments derived from
METABOLISM AND DETOXIFICATION carbohydrates to fats that can then be transported to adipose
tissue for storage. Finally, the liver synthesizes most of the lipo-
The liver contributes in a pivotal way to the biochemical status proteins required by the body, as well as large quantities of cho-
of the body as a whole. It is beyond the scope of this text to lesterol and phospholipids. The liver also serves to detoxify the
provide a comprehensive analysis of all of the metabolic func- blood of substances that originate from the gut or elsewhere in
tions of the liver. Instead, we will focus our discussion on the body. It is highly active in removing particulates from the
broad categories of metabolic functions of the liver that are portal blood, such as small numbers of colonic bacteria that
relevant to the function of the gastrointestinal system, or are of cross the wall of the intestine under normal circumstances. The
particular importance to whole body homeostasis. majority of this “blood cleansing” is provided for by specialized
First, the liver performs four specific functions in carbohy- cells related to blood macrophages, known as Kupffer cells.
drate metabolism: glycogen storage, conversion of galactose and These are highly effective phagocytes that are strategically
fructose to glucose, gluconeogenesis, and the formation of located to be exposed to the majority of the blood flow

559

Ch55_559-564.indd 559 11/26/10 10:29:27 AM


560 SECTION VIII GI Physiology

originating from the gut. Other detoxification functions of the may increase to almost 90% in the period immediately after a
liver are biochemical in nature. Hepatocytes express large num- meal. A schematic diagram of the splanchnic circulation is pro-
bers of cytochrome P450 and other enzymes that can convert vided in Figure 55–1. At a microscopic level, blood perfuses the
xenobiotics (foreign chemicals) to inactive, less lipophilic liver via a series of sinusoids, which are low-resistance cavities
metabolites that can subsequently be excreted into the bile and that receive blood supply both from branches of the portal vein
thereby eliminated from the body. In addition to the metabo- and from the hepatic artery. At rest, many of these sinusoids
lism of xenobiotics, the liver is responsible for the metabolism are collapsed, whereas as portal blood flow to the liver increases
and excretion of a wide variety of hormones and other endoge- coincident with ingestion and absorption of a meal, sinusoids
nous regulators that circulate in the bloodstream. In particular, are gradually recruited to allow the perfusion of the liver with
the liver is responsible for metabolism of the steroid hormones. a much greater volume per unit time but only a minimal
increase in pressure. The liver also has a distinctive morpho-
logic organization that underpins its functions. This organiza-
PROTEIN METABOLISM tion is based on the so-called hepatic triad of branches of the
AND SYNTHESIS portal vein, the hepatic artery, and the bile ducts. Blood flows
into a branch of the portal vein in the center of portal areas,
The liver is also critical for protein metabolism, and the body which are linked by anastomosing cords of cuboidal hepato-
cannot dispense with the liver’s capacity to contribute to pro- cytes to a central venule that in turn drains into the hepatic
tein processing for more than a few days. The liver contributes vein. Branches of the hepatic artery likewise run close to the
the following important aspects of protein metabolism: deam- bile ducts, and likely play an important role in supplying energy
ination of amino acids, formation of urea as a means to dis-
pose of blood ammonia, formation of plasma proteins, and
interconversion of various amino acids, as well as conversion
of amino acids to other intermediates important in the body.
Heart
Likewise, the liver can synthesize all of the nonessential amino
acids that need not be supplied in the diet in their native form Vena
(as will be discussed in more detail in Chapter 58). cava Hepatic veins
The liver also synthesizes proteins that are critical to the cir- 1300 mL/min
500 mL/min
culatory system. With the exception of the immunoglobulins
produced by cells of the immune system, the liver provides t er y * Liver
ar
ic
most of the plasma proteins. Likewise, the liver is also the major
t
pa
He

700 mL/min
site of synthesis of proteins that contribute to blood clotting.
Celiac artery
Spleen
EXCRETION OF LIPID-SOLUBLE

Portal vein
WASTE PRODUCTS Stomach

Pancreas
The liver handles excretion of lipophilic molecules that do not
700 mL/min
enter the renal filtrate, and excretes them in the bile. In turn,
Aorta

Small
the biliary system is designed to convey these substances out intestine
Superior
of the liver and into the intestinal lumen, where they undergo mesenteric artery
little, if any, reabsorption and thus can be eliminated from the Colon
body in the feces. 400 mL/min

ENGINEERING CONSIDERATIONS
Inferior
BLOOD SUPPLY mesenteric artery

Hepatic Macrocirculation Rest of


body
and Microcirculation *Branches of the hepatic artery also supply the stomach,
The liver is unusual in that it receives the vast majority of its pancreas and small intestine
blood supply in the form of venous blood, especially in the FIGURE 55–1 Schematic of the splanchnic circulation under
postprandial period. Even at rest, blood flow to the liver via the fasting conditions. Note that even during fasting, the liver receives
portal vein is at a rate of 1,300 mL/min, compared with only the majority of its blood supply via the portal vein. (Reproduced with
500 mL/min supplied by the hepatic artery. Moreover, the permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of
proportion of blood flow supplied to the liver by the portal vein Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
CHAPTER 55 Functional Anatomy of the Liver and Biliary System 561

Hepatic
Portal vein Liver
Central vein artery
Bile ducts
Sinusoids

Gallbladder
Duodenum

Bile duct

Portal vein

Terminal ileum

FIGURE 55–3 Schematic of the enterohepatic circulation of


conjugated bile acids. Bile acids secreted by hepatocytes enter the
bile and flow through the biliary system to the duodenum.
Conjugated bile acids are selectively reabsorbed in the terminal
Hepatic artery ileum, and flow through the portal vein back to the liver to be
Portal vein Bile duct
reabsorbed by hepatocytes and resecreted. (Reproduced with permission
FIGURE 55–2 Arrangement of blood vessels, bile ducts, from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical Books/
and hepatocytes to form the liver lobule. Branches of the portal McGraw-Hill, Medical Pub. Division, 2006.)
vein and hepatic artery run parallel to bile ducts in the so-called
portal triads. Blood percolates through sinusoids arranged between
the hepatocytes, to be collected eventually in the central vein. Hepatocytes
(Reproduced with permission from Ross MH, Reith EJ: Histology. A Text and Atlas.
New York: Harper and Row, 1985.)
Hepatocytes are the metabolic “factories” of the liver, and are
responsible for most of its characteristic functions. They are
highly specialized polarized epithelial cells. Their apical mem-
and nutrients to the bile duct epithelial cells to support their branes are in the form of grooves between adjacent cells,
transport functions. A diagram showing the interrelationships known as canaliculi (Figure 55–4). The canaliculi form a
of the various cell types that make up the liver is shown in
Figure 55–2.
SINUSOIDAL LUMEN
Stellate
Enterohepatic Circulation cell
Tight junction
The circulatory features of the liver are also notable for the fact
that some substances circulate continuously between the liver
and intestine, in the enterohepatic circulation. This involves Bile canaliculus
passage of solutes through three different environments—the
portal vein and the sinusoids into which it empties, the biliary
system, and the intestinal lumen (Figure 55–3). Most notably, Hepatocyte
this occurs for bile acids that are utilized during intestinal
lipid digestion and absorption. The physiological significance
Endothelium
of this circuit is that it permits the secretion rate to greatly
Kupffer cell
exceed the synthesis or input rate.
SINUSOIDAL LUMEN
Space of
HEPATIC PARENCHYMA Disse
FIGURE 55–4 Interrelationships of the major cell types
AND SINUSOIDS making up the liver. Hepatocytes are arranged in plates joined by
tight junctions, and their apical membranes make up the bile
The most prevalent cell type in the liver is the hepatocyte
canaliculi. They are segregated from the blood-filled sinusoids by
(80% of total cells, approximately 100 billion in an adult human fenestrated endothelial cells without a basement membrane, and by a
liver), whereas the nonparenchymal cell types include the stel- loose connective tissue layer known as the space of Disse. Kupffer cells
late cells, sinusoidal endothelial cells, and previously men- reside in the sinusoidal lumen, whereas stellate cells are found in the
tioned Kupffer cells. In this section, we will review how their space of Disse. (Reproduced with permission from Barrett KE: Gastrointestinal
properties contribute to the physiological functions of the liver. Physiology. New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
562 SECTION VIII GI Physiology

continuous network that drains eventually into the bile From liver
ductules. On the opposing pole of the hepatocyte, the basolat-
Right and left hepatic
eral membrane faces the bloodstream in the form of the ducts from liver
hepatic sinusoids. The apical and basolateral membranes of Cystic duct
hepatocytes are separated by tight junctions that define the
canaliculi. These junctions are relatively permeable, however, Common hepatic duct
permitting the passage of glucose and other small solutes. Common bile duct
Gallbladder
Pancreatic duct
Kupffer Cells
Kupffer cells arise from the macrophage lineage, and line the
sinusoidal epithelium on the bloodstream side (Figure 55–4). Sphincter of Oddi
They are presumed to play a major role in host defense. Their Duodenum
location is such that they are exposed to virtually all of the
portal blood flow. Kupffer cells also express cell-surface recep-
tors for altered proteins, such as Fc immunoglobulin receptors FIGURE 55–5 Functional anatomy of the biliary system.
that can be used to internalize foreign proteins or microorgan- (Modified with permission from Barrett KE: Gastrointestinal Physiology. New York:
isms that have been coated with host antibodies. Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)

Sinusoidal Endothelium system is depicted in Figure 55–5. Bile drains from the liver via
The endothelial cells that line the hepatic sinusoids have two the right and left hepatic ducts that join to form the common
characteristic properties that distinguish them from endothe- hepatic duct. A cystic duct diverts bile for storage into the gall-
lial cells in other organs of the body. First, they are perforated bladder. The anastomosis of the common hepatic duct and the
by large intracellular pores known as fenestrae, which are cystic duct forms the common bile duct, which transfers bile to
100–200 nm in diameter. These are designed to permit the the sphincter of Oddi. At a functional level, the biliary system
passage of even quite large macromolecules out of the blood, can be divided into four components. First, the canaliculi, which
including albumin with bound ligands. Second, sinusoidal are composed of the adjacent apical membranes of hepatocyte
endothelial cells in the healthy liver do not have a basement couplets, form the initial biliary secretion. This secretion is then
membrane. In total, therefore, the sinusoidal endothelium modified as it flows along the biliary ductules, which are anal-
presents virtually no barrier to the efflux of albumin and other ogous to pancreatic ducts. The ductules are made up of colum-
similarly sized molecules from the vascular space. nar epithelial cells (cholangiocytes), and both absorb and
secrete various substances into and out of the bile. The ductules
are perfused by a capillary network arising from the hepatic
Space of Disse artery, rather than from the sinusoids. The majority of this peri-
The space of Disse (pronounced Diss-eh) is a layer of loose ductular capillary plexus drains into the sinusoids. Flow in the
connective tissue that lies between the sinusoidal endothelium periductular capillary plexus is in the opposite direction to bile
and the basolateral membrane of hepatocytes. It is notably flow. The larger bile ductules dilute and alkalinize the bile, again
highly permeable to the bidirectional exchange of solutes analogous to the function of the pancreatic ducts.
between the sinusoidal blood flow and hepatocytes. The bile ducts serve simply as conduits for the bile without
modifying its composition significantly, other than by adding
Hepatic Stellate Cells mucus. Mucus secretion presumably serves to protect the
ductular epithelium from potentially injurious surfactant
Hepatic stellate cells, previously also referred to as Ito cells,
effects of the bile itself. Finally, bile is stored in the gallbladder
are star-shaped cells that reside in the space of Disse. They
between meals, which is a blind sac lined by highly absorptive
play an important role in the normal liver by storing a variety
epithelial cells linked by well-developed tight junctions. The
of lipids. In addition, these cells are contractile, and may be
gallbladder serves not only to store bile, but also to concen-
involved in the regulation of sinusoidal diameter. Stellate cells
trate it. However, the gallbladder is not essential to life and can
also play a critical role in liver injury by producing extracellu-
be removed without compromising nutrition.
lar matrix materials, such as collagen. This collagen is depos-
ited in the space of Disse and impairs hepatic function.

BILIARY TRACT AND GALLBLADDER CLINICAL CORRELATION


A 70-year-old veteran is seen in the primary care clinic of the
The third functional division of the liver is concerned with the
VA hospital. He complains that his girth has increased pro-
production and transport of bile out of the liver and into the
gressively over a period of several months, despite attempts
gastrointestinal lumen. The functional anatomy of the biliary
CHAPTER 55 Functional Anatomy of the Liver and Biliary System 563

to lose weight. His ankles are also noted to be edematous. His CHAPTER SUMMARY
history is notable for service in Korea, where he reports that ■ Functions of the liver and biliary system include glucose
he was diagnosed with acute “liver disease” that subsequently storage and release, protein synthesis, detoxification of
resolved. A needle is inserted into his peritoneal cavity and xenobiotics and ammonia, metabolism of endogenous
several liters of tan-colored fluid drain out. A blood test hormones, initial handling of substances absorbed from
reveals evidence of past exposure to the hepatitis C virus. He the intestine, and excretion of lipophilic molecules and
heavy metals in the bile.
is placed on interferon therapy, and told to return to the GI
■ Functions of the liver are facilitated by its unique circulatory
clinic in 6 weeks for follow-up, but 4 weeks later he presents
features. Blood arrives at the liver via two routes: the portal
at the emergency room vomiting bright red blood. An emer-
vein, which drains blood from the intestine, and the hepatic
gency endoscopic examination reveals a ruptured blood ves- artery.
sel bleeding into his esophagus. Hemostasis is obtained by
■ Blood percolates through the liver via a low-resistance system
placing a band around the bleeding vessel. of sinusoids that maximize exposure of hepatocytes to the
Portal hypertension refers to conditions where the resis- blood’s contents.
tance to blood flow through the liver is increased, which ■ The functions of the liver are also subserved by specific cell
can have several causes and results in a variety of problems. types that assume specific geometric relationships. Hepatocytes
As we have already discussed, the liver has a very low- conduct the majority of the metabolic functions of the liver and
resistance vasculature in health, and pressures increase little produce the initial biliary secretion.
as flow increases since additional sinusoids can be recruited. ■ Kupffer cells line the sinusoids and cleanse the blood of
However, in several liver diseases, inflammatory responses particulates, such as bacteria.
trigger hepatic stellate cells to increase collagen production, ■ The endothelial cells of the liver have large fenestrations that
reducing permeability across the sinusoidal endothelium allow small proteins and other molecules to leave the circula-
and space of Disse and impairing liver function due to the tion, but retain blood cells and intact chylomicrons.
associated fibrosis. The hardening of the liver impedes flow ■ Hepatic stellate cells are contractile and likely regulate
through the sinusoids. Some of the sinusoids and liver sinusoidal caliber. In disease, they play an important role
parenchyma may also be destroyed and replaced by fibrous in generating fibrosis.
tissue, further impairing liver function. The most obvious ■ Liver failure due to damage to the liver cells or to the biliary
clinical consequence of portal hypertension is a condition system, or blockade of biliary drainage, results in a host of
known as ascites. Because the hepatic sinusoids and space systemic problems.
of Disse are very permeable and allow albumin to pass,
large quantities of lymph are produced by the liver even in
health, and are collected by a series of lymph ducts that STUDY QUESTIONS
eventually return the fluid to the blood via the thoracic
1. In a patient with end-stage liver disease, which of the
duct. However, when portal hypertension develops, plasma following combinations of findings would be expected
transudation increases and overwhelms the hepatic lym- in the plasma?
phatics, which may themselves be compromised by liver
fibrosis. The resulting fluid, which contains almost as much Albumin Glucose Ammonia
albumin as the plasma, weeps from the surface of the liver
and accumulates in the peritoneal cavity. In advanced liver A) Increased Increased Increased
disease, many liters of fluid may be found. Another conse- B) Decreased Decreased Decreased
quence of portal hypertension is the development of col- C) Increased Decreased Increased
lateral blood vessels to surrounding structures. These form
in an attempt to bypass the blockage to portal flow posed by D) Decreased Increased Decreased
the hardened liver, and reconnect to the systemic circula- E) Decreased Decreased Increased
tion. If the collateral vessels link to the esophagus, they are
referred to as esophageal varices and are vulnerable to ero- 2. A 60-year-old man comes to his physician complaining of a
sion and rupture, particularly if their internal pressure is progressive increase in girth over several months, despite
high. Rupture of such varices represents a major medical attempts to diet. He is suffering from jaundice (yellowing of the
emergency due to the challenges involved in reestablishing skin and sclera) and also complains of nausea and malaise. When
hemostasis. Ruptured varices are also at high risk for a large needle is inserted into his abdomen, several liters of tan
rebleeding. Variceal pressures can be reduced by construct- fluid drain out. An increase in which of the following does not
account for this accumulation of fluid?
ing a surgical shunt between the portal vein and the sys-
temic circulation, although doing so diverts portal blood A) portal pressure
B) hepatic collagen
from any remaining functional liver parenchyma, and thus
C) plasma albumin
increases complications associated with the loss of the D) stellate cell activity
detoxifying functions of hepatocytes. E) plasma transudation
564 SECTION VIII GI Physiology

3. The liver is responsible for removing the small numbers of 5. What structure in the liver permits protein-bound metabolic
bacteria that enter the portal circulation from the intestines. products to access the basolateral membranes of hepatocytes?
Which cell type fulfills this function? A) canaliculi
A) sinusoidal epithelial cells B) sinusoidal fenestrae
B) cholangiocytes C) Kupffer cells
C) hepatocytes D) bile ducts
D) Kupffer cells E) tight junctions
E) stellate cells
4. A gallstone lodged in which of the following sites will result in
increased bile acid flux through the hepatocytes making up only
the left side of the liver?
A) cystic duct
B) common hepatic duct
C) right hepatic duct
D) left hepatic duct
E) common bile duct
56
C H A P T E R

Bile Formation,
Secretion, and Storage
Kim E. Barrett

O B J E C T I V E S

■ Understand the physiologic functions of bile as a route for excretion and in


aiding in the digestion and absorption of dietary lipids.
■ Understand how bile acids are formed from cholesterol, how they are
modified during gut passage, and their role in driving bile secretion.
■ Describe the major biliary lipids and how they are transported into the
canaliculus.
■ Describe how the composition of bile is modified as the bile moves through
the biliary ductules.
■ Understand the role of the gallbladder in concentrating bile and coordinating
its secretion with ingestion of a meal, and how contraction of the gallbladder
is regulated.
■ Explain why the gallbladder is vulnerable to the formation of cholesterol
gallstones.

BASIC PRINCIPLES OF BILIARY BILE ACID METABOLISM


EXCRETION AND SECRETION Bile secretion in the liver is driven by the active, ATP-dependent
efflux of conjugated bile acids out of the hepatocyte into the
ROLE AND SIGNIFICANCE canaliculus. In this section, we will consider how bile acids
are synthesized, and subsequent modifications to their struc-
The liver fulfills its excretory function by producing bile, a ture that promote their role as biological detergents.
lipid-rich solution designed to promote the elimination of
hydrophobic solutes. Bile consists of a micellar solution in
which bile acids, cholesterol metabolites produced by hepato- FORMATION OF BILE ACIDS
cytes, form mixed micelles with phosphatidylcholine. These FROM CHOLESTEROL
mixed micelles solubilize molecules that would otherwise have
minimal aqueous solubility, such as cholesterol itself and a Bile acids are amphipathic end products of cholesterol
variety of xenobiotics. Bile also plays an important role in metabolism. The term amphipathic refers to the fact that bile
the digestion and absorption of dietary lipids. While bile acids acids have both a hydrophobic and a hydrophilic face, and
are not essential for the uptake of most fatty acids, which have form micelles. Synthesis of bile acids from cholesterol occurs
appreciable aqueous solubility, they do markedly increase in the hepatocyte. Changes to both the steroid nucleus of
the efficiency of this process. On the other hand, insoluble cholesterol and its alkyl side chain are required to convert the
dietary lipids, such as saturated long-chain fatty acids and highly insoluble cholesterol to the water-soluble bile acid
fat-soluble vitamins, are almost entirely dependent on micel- product. The initial, and rate-limiting, step in bile acid
lar solubilization for absorption. formation is the hydroxylation of cholesterol at the 7 position of

565

Ch56_565-574.indd 565 11/26/10 10:27:38 AM


566 SECTION VIII GI Physiology

the steroid nucleus via the enzyme cholesterol 7α-hydroxylase lithocholic acid from chenodeoxycholic acid, and deoxy-
(Figure 56–1). Note that cholesterol already contains a hydroxyl cholic acid from cholic acid. Trace amounts of a third second-
group at the 3 position, and this is retained in all of the bile ary bile acid, ursodeoxycholic acid (so called because it is a
acids. However, the 3-hydroxyl group in cholesterol is in the prominent bile acid in bears), are also generated in humans by
β-orientation, and this is converted to the α-position by a pro- epimerization of the 7α-hydroxyl group. Although only very
cess known as epimerization. After these initial reactions, little ursodeoxycholic acid is formed in humans, it is impor-
downstream pathways diverge to yield the two primary bile tant to know about this compound because it is used therapeu-
acids of humans, chenodeoxycholic acid and cholic acid. tically. The secondary bile acids are less water soluble than the
Note that all of the hydroxyl groups in the mature bile acids are primary bile acids. Lithocholic acid, in particular, is cytotoxic
in the form of α-epimers, and are thus oriented to the same if present at high concentrations, and physiologic mechanisms
face of the molecule. Cholesterol is a flat molecule, insoluble, have developed to limit its toxicity.
and a major membrane constituent. In contrast, bile acids are
kinked molecules that are highly water soluble when ionized.
Bile acid synthesis in healthy humans is at a rate of approxi- BILE ACID CONJUGATION
mately 200–400 mg per day. Synthesis is subject to feedback
inhibition at the level of the 7α-hydroxylase enzyme. Both primary and secondary bile acids are further modified in
the hepatocyte by conjugating them to the amino group of
either glycine or taurine in a stable amide linkage (Figure 56–2).
PRIMARY AND SECONDARY It is these conjugated bile acids that are the substrates for
BILE ACIDS active transport across the canalicular membrane. Conjuga-
tion also renders the bile acids more water soluble, and alters
When the primary bile acids enter the distal small intestine or other physicochemical properties.
the colon, they can be acted on by bacterial enzymes to yield In addition to bacterial conversion of primary to secondary
secondary bile acids. The most important conversion is dehy- bile acids, bacteria can deconjugate both primary and second-
droxylation of the 7 position of the steroid nucleus, to yield ary bile acids, making them more lipophilic. Unconjugated bile

Cholesterol
OH

7α−hydroxylase

OH

12α−hydroxylase

HO OH HO OH

C27 dehydroxylase C27 dehydroxylase


Primary bile acids

OH
COOH COOH

Chenodoxycholic Cholic acid


HO OH acid
HO OH

Bacteria Bacteria Bacteria


FIGURE 56–1 Structures of primary and
Secondary bile acids

secondary bile acids and their precursors. OH


Primary bile acids are synthesized in the liver, COOH COOH
COOH
whereas secondary bile acids are produced in
the colon by bacterial enzymes. (Modified with
permission from Barrett KE: Gastrointestinal Physiology. HO HO OH
New York: Lange Medical Books/McGraw-Hill, Medical Pub. HO
Division, 2006.) Lithocholic acid Ursodeoxycholic acid Deoxycholic acid
CHAPTER 56 Bile Formation, Secretion, and Storage 567

Charged side chain


C OH Unconjugated bile
acid OH group
O pKa = 5.0

C O N CH2 C O−

O H O Simple micelle
Bile acid monomers
Glycine conjugate
pKa = 3.9

C O N (CH2)2 S O−

O H O
Mixed micelle
Taurine conjugate
pKa < 1

FIGURE 56–2 Conjugation of bile acids with either glycine


or taurine reduces their pKa. (Modified with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical
Pub. Division, 2006.)

Phosphatidylcholine
acids can be passively absorbed across the wall of the intestine. Cholesterol
They then travel through the portal vein back to the liver, where FIGURE 56–3 Physical forms adopted by bile acids in
they are reconjugated in the hepatocyte. Thus, all bile acids solution. Micelles are shown in cross-section, and are actually
secreted by the hepatocyte are in their conjugated forms. thought to be cylindrical in shape. Mixed micelles of bile acids
Special handling applies to the potentially toxic bile acid, present in hepatic bile also incorporate cholesterol and
lithocholic acid. In addition to conjugation with glycine or phosphatidylcholine. (Adapted with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill,
taurine, lithocholic acid can be sulfated, particularly if present
Medical Pub. Division, 2006.)
in abnormally high concentrations. This increases the hydro-
philicity of the molecule and reduces its cytotoxic effects. The
sulfated conjugates of lithocholic acid also cannot be absorbed unconjugated forms. Due to this charge, conjugated bile acids
by the intestine, which results in their elimination from the are incapable of crossing cell membranes by passive means,
pool of bile acids that circulate enterohepatically. needing instead an active transport mechanism for their secre-
tion or uptake (Figure 56–4). This allows enterohepatic circu-
lation of bile acids to be coordinated with the period when
PHYSICOCHEMICAL PROPERTIES they are needed to help digest the meal.
OF BILE ACIDS
The amphipathic character of bile acids is vital to the BILE COMPOSITION
physiologic function of these molecules. Above a certain
concentration called the critical micellar concentration CANALICULAR BILE
(CMC), bile acid molecules are spontaneously self-associated
into structures known as micelles, in which the hydrophobic The secretion of bile begins when bile acids are actively
faces are masked from the surrounding aqueous environment secreted across the canalicular membrane. Because the bile
(Figure 56–3). However, simple micelles composed of bile acids are osmotically active, canalicular bile is transiently
acids alone do not exist in bile or intestinal content. In bile, hyperosmotic. However, the canalicular tight junctions are
bile acids form mixed micelles with phosphatidylcholine. In relatively permeable, and so water is drawn into the canalicu-
turn, these mixed micelles can serve as the “solvent” for hydro- lus to balance this, along with plasma cations to maintain elec-
phobic waste products. trical neutrality. Other secondary solutes also enter bile
Conjugated bile acids are present in both bile and intestinal passively from the plasma, including glutathione, glucose,
contents as anions because they have a lower pKa than the amino acids, and urea.
568 SECTION VIII GI Physiology

Hepatocyte phatidylcholine from their normal position in the inner leaflet


of the canalicular membrane, and specifically ejects them into
the canalicular lumen as vesicles. These vesicles then fuse with
secreted bile acids to form mixed micelles. Cholesterol is also
secreted into the bile, particularly in humans, at a ratio of
Tight junction
approximately 0.3 to the amount of phosphatidylcholine (or
one tenth of the amount of bile acids). Such secretion appears
to be mediated by a heterodimer of the transporters ABC5 and
ABC8. Canalicular bile, in health, also contains conjugated
Canaliculus bilirubin, which gives bile its characteristic brown color, and a
Active secretion variety of other organic anions and cations that arise from the
• Bile acids biotransformation of xenobiotics and endogenous hormones.
• Phosphatidylcholine
The membrane transporters that allow these various molecules
• Conjugated bilirubin
to enter the bile are listed in Table 56–1.
• Xenobiotics

DUCTULAR BILE
Passive permeation As the bile moves out of the canaliculi, it is transferred to the
• Water smallest bile ductules via structures known as the canals of
• Glucose Hering. The bile ductules are lined by cholangiocytes, which
• Calcium are columnar epithelial cells specialized to modify bile
• Glutathione composition. The tight junctions linking the cholangiocytes
• Amino acids are much less permeable than those linking hepatocytes. They
• Urea are freely permeable to water, but are only selectively permeable
FIGURE 56–4 Pathways for solute entry into bile by either to electrolytes and impermeable to larger solutes. Because of
active secretion or passive permeation through the tight their permeability to water, bile rapidly becomes isotonic. The
junctions linking adjacent hepatocytes. (Reproduced with permission ductules also serve to scavenge solutes that were filtered into
from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical Books/ the bile at the leaky canaliculus. In particular, glucose is actively
McGraw-Hill, Medical Pub. Division, 2006.)
reabsorbed. Likewise, glutathione is hydrolyzed to its constitu-
ent amino acids by the apically fixed enzyme, gamma-glutamyl
transpeptidase (GGT). The function of the bile ductules is
The composition of canalicular bile is also modified by the also coordinated with ingestion of a meal. In particular, the
active secretion of additional factors from the hepatocytes. cholangiocytes secrete bicarbonate in response to secretin, via
Phosphatidylcholine, a component of the hepatocyte mem- a process involving the coupled activity of the CFTR chloride
brane, enters the bile and forms mixed micelles with the bile channel and chloride/bicarbonate exchange at the apical
acids. The ratio of phosphatidylcholine to bile acids is approxi- membrane. Sodium ions follow paracellularly to maintain
mately 0.3. Although phosphatidylcholine is only one of the electrical neutrality, in turn drawing additional water into the
phospholipids present in the hepatocyte plasma membrane, it bile and increasing its volume and flow. Thus, bile becomes
is selectively secreted into bile. This is mediated by multidrug slightly alkaline. Finally, the ductules secrete IgA molecules
resistance protein 3 (MDR3), which “flips” molecules of phos- into the bile that contribute to host defense.

TABLE 56–1 Hepatocyte transporters.


Name Location Substrate/Function

Sodium taurocholate cotransporting polypeptide (NTCP) Basolateral membrane Uptake of conjugated bile acids from blood

Organic anion transporting protein (OATP) Basolateral membrane Uptake of bile acids and xenobiotics from blood

Bile salt export pump (BSEP) Canalicular membrane Secretion of conjugated bile acids into bile

Multidrug resistance protein 3 (MDR3) Canalicular membrane “Flippase” that adds phosphatidylcholine to bile

Multidrug resistance protein 1 (MDR1) Canalicular membrane Secretion of hydrophobic cationic drugs into bile

ABC5/ABC8 Canalicular membrane Secretion of cholesterol into bile

Multiple organic anion transport protein (cMOAT, MRP2) Canalicular membrane Secretion of sulfated lithocholic acid and
conjugated bilirubin into bile
CHAPTER 56 Bile Formation, Secretion, and Storage 569

HEPATIC BILE cross the wall of the intestine passively. Rather, there is a
specific, sodium-coupled transporter known as the apical
Hepatic bile emerges from the liver in the common hepatic sodium-dependent bile salt transporter (asbt) that
duct, prior to further modification by gallbladder storage. The recognizes bile acid conjugates and reabsorbs them. The
large bile ducts are thought to have little ability to modify bile expression of asbt in the intestine is limited to epithelial cells
composition, other than by adding mucus. Hepatic bile is iso- in the terminal ileum. Thus, conjugated bile acids remain
osmotic with plasma, slightly alkaline, and contains appreciable with the meal in the lumen until the nutrients are absorbed,
quantities of IgA but essentially no glucose or amino acids. whereupon they are reclaimed from the lumen and enter the
portal circulation via a second bile acid transporter, OST, to
be returned to the liver. Only a minor portion of the bile
ENTEROHEPATIC CIRCULATION acid pool spills over into the colon in health. Moreover,
conjugated bile acids that enter the colon are deconjugated
OF BILE ACIDS by the resident bacteria. Deconjugated bile acids are largely
nonionized and can be absorbed passively. The amount of
Unlike the digestive enzymes arising from the pancreas, which
bile acids found in the feces balances daily synthesis at
contribute to nutrient digestion catalytically, bile acids contrib-
steady state (Figure 56–5).
ute to lipid digestion and absorption by mass action. This means
that considerable quantities of bile acids are needed to solubilize
the quantities of products of fat digestion that are derived from HEPATOCYTE TRANSPORT
a typical diet on a daily basis. The liver synthesizes 200–400 mg
of bile acids per day. However, the concentration of bile acids in MECHANISMS
the small intestinal lumen during digestion is much higher than
Bile acids return to the liver bound to albumin, leave the por-
would be predicted, because bile acid secretion with a meal is
tal circulation in the sinusoids, and then are specifically and
about 2,000–3,000 mg/h. This is achieved by recycling the
efficiently taken up across the basolateral membrane of the
majority of the bile acids secreted during a meal, such that a
hepatocytes via a variety of specific transporters (Table 56–1).
large pool (about 2,000 mg) of these molecules is constantly
Probably the best characterized is the sodium taurocholate
cycling between the intestine and liver (Figure 56–5).
cotransporting polypeptide (NTCP), a sodium-coupled
transporter for the taurine conjugate of cholic acid that shares
INTESTINAL UPTAKE MECHANISMS homology with asbt. Other bile acids are apparently trans-
ported by members of the organic anion transporting
Bile acids secreted into the gut lumen are initially in conju- polypeptide (OATP) family, which are sodium-independent.
gated form. Because these bile acids are ionized, they cannot Unconjugated bile acids are reconjugated with either tau-
rine or glycine, and overall the recycled bile acids are handled
in much the same way as bile acids that have been newly
Hepatic synthesis synthesized, being actively transported into the bile canaliculus
Sphincter of Oddi via the bile salt export pump (BSEP). The only exception
Spillover from
applies to secretion of the sulfated forms of conjugated
liver into
systemic Gallbladder lithocholic acid, which enter the bile via the multiple organic
circulation Small intestine anion transporter (MOAT), also referred to as MRP2.
Active
ileal
uptake

Large intestine
REGULATION OF BILE ACID SYNTHESIS
Return AND TRANSPORT
to liver Spillover into colon
Passive uptake The enterohepatic cycling of bile acids also controls the rate
of deconjugated at which they are synthesized and transported. Bile acids exert
bile acids from colon
feedback inhibition on cholesterol 7α-hydroxylase, such that
Fecal loss ( = hepatic synthesis) when return of bile acids from the intestine is high, the
synthesis of new primary bile acids is reduced. Conversely,
FIGURE 56–5 Quantitative aspects of the circulation of bile interruption of the enterohepatic circulation of bile acids for
acids. The majority of the bile acid pool circulates between the small
intestine and liver. A minority of the bile acid pool is in the systemic
any reason will relieve this feedback inhibition, increasing the
circulation (due to incomplete hepatocyte uptake from portal blood) rate at which cholesterol is converted to bile acids. Normal
or spills over into the colon and is lost to the stool. Fecal loss is rates of bile acid synthesis can increase 10–20-fold under these
equivalent to hepatic synthesis of bile acids at steady state. (Adapted conditions. This increased synthetic rate may or may not be
with permission from Barrett KE: Gastrointestinal Physiology. New York: Lange sufficient to maintain the size of the circulating bile acid pool,
Medical Books/McGraw-Hill, Medical Pub. Division, 2006.) depending on fecal losses.
570 SECTION VIII GI Physiology

Dietary
cholesterol 0.2 g/day
FUNCTIONAL ANATOMY
OF THE GALLBLADDER
The gallbladder is a muscular sac with a capacity of approximately
Hepatic and 50 mL in adult humans. It is linked to the biliary system via the
Body Bile
extrahepatic cystic duct, a bidirectional conduit for bile flow. During periods
cholesterol acid
synthesis of fasting, bile secreted by the liver is diverted into the gallbladder.
pools pools
0.8−1 g/day
On the other hand, when the gallbladder receives neurohumoral
cues that fats are present in the small intestine, it contracts and
bile flows out of the gallbladder and into the intestine.

Intact cholesterol Cholesterol


0.8 g/day excreted as EPITHELIUM
bile acids
0.2−0.4 g/day The gallbladder has two functional layers. The inner of these,
facing the bile, is a columnar epithelium that participates
Fecal excretion = Input actively in bile concentration. The tight junctions that link
adjacent epithelial cells are among the best developed any-
FIGURE 56–6 Relationship of bile acid pools to whole body where in the body, making the epithelium highly resistant to
cholesterol homeostasis in health. The combined fecal excretion of
the passive flux of solutes. This “tight” epithelium prevents the
cholesterol and bile acids is equivalent to input of cholesterol from
passive loss of bile acid molecules and thus is essential to the
the diet plus endogenous synthesis of cholesterol. (Modified with
permission from the Undergraduate Teaching Project of the American
ability of the gallbladder to concentrate the bile.
Gastroenterological Association, Unit 11. Copyright 2002.)

MUSCULATURE
The epithelial layers are underlaid by smooth muscle that can
Bile acid synthesis should also be considered in the context
alter the caliber of the gallbladder lumen. The muscle cells
of whole body cholesterol metabolism (Figure 56–6). The cho-
lesterol pools reflect hepatic and extrahepatic synthesis (as
well as a small component derived from absorption of dietary
l Dorsal
cholesterol). At steady state, cholesterol input must be bal- ga vagal
Va rents complex
anced by elimination. Cholesterol is lost from the body in two e
eff
forms by being secreted intact into the bile or after conversion
to bile acids. In either case, the sole excretory pathway is via
the bile as no cholesterol or bile acids are excreted in urine in
healthy individuals. Because it is possible to increase bile acid
synthesis by interrupting the enterohepatic circulation, this ACh Vagal
ACh and CCK
represents a pharmaceutical approach to enhance cholesterol cause smooth Gall-
afferent
elimination. muscle contraction bladder NO
VIP
CCK Sphincter
of Oddi Duodenum
BASIC PRINCIPLES OF Nutrients
GALLBLADDER FUNCTION Via CCK
bloodstream
CCK

ROLE AND SIGNIFICANCE FIGURE 56–7 Neurohumoral control of gallbladder


contraction and biliary secretion. Nutrients in the duodenum
The gallbladder serves to store and concentrate bile coming lead to the release of cholecystokinin (CCK), which acts through
from the liver in the period between meals. Gallbladder func- both endocrine and neurocrine routes to activate gallbladder
tion therefore permits coordination of the secretion of a bolus contraction and relax the sphincter of Oddi, resulting in the
of concentrated bile with the entry of dietary lipids into the secretion of a bolus of concentrated bile into the duodenal lumen.
Secondary neurotransmitters released by the enteric nervous
small intestine. The gallbladder is not essential to normal
system in response to a vagovagal reflex include the excitatory
digestion and absorption of a meal. In the absence of a func- neurotransmitter acetylcholine (ACh) and the inhibitory transmitters
tioning gallbladder, the bile acid pool continues to cycle vasoactive intestinal polypeptide (VIP) and nitric oxide (NO).
through the enterohepatic circulation, and the majority of the (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks
bile acid pool is stored in the small intestine. H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
CHAPTER 56 Bile Formation, Secretion, and Storage 571

receive cholinergic input from the vagus nerve. They also The solubility of cholesterol in bile depends on its concen-
express receptors for CCK. CCK additionally activates nerves tration relative to that of bile acids and phosphatidylcholine.
that innervate the gallbladder (Figure 56–7). As bile is concentrated, bile acids cannot leave the gallblad-
der since they are too large to pass through the tight junc-
tions linking adjacent epithelial cells, and they are also not
GALLBLADDER STORAGE OF BILE actively transported by the gallbladder epithelium. Accord-
ingly, the proportion of bile acids to cholesterol plus phos-
EFFECTS ON COMPOSITION phatidylcholine does not change, or even increases slightly,
because the gallbladder can absorb cholesterol. As a result,
Hepatic bile is isotonic with plasma, with sodium as its bile becomes slightly less saturated in cholesterol as it is
major cation and chloride as its major anion. After gallblad- stored. Gallstone disease is nevertheless quite prevalent in
der storage, water is removed from the lumen and the con- humans because we secrete relatively high concentrations of
centration of bile acids is increased by approximately 10-fold, cholesterol in hepatic bile.
whereas chloride and bicarbonate concentrations decrease.
On the other hand, the concentrations of all cations in the
bile increase, although to a lesser degree than those of the MECHANISM FOR BILE
bile acids, indicating that cations are also subject to net CONCENTRATION
absorption by the gallbladder epithelium. A comparison of
the compositions of hepatic and gallbladder bile is shown as Bile is concentrated in the gallbladder via the active transport
Figure 56–8. of ions across the tight gallbladder epithelium (Figure 56–9).
Despite the dramatic increase in the sum of anions and Sodium in the bile is exchanged for protons via members of
cations during gallbladder storage of bile, bile remains iso- the NHE family of exchangers that we encountered in the gas-
tonic. How is this possible? The answer lies in the fact that the trointestinal tract. Protons that are secreted into the stored bile
majority of the bile acid molecules are physically in the form of react with bicarbonate ions, yielding CO2 and water. The CO2
mixed micelles that also contain cholesterol and phosphatidyl- diffuses out of the gallbladder lumen passively, and water can
choline. Once the CMC is reached, the monomeric concentra- be reabsorbed. In addition to sodium absorption, moreover,
tion of bile acids does not change. Any additional bile acid chloride is also actively absorbed by the gallbladder epithe-
molecules are immediately incorporated into existing micelles. lium. Finally, water leaves across the gallbladder epithelium to
Each particle in a solution contributes the same amount of follow the osmotic effect of absorbed sodium chloride.
osmotic force, be it a molecule, ion, or micelle. This allows the
osmolality of bile to remain constant despite its concentration.
Bile also changes from being slightly alkaline (a result of bicar-
bonate secretion in the ductules) to slightly acidic.
MOTOR FUNCTIONS OF
THE GALLBLADDER AND
Hepatic bile
BILIARY SYSTEM
Gallbladder bile
GALLBLADDER CONTRACTION
300
Na+
Postprandial gallbladder contraction coincides with gastric
250 emptying. The entry of the meal into the duodenum triggers
the release of a series of neurohumoral messengers that
Concentration (mM)

200 increase gallbladder tone (Figure 56–7). Cholecystokinin


Bile acid anions (CCK) released from cells lining the duodenal lumen travels
150 through the bloodstream, and binds directly to CCK-A recep-
Cl− tors on gallbladder smooth muscle cells. In addition, CCK
100 activates vagal afferents in the wall of the duodenum, and
HCO3− these in turn initiate a vagovagal reflex that releases ACh at
50 gallbladder synapses, further increasing contractile activity.

0 1 2 3 4 5 6
SPHINCTER OF ODDI FUNCTION
Storage time (h)

FIGURE 56–8 Changes in bile composition during Even if the gallbladder contracts fully, this does not allow
gallbladder storage. (Reproduced with permission from Barrett KE: for bile secretion into the duodenum if pressure at the sphinc-
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical ter of Oddi remains high. Therefore, relaxation of the sphinc-
Pub. Division, 2006.) ter is normally coordinated with gallbladder contraction. The
572 SECTION VIII GI Physiology

LUMEN BLOODSTREAM

CO2 + H2O

Na+ CA 3Na+
NHE
H+ H+ + HCO3− 2K+
FIGURE 56–9 Mechanism of bile concentration
by gallbladder epithelial cells. Sodium is reabsorbed
via a sodium/hydrogen exchanger on the apical H+ + HCO3−
membrane (NHE), in exchange for protons generated
intracellularly by carbonic anhydrase (CA).The pathway
for chloride absorption is less well characterized, but H2O + CO2
may involve the cystic fibrosis transmembrane CFTR? ?
conductance regulator (CFTR) chloride channel and/or Cl− Cl−
a chloride/bicarbonate exchanger (not shown). ?
Gallbladder epithelial tight junctions have extremely
low permeability, and resist the passage of bile acid
anions (BA−) out of the lumen. (Reproduced with permission Reabsorbed
from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical BA−
Books/McGraw-Hill, Medical Pub. Division, 2006.)

reduction in sphincter of Oddi tone is primarily brought about


terol stones account for the majority of gallstones in most
by CCK. CCK activates predominantly a neural mechanism to
Western countries.
initiate this physiologic response, transmitted through the
Human bile is unusually rich in cholesterol. In choles-
enteric nervous system. The final mediators acting at the level
terol gallstone disease, the balance between the normal
of the sphincter smooth muscle are VIP and nitric oxide,
ratios of cholesterol to the other biliary lipids is disrupted,
released from sphincter of Oddi nerve ganglia.
either due to cholesterol hypersecretion, relative hyposecre-
tion of bile acids or phospholipids, or some combination of
these. Obesity, the use of oral contraceptives, estrogen, old
age, sudden weight loss, and genetic factors may lead to
CLINICAL CORRELATION cholesterol hypersecretion. Conversely, a diminished bile
acid pool can occur if the enterohepatic circulation is inter-
A 43-year-old woman who is a mother of three comes to rupted. In either case, patients are at risk for supersatura-
her primary care physician complaining of sharp, colicky tion of cholesterol and thus for the development of
abdominal pain, usually when she enjoys a pepperoni gallstones. Supersaturation is not necessarily sufficient for
pizza or other large, fatty meals. Recently, she has had stone formation, however, since nucleation must also
pain with any meal. Her physical examination is unre- occur. Some patients may be genetically predisposed to
markable other than a finding that she is overweight. secrete proteins that can act as nucleating agents, whereas
Nevertheless, having ruled out any cardiac disease, her other proteins in bile may retard nucleation. Two thirds
physician refers her for an imaging study of her gallblad- of patients with gallstones will have no associated symp-
der. This reveals several stones. The patient undergoes a toms. Others present with episodic pain in the epigastric
laparoscopic removal of her gallbladder (cholecystec- region. In most patients, the pain is biliary colic, thought to
tomy) and her symptoms subside, other than the fact that reflect a tonic spasm resulting from transient obstruction
she still cannot tolerate pepperoni pizza or fatty meals of the cystic duct by a stone, and sometimes precipitated by
without some discomfort and bloating. eating a large meal. Biliary colic, while severe, usually sub-
The formation of stones in the gallbladder is a common sides within a few hours, which serves to distinguish it from
disease that has afflicted humans for millennia. More than acute cholecystitis, where obstruction of the cystic duct
20 million Americans have gallstones. Gallstone-related leads to inflammation of the gallbladder. In some patients,
symptoms and complications are among the most com- acute inflammation may progress to chronic cholecystitis,
mon gastroenterologic disorders requiring hospitaliza- resulting in a thickened and fibrotic gallbladder. The defin-
tion, at great cost to the health care system. Gallstones are itive treatment for symptomatic gallstone disease is chole-
of two types, related to the deposition of either cholesterol cystectomy, which is usually simple, safe, and curative,
(cholesterol stones) or bilirubin (pigment stones). Choles- particularly with the advent of laparoscopic approaches.
CHAPTER 56 Bile Formation, Secretion, and Storage 573

CHAPTER SUMMARY 2. The composition of bile is modified as it flows through the biliary
ductules. Which of the following is expected to increase in
■ Bile is secreted by the liver as a vehicle to excrete lipid-soluble concentration during this transit?
waste products of metabolism as well as xenobiotics, and it also
A) glucose
aids in fat digestion and absorption.
B) bile acid monomers
■ The major solutes driving the primary secretion of bile are the C) alanine
bile acids, which are amphipathic molecules synthesized from D) glutathione
cholesterol in the hepatocyte. E) IgA
■ Bile acids are actively secreted into the bile canaliculus in 3. A scientist studying the enterohepatic circulation measures the
conjugated forms by an energy-dependent transporter. portal concentration of conjugated bile acids in rats treated with
■ Bile also contains cholesterol and phosphatidylcholine, which various drugs. An inhibitor of which of the following ion
are actively transported into the primary secretion, as well as transport proteins would be expected to reduce the uptake of
solutes filtered from the plasma, such as calcium and glucose. sodium taurocholate from the small intestinal lumen?
■ Bile acids recycle several times daily from the intestine to the A) Na+,K+-ATPase
liver in the enterohepatic circulation; in their conjugated forms, B) CFTR
they are actively reabsorbed in the terminal ileum, thereby C) ENaC
generating a recycling pool of bile acids. D) NKCC1
■ The gallbladder serves to store bile between meals and to E) chloride/bicarbonate exchanger
coordinate the release of a concentrated bolus of bile with the 4. Compared with hepatic bile, bile that has been stored in the
presence of the meal in the duodenum. gallbladder for several hours would be expected to display which
■ Gallbladder storage of bile results in changes in its composi- of the following changes in concentration?
tion, such that bile acids become the dominant anions.
■ Bile remains isotonic during this process as bile acid monomers Cholesterol Base Equivalents Calcium
are rapidly incorporated into mixed micelles.
A) Increased Increased Increased
■ Concentration of bile results from active transport processes
taking place in the lining epithelial cells. B) Decreased Decreased Decreased

C) Increased Decreased Increased

D) Decreased Increased Decreased


STUDY QUESTIONS
E) Decreased Decreased Increased
1. A patient with Crohn’s disease undergoes surgical resection of
her diseased terminal ileum. After recovering from the surgery, F) Increased Increased Decreased
she is noted to have moderate steatorrhea. What level of bile acid
synthesis by the patient’s hepatocytes would be expected 5. Sphincter of Oddi relaxation is normally coordinated with
compared with a normal individual? gallbladder contraction to allow bile outflow into the duodenum.
A) 10-fold higher Which of the following mediators circulates through the
B) 10% higher bloodstream to mediate this coordination when the meal is
C) unchanged in the duodenum?
D) 10% less A) gastrin
E) 10-fold less B) motilin
C) acetylcholine
D) cholecystokinin
E) nitric oxide
This page intentionally left blank
57
C H A P T E R

Handling of Bilirubin and


Ammonia by the Liver
Kim E. Barrett

O B J E C T I V E S

■ Understand the origins of bilirubin in the plasma, the need to excrete this
substance, and how it is transported through the body.
■ Describe the pathway of bilirubin handling, and further metabolic
modifications that occur.
■ Define the contributors to the level of ammonia in the circulation, and
explain why a mechanism for disposal of this metabolite is needed.
■ Describe the metabolic steps involved in the conversion of ammonia to urea
in the hepatocyte.
■ Understand the routes for eventual disposal of urea.

BASIC PRINCIPLES OF PATHWAYS OF BILIRUBIN


BILIRUBIN METABOLISM SYNTHESIS AND METABOLISM
Bilirubin is a metabolite of heme, a compound that serves to Bilirubin derives from two main sources. The majority (80%)
coordinate iron in various proteins. Very recently, bilirubin has of the bilirubin formed in the body comes from the heme
been shown to possess important functions as an antioxidant, released from senescent red blood cells. The remainder origi-
but it also serves simply as a means to excrete unwanted heme, nates from various heme-containing proteins found in other
derived from various heme-containing proteins such as hemo- tissues, notably the liver and muscles.
globin, myoglobin, and various P450 enzymes. Bilirubin and its
metabolites are also notable for the fact that they provide color
to the bile and stool, as well as, to a lesser extent, the urine. CELLULAR HEME METABOLISM
Bilirubin is produced by a two-stage reaction that occurs in
ROLE AND SIGNIFICANCE cells of the reticuloendothelial system, including phagocytes,
the Kupffer cells of the liver, and cells in the spleen and bone
It is important for the body to be able to excrete bilirubin as it is marrow. Heme is taken up into these cells and acted on by
potentially toxic. Excessive levels of bilirubin in the bloodstream heme oxygenase, liberating the chelated iron from the heme
can lead to accumulation of bilirubin in the brain due to its abil- structure and releasing an equimolar amount of carbon mon-
ity to cross the blood–brain barrier, a condition known as ker- oxide, which is eliminated via the lungs. The reaction yields a
nicterus (meaning “stained nuclei”). The development of this green pigment known as biliverdin (Figure 57–1). Biliverdin
condition impairs brain function and can be fatal if untreated. is then acted on by the enzyme biliverdin reductase, produc-
Bilirubin is also notable for its yellow coloration. Accumulation ing the yellow bilirubin.
of this substance in the blood is the basis for jaundice, or a yellow Bilirubin is almost insoluble in aqueous solutions at neutral
discoloration of the skin and eyes, which is a common symptom pH. Thus, after its release into the plasma, it is taken up by
of liver diseases. Thus, measurement of bilirubin in the plasma albumin and transported throughout the body. The binding
can be a useful marker of such conditions. affinity of unconjugated bilirubin for albumin is extremely

575

Ch57_575-582.indd 575 11/26/10 10:30:16 AM


576 SECTION VIII GI Physiology

COO− COO− Once inside the hepatocyte, bilirubin requires special han-
dling to maintain its solubility and traffic it appropriately.
Thus, it is believed to bind to a variety of intracellular pro-
CH3
teins, including fatty acid–binding proteins that direct the
H3C molecule to the microsomal compartment for conjugation.
N N These proteins are likely also responsible for vectorial trans-
Heme
Fe port of the conjugated bilirubin to the canalicular membrane
N N for transport into the bile.
H2 C CH3 Following its conjugation, bilirubin exits the hepatocyte
into the bile via a membrane transport protein known as
CH3 CH2 MRP2. While this transporter has a relatively broad specific-
ity, transporting additional metabolic products as well as
some drug conjugates, it appears that its main physiological
NADPH + O2 substrate is conjugated bilirubin. Insights into the role and
Heme oxygenase significance of this transporter have been gained from a
CO + Fe3+ + NADP+ genetic disorder known as Dubin–Johnson syndrome, where
mutations in the MRP2 gene lead to the absence of the trans-
porter. However, even in health, transport of either unconju-
M V M P P M M V
gated or conjugated bilirubin through the hepatocyte cytosol
Biliverdin is not entirely efficient, and some escapes back into the plasma
where it binds once more to albumin and can be transported
O N N N N O around the body. On the other hand, only conjugated biliru-
H H H
bin is able to enter the bile via MRP2. It is primarily present in
the aqueous fraction of the bile and is not believed to associ-
NADPH ate to any significant extent with the mixed micelles formed
Biliverdin
reductase
by the biliary lipids. Conjugated bilirubin is also neither fur-
NADP+ ther metabolized nor absorbed during its passage along the
biliary tree.
M V M P P M M V

Bilirubin
HEPATOCYTE CONJUGATION
O N N N N O The hepatocyte plays an important role in bilirubin handling
H H H H
H H by conjugating the molecule to glucuronic acid. This reaction
FIGURE 57–1 Conversion of heme to bilirubin is a two-step is catalyzed by UDP glucuronyl transferase (UGT), and
reaction catalyzed by heme oxygenase and biliverdin reductase. results in the sequential esterification of two glucuronide moi-
M, methyl; P, proprionate; V, vinyl. (Modified with permission from Barrett KE: eties to the propionic acid side chains of bilirubin (Figure 57–2).
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical
Under normal conditions, most molecules of bilirubin are
Pub. Division, 2006.)
modified with two glucuronide groups, forming bilirubin
diglucuronide. Conjugation has several important effects on
high; there is usually very little free unconjugated bilirubin in the physicochemical properties of bilirubin. First, it markedly
the plasma. When the bilirubin-laden albumin reaches the increases its water solubility, allowing it to be transported in
liver, the high permeability of the hepatic microcirculation, as the bile without a protein carrier. Second, as a result of this
discussed in Chapter 55, allows the complex to enter the space increase in hydrophilicity, and increased molecular size, con-
of Disse such that it encounters the basolateral aspect of hepa- jugated bilirubin cannot be passively reabsorbed from the
tocytes. At this site, bilirubin is taken up by a specific transport intestinal lumen. There are also no specific transporters for
mechanism to enter the hepatocyte. However, this process is the uptake of conjugated bilirubin in the intestine. Thus, con-
relatively inefficient, with the first-pass clearance of bilirubin jugation serves to promote the elimination of this metabolic
being only about 20%. waste product. Finally, conjugation modestly decreases the
affinity of bilirubin for albumin. Normal levels of total serum
bilirubin are approximately 1–1.5 mg/dL in human adults,
HEPATIC TRANSPORT MECHANISMS consisting of approximately 90% unconjugated and 10% con-
jugated bilirubin. The relative proportions of conjugated and
The transporter responsible for uptake of the bilirubin into unconjugated bilirubin in disease states provide important
the hepatocyte is thought to be a member of the organic anion clues about the level of any dysfunction in the bilirubin export
transporting polypeptide (OATP) family (Figure 57–2). pathway.
CHAPTER 57 Handling of Bilirubin and Ammonia by the Liver 577

Alb B BILIRUBIN HOMEOSTASIS


Reflux to plasma
BACTERIAL METABOLISM
Alb + B
OATP Space of Diss
e In the small intestine, there appears to be little deconjugation
or additional metabolism of bilirubin in the healthy gut.
However, when conjugated bilirubin enters the colon, it can
B
be rapidly deconjugated by the enteric flora, releasing it for
UDP glucuronyl UDP-G further metabolism by anaerobic bacteria (Figure 57–3). Bili-
transferase UDP rubin is extensively metabolized in this site, producing mol-
BG ecules known as urobilinogens and stercobilinogens. These
BG Canaliculus
BG2
MRP2 BG2 are further metabolized to urobilins and stercobilins, which
give color to the stool. Urobilinogen, which is able to cross
the colonic epithelium passively, also enters the enterohepatic
circulation. In turn, it can be conjugated by the hepatocyte
and secreted into bile.
Hepatocyte

FIGURE 57–2 Handling of bilirubin by hepatocytes. Albumin


URINARY ELIMINATION
(Alb)-bound bilirubin (B) enters the space of Disse, and bilirubin is
Even under conditions of hyperbilirubinemia, little, if any,
selectively transported into the hepatocyte, where it is either
monoconjugated or diconjugated with glucuronic acid (G).The
unconjugated bilirubin is able to enter the urine because of
conjugates are secreted into bile via the multidrug resistance protein its tight binding to albumin. Urobilinogen, on the other
2 (MRP2). Some unconjugated and conjugated bilirubin may also hand, has appreciable aqueous solubility, and a small frac-
reflux into the plasma. OATP, organic anion transporting polypeptide. tion (typically less than 5% of the urobilinogen pool) is lost
(Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: to the urine on a daily basis, and is thought to contribute to
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) the color of urine.

Bilirubin UroB

Liver
Systemic UroBG2
circulation BG2

Enterohepatic
ne circulation
intesti
UroB Small

Urine B
(1−3%) Bacteria

UroB

Colon

Fecal urobilins and stercobilins


(97−99%)
FIGURE 57–3 Cycling of bilirubin and its products through the liver, intestines, portal and systemic circulations, and kidneys.
B, bilirubin; UroB, urobilinogen; G, glucuronide. Most UroB reabsorbed from the colon is taken up by the liver, but a small portion (indicated by
the dashed arrow) spills over into the systemic circulation, from where it can be excreted by the kidneys. (Adapted with permission from Barrett KE:
Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
578 SECTION VIII GI Physiology

Jaundice

Bilirubin No bilirubin
in urine in urine

Genetic Hepatobiliary Overproduction Impaired


defects in disease conjugation
FIGURE 57–4 Differential diagnosis of excretion
jaundice depending on whether bilirubin is or is
not present in the urine. (Reproduced with permission
from Barrett KE: Gastrointestinal Physiology. New York: Lange Extrahepatic Intrahepatic Impaired
Medical Books/McGraw-Hill, Medical Pub. Division, 2006.) cholestasis cholestasis hepatic uptake

HYPERBILIRUBINEMIA AMMONIA FORMATION


Hyperbilirubinemia simply refers to excessive levels of biliru- AND DISPOSITION
bin in the blood. This can be detected during clinical examina-
Ammonia in the circulation originates in a number of differ-
tion by the yellowish coloration it imparts to the skin (jaundice),
ent sites. A diagram showing the major contributors to ammo-
and is an important marker of several disease states, some of
nia levels is shown in Figure 57–5. Note that the liver is
which center on the liver. It is important to distinguish whether
efficient in taking up ammonia from the portal blood in
the plasma contains increased levels of conjugated bilirubin,
health, leaving only approximately 15% to spill over into the
unconjugated bilirubin, or both, in order to define the mecha-
systemic circulation.
nism of disease. This can be assessed directly, but clues can
also be provided by the presence of bilirubin in the urine,
which imparts a brown coloration. Because of its less avid INTESTINAL PRODUCTION
binding to albumin, an increase in urinary bilirubin reflects
almost exclusively the conjugated molecule. In general, because The major contributor to plasma ammonia is the intestine,
conjugated bilirubin can be excreted in the urine, increases in supplying about 50% of the plasma load. Intestinal ammonia
its plasma levels represent a less serious state than those asso- is derived via two major mechanisms. First, ammonia is
ciated with severe unconjugated hyperbilirubinemia. An algo-
rithm for the differential diagnosis of hyperbilirubinemia that
takes account of these factors is presented in Figure 57–4.

BASIC PRINCIPLES OF
AMMONIA METABOLISM
Ammonia (NH3) is a small metabolite that results predominantly
from protein degradation. It is highly membrane permeable and
readily crosses epithelial barriers in its nonionized form. Renal
Intestinal
production
production
40%
(mostly colonic)
ROLE AND SIGNIFICANCE 50%

Ammonia is important clinically because it is highly toxic to


the nervous system. Because ammonia is being formed con-
stantly from the deamination of amino acids, it is important
that mechanisms exist to provide for its timely and efficient Red
disposal. The liver is critical for ammonia catabolism because blood Muscle
cells 5%
it is the only tissue in which all elements of the urea cycle, also
5%
known as the Krebs–Henseleit cycle, are expressed, providing
for the conversion of ammonia to urea. Ammonia is also con- FIGURE 57–5 Sources of ammonia production. (Reproduced
sumed in the synthesis of nonessential amino acids, and in with permission from Barrett KE: Gastrointestinal Physiology. New York: Lange
various facets of intermediary metabolism. Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
CHAPTER 57 Handling of Bilirubin and Ammonia by the Liver 579

Net reaction
Hepatocyte 2NH3 + CO2 = Urea + H2O

NH3 O NH3+

ATP H2N C NH (CH2)3 CH COO−


P
HCO3− ADP O O
Citrulline
Aspartate
1
+
NH4 H2N C O P O− 2
1
Carbamoyl AMP
Mitochondrion O−
phosphate
COO− NH2+ NH3+
OOC CH2 CH NH C NH (CH2)3 CH COO−
Arginine succinate
3
Ornithine NH3+ Fumarate
+

H3N (CH2)2 CH COO
NH2+ NH3+
H2N C NH (CH2)3 CH COO−
Arginine
Urea cycle
4 H 2O

O
Cytosol
H2N C NH2
Urea
To circulation
1 Carbamoyl synthetase 2 Arginosuccinate synthetase 3 Arginine succinate lyase 4 Arginase
FIGURE 57–6 Whole body ammonia homeostasis in health. The majority of ammonia produced by the body is excreted by the kidneys in
the form of urea. (Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

liberated from urea in the intestinal lumen by enzymes UREA CYCLE


known as ureases. Ureases are not expressed by mammalian
cells, but are products of many bacteria, and convert urea to The most important site for ammonia catabolism is the liver,
ammonia and carbon dioxide. Second, after proteins are where the elements of the urea cycle are expressed in hepato-
digested by either host or bacterial proteases, further break- cytes. A depiction of the urea cycle is provided in Figure 57–6.
down of amino acids generates free ammonia. Ammonia in Ammonia is converted in the mitochondria to carbamoyl
its nonionized form crosses the intestinal epithelium freely, phosphate, which in turn reacts with ornithine to generate cit-
and enters the portal circulation to travel to the liver; how- rulline. Citrulline, in turn, reacts in the cytosol with aspar-
ever, depending on the pH of the colonic contents, a portion tate, produced by the deamination of glutarate, to yield
of the ammonia will be protonated to ammonium ion. sequentially arginine succinate, and then arginine itself. The
Because the colonic pH is usually slightly acidic, the ammo- enzyme arginase then dehydrates arginine to yield urea and
nium is thereby trapped in the lumen and can be eliminated ornithine, the latter of which returns to the mitochondria and
in the stool. can reenter the cycle to generate additional urea. The net reac-
tion is the combination of two molecules of ammonia with one
of carbon dioxide, yielding urea and water.
EXTRAINTESTINAL PRODUCTION
The second largest contributor to plasma ammonia levels is UREA DISPOSITION
the kidney. You will recall from renal physiology that ammo-
nia transport by tubular epithelial cells is an important part A “mass balance” for the disposition of ammonia and urea is
of the response to whole body acid–base imbalances (see presented in Figure 57–7. As a small molecule, urea can cross
Chapter 47). Ammonia is also produced in the liver during cell membranes readily. Likewise, it is filtered at the glomeru-
the deamination of amino acids. Minor additional compo- lus and enters the urine. While urea can be passively reab-
nents of plasma ammonia derive from adenylic acid metabo- sorbed across the renal tubule as the urine is concentrated, its
lism in muscle cells, as well as glutamine released from permeability is less than that of water such that only approxi-
senescent red blood cells. mately half of the filtered load can be reabsorbed. Because of
580 SECTION VIII GI Physiology

Systemic
circulation ening of the liver alters several aspects of structure and func-
a l c ir c u l a ti o
tion. Indeed, alcohol abuse is one of the most important
15% P o rt n causes of chronic liver disease, and cirrhosis (irreversible
85% deposition of excess collagen in the liver) accounts for the
majority of all medical deaths among alcoholics. Most
NH3 ingested ethanol is metabolized rapidly in the liver. Products
of ethanol metabolism, most notably acetaldehyde, impair
n Urea several aspects of hepatocyte metabolic function, as well as
latio
cu producing oxidative stress and forming protein adducts that
cir
may trigger adverse immune reactions that lead to cell death.
c
mi
Syste

In its initial stages, alcoholic liver disease involves the accu-


mulation of fat in the liver. Ultimately, hepatic stellate cells
25% Urea NH3 H
+ are activated to produce collagen, and this occurs chroni-
75% cally if ingestion of excessive amounts of alcohol continues.
In a subset of patients, hepatitis and fibrosis will progress to
NH4+ cirrhosis, characterized by fibrous bands connecting the
Proteins
+ portal triads with central veins, and small, regenerative nod-
Amino ules. Patients with alcoholic liver disease that has progressed
acids
at least to hepatitis and fibrosis present with a spectrum of
Urinary symptoms of chronic liver failure, including jaundice, nau-
excretion sea, and malaise. Male patients may have hypogonadism
as urea Fecal excretion
as ammonium ion
and feminization, ascribable to both the direct toxic effects
FIGURE 57–7 The urea cycle, which converts ammonia to of ethanol on testicular Leydig cells and effects on estrogen
urea, takes place in the mitochondria and cytosol of hepatocytes. production and reduced estrogen breakdown. In severe
(Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: cases, there can be a collection of fluid in the abdominal
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) cavity known as ascites that may become infected (see
Chapter 55), hepatic encephalopathy, renal failure, and
eventually death. The primary treatment is to secure absti-
this, the kidney serves as the site where the majority of the nence from alcohol, although some liver changes may be
urea produced by the liver is excreted. However, some circu- irreversible even after drinking has stopped.
lating urea may also passively back diffuse into the gut, where When ammonia degradation is reduced, it can accumu-
it is acted on by bacterial ureases to again yield ammonia and late in the plasma to levels that become toxic to the central
water. Some of the ammonia generated is excreted in the form nervous system. Remember that ammonia, as a small, neu-
of ammonium ion; the remainder is again reabsorbed to be tral molecule, is relatively permeable across cell membranes
handled by the liver once more. and can easily traverse the blood–brain barrier. Patients
will experience a gradual decline in mental status with con-
fusion and dementia, followed eventually by coma if the
condition is untreated. The increase in plasma ammonia in
CLINICAL CORRELATION liver disease occurs by two mechanisms. First, if hepatocyte
A 55-year-old homeless man is brought to the emergency function is compromised, there is less capacity to degrade
room by the police because he has been found unrespon- ammonia coming from the intestine and extraintestinal
sive in the street. His friends state that he has been drinking sites. Second, if blood flow through the liver is impaired by
heavily. On physical examination, he is found to show signs cirrhosis and portal hypertension has set in (see also Chap-
of malnutrition, and his skin and the sclera of his eyes have ter 55), collateral blood vessels may form that shunt the
a yellowish coloration. His abdomen is markedly distended, portal blood flow around the liver, bypassing the residual
and palpation shows that his liver is enlarged. Over the next capacity of the liver to degrade ammonia. It is likely that
hour, he regains consciousness, but still shows signs of sig- both mechanisms contribute to the rise in plasma ammo-
nificant confusion, and cannot supply his name, or identify nia in the setting of long-standing liver disease.
the day of the week or the current President. A counselor Because the intestine supplies the largest load of ammo-
comes to see him to discuss the likely adverse outcomes of nia to the circulation, treatments for hepatic encephalo–
continued alcohol ingestion, and he is referred to a shelter pathy focus primarily on reducing the delivery of ammonia
and a treatment program. into the portal circulation. A common technique is to give
Chronic ingestion of excessive quantities of alcohol can a sugar, lactulose, which cannot be degraded by mamma-
have insidious effects on hepatic function, as fibrotic hard- lian digestive enzymes but is broken down by bacteria in
CHAPTER 57 Handling of Bilirubin and Ammonia by the Liver 581

2. A 2-year-old boy is brought to the pediatrician because his


the colon to form short-chain fatty acids. In turn, the pH of mother has noted a persistent, dark brown coloration of his
the colonic lumen is decreased, and more of the ammonia urine. He is otherwise healthy, and his mother notes that a
being formed in that site is protonated and “trapped” as cousin displayed similar symptoms. Tests reveal conjugated
ammonium ion to be lost to the stool. Similarly, patients hyperbilirubinemia. Bile produced by this child would be
can be given a nonabsorbable antibiotic such as neomycin expected to display which of the following changes in
that reduces the level of bacterial colonization in the intes- composition compared with that of a normal child?
tine, thereby reducing ammonia production. Finally,
patients with liver disease are often advised to follow a low- Bilirubin Urobilinogen Bile Acids
protein diet, again in an effort to reduce ammonia produc- A) Decreased Decreased Decreased
tion in the intestine. Ultimately, however, the only lasting
B) Increased Increased Increased
treatment for hepatic encephalopathy is a liver transplant,
and mental symptoms often are reversible if they have not C) Decreased Increased Decreased
been too long-standing. Nevertheless, transplantation D) Increased Decreased Increased
remains controversial in alcoholic patients.
E) Decreased Decreased Unchanged

F) Increased Increased Unchanged

CHAPTER SUMMARY 3. In health, ammonia formed in the colon is partially excreted in


■ Bilirubin is a highly insoluble antioxidant produced by the the stool. Which of the following allows for this excretion?
metabolism of heme. It is derived mostly from senescent red A) limited diffusion of ammonia across colonocytes
blood cells and circulates with albumin. B) short-chain fatty acid production
■ Bilirubin is taken up into hepatocytes, conjugated with C) active secretion of ammonia by colonocytes
glucuronide, and transported across the bile canaliculus by D) absorption of ammonium ions
MRP2. Conjugation increases the solubility of bilirubin and E) uptake by bacteria
prevents its reuptake from the intestinal lumen. 4. A 70-year-old man with long-standing alcoholic liver disease is
■ Only conjugated bilirubin is transported into the bile, but both noted to have progressively worsening confusion and
conjugated and unconjugated bilirubin may regurgitate from disorientation. Loss of the function of which cell type accounts
the hepatocyte into the plasma. for his altered mental state?
■ Bilirubin is deconjugated and further metabolized by colonic A) Kupffer cells
bacteria; some of the products may circulate enterohepatically— B) hepatocytes
notably urobilinogen, which also enters the urine. C) colonocytes
■ Hyperbilirubinemia can arise from an increase in the plasma D) vascular endothelial cells
levels of conjugated bilirubin, unconjugated bilirubin, or both, E) stellate cells
and often reflects liver disease. 5. A patient with severe portal hypertension is noted to have
■ Ammonia in plasma is derived from protein degradation and bulging veins that protrude into his esophagus (esophageal
deamination of amino acids, as well as from metabolism of varices) on endoscopic examination. He is treated surgically by
urea by bacterial ureases. The intestine supplies the majority of the placement of a shunt connecting the portal vein to the vena
plasma ammonia. cava. Which of the following will pertain after the surgery
compared with before?
■ Excessive amounts of ammonia in the circulation are toxic to
the central nervous system, so circulating levels are carefully
regulated in health. Risk of Encephalopathy Risk of Variceal Bleeding
■ The liver is the site of ammonia catabolism via the Krebs– A) Increased Decreased
Henseleit, or urea, cycle. The urea produced is mostly excreted
B) Decreased Decreased
by the kidneys.
C) Unchanged Decreased

D) Increased Increased
STUDY QUESTIONS E) Decreased Increased
1. A newborn infant is noted to be suffering from mild jaundice,
F) Unchanged Increased
but no bilirubin is found in the urine. The child’s symptoms are
most likely attributable to a developmental delay in the
expression or establishment of which of the following?
A) colonic bacterial colonization
B) MDR2
C) UGT
D) heme oxygenase
E) biliverdin reductase
This page intentionally left blank
58
C H A P T E R

Digestion and Absorption


of Carbohydrates, Proteins,
and Water-soluble Vitamins
Kim E. Barrett

O B J E C T I V E S

■ Understand the barriers to assimilation of water-soluble macromolecules


into the body.
■ Describe dietary sources of carbohydrates, and the pathways involved in the
digestion and absorption of carbohydrate polymers, dietary disaccharides,
and monosaccharides.
■ Compare and contrast protein assimilation with that of carbohydrates.
■ Identify essential amino acids, and understand why they must be provided
in the diet.
■ Describe pathways involved in the digestion and absorption of proteins,
peptides, and amino acids.
■ Understand how protein assimilation is regulated.
■ Define how key water-soluble vitamins are taken up into the body.

BASIC PRINCIPLES OF TABLE 58–1 Nutritionally important carbohydrates.


CARBOHYDRATE AND Starch
Amylose
PROTEIN ASSIMILATION Amylopectin

ROLE AND SIGNIFICANCE Disaccharides


Sucrose
Lactose
Carbohydrates and proteins are water-soluble macromolecules
Monosaccharides
of nutritional significance. Together with lipids, discussed in
Glucose
the next chapter, they represent the major sources of calories Galactose
in the diet, and each supplies specific building blocks for mol- Fructose
ecules needed for the physiologic function of the body as a Dietary fiber
whole. Dietary carbohydrates are the major exogenous source
of glucose, which is utilized by cells as an energy source. Nutri-
tionally significant carbohydrates include both large polymers While the body can synthesize glucose de novo from a variety
and disaccharides (Table 58–1). Proteins supply amino acids, of substrates, some amino acids (essential amino acids) can-
which are resynthesized into new proteins needed by the body. not be synthesized by the body.

583

Ch58_583-592.indd 583 11/26/10 10:27:03 AM


584 SECTION VIII GI Physiology

BARRIERS TO THE ASSIMILATION OF subject to digestion by mammalian enzymes, it can be broken


WATER-SOLUBLE MACROMOLECULES down by hydrolases expressed by certain colonic bacteria.
These reactions generate short-chain fatty acids, which are
Due to their hydrophilicity, proteins and carbohydrates are “at important energy sources for colonocytes.
home” in the aqueous environment of the intestinal lumen.
However, neither they nor the water-soluble end products of LUMINAL DIGESTION
their digestion can readily traverse the membranes of the epi-
thelial cells that line the small intestine. Moreover, intact
OF CARBOHYDRATE
dietary polymers are too large to be transported into cells.
Thus, an ordered series of chemical reactions breaks down
Salivary Digestion
both proteins and carbohydrate polymers to their component Digestion of carbohydrates begins in the mouth. Saliva con-
monomers or short oligomers. Digestion of both carbohy- tains a 56-kd amylase enzyme that is closely related to the
drates and proteins takes place in two sites. First, enzymes 55-kd amylase that is secreted into the pancreatic juice. As its
secreted into the intestinal lumen begin the digestive process. name implies, salivary amylase is capable of digesting amy-
Second, membrane-bound hydrolases localized to the lose, the straight-chain component of starch. Salivary amylase
microvillous membrane (“brush border”) of the epithelial is not essential for the normal digestion of carbohydrates,
cells lining the villus tips in the small intestine mediate the since all of the enzymes in the pancreatic juice are present in
next stage of digestion. The epithelium is only capable of trans- considerable excess of requirements. However, the salivary
porting monosaccharides, so even dietary disaccharides must enzyme likely does assume an important role in infants, where
be digested at the brush border before they can be absorbed. there is a developmental delay in the production of pancreatic
For proteins, on the other hand, the epithelium expresses enzymes, and as a backup in patients with pancreatic insuffi-
transporters that can take up short peptides, as well as those ciency, such as in those with cystic fibrosis.
specific for monomeric amino acids. Thus, peptides taken up Salivary amylase is quite sensitive to acidic pH. However, its
into the enterocytes undergo a third stage of digestion in the activity can be protected if its substrate occupies the active site
cytosol, mediated by intracellular hydrolases. of the enzyme. Thus, while starch is present in the gastric
lumen, it is likely that its digestion mediated by salivary amy-
lase can continue, until the task is assumed by pancreatic amy-
lase. The latter is also sensitive to acid, but acts in an
CARBOHYDRATE ASSIMILATION environment where gastric juices have been neutralized by
SOURCES OF duodenal, pancreatic, and biliary bicarbonate secretion.
The synthesis and secretion of salivary amylase in the serous
CARBOHYDRATE IN THE DIET cells of the salivary glands are regulated by neurohumoral sig-
nals coincident with ingestion of a meal. Interestingly, in com-
Carbohydrates represent a major component of most human
mon with the pancreatic isoform, the synthesis of salivary
diets, and assume particular importance in specific popula-
amylase is upregulated by carbohydrate ingestion. Thus, the
tions (Table 58-1). There are three main forms of carbohydrate
substrate controls the availability of the means of its digestion.
that have nutritional significance—starch, sucrose, and lac-
tose. Starch is the name given to a complex mixture of dietary
polymers of glucose derived from plant sources, such as cere- Intestinal Digestion
als and starchy vegetables. There are two different types of glu- In health, the majority of starch digestion likely involves the
cose polymers in starch, which is significant because they 55-kd amylase that is secreted as an active enzyme into the
require different enzymes to digest them fully. About 25% of pancreatic juice by pancreatic acinar cells (see Chapter 51).
starch consists of amylose, which is composed of simple, Both the pancreatic and salivary enzymes act rapidly to cleave
straight-chain polymers of glucose (Figure 58–1A). The starch into a mixture of products, depending on whether amy-
remainder of the nutritional portion of starch consists of amy- lose or amylopectin is the substrate. The amylase enzymes tar-
lopectin, which is composed of complex, branched polymers get internal α-1,4 bonds of both molecules, but the terminal
of glucose (Figure 58–1A). bonds, as well as the α-1,6 bonds that provide for the branched-
Sources of starch also supply other carbohydrate polymers, chain structure of amylopectin, are resistant (Figure 58–1A).
as well as noncarbohydrate polymers, that collectively are This means that while the action of amylase is rapid, none of
known as dietary fiber. Fiber is characterized by the fact that the products it generates can immediately be absorbed by the
constituent polymers cannot be degraded by mammalian enterocytes, since the epithelium can only transport monosac-
digestive enzymes. It is critical for intestinal health because, charides. By the time the meal reaches the proximal small
being indigestible in the small intestine, it remains in the intestine, therefore, digestion of starch will generate a mixture
lumen and provides bulk to the stool, retaining fluid and aid- of maltose (a dimer of glucose), maltotriose (a trimer of glu-
ing passage of the fecal material through the colon. Fiber has cose), and α-limit dextrins, which are the simplest structures
additional nutritional significance in that, although it is not that can be derived from the branch points in amylopectin.
CHAPTER 58 Digestion and Absorption of Carbohydrates, Proteins, and Water-soluble Vitamins 585

A B
Glucose Maltose
α1,4 bond 1
Maltotriose
Amylose
Glucoamylase
Sucrase
Isomaltase

Amylase

α1,6 bond 2
Amylopectin
α-limit dextrin

Glucoamylase

Maltose

Maltotriose +
Glucose oligomers
Isomaltase

+
α-limit dextrin Glucoamylase
Sucrase
Isomaltase

FIGURE 58–1 A) Structure of amylose and amylopectin, which are polymers of glucose (indicated by circles). These molecules are
partially digested by the enzyme amylase, yielding the products shown at the bottom of the figure. B) Brush border hydrolases responsible for
the sequential digestion of the products of luminal starch digestion. Glucose monomers are indicated by circles. Panel 1 depicts the digestion of
linear oligomers of glucose; panel 2 shows the final steps in digestion of α-limit dextrins. (Reproduced with permission from Barrett KE, Barman SM, Boitano
S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

Both maltose and maltotriose are resistant to amylase as they along the crypt–villus axis, with highest expression in the
contain only terminal, and no internal, α-1,4 bonds. villus tips as well as in the proximal part of the small intestine.
The enzymes are trafficked specifically to the apical mem-
brane of the cells in these sites and anchored in the membrane
BRUSH BORDER DIGESTION by a single transmembrane segment. Brush border hydrolases
OF OLIGOSACCHARIDES are also heavily glycosylated. This may protect them, to some
AND DISACCHARIDES degree, from proteolysis. The enzymatic activities involved in
brush border hydrolysis include sucrase, isomaltase, glu-
The products of luminal starch digestion, as well as dietary coamylase, and lactase. Sucrase and isomaltase activities are
disaccharides, then are acted on by specific hydrolases present actually encoded in a single polypeptide chain with two dis-
on the enterocyte brush border. Brush border digestion is an tinct active sites, and thus the complete protein is referred to
essential component of the pathways leading to assimilation of as sucrase-isomaltase. Overall, the brush border hydrolases
all dietary carbohydrates, with the exception of glucose. Brush cooperate to facilitate the complete digestion of dietary car-
border hydrolysis of carbohydrates, as well as other dietary bohydrates and the products derived from their luminal
components, likely increases the efficiency of carbohydrate digestion.
absorption because the monosaccharides generated are pro-
duced in close proximity to the transporters that are then
required for their uptake. Likewise, this may also sequester Glucose Oligomers and α-Limit Dextrins
digested monosaccharides from the limited numbers of small The final digestion of these products of amylose and amylo-
intestinal bacteria. pectin digestion involves the concerted action of several brush
Brush border hydrolysis is catalyzed by a series of enzymes border enzymes (Figure 58–1B). Glucoamylase, sucrase, and
that are synthesized in the enterocytes as they differentiate isomaltase activities are all capable of digesting the bonds
586 SECTION VIII GI Physiology

1
Sucrose

Sucrase

Isomaltase
Na+

Brush border
membrane SGLT-1 GLUT5

Cytosol
Glucose

Fructose

Lactose

2
Lactase
Na+

SGLT-1
FIGURE 58–2 Brush border digestion
and assimilation of the disaccharides sucrose
(panel 1) and lactose (panel 2). SGLT-1,
Cytosol
sodium-glucose cotransporter-1. (Reproduced with
permission from Barrett KE, Barman SM, Boitano S, Brooks Glucose
H: Ganong’s Review of Medical Physiology, 23rd ed.
McGraw-Hill Medical, 2009.)
Galactose

contained in linear, short-chain (2–9 sugar units) oligomers of across the apical membrane of the enterocyte. This is particu-
glucose, which include maltose and maltotriose. However, larly the case for fructose, which enters the cytosol not via the
isomaltase is critical for the full digestion of starch, since it is SGLT-1 transporter, but rather via a sodium-independent,
unique among the listed activities in being able to cleave not facilitated diffusion pathway (GLUT5).
only the α-1,4 bonds of linear glucose oligomers, but also the
α-1,6 bonds of the α-limit dextrins. Glucose uptake is then
mediated by SGLT-1, the sodium/glucose cotransporter.
Lactose
This cotransporter takes advantage of the low intracellular Lactose is an important nutrient in those who consume large
sodium concentration established by the basolateral Na,K- quantities of milk, such as infants. It is a disaccharide that con-
ATPase to accumulate glucose in the cytosol against a concen- sists of glucose and galactose and is broken down at the brush
tration gradient (i.e., uphill transport). border by lactase, an enzyme that contains two identical active
sites within a single polypeptide chain. The products of this
hydrolysis reaction are, in turn, both substrates for SGLT-1
Sucrose and thus can be accumulated against a concentration gradient
Sucrose (table sugar) is a prominent carbohydrate in many (Figure 58–2).
Western diets and requires no luminal digestion because it is a Lactose assimilation is limited in two important ways. First,
simple disaccharide consisting of glucose and fructose. Rather, there is a developmental decline in lactase expression, meaning
it is specifically digested at the level of the brush border by the that levels of this enzyme in adulthood may be inadequate to
enzyme sucrase, yielding the respective monosaccharides hydrolyze all of the substrate presented to them. Thus, lactose
(Figure 58–2). Expression of sucrase-isomaltase is usually in hydrolysis, rather than transport of the products of this reac-
excess of the requirements for this enzyme, at least in Western tion, is usually rate limiting for assimilation. Second, the activity
populations that emphasize sucrose in the diet. This means of lactase is inhibited by glucose, in a process known as “end-
that the rate-limiting step for sucrose assimilation is not product inhibition.” If glucose levels increase in the vicinity of
its hydrolysis, but rather the uptake of the released products the enzyme, breakdown of lactose will further be inhibited.
CHAPTER 58 Digestion and Absorption of Carbohydrates, Proteins, and Water-soluble Vitamins 587

MONOSACCHARIDE UPTAKE and long terms. Acutely, brush border hydrolases on the sur-
PATHWAYS face of enterocytes are degraded at the end of the meal, when
dietary protein is no longer available to compete for the activ-
The final steps in carbohydrate assimilation involve specific ity of pancreatic proteases. These enzymes are then resynthe-
membrane transport pathways that permit uptake of these sized by the enterocyte to ready the epithelium to handle
hydrophilic molecules across the enterocyte apical membrane, carbohydrates in the next meal. This cycle of degradation and
as well as mediating their transfer out of the enterocyte across resynthesis is not specific for the enzymes involved in carbo-
the basolateral membrane and thence into the portal circula- hydrate digestion, but occurs for the entire complement of
tion. We have already discussed two of these transporters, brush border proteins needed for nutrient assimilation. On
SGLT-1 and GLUT5. SGLT-1 is synthesized by villus entero- the other hand, and on a longer time scale, if carbohydrates are
cytes but not by those in the crypts, likely as a result of tran- specifically withheld from the diet, there is a gradual decline in
scriptional regulatory mechanisms that parallel those involved the expression of the hydrolases and transporters that are
in establishing brush border hydrolase expression. It exists in involved in the assimilation of this class of nutrients, and likely
the membrane as a homotetramer, which appears to be impor- also in the expression of amylase. All of these components are,
tant for its function. The protein mediates the ordered transfer in turn, upregulated if carbohydrate is then returned to the
of both sodium and glucose across the membrane. Sodium diet. These long-term changes are specific for the nutrient
binds first to an extracellular site on the transporter, followed withdrawn from the diet. There are also hormonal influences
by glucose, which triggers a conformational change in the pro- on the expression of brush border hydrolases and transporters
tein. This transfers these substrates to the cytoplasmic face of that match the capacity for carbohydrate assimilation with the
the membrane, where first glucose, and then sodium, can dis- body’s needs. Insulin, in particular, appears to suppress the
sociate into the cytosol. Transporters of the GLUT family also levels of these molecules, meaning that glucose assimilation
play important roles in carbohydrate assimilation. The portion can be enhanced in the setting of type I diabetes mellitus.
of absorbed glucose that is not needed for immediate energy
needs of the enterocyte exits the cell across the basolateral
membrane via a facilitated diffusion pathway (GLUT2). GLUT2 PROTEIN ASSIMILATION
is sodium-independent, and thus the movement of glucose
through this transporter will depend only on the relative con- COMPARISON WITH CARBOHYDRATE
centrations of the sugar inside and outside the cell. It is also ASSIMILATION
expressed in many other cell types throughout the body, where
it participates in glucose uptake. A related molecule, GLUT5, The digestion and absorption of proteins and carbohydrates
provides for the brush border uptake of the fructose that is gen- shares many similar features, since both are water-soluble
erated from the hydrolysis of sucrose (Figure 58–2). GLUT5 is macromolecules, including a role for both luminal and brush
also present in the basolateral membrane and thus can mediate border digestion and the presence of specific transporters in
transfer of fructose to the bloodstream, although there is evi- the apical membranes of enterocytes that take up the products
dence that fructose is additionally a substrate for GLUT2. of these digestion reactions. However, there are also important
differences. First, the 20 naturally occurring amino acids,
compared with the 3 nutritionally significant monosaccha-
REGULATION OF CARBOHYDRATE rides, mean that proteins represent a significantly more diverse
ASSIMILATION set of substrates and require a broader spectrum of peptidases
and transporters to mediate their digestion and uptake. Sec-
Developmental ond, the intestine is capable of transporting not only single
In humans, the machinery for brush border digestion and amino acids, but also short oligomers, encompassing dipep-
absorption of carbohydrates is all in place before birth. How- tides, tripeptides, and perhaps even tetrapeptides. In fact,
ever, the capacity for luminal digestion of carbohydrates is regu- some amino acids are absorbed much more efficiently in the
lated in the postnatal period. Expression of pancreatic amylase form of peptides than as the single molecules. Finally, the exis-
is low in infants below the age of 1 and is gradually induced as tence of peptide transport in the intestine implies that these
starch is added to the diet. Conversely, lactase levels in the brush molecules must eventually be digested to their component
border decline after weaning. However, both of these responses amino acids in order for them to be useful to other body tis-
likely do not reflect strict developmental regulation, but rather sues. This final stage of protein digestion takes place in the
are appropriate adaptive responses to the appearance or disap- cytosol of the enterocyte.
pearance of the relevant substrates in the normal diet.
Essential Amino Acids
Dietary Another important concept when considering protein assimi-
The various components of the systems involved in carbohy- lation is that of the essential amino acid. While the body (prin-
drate assimilation are regulated by the diet in both the short cipally the liver) is able to convert one amino acid to another,
588 SECTION VIII GI Physiology

Amino acids

Basic Acidic
Neutral
Arg
FIGURE 58–3 Naturally occurring amino acids Lys , His
Glu, Gln
Asp, Asn
organized on the basis of their physicochemical
properties. Residues that are boxed are essential
amino acids that must be obtained from dietary
Aliphatic Aromatic Hydroxyl Sulfur Imino
sources by humans. (Reproduced with permission from Gly, Ala Tyr Ser Cys Pro
Barrett KE: Gastrointestinal Physiology. New York: Lange Medical
Val , Leu , Ile Phe , Try Thr Met Hydroxypro
Books/McGraw-Hill, Medical Pub. Division, 2006.)

nine of the naturally occurring amino acids cannot be synthe- amino acid located at the C-terminus. Despite their various
sized de novo, and therefore must be obtained from the diet specificities, however, all of the pancreatic peptidases have one
and absorbed either as the amino acid itself or in peptide form important feature in common. They are all stored in the pan-
(Figure 58–3). Proteins derived from animal sources contain creatic acinar cells as inactive precursors, which apparently is
all of the essential amino acids. However, proteins from vege- important to prevent autodigestion of the pancreas.
table sources are “incomplete,” meaning that they lack one or How then are these inactive enzymes converted to their
more of the essential amino acids. active forms only when they are in small intestinal lumen? The
answer lies in yet another proteolytic enzyme—enterokinase—
expressed on the apical membrane of small intestinal epithe-
LUMINAL PROTEOLYSIS lial cells. When the pancreatic juice is secreted into the
intestine, it comes into contact with enterokinase, which
Gastric cleaves an N-terminal hexapeptide from trypsinogen, yield-
Digestion of dietary proteins begins in the stomach—there are ing active trypsin. Trypsin, in turn, can then activate addi-
no nutritionally significant proteolytic enzymes found in the tional trypsin molecules as well as all of the other inactive
saliva. As we learned in Chapter 50, on the other hand, the pancreatic peptidases only once they are in the intestinal
chief cells of the gastric glands synthesize and store pepsino- lumen (Figure 58–4).
gens, inactive precursors of pepsins, which are a group of The pathways of luminal proteolysis are shown in diagram-
related proteolytic enzymes especially suited to action in the matic form in Figure 58–5. Large peptides derived from gastric
stomach. At low pH, there is autocatalytic cleavage of an proteolysis are sequentially cleaved by the endopeptidases
N-terminal peptide from pepsinogen that yields the active (trypsin, chymotrypsin, and elastase). These reactions yield
form. Pepsins preferentially cleave dietary proteins at neutral shorter peptides with either neutral or basic amino acids at
amino acids, with a preference for large aliphatic or aromatic their C-termini, which can be acted on in turn by carboxypep-
side chains. They are also sensitive to the pH of their environ- tidase A or carboxypeptidase B, respectively. Thus, the prod-
ment, and are inactivated above a pH of 4.5. This means that ucts of proteolysis in the intestinal lumen consist of free basic
gastric pepsins are quickly inactivated once they enter the and neutral amino acids as well as short peptides that cannot
small intestine, which may be important to prevent digestion be cleaved further due to the lack of an appropriate amino acid
of the epithelium. at their C-terminus. Approximately 60–70% of dietary protein
Because of the relatively limited specificity of pepsins, gas- is in the form of small oligopeptides following luminal hydro-
tric proteolysis results in incomplete digestion with only a few lysis; the remainder is in the form of amino acids.
free amino acids; the products are mostly large, nonabsorbable
peptides. And in common with other aspects of the gastroin-
testinal system that are either redundant or present in excess, BRUSH BORDER HYDROLYSIS
gastric proteolysis does not appear to be essential for normal
Like carbohydrate assimilation, the degradation of proteins in
levels of protein assimilation.
the lumen is incomplete and they also undergo a process of
brush border hydrolysis. However, because of the diversity of
Intestinal possible substrates, there is the requirement for a much larger
The bulk of proteolysis occurs in the small intestinal lumen. number of brush border hydrolases. These membrane-bound
This is a highly ordered process mediated by two families of enzymes comprise both endopeptidases and ectopeptidases,
pancreatic proteases, the secretion of which we discussed in and are expressed by villus, but not crypt, enterocytes. The
Chapter 51. Endopeptidases cleave proteins and peptides at activity of these enzymes yields free amino acids in the vicinity
internal amide bonds. Ectopeptidases cleave at the terminal of the enterocyte apical membrane, although some peptides
amino acid. All of the ectopeptidases secreted by the pancreas remain relatively resistant to hydrolysis, and are taken up in
are carboxypeptidases—that is to say, they cleave off the their unhydrolyzed form.
CHAPTER 58 Digestion and Absorption of Carbohydrates, Proteins, and Water-soluble Vitamins 589

Pancreatic juice

Enterokinase

Trypsinogen
Trypsin

Trypsinogen Trypsin

Chymotrypsinogen Chymotrypsin FIGURE 58–4 Mechanism to avoid


activation of pancreatic proteases until they
Proelastase Elastase are in the duodenal lumen. Trypsinogen is
cleaved by the enzyme enterokinase, which is
Procarboxypeptidase A Carboxy- expressed on the apical membrane of
peptidase A duodenal epithelial cells (depicted as the pair
of scissors).The trypsin that is thereby liberated
Procarboxypeptidase B Carboxy-
peptidase B can activate all of the other pancreatic
proteases. (Reproduced with permission from Barrett
KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of
Lumen Epithelium Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)

UPTAKE MECHANISMS FOR substrate specificity, accommodating peptides of various sizes


OLIGOPEPTIDES AND AMINO ACIDS and charges. Moreover, the stoichiometry of proton coupling can
change, depending on the net charge of the peptide being trans-
ported. On the other hand, transport is strictly stereospecific—
Peptide Transporters transport of peptides comprised of d-amino acids is negligible.
Perhaps the most fascinating aspect of protein assimilation is its The activity of this transporter is nutritionally significant,
dependence, in part, on a remarkable peptide transporter desig- because it mediates the uptake of peptides resistant to brush bor-
nated as peptide transporter 1 (PEPT1) (Figure 58–6). This der hydrolysis and thus increases the efficiency of protein assim-
protein is expressed on the apical membrane of enterocytes, and ilation from the gut. Indeed, certain amino acids are much more
mediates the proton-coupled uptake of a broad variety of short effectively absorbed in peptide form than when presented as
peptides. What makes PEPT1 so intriguing is its extremely broad individual molecules, especially glycine and proline.

Ser
Carboxypeptidase A
Chymotrypsin Peptide with
Elastase C-terminal
neutral AA

Large Arg
peptides Ser
Short peptides
free neutral and
basic AA’s
FIGURE 58–5 Luminal digestion of
Trypsin
peptides resulting from partial proteolysis
Carboxypeptidase B
in the stomach. Individual amino acid
Arg residues are shown as squares. (Reproduced with
Peptide with permission from Barrett KE, Barman SM, Boitano S,
C-terminal Brooks H: Ganong’s Review of Medical Physiology, 23rd
basic AA ed. McGraw-Hill Medical, 2009.)
590 SECTION VIII GI Physiology

and amino tripeptidases. A cytosolic propeptidase, which


hydrolyzes absorbed proteins that contain proline, has also
Na+ 3Na+ been identified. Recall that peptides containing proline (and
NHE glycine) are relatively resistant to luminal or brush border
H+ 2K+ hydrolysis.
Cytosolic
digestion
H+
PEPT1 REGULATION OF PROTEIN
Basolateral ASSIMILATION
amino acid
Di-, tripeptides transporters
Many of the factors that regulate carbohydrate assimilation
FIGURE 58–6 Disposition of short peptides in intestinal have analogous effects on protein assimilation. For example,
epithelial cells. Peptides are absorbed together with a proton brush border hydrolases, peptide transporters, and amino acid
supplied by an apical sodium/hydrogen exchanger (NHE) by the transporters can be degraded by proteolytic enzymes; these
peptide transporter 1 (PEPT1). Absorbed peptides are digested by molecules are then resynthesized by the enterocyte to partici-
cytosolic proteases, and any amino acids that are surplus to the needs pate in the digestion and absorption of the next meal. Simi-
of the epithelial cell are transported into the bloodstream by a series larly, the membrane proteins involved in protein assimilation
of basolateral transport proteins. (Reproduced with permission from Barrett are expressed in gradients along the proximal to distal and
KE, Barman SM, Boitano S, Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. crypt to villus axes.
McGraw-Hill Medical, 2009.)

WATER-SOLUBLE VITAMIN
Amino Acid Transporters
Despite the significance of peptide uptake in the intestine,
ASSIMILATION
many amino acids are absorbed predominantly in molecular Vitamins are molecules that cannot be synthesized by the
form. The physiology of amino acid transport in the intestine body, but which are essential to normal metabolism. Many act
was at one time a complex topic, given the diversity of trans- as cofactors for specific biochemical reactions, or play other
porters that had been defined only functionally as well as some critical roles in the body. Like essential amino acids, most vita-
overlapping specificities. Moreover, some, but not all, amino mins must be obtained from dietary sources. We will consider
acid transporters are sodium-dependent, analogous to SGLT-1 here the uptake of two key water-soluble vitamins, since this
that we considered earlier for glucose uptake as well as the process is analogous to the assimilation of the end products of
sodium-dependent bile acid transporter that recovers these carbohydrate and protein digestion. Lipid-soluble vitamins
molecules in the distal ileum. Others, analogous to PEPT1, will be discussed in the next chapter.
may transport specific amino acids in concert with one or
more protons. Finally, some amino acid transporters clearly
have properties that classify them as facilitated diffusion path- VITAMIN C
ways or even channels.
More recently, some clarity has been brought to this field by Vitamin C (ascorbic acid) acts as an antioxidant in the
molecular cloning of a large number of amino acid transport body as well as participates in a number of hydroxylation
proteins from the gut, and their organization into families. We reactions. It is obtained from a variety of dietary sources,
now know that there are multiple transport systems for neu- including citrus fruits and several vegetables. Having a pKa
tral, cationic, and anionic amino acids, with each system being of 4.2, it is ionized at the pH of the small intestinal lumen
distinct but exhibiting overlapping specificity. However, it is and thus its passive diffusion across the epithelium is negli-
beyond the scope of an introductory course in physiology to gible. Thus, specific transport mechanisms exist to ensure
discuss all of these transporters in detail. its assimilation.
Uptake of vitamin C is predominantly localized to the ileum.
Ascorbic acid is transported across enterocyte apical mem-
CYTOSOLIC PROTEOLYSIS branes via a family of sodium-coupled cotransporters (SVCT1
and SVCT2), and uptake is controlled by intracellular signals.
Peptides absorbed into the enterocyte cytosol are further Vitamin C uptake is also regulated by its own levels in the
degraded before passing into the portal circulation. A portion body. Thus, supplementation with this vitamin, either orally
of the released amino acids, as well as those taken up in molec- or via injection, leads to a decrease in intestinal capacity for its
ular form, may also be utilized locally for the enterocyte’s own transport. This finding implies that, unlike nutrient uptake,
needs for protein synthesis. Peptide breakdown is mediated by the intestine displays a capacity to allow for “vitamin homeo-
a series of cytosolic peptidases that act to cleave N-terminal stasis,” maintaining whole body levels of vitamins at a rela-
amino acids from these molecules, such as amino dipeptidases tively stable level.
CHAPTER 58 Digestion and Absorption of Carbohydrates, Proteins, and Water-soluble Vitamins 591

VITAMIN B12 (COBALAMIN) dietary proteins and binds initially to the R-binding protein
(also known as haptocorrin), forming a complex that is stable
Another water-soluble vitamin of note is vitamin B12, which is at the acidic pH at this site. R-binding protein derives pre-
utilized by all cells in its coenzyme form in various metabolic dominantly from the salivary glands. Intrinsic factor cannot
reactions. Unlike vitamin C, whose uptake occurs via a simple, bind to cobalamin at low pH. Rather, it follows the cobala-
sodium-coupled mechanism, the uptake of cobalamin is more min–R-binding protein complex into the duodenum, where
complex, and requires the participation of a specific binding the R-binding protein is degraded by pancreatic proteases and
factor secreted by parietal cells, known as intrinsic factor. the cobalamin is transferred to intrinsic factor at the increased
Intrinsic factor secretion is triggered in the stomach by the pH that is brought about as gastric secretions are neutralized.
same neurohumoral cues that result in gastric acid secretion. The N-terminus of intrinsic factor also contains a binding
Intrinsic factor then binds to dietary cobalamin and later pro- sequence recognized by an apical receptor for this molecule
vides for the uptake of this vitamin. In an analogous fashion, that is expressed by enterocytes, known as the intrinsic
cobalamin in the circulation is bound to a separate protein, factor-cobalamin receptor (IFCR). IFCR is highly expressed
plasma transcobalamin II (TC II). Specific receptors exist to in the terminal ileum. When the complex of intrinsic factor
mediate the endocytosis of cobalamin bound to each of these and cobalamin binds to IFCR, the receptor plus the bound
carrier proteins. ligand is internalized, and these are then directed to a vesicu-
Here, of course, we are chiefly concerned with the processes lar pathway for sorting and trafficking. Intrinsic factor is
mediating cobalamin uptake from the intestinal lumen (Fig- degraded, and the released cobalamin is then bound to TC II,
ure 58–7). In the gastric lumen, cobalamin is released from synthesized by the enterocyte. In turn, this new complex is
trafficked to the basolateral membrane, and released into the
circulation in this form. Cobalamin bound to TC II can in
turn be taken up via the specific receptor for this complex that
Food-bound
Cbl is ubiquitously expressed throughout the body.

Cbl bound
Acid pH CLINICAL CORRELATION
Biliary
Cbl Cbl free An 18-year-old, Asian American college freshman is living
R away from home for the first time. Having been brought up
in a household that emphasized a traditional Chinese diet,
R-Cbl and having been forbidden by his parents from indulging
+ Parietal
cells
in snacks and junk food, he is particularly excited by the
IF
secrete IF broad range of dishes to choose from in the cafeteria. He
becomes particularly enamored of cheese pizza and choco-
late ice cream. However, 2 weeks after the start of term, he
R∗ comes to the student health center complaining of frequent
Proteolytic Pancreatic proteases diarrhea, gas, and bloating. A physical exam reveals that he
degradation
of R protein is otherwise healthy and well nourished, and the patient
Cbl
denies any fever or bloody stools. Based on his history and
Cbl released
after R protein +IF IF-Cbl symptoms, a diagnosis of lactose intolerance is made.
degradation A deficiency in the ability to assimilate dietary lactose is
IF-Cbl complex forms a common disorder, particularly in specific ethnic groups
IF-Cbl
such as African-Americans and Asians who have not tradi-
tionally emphasized milk as a component of the adult diet.
lleal enterocyte
with receptor for Lactose intolerance arises secondary to the normal devel-
Enterocyte
IF-Cbl complex opmental decline in lactase-phlorizin hydrolase levels that
occurs after weaning, which occurs to a greater extent in
FIGURE 58–7 Sequential steps in the gastrointestinal some people than in others. In susceptible individuals,
absorption of vitamin B12 (cobalamin, Cbl). In the stomach, Cbl
ingestion of lactose in dairy products overwhelms the
binds salivary R protein and intrinsic factor (IF) secreted by parietal
cells. Proteolytic degradation of the R protein in the intestinal lumen capacity of the brush border lactase-phlorizin hydrolase to
yields a complex of only Cbl and IF, which then binds to a specific digest this disaccharide, leaving the undigested material in
receptor located in the apical membrane of epithelial cells lining the the small intestinal lumen from where it passes to the colon.
terminal ileum. (Reproduced with permission from Halsted CH, Lonnerdal BL: In the colon, commensal bacteria are highly active in
Vitamin and mineral absorption. In: Textbook of Gastroenterology, 4th ed. Yamada T, degrading lactose as an energy source, leading to symptoms
Alpers DH, Kaplowitz N, Laine L, Owyang C, Powell DW (editors). Philadelphia: of abdominal pain and bloating from the hydrogen and
Lippincott Williams and Wilkins, 2003.)
592 SECTION VIII GI Physiology

2. An infant with diarrhea is given a glucose and electrolyte solution


CO2 gases that are produced. The delayed production of orally. What membrane protein accounts for the ability of this
hydrogen (or labeled CO2) in the breath following lactose solution to provide rapid hydration?
ingestion can accordingly be used as a test for the presence A) sucrase-isomaltase
of lactose intolerance. B) SGLT-1
Patients with lactose intolerance who wish to consume C) CFTR
dairy products can do so without major discomfort if they D) chloride–bicarbonate exchanger
also take oral supplements of a lactase enzyme derived from E) lactase-phlorizin hydrolase
bacteria. This supplemental lactase is resistant to degrada- 3. Glucose–galactose malabsorption is a rare disorder caused by
tion by gastric acid, and thus is available in the small intes- mutations in SGLT-1. Infants with this disorder develop severe
tinal lumen to cleave dietary lactose into its component osmotic diarrhea if they consume certain carbohydrates. Of the
monosaccharides, which can then be absorbed by the small following, which would not be expected to cause symptoms in
intestine. these patients?
A) sucrose
B) glucose
C) amylopectin
CHAPTER SUMMARY D) lactose
E) fructose
■ Carbohydrates and proteins are water-soluble polymers that are
4. A child is brought to the pediatrician because of severe failure to
important sources of nutrition.
thrive, diarrhea, and edema of the extremities. Blood tests reveal
■ The end products of digestion are hydrophilic, and thus that he has low plasma protein concentration (hypoproteinemia).
specific transporters are needed to translocate them across the Duodenal aspirates are obtained at endoscopy after intravenous
plasma membrane of enterocytes. administration of cholecystokinin, and are found to be incapable
■ Both carbohydrates and proteins, with the exception of glucose of protein hydrolysis at neutral pH unless a small amount of
monomers, must be digested to allow for their uptake across trypsin is added. The patient is likely suffering from a congenital
the intestinal epithelium. lack of which of the following?
■ Only monosaccharides can be absorbed by the intestine, A) pepsinogen
whereas the intestine can assimilate short peptides in addition B) PEPT1
to free amino acids. C) trypsinogen
■ Digestion of both carbohydrates and proteins occurs in an D) carboxypeptidases
ordered series of distinct stages. E) enterokinase
■ Both luminal and brush border hydrolysis are important; the 5. A 75-year-old woman comes to her physician complaining of
latter may increase efficiency. progressively worsening fatigue and numbness in her fingers.
■ Absorbed peptides also undergo cytosolic digestion in the A blood test reveals that she is anemic, despite adequate iron
enterocyte. intake. Her symptoms resolve following a series of vitamin
injections. Her symptoms are consistent with an age-related
■ Water-soluble vitamins also undergo selective transport across
decline in synthesis of which of the following proteins?
the intestinal epithelium.
A) haptocorrin
B) intrinsic factor
C) transcobalamin
STUDY QUESTIONS D) SVCT1
1. Digestive enzymes can be secreted by the pancreas in active or E) PEPT1
inactive forms. Enzymes capable of digesting which of the
following nutrients are only secreted as inactive precursors?
A) starch
B) nucleic acids
C) proteins
D) triglyceride
E) cholesterol esters
59
C H A P T E R

Lipid Assimilation
Kim E. Barrett

O B J E C T I V E S

■ Understand the special barriers to absorption of lipids supplied in the diet.


■ Describe the phases of lipid digestion.
■ Describe events at the level of the intestinal epithelium that govern uptake
of different classes of lipids.
■ Understand how the products of lipolysis cross the brush border.
■ Delineate pathways for lipid processing in the enterocyte.
■ Describe how chylomicrons are formed and their eventual disposition.

GENERAL PRINCIPLES dispersed surface area to allow for lipolysis at the oil/water inter-
face. Additional phase transitions allow for efficient trafficking
OF LIPID ASSIMILATION of lipids to the enterocyte surface, where they can be absorbed.

ROLE AND SIGNIFICANCE


Lipids are organic substances that are hydrophobic, and thus DIETARY AND ENDOGENOUS SOURCES
are more soluble in organic solvents (or cell membranes) than OF LIPIDS IN INTESTINAL CONTENT
in aqueous solutions. They form an important part of most
human diets. First, they are denser in calories than either pro- Lipids represent a major source of calories in most Western
teins or carbohydrates, increasing the nutritional content of a diets, with an average of 120–150 g consumed daily by a typi-
given meal. Second, the fat-soluble vitamins are lipids. Third, cal adult. On a daily basis, the intestine is also presented with
many of the compounds that account for the flavor and aroma 40–50 g of endogenous lipids arising from the biliary system.
of foods are volatile hydrophobic molecules, meaning that Despite their hydrophobicity, the process of lipid assimilation
lipids serve as an important vehicle to render food palatable. has evolved to be highly efficient, with significant reserve
capacity also present. The ready availability of lipid-rich foods
in developed countries is an important contributor to the bur-
BARRIERS TO ASSIMILATION geoning problem of obesity.
OF HYDROPHOBIC MOLECULES Lipids in the diet as well as in endogenous pools are com-
posed of several distinct molecular classes. The majority of
The products of lipid digestion (lipolysis) are, in large part, lipid in the diet is in the form of long-chain triglycerides (i.e.,
readily able to cross cell membranes to allow for absorption into fatty acids with at least 12 carbon atoms each, esterified to glyc-
the body. However, lipids are not “at home” in the aqueous erol). Phospholipids, which are components of cell mem-
milieu of the intestinal contents. Likewise, they must interact branes, are also significant contributors. Other, more minor
with lipolytic enzymes that are themselves soluble proteins. sources of dietary lipids include plant sterols (whose absorp-
Finally, the products of lipolysis must arrive at the brush border tion may be inefficient) and cholesterol, another membrane
at a sufficient rate to allow for uptake before being propelled constituent that is present in the diets of all except vegans.
along and out of the gut. Systems therefore exist to maintain However, even vegans will encounter cholesterol in the intesti-
lipids in suspension in the gut contents with a sufficiently nal contents because it is secreted into the bile, along with the

593

Ch59_593-600.indd 593 11/26/10 10:27:50 AM


594 SECTION VIII GI Physiology

other biliary lipid, phosphatidylcholine. In fact, the endoge- 1


Gastric lipase
nous secretion of cholesterol of 1–2 g per day usually exceeds
dietary intake of 200–500 mg that is typical of most individuals. H2O
While fat-soluble vitamins are only present in trace amounts, O
their absorption is critical for a variety of body processes. The CH2 O C
fat-soluble vitamins are A, D, E, and K. Vitamin A (retinoic
O
acid) is an important regulator of gene transcription. The active
metabolite of vitamin D (calciferol) regulates calcium and CH O C
phosphate absorption by the intestine, and homeostasis of these O
ions throughout the body (see Chapters 48 and 64). Vitamin E
CH2 O C
(tocopherol) is a vital antioxidant. Finally, vitamin K is utilized
by the liver to catalyze the posttranslational modification of Fatty acid plus
several blood clotting factors. The fat-soluble vitamins, as a diglyceride
group, have negligible aqueous solubility. 2 Pancreatic lipase
H2O

INTRALUMINAL DIGESTION O

CH2 O C
Like in the assimilation of protein and carbohydrate, the initial
O
stages of lipid assimilation take place in the intestinal lumen.
Luminal events include dispersion of the lipid, which is liquid CH O C
at body temperature, into an emulsion, thereby maximizing O
the area of the oil/water interface at which lipolysis occurs.
CH2 O C
Luminal events also include lipolysis, mediated by a series of
Fatty acids plus
pancreatic and other enzymes, and uptake of the products of monoglyceride
lipolysis into micelles that can then transfer these molecules H2O
to the epithelial surface. Indeed, there is an ordered series of Pancreatic lipase
phase transitions that facilitate lipid assimilation. Oil droplets
are converted to lamellar, vesicular, and liquid crystalline FIGURE 59–1 Positional specificity of gastric (1) and
pancreatic (2) lipases. Both enzymes can digest triglycerides
product phases, and finally to micelles that contain the
but the products differ. (Modified with permission from Barrett KE:
products of lipolysis together with bile acids. Gastrointestinal Physiology. New York: Lange Medical Books/McGraw-Hill,
Medical Pub. Division, 2006.)

GASTRIC DIGESTION
Digestion of the lipid components of the diet begins in the the first position of triglyceride (Figure 59–1) and is subject to
stomach. Gastric peristalsis and mixing patterns provide a end-product inhibition such that gastric lipolysis is largely
shearing action that disperses triglyceride and phospholipids incomplete because the triglyceride molecule is not fully bro-
into a fine emulsion. The oil droplets can then be acted upon ken down to its component parts.
by gastric lipase, which binds to the surface of the oil droplets
where it can act on triglyceride molecules to generate free fatty INTESTINAL DIGESTION
acids and diglycerides. However, at the low pH of the gastric
lumen, the fatty acids become protonated and move into the As the pH increases in the small intestine, the fatty acids that
center of oil droplets. Thus, overall, gastric lipolysis is incom- were liberated by gastric lipase become ionized and orient
plete, and fails to generate products that are free to diffuse to themselves to the outside of the oil droplets. This surrounds
the mucosal surface. In general, 10–30% of overall lipolysis the droplet with a layer of ionized fatty acids that stabilizes the
takes place in the stomach in a healthy adult, and gastric fat emulsion. Because even long-chain fatty acids have some
digestion of lipids is not essential to their normal uptake. Gas- solubility in water, some will dissociate from the droplet and
tric lipase, the enzyme that initiates digestion in the lumen traverse the lumen to the intestinal epithelium. Fatty acids are
of the stomach, is specialized for activity in the unique condi- potent stimuli of cholecystokinin (CCK) release. CCK has a
tions that pertain in this segment of the gastrointestinal tract. number of actions that are pertinent to lipid digestion and
Most notably, the enzyme displays a pH optimum consistent absorption. First, it causes an increase in secretion of pancre-
with that of the gastric contents—4.0–5.5. The enzyme is also atic enzymes. Second, it relaxes the sphincter of Oddi, allow-
relatively resistant to the action of pepsin. Further, gastric ing outflow of the pancreatic juice into the intestinal lumen,
lipase is independent of the presence of any specific cofactors, and finally, it contracts the gallbladder, providing a bolus of
but is inhibited by bile acids in the duodenum. Finally, gastric concentrated bile that contains the bile acids needed eventu-
lipase acts preferentially to hydrolyze the fatty acid linked to ally to dissolve the products of lipolysis in mixed micelles.
CHAPTER 59 Lipid Assimilation 595

Enzymes and Other Factors 3


Involved in Digestion
Pancreatic acinar cells secrete a number of proteins that are C
L
important in fat digestion (Table 59–1). The first of these is
3
pancreatic lipase. This enzyme is functionally related to gastric

Fatty acids released


2 Bile acids
lipase, but it displays a number of important differences. First, Bile acids added
it has a different positional specificity, acting on both the 1 and L
2
3 positions of the glycerol molecule to liberate esterified fatty
acids (Figure 59–1). Thus, the products of pancreatic lipase are
fatty acids and monoglycerides. Second, it displays a pH opti- 1 Colipase added 1
mum in the neutral range. However, both gastric and pancre-
atic lipases share the property of being inhibited by bile acids. Fat L
This is not a major issue in the stomach, which is proximal to droplet
the entry of bile. How to solve this conundrum, on the other
hand, for pancreatic lipase? The answer lies in the presence of Time
a second product of pancreatic acinar cells, colipase. Colipase FIGURE 59–2 Role of colipase in promoting lipase activity in
is synthesized as an inactive precursor (procolipase) that is the intestinal lumen. Lipase (L) can absorb to the surface of fat
secreted in approximately equimolar amounts with lipase, and droplets (1), but is displaced by the binding of amphipathic bile acids
is activated by proteolytic cleavage in the intestinal lumen. that arrange themselves around the exterior of the oil droplet (2).
Colipase is capable of binding to both bile acids and lipase, Colipase (C) can bind to both bile acids and lipase, again bringing
which stabilizes the presence of lipase on the surface of oil lipase in proximity to its substrates in the oil droplet (3). (Modified with
permission from Barrett KE: Gastrointestinal Physiology. New York: Lange Medical
droplets. The significance of this interaction is shown in
Books/McGraw-Hill, Medical Pub. Division, 2006.)
Figure 59–2. If lipase alone is present, it adsorbs to the surface
of the oil droplets and generates free fatty acids. With addition
of bile acids, however, these array themselves on the surface of
including those of pancreatic duct cells, this delayed activation
the oil droplets and displace lipase, halting its enzymatic activ-
of the enzyme is important in the prevention of autodigestion of
ity. However, if colipase is also present, it can anchor lipase to
the pancreas. Phospholipase A2 breaks down dietary phospho-
the oil droplet, and thus lipolytic action is restored.
lipids by cleaving the fatty acid located in the 2 position of glyc-
Additional pancreatic exocrine products also contribute to
erol, as well as degrading (and thereby reclaiming) the
lipid digestion. Phospholipase A2 is also stored in pancreatic
phosphatidylcholine that is present in biliary secretions. The
acinar cells as an inactive proform that is activated by prote-
activity of this enzyme is dependent on luminal calcium. A final
olytic cleavage once it reaches the intestinal lumen. Because
lipolytic enzyme in the pancreatic juice is cholesterol esterase,
phospholipids are the major constituent of cell membranes,
also referred to as nonspecific esterase. This enzyme is capable
of degrading not only esters of cholesterol derived from dietary
sources, but also the esters of vitamins A, D, and E. Likewise,
TABLE 59–1 Mediators of intestinal lipolysis.
the broad specificity of this enzyme renders it capable of com-
Protein Source Activity Comments plete digestion of triglycerides, since it is capable of hydrolyzing
the 2-position fatty acid that is left untouched by both the gas-
Pancreatic Pancreatic Hydrolyzes 1 Inhibited by
tric and pancreatic lipases. Cholesterol esterase acts in the
lipase acinar cells and 3 positions bile acids
of triglyceride lumen as a tetramer, and, interestingly, the formation of this
complex depends on the presence of bile acids, distinguishing
Colipase Proform secreted Cofactor for Binds to lipase
by pancreatic lipase and to bile
this enzyme from both acid (i.e., gastric) and pancreatic lipases.
acinar cells acids It is also important to note that an enzyme closely related to
cholesterol esterase, known as breast milk lipase, is produced
Secretory Proform secreted Hydrolyzes fatty Requires
phospholipase by pancreatic acid in 2 position calcium for in the mammary gland of lactating females. This enzyme may
A2 acinar cells of phospholipids activity “predigest” the lipid components of breast milk, increasing the
Cholesterol Pancreatic Broad substrate Requires bile
efficiency of their uptake in the neonatal period. Breast milk
esterase acinar cells specificity— acids for lipase shares the broad specificity of cholesterol esterase.
cholesterol and activity
vitamin esters; 1,
2, and 3 positions Phase Transitions Involved
of triglyceride in Product Solubilization
Breast milk Mammary Related to Important Not only must dietary and endogenous lipids be digested by
lipase gland cholesterol in neonates
esterase
enzymes to allow for their assimilation into the body, but they
must also be trafficked across the intestinal lumen. This is
596 SECTION VIII GI Physiology

accomplished in the first instance by “phase transitions” that such limited aqueous solubility that they are essentially inca-
move the products of lipolysis from the oil droplets where they pable of absorption unless dissolved in micelles. This is the
are generated to the epithelial surface. case for cholesterol, fat-soluble vitamins, and plant sterols.
The phases involved in lipid absorption can actually be visu-
alized by mixing an emulsion of fat with lipase, colipase, and
bile acids on a microscope slide. The products of lipolysis that EPITHELIAL EVENTS IN
are released from the surface of the oil droplet form a lamellar
phase similar to the lipid bilayer that surrounds cells. Vesicles LIPID ASSIMILATION
bud off from this lamellar phase, which can be observed under
the microscope as a liquid crystalline phase that is clearly dis- BRUSH BORDER EVENTS
tinct from the oil droplets. Eventually, micelles are formed, or
a true solution of lipids in water. The phase transitions in the Mechanisms of Absorption
small intestine are directed to a single end—to transfer the of Lipolytic Products
products of lipolysis ultimately to the absorptive epithelium. In theory, the products of lipolysis have sufficient hydrophobic-
ity that they should be able to passively “flip” across the
enterocyte apical membrane by simply partitioning into the
ROLE OF BILE ACIDS/MICELLES lipid bilayer. Such a mechanism may indeed contribute to the
intestinal uptake of the various products of the mixed micelle,
As the gallbladder contracts, the meal encounters a bolus of with the exception of conjugated bile acids, which are exclu-
concentrated bile. The bile acids have an important role in sively absorbed via an active transport mechanism localized
solubilizing the end products of lipolysis and promoting their to the terminal ileum. However, there are likely also to be car-
transfer to the absorptive epithelium. As the ratio of bile acids rier-mediated mechanisms that assist in transporting the prod-
to lipolytic products increases, the relatively insoluble lipids ucts of lipid digestion across the microvillous membrane.
are incorporated into mixed micelles. The formation of these Uptake of plant sterols from the diet is inefficient. At least
structures is dependent on the physicochemical properties of part of the reason for this was revealed by studying the disease
bile acids. Likewise, micelle formation depends on the concen- known as sitosterolemia, in which patients accumulate
tration of bile acids in the lumen, since these structures cannot abnormally high levels of plant sterols in the plasma. This dis-
form unless bile acid concentrations exceed the “critical micel- ease is caused by mutations in two transporters, ABCG5 and
lar concentration” (CMC). Lipolytic products that are cap- ABCG8. Together, these proteins normally come together to
tured in micelles are truly in solution, compared with the form an efflux pump that transports any plant sterols that are
emulsion of oil droplets that has been their physical form hith- taken up into the enterocyte back out into the intestinal lumen
erto. The micelles that form in small intestinal content are piv- (Figure 59–3). The transporter so formed also can export cho-
otally dependent on the amphipathic nature of bile acids, with lesterol, albeit with less efficiency, explaining why the absorp-
both a hydrophilic and a hydrophobic face. The structure of tion of either dietary or biliary cholesterol is incomplete.
the mixed micelles in the intestine is analogous to that of the Enterocytes also express at least one specific pathway for choles-
biliary mixed micelles we discussed in Chapter 56 (Figure terol uptake, known as the Niemann–Pick C1-like 1 (NPC1L1)
56–3). The bile acids assemble such that their hydrophilic faces gene product (Figure 59–3).
are opposed to the aqueous environment. At the same time, In summary, uptake of the products of lipolysis appears to
the hydrophobic faces of the bile acid molecules can sequester involve a combination of passive transfer across the brush bor-
the products of lipolysis such that their solubility is measurably der membrane, as well as facilitated diffusion for some lipids
increased. The resulting mixed micelles carry fatty acids and that is mediated by specific protein carriers. Intestinal absorp-
other lipolytic products through the aqueous lumen, markedly tion of at least some lipids is additionally compromised by
increasing their rate of diffusion. Indeed, the rate-limiting step pumps that can efflux lipid substrates from the enterocyte
for lipid assimilation is the ability to present lipid molecules at cytosol, limiting their ability to enter the body.
a sufficient concentration at the brush border to provide for
uptake. While bile acids increase the efficiency of uptake of the
products of lipolysis, the majority of such products are not
Special Considerations for
dependent on micellar solubilization for their assimilation into Medium-chain Fatty Acids
the body. Fatty acids and monoglycerides have measurable Fatty acid chain length, likely via effects on aqueous solubility,
aqueous solubility and can diffuse in molecular form to also appears to have an important influence on the molecular
the brush border, where they can then be absorbed. This raises mechanisms that govern uptake of these molecules. Impor-
the concept of the “anatomic reserve” of the small intestine. tantly, medium-chain fatty acids, with 6–12 carbon atoms,
The normal surface area of the small intestine, and its length, have increased water solubility, and this means that they have
is sufficient to provide for molecular uptake of fatty acids and measurable absorption via the paracellular route. Medium-
monoglycerides even if micelles are absent, although transport chain fatty acids therefore appear to bypass the intracellular
rates are slower. On the other hand, some dietary lipids have processing events that are encountered by the long-chain fatty
CHAPTER 59 Lipid Assimilation 597

Cholesterol prior to export to the systemic circulation. Long-chain fatty


acids and monoglycerides are reesterified into triglyceride in
LUMEN the smooth endoplasmic reticulum; phospholipids, choles-
terol, and fat-soluble vitamins are also selectively trafficked

NPC1L1
ABC G5

ABC G8
and reesterified. Approximately 75% of absorbed fatty acids is
reassembled into triglyceride; the remainder is retained within
ATP the enterocyte for local needs.
The various reassembled lipids are then coated with proteins
ADP known as apoproteins for export from the enterocyte
(Figure 59–4). Apoproteins are synthesized in the rough endo-
plasmic reticulum, and undergo glycosylation in the Golgi appa-
ratus where they also encounter the reesterified lipids taken up
from the intestinal lumen. The particles that are formed via this
process are referred to as chylomicrons, and have a core of trig-
Utilized Chylomicron lyceride surrounded by phospholipids, cholesterol esters, and the
by enterocyte assembly apoproteins. The chylomicron is the structure used to transport
dietary lipids to other locations in the body. Approximately
80–90% (w/w) of the chylomicron is composed of triglyceride,
Exocytosis with 8–9% phospholipids and trace amounts of cholesterol, fat-
soluble vitamins, and protein. The resulting structure is exported
across the basolateral membrane by exocytosis.

LYMPHATIC UPTAKE
FIGURE 59–3 Intestinal handling of cholesterol. Cholesterol OF ABSORBED LIPID
in the intestinal lumen is taken up via the NPC1L1 transporter.
Absorbed cholesterol can be effluxed back out of the cell via the The physical form of lipid exported from the enterocyte deter-
ABCG5/ABCG8 efflux pump at the expense of ATP hydrolysis, can be mines the subsequent route this nutrient can take to leave the
retained for use in the enterocyte, or can be packaged with other gut. Chylomicrons range in size from 750 to 5,000 Å in diam-
absorbed lipids into chylomicrons, which leave the epithelial cell via eter. They are therefore too large to cross the intercellular
exocytosis. (Modified with permission from Barrett KE: Gastrointestinal Physiology. junctions linking capillary endothelial cells. This means that
New York: Lange Medical Books/McGraw-Hill, Medical Pub. Division, 2006.)
the only way for them to leave the intestine is via the lymphatics,
which have leakier junctions. However, eventually these chy-
lomicrons will enter the systemic circulation via the thoracic
acids that predominantly enter the enterocyte cytosol, dis-
cussed in greater detail later. As a result, such fatty acids also
follow a different route out of the gut, being exported chiefly
via the portal circulation rather than the lymphatic route used
by other lipids.
Smooth ER FA/MG Synthesis of TG and
TG
phospholipids
INTRACELLULAR PROCESSING
Synthesis of
apolipoproteins
Role of Fatty Acid Binding Proteins
Rough ER Apolipoprotein
The small intestinal epithelium expresses a family of low-molec- glycosylation
ular-weight binding proteins that are capable of binding fatty
Exocytosis
acids and other dietary lipids. The best studied of these are the Golgi
ileal-fatty acid binding protein (I-FABP) and the liver-type
(hepatic) FABP. These and related proteins participate in the Chylomicrons
directed trafficking of absorbed lipids to the smooth endoplas-
FIGURE 59–4 Secretion of chylomicrons by intestinal
mic reticulum, which is the site of intracellular lipid processing. epithelial cells. Absorbed fatty acids (FA) and monoglycerides (MG)
are reesterified to form triglyceride (TG) in the smooth endoplasmic
reticulum. Apolipoproteins are synthesized in the rough endoplasmic
Chylomicron Formation
reticulum and glycosylated, and then coated around lipid cores and
Unlike the products of protein and carbohydrate digestion, secreted from the basolateral pole of the enterocyte via a mechanism
which are exported to the body in their digested forms, the of exocytosis. (Reproduced with permission from Barrett KE, Barman SM, Boitano S,
products of lipid digestion are reassembled in the enterocyte Brooks H: Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.)
598 SECTION VIII GI Physiology

duct. The lipid-bearing chylomicrons serve to carry lipids to


ileum, the situation will be complicated further by a
various body tissues. They are too large to exit the bloodstream
decrease in the bile acid pool, in the absence of an anatomic
across the fenestrae of hepatic endothelial cells, and thus are
reserve that might otherwise compensate for this. Short
retained in the plasma, although components may leave the
bowel syndrome is usually the consequence of surgery—
chylomicron as needed at this and other body sites. Eventually,
either to remove segments of necrotic bowel in a pediatric
the chylomicrons are reduced to remnants that are small
condition known as necrotizing enterocolitis, which may
enough to pass into the space of Disse, and can finally have
have an infectious pathogenesis in some cases, or in patients
their remaining components recycled by hepatic metabolism.
who have undergone resections for extensive Crohn’s dis-
ease. Eventually, small bowel transplantation may offer
hope that such patients will be able to dispense with paren-
ABSORPTION OF teral nutrition, although problems with conserving bowel
FAT-SOLUBLE VITAMINS in a viable state prior to transplantation, as well as the usual
problems of rejection, have not yet fully been resolved.
Despite their importance, relatively little is understood about Likewise, some exciting experimental treatments that
the specific molecular basis of the absorption and handling of employ growth factors to increase mucosal mass may hold
fat-soluble vitamins by intestinal epithelial cells. It is known promise, but concerns that these therapies might predis-
that such vitamins are reesterified in the enterocyte, and incor- pose to intestinal malignancies must be resolved. This is
porated into the developing chylomicron. Presumably this is particularly the case in inflammatory bowel disease, where
the form in which they are trafficked to sites of need. Traffick- the risk of intestinal malignancy is already increased.
ing of fat-soluble vitamins to the brush border membrane is
essentially entirely dependent on micelles. A failure to form
micelles in the intestinal lumen will almost inevitably be fol-
lowed by deficiencies in fat-soluble vitamins in the body as a CHAPTER SUMMARY
whole, which may manifest clinically as rickets and osteomal- ■ Lipids are important constituents of most diets, make food
acia (see Chapter 64), night blindness (see Chapter 15), or an more palatable, and are energy-dense.
inability to effectively clot the blood, among others. Many ■ Lipid digestion and absorption involves a complex series of
fat-soluble vitamins are now available in more water-soluble events designed to emulsify, digest, and solubilize these
forms that can be used to treat such problems, such as prior to hydrophobic dietary constituents.
elective surgery. ■ Biliary excretion of cholesterol may substantially exceed dietary
intake.
■ Micelles increase the efficiency of absorption of the products of
triglyceride digestion, but they are not required for this process
in healthy individuals.
CLINICAL CORRELATION ■ Absorption of fat-soluble vitamins and cholesterol is entirely
dependent on micellar solubilization.
A 30-year-old woman suffering from inflammation of the ■ Absorbed lipids are esterified and repackaged in the enterocyte
ileum (Crohn’s disease) undergoes a surgical resection of into a structure known as a chylomicron, which then traffics
a long segment of her small bowel, including the terminal absorbed lipids around the body, initially bypassing the portal
ileum. The remaining proximal segment of small bowel is circulation.
connected directly to the ileocecal valve. After recovering ■ Chylomicrons consist predominantly of resynthesized
from her surgery and resuming oral feeding, she notes triglyceride, but apoproteins are essential components.
that diarrheal symptoms have returned, and fears a recur- ■ Lipid digestion is a highly efficient process and significant
rence of her bowel inflammation. However, in contrast to excess capacity exists in the system.
the bloody diarrhea with cramping that she experienced
previously, now her stools are large, bulky, and foul-
smelling, but with no blood. She also experiences some STUDY QUESTIONS
abdominal bloating. Examination of a fecal smear under
1. A patient with obstructive jaundice who is scheduled for
the microscope reveals prominent lipid droplets. gallbladder surgery is found to have an increased prothrombin
Short bowel syndrome is defined as being present in a time, which indicates an increase in blood clotting time. This
patient who no longer has sufficient mucosal area for the laboratory finding is most likely due to malabsorption of which
adequate uptake of nutrients via the enteric route, and of the following vitamins?
thus such patients may require parenteral nutritional sup- A) A
port (i.e., via the intravenous route). Even if sufficient B) B12
bowel remains for absorption of the majority of nutrients, C) D
if the segment of missing bowel includes the terminal D) E
E) K
CHAPTER 59 Lipid Assimilation 599

2. A patient is treated for hypercholesterolemia with 4. A patient with a long-standing history of mild cystic fibrosis
cholestyramine, a resin that binds bile acid molecules. notices that his stools are becoming bulky and oily. Laboratory
Absorption of which of the following is likely to be abnormal tests confirm steatorrhea. Which of the following is not involved
in this patient? in the apparently decreased fat assimilation in this patient?
A) long-chain triglyceride A) lipase inactivation
B) medium-chain triglyceride B) decreased pancreatic lipase output
C) starch C) reduced pancreatic bicarbonate secretion
D) vitamin D D) loss of the anatomic reserve
E) vitamin C E) decreased colipase synthesis
3. A mouse was constructed in which the expression of NPC1L1 5. An otherwise healthy child is found to have mild, intermittent
was knocked out by genetic targeting. Assimilation of which of steatorrhea but no evidence of malabsorption of fat-soluble
the following substances from the diet would be expected to be vitamins. There are also no defects in protein or carbohydrate
abnormal in this animal? assimilation. His brother was similarly affected. Lack of which
A) triglyceride of the following is most consistent with the clinical picture?
B) vitamin D A) bile acid micelles
C) vitamin E B) cholesterol esterase
D) cholesterol C) L-FABP
E) phospholipids D) gastric lipase
E) pancreatic lipase
This page intentionally left blank
SECTION IX ENDOCRINE AND
METABOLIC PHYSIOLOGY

60
C H A P T E R

General Principles of
Endocrine Physiology
Patricia E. Molina

O B J E C T I V E S

■ Contrast the terms endocrine, paracrine, and autocrine.


■ Define the terms hormone, target cell, and receptor.
■ Understand the major differences in cellular mechanisms of action of
peptides, steroid, and thyroid hormones.
■ Understand the role of hormone-binding proteins.
■ Understand the feedback control mechanisms of hormone secretion.
■ Explain the effects of secretion, degradation, and excretion on plasma
hormone concentrations.

THE ENDOCRINE SYSTEM: hormones ranging from small peptides to glycoproteins, which
exert their effects in either neighboring or distant target cells.
PHYSIOLOGIC FUNCTIONS This endocrine network of organs and mediators does not
work in isolation, and is closely integrated with the central and
AND COMPONENTS peripheral nervous systems as well as with the immune sys-
Some of the key functions of the endocrine system include: tems, leading to currently used terminology such as “neuroen-
docrine” or “neuroendocrine–immune” systems for describing
• regulation of sodium and water balance and control of blood their interactions. Three basic components make up the core
volume and pressure; of the endocrine system:
• regulation of calcium and phosphate balance to preserve ex-
tracellular fluid concentrations required for cell membrane • Endocrine glands: The classic endocrine glands are duct-
integrity and intracellular signaling; less and secrete their chemical products (hormones) into
• regulation of energy balance and control of fuel mobilization, the interstitial space from where they reach the circulation.
utilization, and storage to ensure that cellular metabolic Unlike the cardiovascular, renal, and digestive systems, the
demands are met; endocrine glands are not anatomically connected and are
• coordination of the responses to stress; scattered throughout the body (Figure 60–1). Communica-
• regulation of reproduction, development, growth, and se- tion among the different organs is ensured through the
nescence. release of hormones or neurotransmitters.
• Hormones— Hormones are chemical products, released
The endocrine system is an integrated network of multiple in very small amounts from the cell, that exert a biologic
organs derived from different embryologic origins that release action on a target cell. Hormones can be released from

601

Ch60_601-612.indd 601 11/26/10 10:31:12 AM


602 SECTION IX Endocrine and Metabolic Physiology

Hypothalamus
Releasing hormones:
GHRH, CRH, Pituitary gland
TRH, GnRH Growth hormone,
Inhibitory hormones: Prolactin
Somatostatin, ACTH, MSH
Dopamine TSH, FSH, & LH
Projecting to Posterior Pituitary:
Antidiuretic Hormone
Oxytocin
Parathyroid glands
Parathyroid hormone
Thyroid gland
T3, T4, & calcitonin
FIGURE 60–1 The classic endocrine system.
Endocrine organs are located throughout the body,
and their function is controlled by hormones delivered Adrenal glands
Cortisol
through the circulation or produced locally or by direct Aldosterone
neuroendocrine stimulation. Integration of hormone Adrenal androgens Pancreas
production from endocrine organs is under regulation Epinephrine Insulin
by the hypothalamus. Many other tissues are now Norepinephrine Glucagon
known to produce hormones and can thus be Somatostatin
considered part of the endocrine system. Examples
include adipose tissue (e.g., leptin), the gastrointestinal
tract (e.g., ghrelin), skeletal muscle (myokines), the
kidneys (erythropoietin), and the heart (e.g., atrial Ovaries
natriuretic peptide). GHRH, growth hormone–releasing Estrogens
Progesterone
hormone; CRH, corticotropin-releasing hormone; TRH,
thyrotropin-releasing hormone; GnRH, gonadotropin-
releasing hormone; ACTH, adrenocorticotropic
hormone; MSH, melanocyte-stimulating hormone; TSH, Testes
thyroid-stimulating hormone; FSH, follicle-stimulating Testosterone
hormone; LH, luteinizing hormone; T3, triiodothyronine;
T4, thyroxine. (Modified with permission from Molina PE:
Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

the endocrine glands (i.e., insulin, cortisol); the brain (i.e., PROTEIN OR PEPTIDE HORMONES
corticotropin-releasing hormone, oxytocin, and antidiuretic
hormone); and other organs such as the heart (atrial natriuretic Protein or peptide hormones constitute the majority of hor-
peptide), liver (insulin-like growth factor-I), and adipose tis- mones. Most are synthesized as preprohormones and
sue (leptin). undergo posttranslational processing. They are stored in
• Target organ: The target organ contains cells that express secretory granules before being released by exocytosis
hormone-specific receptors and that respond to hormone (Figure 60–2). Examples of peptide hormones include insu-
binding by a demonstrable biologic response. lin, glucagon, and adrenocorticotropic hormone (ACTH).
Some hormones in this category, such as the gonadotropic
hormone, luteinizing hormone (LH), and follicle-stimulating
HORMONE CHEMISTRY AND hormone (FSH), together with thyroid-stimulating hormone
MECHANISMS OF ACTION (TSH) and human chorionic gonadotropin (hCG), contain
carbohydrate moieties, leading to their designation as glyco-
Based on their chemical structure, hormones can be classified proteins.
into proteins (or peptides), steroids, and amino acid derivatives
(amines). Hormone structure, to a great extent, dictates the loca-
tion of the hormone receptor, with amines and peptide hormones STEROID HORMONES
binding to receptors in the cell surface and steroid hormones
able to cross plasma membranes and bind to intracellular recep- Steroid hormones are derived from cholesterol and are syn-
tors. An exception to this generalization is thyroid hormone, an thesized in the adrenal cortex, gonads, and placenta. They are
amino acid–derived hormone that is transported into the cell in lipid soluble, require binding proteins to circulate in plasma,
order to bind to its nuclear receptor. Hormone structure influ- and cross the plasma membrane to bind to intracellular
ences the half-life of the hormone as well, with peptides having cytosolic or nuclear receptors. Vitamin D and its metabolites
shorter half-lives than steroid hormones. are also steroid hormones.
CHAPTER 60 General Principles of Endocrine Physiology 603

Endocrine cell

Granular endoplasmic
reticulum Synthesis

Preprohormone

Nucleus Prohormone

Packaging

Golgi Prohormone
apparatus
Hormone

Storage
Secretory Hormone
vesicles

Plasma
membrane
Secretion
Hormone
(and any
Cytosol "pro" fragments)

Interstitium
Ca2+
FIGURE 60–2 Peptide hormone synthesis. Peptide hormones are synthesized as preprohormones in the ribosomes and processed
to prohormones in the endoplasmic reticulum (ER). In the Golgi apparatus, the hormone or prohormone is packaged in secretory vesicles,
the contents of which are released from the cell in response to an influx of Ca2+. The increase in cytoplasmic Ca2+ is required for docking of the
secretory vesicles in the plasma membrane and for exocytosis of the vesicular contents. The hormone and the products of the posttranslational
processing that occurs inside the secretory vesicles are released into the extracellular space. Examples of peptide hormones are ACTH, insulin,
growth hormone, and glucagon. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

AMINO ACID–DERIVED HORMONES duces a biologic effect on the same cell that released it, and intra-
crine when the hormone has an intracellular effect without first
Amino acid–derived hormones are synthesized from the being released into the extracellular space.
amino acid tyrosine and include the catecholamines norepi-
nephrine, epinephrine, and dopamine, as well as the thyroid HORMONE TRANSPORT
hormones, derived from the combination of two iodinated
tyrosine amino acid residues. Hormones released into the circulation can circulate either
freely or bound to carrier proteins, also known as binding
HORMONE EFFECTS proteins. The binding proteins serve as a reservoir for the
hormone and prolong the hormone’s half-life, the time during
The biologic effect of a hormone can be classified in many ways which the concentration of a hormone decreases to 50% of its
(Figure 60–3). The effect is endocrine when a hormone is initial concentration. The free or unbound hormone is the
released into the circulation and then travels in the blood to pro- active form of the hormone, which binds to the specific
duce a biologic effect on distant target cells. The effect is para- hormone receptor. Hormone binding to its carrier protein
crine when a hormone released from one cell produces a biologic serves to regulate the activity of the hormone by determining
effect on a neighboring cell, which is frequently a cell in the same how much hormone is free to exert a biologic action. Most
organ or tissue. The effect is autocrine when a hormone pro- carrier proteins are globulins and are synthesized in the liver.
604 SECTION IX Endocrine and Metabolic Physiology

Interstitial space

Endocrine cell

Hormone

Circulatory Paracrine signaling


system

Hormone-specific
receptor

Endocrine signaling Autocrine


signaling

Intracrine
signaling

FIGURE 60–3 Mechanisms of hormone action. Depending on where hormones exert their effects, they can be classified into endocrine,
paracrine, autocrine, or intracrine mediators. Hormones that enter the bloodstream and bind to hormone receptors in target cells in distant organs
mediate endocrine effects. Hormones that bind to cells near the cell that released them mediate paracrine effects. Hormones that produce their
physiologic effects by binding to receptors on the same cell that produced them mediate autocrine or intracrine effects. (Modified with permission from
Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

As a result, alterations in hepatic function may result in abnor- cortisol synthesis and release from the adrenal glands. This
malities in binding protein levels and may indirectly affect feedback mechanism restores free cortisol levels and prevents
total hormone levels. The majority of amines, peptides, and manifestation of cortisol deficiency.
protein (hydrophilic) hormones circulate in their free form. The half-life of a hormone is inversely related to its rate of
Steroid and thyroid (lipophilic) hormones circulate bound to removal from the circulation. Once hormones are released
specific transport proteins. into the circulation, they can bind to their specific receptor in
The interaction between a given hormone and its carrier a target organ, they can undergo metabolic transformation by
protein is in a dynamic equilibrium and allows adjustments the liver, or they can undergo urinary excretion. In the liver,
that prevent clinical manifestations of hormone deficiency or hormones can be inactivated through Phase I (hydroxylation
excess. Secretion of the hormone is adjusted rapidly following or oxidation) and/or Phase II (glucuronidation, sulfation, or
changes in the levels of carrier proteins. For example, plasma reduction with glutathione) reactions, and then excreted by
levels of cortisol-binding protein increase during pregnancy. the liver through the bile or by the kidney. In some instances,
The increase in circulating levels of cortisol-binding protein the liver can actually modify a hormone precursor, as is the
leads to an increased binding capacity for cortisol (a steroid case for vitamin D synthesis. Hormones can be degraded at
hormone produced in the adrenal glands), resulting in a their target cell through internalization of the hormone–
decrease in free cortisol levels. This decrease in free cortisol receptor complex followed by lysosomal degradation of the
stimulates the hypothalamic release of CRH, which stimulates hormone. Only a very small fraction of total hormone produc-
ACTH release from the anterior pituitary and consequently tion is excreted intact in the urine and feces.
CHAPTER 60 General Principles of Endocrine Physiology 605

HORMONE CELLULAR EFFECTS onists are widely used clinically to restore endocrine function
in patients with hormone deficiency or excess.
The biologic response to hormones is elicited through binding
to hormone-specific receptors at the target organ. Hormones
circulate in very low concentrations (10−7 to 10−12 M), so the HORMONE RECEPTORS AND
receptor must have high affinity and specificity for the hor-
mone to produce a biologic response.
SIGNAL TRANSDUCTION
Affinity is determined by the rates of association and Hormones produce their biologic effects by binding to specific
dissociation for the hormone–receptor complex under equi- hormone receptors in target cells, and the type of receptor to
librium conditions. It is a reflection of how tight the hor- which they bind is largely determined by the hormone’s chem-
mone–receptor interaction is. Specificity is the ability of a ical structure. Hormone receptors are classified depending on
hormone receptor to discriminate between hormones with their cellular localization, as cell membrane (Figures 60–4
related structures. and 60–5) or intracellular receptors (Figure 60–6).
The binding of hormones to their receptors is saturable,
with a finite number of hormone receptors to which a hor-
mone can bind. Abnormal endocrine function is the result of CELL MEMBRANE RECEPTORS
either excess or deficiency in hormone action. This can result
from abnormal production of a given hormone (either in These receptor proteins are located within the phospholipid
excess or in insufficient amounts) or from decreased receptor bilayer of the cell membrane of target cells (Figures 60–4 and
number or function. Hormone–receptor agonists and antag- 60–5). Functionally, cell membrane receptors can be divided

Amino acid derived: Peptide and protein:


Epinephrine, norepinephrine Glucagon, Angiotensin, GnRH, SS,
GHRH, FSH, LH, TSH, ACTH
N

E1 E2 E3

α γ γ
β β Ion channels,
GDP PI3Kγ, PLC-β, Biological responses
I1 I2 I3
adenylate cyclases Proliferation, differentiation,
C development, cell survival,
angiogenesis, hypertrophy,
cancer
G-protein−independent
effector molecules

αi αq αs α12

GTP GTP GTP GTP Gene expression


Adenylate PLC-β Adenylate RhoGEFs regulation
P
cyclase DAG cyclase Rho
cAMP Ca++ cAMP P
PKC

Transcription
Nucleus
factors

FIGURE 60–4 G protein–coupled receptors. Peptide and protein hormones bind to cell surface receptors coupled to G proteins. Binding
of the hormone to the receptor produces a conformational change that allows the receptor to interact with the G proteins. This results in the
exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) and activation of the G protein. The second-messenger systems
that are activated vary depending on the specific receptor, the α-subunit of the G protein associated with the receptor, and the ligand it binds.
Examples of hormones that bind to G protein–coupled receptors are shown. DAG, diacylglycerol; PLC, phospholipase C; cAMP, cyclic adenosine
monophosphate; RhoGEFs, Rho guanine nucleotide exchange factors; PI3K, phosphatidyl-3-kinase; PKC, protein kinase C; GnRH, gonadotropin-
releasing hormone; SS, somatostatin; GHRH, growth hormone–releasing hormone; FSH, follicle-stimulating hormone; LH, luteinizing hormone;
TSH, thyroid-stimulating hormone; ACTH, adrenocorticotropin hormone. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York:
McGraw-Hill Medical, 2010.)
606 SECTION IX Endocrine and Metabolic Physiology

N N
N N Growth
factor

Plasma Plasma
membrane membrane

Tyrosine Non-
kinase receptor Cytosol
domain tyrosine
kinase
C C

ATP Kinase
PO4
ADP JAK kinase
Docking
protein
Docking Protein +ATP Protein-PO4 + ADP
protein

Recruitment & Protein activation


activation of proteins
FIGURE 60–5 Receptor kinase and receptor-linked kinase receptors. Receptor kinases are receptors that have intrinsic tyrosine or
serine kinase activity that is activated by binding of the hormone to the amino terminal of the cell membrane receptor. The activated kinase
recruits and phosphorylates downstream proteins producing a cellular response. An example of hormones that utilize this receptor pathway
is insulin. Receptor-linked tyrosine kinase receptors do not have intrinsic activity in their intracellular domain. They are closely associated with
kinases that are activated with binding of the hormone. Examples of hormones using this mechanism are growth hormone and prolactin.
ATP, adenosine triphosphate; ADP, adenosine diphosphate; JAK, janus kinase. (Modified with permission from Cooper GM: The Cell: A Molecular Approach,
4th ed. Sinauer, 2007.)

Extracellular Cytoplasm Nucleus


fluid

Steroid Activated
Steroid enters target
cell by diffusion receptor dimer

Gene
transcription

Steroid DNA
receptor Steroid response
element

Nuclear
Plasma envelope
membrane

FIGURE 60–6 Intracellular receptors. The general scheme for the mechanism of action of steroid receptors. The ligand (steroid) diffuses
into the cell and binds to the cytosolic receptor. Once binding occurs, the receptors dimerize (pair up) and are translocated to the nucleus where
they bind to a steroid response element on the DNA. This activates gene transcription to mRNA, and ultimately, through increased mRNA
translation to specific proteins, results in a cellular response. Some intracellular receptors, rather than being in the cytosol in the unbound state,
reside in the nucleus (e.g. thyroid hormone receptors), but their ultimate mechanisms of activation of gene transcription and translation are
similar. (Reproduced with permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)
CHAPTER 60 General Principles of Endocrine Physiology 607

into ligand-gated ion channels and receptors that regulate producing cellular responses. The action of cAMP is termi-
activity of intracellular proteins. nated by the breakdown of cAMP by the enzyme phosphodi-
esterase. In addition, the cascade of protein activation can also
be controlled by phosphatases, which dephosphorylate pro-
Ligand-gated Ion Channels teins. Phosphorylation of proteins does not necessarily result
These receptors are functionally coupled to ion channels. Hor- in activation of an enzyme. In some cases, phosphorylation of
mone binding to this receptor produces a conformational a given protein results in inhibition of its activity.
change that opens ion channels on the cell membrane, pro- Gαq activation of phospholipase C results in the hydrolysis
ducing ion fluxes in the target cell. The cellular effects occur of phosphatidylinositol bisphosphate and the production of
within seconds of hormone binding. diacylglycerol (DAG) and inositol trisphosphate (IP3). DAG
activates protein kinase C, which phosphorylates effector pro-
Receptors that Regulate Activity teins. IP3 binds to calcium channels in the endoplasmic reticu-
of Intracellular Proteins lum, leading to an increase of Ca2+ influx into the cytosol. Ca2+
can also act as a second messenger by binding to cytosolic pro-
These receptors are transmembrane proteins that transmit sig- teins such as calmodulin. Binding of Ca2+ to calmodulin
nals to intracellular targets when activated. Ligand binding to results in the activation of kinases, leading to a cascade of
the receptor on the cell surface and activation of the associated phosphorylation of effector proteins and cellular responses.
protein initiate a signaling cascade of events that activates
intracellular proteins and enzymes and that can include effects Receptor protein tyrosine kinases
on gene transcription and expression. The main types of cell
Receptor protein tyrosine kinases are usually single trans-
membrane hormone receptors in this category are the G pro-
membrane proteins that have intrinsic enzymatic activity that
tein–coupled receptors (Figure 60–4) and the receptor protein
is activated by hormone binding. This results in phosphoryla-
tyrosine kinases (Figure 60–5). An additional type of receptor,
tion of tyrosine residues on the catalytic domain of the recep-
the receptor-linked kinase receptor, activates intracellular
tor itself, increasing its kinase activity. Phosphorylation outside
kinase activity following binding of the hormone to the plasma
the catalytic domain creates specific binding or docking sites
membrane receptor. This type of receptor is used in producing
for additional proteins that are recruited and activated, initiat-
the physiologic effects of growth hormone (Figure 60–5).
ing a downstream signaling cascade.
Hormone binding to cell surface receptors results in rapid
G protein–coupled receptors activation of cytosolic proteins and cellular responses. Through
G protein–coupled receptors are transmembrane proteins cou- protein phosphorylation, hormone binding to cell surface
pled to heterotrimeric guanine-binding proteins (G proteins) receptors can also alter the transcription of specific genes
consisting of three subunits: α, β, and γ. Hormone binding to through the phosphorylation of transcription factors. An
the G protein–coupled receptor produces a conformational example of this mechanism of action is the phosphorylation of
change that induces interaction of the receptor with the regula- the transcription factor cAMP response element-binding
tory G protein, stimulating the release of guanosine diphos- protein (CREB) by protein kinase A in response to receptor
phate (GDP) in exchange for guanosine triphosphate (GTP), binding and adenylate cyclase activation. This same transcrip-
resulting in activation of the G protein (Figure 60–4). The acti- tion factor (CREB) can be phosphorylated by calcium–calm-
vated G protein (bound to GTP) dissociates from the receptor odulin following hormone binding to receptor tyrosine kinase
followed by dissociation of the α from the βγ subunits. The and activation of phospholipase C. Therefore, hormone bind-
subunits activate intracellular targets, which can be either an ing to cell surface receptors can elicit immediate responses
ion channel or an enzyme. Hormones that use this type of when the receptor is coupled to an ion channel or through the
receptor include TSH, antidiuretic hormone or arginine vaso- rapid phosphorylation of preformed cytosolic proteins, and it
pressin, and catecholamines. can also activate gene transcription through phosphorylation
On the basis of the Gα subunit, G proteins can be classified of transcription factors.
into four families associated with different effector proteins.
The signaling pathways of three of these have been extensively
studied. The Gαs activates adenylate cyclase, Gαi inhibits INTRACELLULAR RECEPTORS
adenylate cyclase, and Gαq activates phospholipase C; the
second-messenger pathways used by Gα12 have not been com- Receptors in this category belong to the steroid receptor
pletely elucidated. superfamily (Figure 60–6). These receptors are transcription
The interaction of Gαs with adenylate cyclase and its activa- factors that have binding sites for the hormone (ligand) and
tion result in increased conversion of adenosine triphosphate for DNA and function as ligand (hormone)-regulated tran-
to cyclic 3′,5′-adenosine monophosphate (cAMP), with the scription factors. Hormone–receptor complex formation and
opposite response elicited by binding to Gαi-coupled receptors. binding to DNA result in either activation or repression of
The increase in intracellular cAMP activates protein kinase A, gene transcription. Binding to intracellular hormone recep-
which in turn phosphorylates effector proteins, responsible for tors requires that the hormone be hydrophobic and cross the
608 SECTION IX Endocrine and Metabolic Physiology

plasma membrane. Steroid hormones and the active steroid cyte adrenergic receptors following sustained elevations in
derivative of vitamin D fulfill this requirement (Figure 60–6). thyroid hormone levels.
Thyroid hormones must be actively transported into the cell.
The distribution of the unbound intracellular hormone
receptor can be cytosolic or nuclear. Hormone–receptor com- CONTROL OF HORMONE RELEASE
plex formation with cytosolic receptors produces a conforma-
tional change that allows the hormone–receptor complex to The secretion of hormones involves synthesis or production of
enter the nucleus and bind to specific DNA sequences to regu- the hormone and its release from the cell. In general, the dis-
late gene transcription. Once in the nucleus, the receptors cussion of regulation of hormone release in this section refers
regulate transcription by binding, generally as dimers, to hor- to both synthesis and secretion; specific aspects pertaining to
mone response elements normally located in regulatory the differential control of synthesis and release of specific hor-
regions of target genes. In all cases, hormone binding leads to mones will be discussed in the respective chapters when they
a nearly complete nuclear localization of the hormone– are considered of relevance.
receptor complex. Unbound intracellular receptors may be Plasma levels of hormones oscillate throughout the day,
located in the nucleus, as in the case of thyroid hormone recep- showing peaks and troughs that are hormone specific
tors. The unoccupied thyroid receptor represses transcription (Figure 60–7). This variable pattern of hormone release is
of genes. Binding of thyroid hormone to the receptor activates determined by the interaction and integration of multiple con-
gene transcription. trol mechanisms, which include hormonal, neural, nutritional,
and environmental factors that regulate the constitutive (basal)
HORMONE–RECEPTOR REGULATION and stimulated (peak levels) secretion of hormones. The peri-
odic and pulsatile release of hormones is critical in maintain-
Hormones can influence responsiveness of the target cell by ing normal endocrine function and in exerting physiologic
modulating receptor function. Target cells are able to detect effects at the target organ. The hypothalamus plays an impor-
changes in hormone signal over a very wide range of stimulus tant role in control of hormone pulsatility. Although the
intensities. This requires the ability to undergo a reversible mechanisms that determine the pulsatility and periodicity of
process of adaptation or desensitization, whereby a pro- hormone release are not completely understood for all the dif-
longed exposure to a hormone decreases the response to that ferent hormones, three general mechanisms can be identified
level of hormone. This allows cells to respond to changes in as common regulators of hormone release.
the concentration of a hormone (rather than to the absolute
concentration of the hormone) over a very wide range of hor-
mone concentrations. Several mechanisms can be involved in NEURAL CONTROL
desensitization to a hormone. Hormone binding to cell-
Control and integration by the central nervous system is a key
surface receptors, for example, may induce their endocytosis
component of hormonal regulation and is mediated by direct
and temporary sequestration in endosomes. Such hormone-
neurotransmitter control of endocrine hormone release
induced receptor endocytosis can lead to the destruction of
(Figure 60–8). Neural control plays an important role in the
the receptors in lysosomes, a process that leads to receptor
regulation of peripheral endocrine hormone release. Endo-
downregulation. In other cases, desensitization results from
crine organs such as the pancreas receive sympathetic and
a rapid inactivation of the receptors, for example, as a result of
parasympathetic input, which contributes to the regulation of
a receptor phosphorylation. Desensitization can also be
insulin and glucagon release.
caused by a change in a protein involved in signal transduc-
tion following hormone binding to the receptor or by the pro-
duction of an inhibitor that blocks the transduction process. HORMONAL CONTROL
In addition, one hormone can downregulate or decrease the
expression of receptors for another hormone and reduce that Hormone release from an endocrine organ is frequently con-
hormone’s effectiveness. Hormone receptors can also undergo trolled by another hormone. When the outcome is stimulation
upregulation. of hormone release, the hormone that exerts that effect is
Upregulation of receptors involves an increase in the num- referred to as a tropic hormone, as is the case for most of the
ber of receptors for the particular hormone and frequently hormones produced and released from the anterior pituitary.
occurs when the prevailing levels of the hormone have been One example of this type of hormone release control is the
low for some time. The result is an increased responsiveness to regulation of glucocorticoid release by ACTH. Hormones can
the physiologic effects of the hormone at the target tissue also suppress another hormone’s release. An example of this is
when the levels of the hormone are restored or when an ago- the inhibition of growth hormone release by somatostatin
nist to the receptor is administered. A hormone can also from the hypothalamus.
upregulate the receptors for another hormone, increasing the Hormonal inhibition of hormone release plays an important
effectiveness of that hormone at its target tissue. An example role in the process of negative feedback regulation of hormone
of this type of interaction is the upregulation of cardiac myo- release, described below and in Figure 60–9. In addition, hor-
CHAPTER 60 General Principles of Endocrine Physiology 609

Cortisol
600

500

Cortisol (nmoI/L)
400

300

200

100

0
09 11 13 15 17 19 21 23 01 03 05 07 09
Clock time

Growth Hormone
30
Sleep

25
Plasma growth hormone

20
(mIU/L)

15

10

0
09 11 13 15 17 19 21 23 01 03 05 07 09
Clock time
FIGURE 60–7 Patterns of hormone release. Plasma hormone concentrations fluctuate throughout the day. Therefore, plasma hormone
measurements do not always reflect the function of a given endocrine system. Both cortisol and growth hormone show considerable variations
in blood levels throughout the day. These levels can also be affected by sleep deprivation, light, stress, and disease, and are dependent on their
secretion rate, rates of metabolism and excretion, metabolic clearance rate, circadian pattern, fluctuating environmental stimuli, and internal
endogenous oscillators. Biologic influences include illness, night work, sleep patterns, changes in longitude, and prolonged bed rest. (Modified
with permission from Molina PE: Endocrine Physiology, 2nd ed. New York: McGraw-Hill Medical, 2007.)

mones can stimulate the release of a second hormone in what plasma glucose levels and the control of parathyroid hormone
is known as a feedforward mechanism. release by plasma calcium and phosphate levels.
Release of one hormone can be influenced by more than one
of these mechanisms. For example, insulin release is regulated
NUTRIENT OR ION REGULATION by nutrients (plasma levels of glucose and amino acids), neural
(sympathetic and parasympathetic stimulation), and hormonal
Plasma levels of nutrients or ions can also regulate hormone (somatostatin) mechanisms. The ultimate function of these con-
release (Figure 60–9). In all cases, the particular hormone reg- trol mechanisms is to allow the neuroendocrine system to adapt
ulates the concentration of the nutrient or ion in plasma either to a changing environment, integrate signals, and maintain
directly or indirectly. Examples of nutrient and ion regulation homeostasis. The responsiveness of target cells to hormonal
of hormone release include the control of insulin release by action leading to regulation of hormone release constitutes a
610 SECTION IX Endocrine and Metabolic Physiology

Preganglionic

Neuron Neuron

ACh

Postganglionic
SNS or PSNS Neuron

FIGURE 60–8 Neural control of hormone


release. Endocrine function is under
tight regulation by the nervous system, hence the
term “neuroendocrine.” Hormone release by ACh or NE ACh
endocrine cells can be modulated by
postganglionic neurons from the sympathetic
nervous system (SNS) or parasympathetic
nervous system (PSNS) using acetylcholine
(ACh) or norepinephrine (NE) as
neurotransmitters, or by preganglionic neurons Endocrine Adrenomedullary
using acetylcholine as a neurotransmitter. cell cell
Therefore, pharmacologic agents that interact
with the production or release of
neurotransmitters affect endocrine function.
(Modified with permission from Molina PE: Endocrine
Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.) Hormone release Hormone release

feedback control mechanism. A dampening or inhibition of the INTERPRETATION OF


initial stimulus is called negative feedback. Stimulation or
enhancement of the original stimulus is called positive feedback
HORMONE MEASUREMENTS
(Figure 60–9). Negative feedback is the most common control Because of the variability in circulating hormone levels result-
mechanism regulating hormone release. The integrity of the sys- ing from pulsatile release, circadian rhythms, sleep/wake
tem ensures that adaptive changes in hormone levels do not lead cycle, and nutritional status, interpretation of isolated plasma
to pathologic conditions. Furthermore, the control mechanism hormone measurements should always be done with caution
plays an important role in short- and long-term adaptations to and with understanding of the integral components of the
changes in the environment. Three levels of feedback can be hormone axis in question.
identified: long loop, short loop, and ultrashort loop. Some general aspects that should be considered when inter-
preting hormone measurements are as follows:
ASSESSMENT OF • Hormone levels should be evaluated with their appropriate
regulatory factors (e.g., insulin with glucose, calcium with
ENDOCRINE FUNCTION parathyroid hormone, thyroid hormone with TSH, etc.).
In general, disorders of the endocrine system result from • Simultaneous increase of pairs (increase in both the hor-
alterations in hormone secretion or target cell responsiveness to mone and the substrate that it regulates, such as increased
hormone action. Alterations in target cell response can be due plasma glucose and insulin levels) can indicate a hormone
to increased or decreased biologic responsiveness to a particular resistant state.
hormone (Figure 60–10). The initial approach to assessment of • Urinary excretion of hormone or hormone metabolites over
endocrine function is measurement of plasma hormone levels. 24 hours may be a better estimate of hormone secretion
CHAPTER 60 General Principles of Endocrine Physiology 611

a. Negative feedback A. Decreased hormone responsiveness


150
TSH Maximal response

Hormone effect (%)


Effect produced by
saturating dose
100
of hormone
Anterior Thyroid
pituitary gland
50

Thyroid 0
hormone Log [hormone]

b. Positive feedback
B. Decreased hormone sensitivity
LH 150

Hormone effect (%)


100
Hypothalamus
Ovary
Pituitary Sensitivity
50 Hormone concentration
required to elicit half-
maximal response
Estradiol 0
Log [hormone]
c. Product regulation
FIGURE 60–10 Receptor function. A. Hormone
PTH responsiveness. Decreased responsiveness to hormone effects can
be due to a decreased number of hormone receptors, a decreased
concentration of enzyme activated by the hormone, an increased
Parathyroid concentration of noncompetitive inhibitor, or a decreased number of
Bone
gland target cells. When responsiveness is decreased, then no matter how
high the hormone concentration is, a maximal response will not be
achieved. B. Hormone sensitivity. A decrease in hormone sensitivity
requires higher hormone concentrations to produce 50% of the
Ca++
maximal response. Decreased sensitivity can be due to decreased
FIGURE 60–9 Mechanisms regulating hormone release hormone–receptor affinity, decreased hormone–receptor number,
a. Negative feedback regulation. In some cases, the endocrine gland is increased rate of hormone degradation, and increased levels of
itself a target organ for another hormone. In this case, endocrine cells antagonistic or competitive hormones. (Modified with permission from
Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
from organ 1 produce a hormone that stimulates the target organ to
produce another hormone (hormone 2). Hormone 2 decreases the
production and release of the hormone that stimulated its release, also
known as a tropic hormone. An example is the regulation of anterior
pituitary release of thyroid-stimulating hormone (TSH) by thyroid TABLE 60–1 Interpretation of hormone levels.
hormones produced by the thyroid gland. b. Positive feedback
regulation, occurs when the release of a hormone stimulates a second Pituitary Target Hormone Level
hormone that then stimulates the first hormone, resulting in a vicious Hormone
cycle. An example is the stimulation of LH release by estradiol during Level Low Normal High
the middle of the menstrual cycle. c. Product control of hormone High Primary failure Autonomous
release. The production and release of a hormone can be regulated by of target secretion of
the circulating levels of the substrate that it controls. An example is the endocrine pituitary
regulation of parathyroid hormone release from the parathyroid glands organ hormone or
by the prevailing serum levels of Ca2+. (Modified with permission from Molina resistance to
target hormone
PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
action

Normal Normal range


than one-time plasma-level measurement. However, these Low Pituitary failure Autonomous
measures rely on adequate renal function. secretion by
• Target hormone excess should be evaluated with the target
endocrine
appropriate tropic hormone. organ
The possible interpretations of altered hormone and regula- Reproduced with permission from Kibble J and Halsey CR: The Big Picture, Medical
tory factor pairs are summarized in Table 60–1. Physiology. New York: McGraw-Hill, 2009
612 SECTION IX Endocrine and Metabolic Physiology

CHAPTER SUMMARY 2. Which of the following would be expected to alter hormone levels?
A) changes in mineral and nutrient plasma levels
■ Hormones are classified into protein, amino acid derived, and
B) pituitary tumor
steroid based on their chemistry.
C) transatlantic flight
■ Binding proteins regulate hormone availability and physiologic D) training for the Olympics
function. E) all of the above
■ Physiologic effects of hormones require binding to specific 3. Which of the following statements concerning hormonal
receptors in target organs. regulation is correct?
■ Hormone release is under neural, hormonal, and product A) A hormone does not inhibit its own release.
regulation. B) The substrate a hormone regulates does not affect that
■ Hormones can control their own release through feedback hormone’s release.
regulation. C) Negative feedback regulation occurs only at the level of the
■ Interpretation of hormone levels requires consideration of anterior pituitary.
hormone pairs or of the nutrient or factor controlled by the D) Feedback inhibition may be exerted by nutrients and
hormone. hormones.
4. The structure of a newly discovered hormone shows that it is a
large peptide with a glycosylated subunit. The hormone is
STUDY QUESTIONS likely to
A) bind to DNA and affect gene transcription
1. Which of the following statements concerning a particular B) bind to adenylate cyclase and stimulate protein kinase C
hormone is correct? C) bind to a cell membrane receptor
A) It will bind to cell membrane receptors in all cell types. D) be secreted intact in the urine
B) It is lipid soluble and has an intracellular receptor.
C) It circulates bound to a protein, and this shortens its
half-life.
D) It is a small peptide; therefore, its receptor localization will
be in the nucleus.
61
C H A P T E R

The Hypothalamus and


Posterior Pituitary Gland
Patricia E. Molina

O B J E C T I V E S

■ Describe the physiologic and anatomic relationships between the


hypothalamus and the anterior and the posterior pituitary.
■ Understand the integration of hypothalamic and pituitary function, and identify
the two different pathways used for hypothalamic–pituitary interactions.
■ Identify the appropriate hypothalamic releasing and inhibitory factors
controlling the secretion of each of the anterior pituitary hormones.
■ Differentiate between the routes of transport of hypothalamic neuropeptides
to the posterior and anterior pituitary.
■ Identify the mechanisms that control the release of oxytocin and antidiuretic
hormone (ADH; arginine vasopressin [AVP]).
■ Understand the physiologic target organ responses and the cellular
mechanisms of oxytocin and ADH action.

The hypothalamus is the region of the brain involved in coordi- third ventricle. At the floor of the third ventricle, the two
nating the physiologic responses of different organs that together halves of the hypothalamus are rejoined to form a bridgelike
maintain homeostasis. It does this by integrating signals from the region known as the median eminence (Figure 61–1). The
environment, other brain regions, and visceral afferents, and then median eminence is important because this is where axon ter-
stimulating the appropriate neuroendocrine responses. In doing minals of hypothalamic neurons release neuropeptides
so, the hypothalamus influences many aspects of daily function, involved in the control of anterior pituitary function. In addi-
including food intake, energy expenditure, body weight, fluid tion, the median eminence is traversed by the axons of hypo-
intake and balance, blood pressure, thirst, body temperature, and thalamic neurons ending in the posterior pituitary. The median
the sleep cycle. Most of these hypothalamic responses are medi- eminence funnels down to form the infundibular portion of
ated through hypothalamic control of pituitary function the neurohypophysis or posterior pituitary.
(Figure 61–1). This control is achieved by the following two
mechanisms: (1) release of hypothalamic neuropeptides synthe-
sized in hypothalamic neurons and transported through the HYPOTHALAMIC NUCLEI
hypothalamohypophysial tract to the posterior pituitary gland
and (2) neuroendocrine control of the anterior pituitary through In the hypothalamus, the neuronal bodies are clustered in
the release of peptides that mediate anterior pituitary gland nuclei with projections reaching other brain regions as well as
hormone release (hypophysiotropic hormones) (Figure 61–2). ending in other hypothalamic nuclei. This intricate system of
neuronal connections allows continuous communication
between the hypothalamic neurons and other brain regions.
FUNCTIONAL ANATOMY Some of the neurons that make up the hypothalamic nuclei
are neurohormonal in nature. Neurohormonal refers to the
The hypothalamus is the part of the diencephalon located ability of these neurons to synthesize neuropeptides that func-
below the thalamus and between the lamina terminalis and tion as hormones and to release these neuropeptides from axon
the mamillary bodies forming the walls and the floor of the terminals in response to neuronal depolarization. Two types of

613

Ch61_613-622.indd 613 11/26/10 10:36:07 AM


614 SECTION IX Endocrine and Metabolic Physiology

Small diame
ter ne
u rons
TRH GnRH
Hypothalamus CRH
GHRH
Somatostatin
Dopamine

Hypothalamus
Optic PVN Preprooxyphysin
chiasm Mamillary
SON Prepropressophysin
body
Superior hypophyseal
artery Optic
chiasm Mamillary
body
Posterior
pituitary
Anterior
pituitary
Oxytocin

Anterior ADH
pituitary
Posterior pituitary
ACTH TSH
GH FSH LH Inferior hypophysial
Prolactin artery

FIGURE 61–1 Anatomic and functional relationship between the hypothalamus and the pituitary. The hypothalamus is
anatomically and functionally linked with the anterior and posterior pituitary. They are closely related because of the portal system of blood
supply. The superior, medial, and inferior hypophyseal arteries provide arterial blood supply to the median eminence and the pituitary.
Magnocellular neurons of the supraoptic and paraventricular nuclei have long axons that terminate in the posterior pituitary. Antidiuretic
hormone (ADH, also known as arginine vasopressin) and oxytocin are synthesized in magnocellular neurons as precursors (preprohormones),
post-translationally processed, and released from the posterior pituitary into the blood. The axons of parvocellular neurons terminate in
the median eminence where they release their neuropeptides. The long portal veins drain the median eminence, transporting the peptides
from the primary capillary plexus to the secondary plexus that provides blood supply to the anterior pituitary. TRH, thyrotropin-releasing
hormone; CRH, corticotropin-releasing hormone; GnRH, gonadotropin-releasing hormone, GHRH, growth-hormone releasing hormone; PVN,
paraventricular nucleus; SON, supraoptic nucleus; ADH, antidiuretic hormone; ACTH, adrenocorticotropin; TSH, thyroid-stimulating hormone;
GH, growth hormone; FSH, follicle stimulating hormone; LH, luteinizing hormone. (Reproduced with permission from Kibble J, Halsey CR: The Big Picture,
Medical Physiology. New York: McGraw-Hill, 2009.)

FIGURE 61–2 Magnocellular neurons are larger in size Magnocellular neuron Parvocellular neuron
and produce large quantities of neurohormones. Located
predominantly in the paraventricular and supraoptic nuclei of
the hypothalamus, their unmyelinated axons form the
hypothalamohypophyseal tract that traverses the median Axonal transport
eminence ending in the posterior pituitary. They synthesize the
neurohormones oxytocin (OT), and antidiuretic hormone (ADH),
and neurophysin (NP), that are transported in neurosecretory Median eminence
Axonal transport CRH, TRH, GnRH, GHRH, SST, DA
vesicles down the hypothalamohypophyseal tract and stored
in varicosities at the nerve terminals in the posterior pituitary. Long portal veins
Parvocellular neurons are small in size and have projections
that terminate in the median eminence, brainstem, and spinal
Anterior pituitary
cord. They release small amounts of releasing or inhibiting
neurohormones (hypophysiotropic hormones - CRH, TRH, Posterior pituitary
GnRH, GHRH, SST, DA) that control anterior pituitary function OT, ADH, NP
(discussed in the next chapter). These are transported in the ACTH, TSH, LH/FSH, GH, Prl
long portal veins to the anterior pituitary where they stimulate
the release of pituitary hormones (ACTH, TSH, LH/FSH, GH, Prl)
into the systemic circulation. (Modified with permission from Molina PE:
Systemic circulation Systemic circulation
Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
CHAPTER 61 The Hypothalamus and Posterior Pituitary Gland 615

neurons are important in mediating the endocrine functions REGULATION OF HORMONE RELEASE
of the hypothalamus: the magnocellular and the parvocellu-
lar neurons (Figure 61–2). The magnocellular neurons are Because the hypothalamus receives and integrates afferent
predominantly located in the paraventricular and supraoptic signals from multiple brain regions, it does not function in
nuclei of the hypothalamus and produce large quantities of the isolation from the rest of the central nervous system. Some
neurohormones oxytocin and antidiuretic hormone (ADH) of these afferent signals convey sensory information about
also known as arginine vasopressin (AVP). The unmyelinated the individual’s environment such as light, heat, cold, and
axons of these neurons form the hypothalamohypophysial noise. Among the environmental factors, light plays an
tract, the bridgelike structure that traverses the median emi- important role in generating the circadian rhythm of hor-
nence and ends in the posterior pituitary. Oxytocin and ADH mone secretion. This endogenous rhythm is generated
are released from the posterior pituitary in response to an through the interaction between the retina, the hypotha-
action potential. Parvocellular neurons have projections that lamic suprachiasmatic nucleus, and the pineal gland
terminate in the median eminence, brainstem, and spinal cord. through the release of melatonin. Melatonin is a hormone
These neurons release small amounts of releasing or inhibiting synthesized and secreted by the pineal gland at night that
neurohormones (hypophysiotropic hormones) that control conveys information concerning the daily cycle of light and
anterior pituitary function. darkness to body physiology and participates in the organi-
zation of circadian rhythms. Its rhythm of secretion is
HYPOTHALAMIC NEUROPEPTIDES entrained to the light/dark cycle. Other signals perceived by
the hypothalamus are visceral afferents that provide infor-
Two general types of neurons constitute the endocrine hypo- mation to the central nervous system from peripheral organs
thalamus: the magnocellular neurons, with axons terminating such as the intestines, the heart, the liver, and the stomach.
in the posterior pituitary, and the parvocellular neurons, with The neuronal signals are transmitted by various neurotrans-
axons terminating in the median eminence. Hypophysiotropic mitters released from the afferent fibers, including gluta-
peptides released near the median eminence are transported mate, norepinephrine, epinephrine, serotonin, acetylcholine,
down the infundibular stalk to the anterior pituitary, where histamine, γ-aminobutyric acid, and dopamine. In addi-
they bind to specific cell membrane receptors in cells of the tion, circulating hormones produced by endocrine organs
anterior pituitary, activating intracellular second-messenger and substrates such as glucose can regulate hypothalamic
cascades that result in the release of anterior pituitary hor- neuronal function. Together, these neurotransmitters, sub-
mones into the systemic circulation (Table 61–1). Peptides strates, and hormones influence hypothalamic hormone
released from the anterior pituitary (adrenocorticotropic release. Therefore, hypothalamic hormone release is under
hormone [ACTH], prolactin, growth hormone [GH], environmental, neural, and hormonal regulation. The ability
luteinizing hormone [LH], follicle-stimulating hormone of the hypothalamus to integrate these signals makes it a
[FSH], and thyroid-stimulating hormone [TSH]) and the center of command for regulating endocrine function and
axons of magnocellular neurons terminating in the posterior maintaining homeostasis.
pituitary (oxytocin and ADH) are transported in the venous Hormones can signal the hypothalamus to either inhibit or
blood draining the pituitary that enters the intercavernous stimulate hypophysiotropic hormone release. This control
sinus and the internal jugular veins to reach the systemic mechanism of negative (or positive) feedback regulation
circulation. consists of the ability of a hormone to regulate its own cascade

TABLE 61–1 Key aspects of hypophysiotropic hormones.


Hypophysiotropic Hormone Predominant Hypothalamic Nuclei Anterior Pituitary Hormone Controlled Target Cell

Thyrotropin-releasing hormone Paraventricular nuclei Thyroid-stimulating hormone and prolactin Thyrotroph

Gonadotropin-releasing hormone Anterior and medial hypothalamus; Luteinizing hormone and follicle-stimulating Gonadotroph
preoptic septal areas hormone

Corticotropin-releasing hormone Medial parvocellular portion of Adrenocorticotropic hormone Corticotroph


paraventricular nucleus

Growth hormone–releasing hormone Arcuate nucleus, close to median Growth hormone Somatotroph
eminence

Somatostatin or growth hormone– Anterior paraventricular area Growth hormone Somatotroph


inhibiting hormone

Dopamine Arcuate nucleus Prolactin Lactotroph


The six recognized hypophysiotropic factors and the predominant locations of their cells of origin are listed in the left columns. The right columns list the anterior pituitary
hormone that each hypophysiotropic factor regulates and the cell that releases the specific hormones.
616 SECTION IX Endocrine and Metabolic Physiology

of release (see Figure 60–9). For example, cortisol produced with the needs of the organism. Disruption of these factors can
from the adrenal gland can inhibit the release of CRH, thus alter the patterns of hormone release.
inhibiting the production of proopiomelanocortin and ACTH
and consequently decreasing adrenal gland synthesis of corti-
sol. This loop of hormonal control and regulation of its own HORMONES OF THE
synthesis is critical in maintaining homeostasis and prevent- POSTERIOR PITUITARY
ing disease. A shorter loop of negative feedback inhibition
also exists, which depends on the inhibition of hypophysio- The neuropeptides produced by the magnocellular neurons,
tropic neuropeptide release by the pituitary hormone that it and consequently released from the posterior pituitary, are
stimulates. In this case, an example would be the ability of oxytocin and ADH. Oxytocin and ADH are closely related
ACTH to inhibit CRH release by the hypothalamus. Some peptides consisting of nine amino acids with ring structures.
neuropeptides also possess an ultrashort feedback loop, in They are synthesized as large precursor molecules in the
which the hypophysiotropic neuropeptide itself is able to magnocellular neurons and packaged into neurosecretory
modulate its own release. As an example, oxytocin stimulates vesicles (Figure 61–3). Within the neurosecretory vesicles, the
its own release, creating a positive feedback regulation of neu- precursor hormone undergoes additional posttranslational
ropeptide release. processing during axonal transport, producing the biologi-
This continuous regulation of hormonal release is dynamic; cally active peptides ADH and oxytocin as well as small pep-
it is continuously adapting to changes in the environment and tide products of hormone processing called neurophysins.
in the internal milieu of the individual. Throughout a given Following neuronal depolarization, neuropeptides released
day, the hypothalamus integrates a multitude of signals to enter the systemic circulation through venous drainage of the
ensure that the rhythms of hormone release are kept in pace posterior pituitary into the intercavernous sinus and internal

Nucleus
Cell body Pre-prohormone synthesis
in ER followed by packaging
into secretory granules
in GA occurs in the cell
body of magnocellular neurons.

Axon

Neurosecretory vesicles are transported


down the hypothalamo-hypophysial
tract. Hormone processing occurs
FIGURE 61–3 Synthesis and processing of during this stage yielding hormone
oxytocin and antidiuretic hormone. Oxytocin and and neurophysins.
antidiuretic hormone are synthesized in the endoplasmic
reticulum (ER) of hypothalamic magnocellular neurons
as preprohormones. In the Golgi apparatus (GA) they are
packaged in secretory granules and are transported
down the hypothalamohypophyseal tract. During their Contents of neurosecretory vesicles
are released from nerve terminals in Mitochondria
transport the precursor hormones are processed, yielding
the posterior pituitary. Exocytosis is
the final hormone and the respective neurophysins. The triggered by Ca2+ influx through
contents of the neurosecretory vesicles are released by Synaptic vesicle
voltage-gated channels opened
exocytosis from the axon terminals in the posterior during neuronal depolarization.
pituitary. Exocytosis is triggered by the influx of Ca2+
through voltage-gated channels that are opened during
the neuronal depolarization. The rise in Ca2+ produces
the docking of the secretory vesicles on the axonal plasma
membrane and the release of the neuropeptides into the
interstitial space. (Modified with permission from Molina PE:
Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
CHAPTER 61 The Hypothalamus and Posterior Pituitary Gland 617

jugular vein. In the systemic circulation, oxytocin and ADH The physiologic effects of oxytocin in the pregnant uterus
circulate unbound. They are rapidly cleared from the circula- are augmented by a dramatic increase in sensitivity to the hor-
tion by the kidney and, to a lesser extent, by the liver and brain. mone during the onset of labor, due to an increased density
Their half-life is short and is estimated to range between 1 and (upregulation) of oxytocin receptors in the uterine muscle,
5 minutes. increased gap junction formation between smooth muscle
cells, and by increased synthesis of prostaglandins.

OXYTOCIN Control of Oxytocin Release


The principal stimulus for oxytocin release is mechanical
The neuropeptide oxytocin is released in response to sensory stimulation of the uterine cervix by the fetus near the end of
stimulation during breastfeeding (lactation) and childbirth gestation and by the forceful contractions of the uterus during
(parturition) (Figure 61–4). the fetal expulsion reflex. In addition, oxytocin release is also
triggered by stimulation of tactile receptors in the nipples of
Physiologic Effects of Oxytocin the lactating breast during suckling (Figure 61–4).
The role of oxytocin in males is not clear, although recent
The two main target organs for oxytocin’s physiologic effects
studies have suggested that it may participate in ejaculation.
are the lactating breast and the uterus during pregnancy
(Figure 61–4). The physiologic effects of oxytocin are achieved
by binding to cell membrane Gq/11 protein–coupled oxytocin ANTIDIURETIC HORMONE
receptors expressed in the uterus, mammary glands, and
brain. In the lactating breast, oxytocin stimulates milk ejection ADH, also known as arginine vasopressin (AVP), is the other
by producing contraction of the myoepithelial cells that line neuropeptide produced by magnocellular neurons of the
the alveoli and ducts in the mammary gland. In the pregnant hypothalamus and released from the posterior pituitary. The
uterus, oxytocin produces rhythmic contractions to help principal effect of ADH is to increase water reabsorption by
induce labor and to promote regression of the uterus following enhancing permeability to water in the distal convoluted
delivery. Oxytocin analogs are used clinically during labor and tubules and the medullary collecting ducts in the kidney (see
delivery to promote uterine contractions and during the Chapter 45). The result is the production of smaller volumes
postpartum period to help decrease bleeding and return the of concentrated urine. In addition, ADH increases vascular
uterus to its normal size (uterine involution) (Table 61–2). resistance. This function of ADH may be important during

PVN & SON

Fear, pain,
noise, fever

Posterior Stretch of cervix FIGURE 61–4 Physiologic effects and regulation of


pituitary end of pregnancy
oxytocin release. The release of oxytocin is stimulated by
the distention of the cervix at term of pregnancy and by
Oxytocin release the contraction of the uterus during parturition. The signals
Sucking of are transmitted to the paraventricular nuclei (PVN) and
lactating breast supraoptic nuclei (SON) of the hypothalamus, where they
Contraction of provide a positive feedback regulation of oxytocin release.
myoepithelial cells The increased number of oxytocin receptors, the increased
number of gap junctions between smooth muscle cells,
and the increased synthesis of prostaglandins enhance the
responsiveness of the uterine muscle. Oxytocin release
causes an increase in uterine contractility, aiding in the
delivery of the baby, and involution of the uterus after
parturition. Suckling of the nipple of the lactating breast
also stimulates oxytocin release. The afferent sensory
signals elicit a rise in oxytocin release into the circulation.
Oxytocin produces contraction of the myoepithelial cells
Uterine lining the breast ducts, resulting in milk ejection. (Modified
contraction with permission from Molina PE: Endocrine Physiology, 3rd ed. New York:
McGraw-Hill Medical, 2010.)
618 SECTION IX Endocrine and Metabolic Physiology

TABLE 61–2 Key aspects of posterior pituitary hormones.


Oxytocin Antidiuretic Hormone

Receptor Gq/11 protein–coupled G protein–coupled (V1R, Gq/11, V2R, Gs)

Second messenger Phospholipase C ↑ [Ca2+]i (V1R) Phospholipase C ↑ [Ca2+]i

(V2R) Adenylate cyclase cAMP

Target organ or cells Uterus V2R: kidney collecting ducts

Mammary myoepithelial cells V1R: smooth muscle cells

Physiologic effects Uterine contraction Increased H2O permeability

Milk ejection Vasoconstriction


cAMP, cyclic adenosine monophosphate.

periods of severe lack of responsiveness to other vasoconstric- second-messenger systems that they activate. The main
tors, as may occur during severe blood loss (hemorrhagic effects of ADH are mediated through the V2R receptor.
shock) or systemic infection (sepsis). The main target site of ADH is the collecting duct in the
kidney. Water permeability of the collecting duct can be
dramatically increased (within a few minutes) through the
Physiologic Effects of ADH production of cyclic adenosine monophosphate (cAMP)
The cellular effects of ADH are mediated by binding following ADH binding to V2 receptors in the basolat-
to G protein–coupled membrane receptors. Three ADH eral membrane of the principal cells in the collecting duct
receptors have been characterized thus far, which differ in (Figure 61–5).
terms of where they are expressed as well as in the specific The increase in cAMP activates protein kinase A and subse-
G proteins to which they are coupled and, thus, in the quently the phosphorylation of an aquaporin (AQP2) leading

Interstitial space

ADH
H2O H2O
V2R Adenylate cyclase
AQP3
AQP4
Basolateral
α
membrane βγ α α

FIGURE 61–5 Cellular mechanism of ATP cAMP

ADH water conservation. The principal


function of antidiuretic hormone (ADH) is to
increase water reabsorption and to conserve PKA
water. ADH binds to the V2 G protein–coupled
receptor (V2R) in the principal cells of the
Principal cell
distal tubule. This triggers the activation of
distal tubule
adenylate cyclase and the formation of
AQP2
cAMP, leading to activation of protein kinase AQP2
A (PKA). PKA phosphorylates the water
channel aquaporin 2 (AQP2), leading to the Cytosolic
insertion of AQP2 into the luminal cell H2O pool of
AQP2 AQP2
membrane. The insertion of water channels Apical
AQP2
into the membrane increases the permeability membrane
to water. Water reabsorbed through these
water channels leaves the cell through
aquaporins 3 (AQP3) and 4 (AQP4), which are
constitutively expressed in the basolateral
H2O
membrane of the principal cells. (Modified with
permission from Molina PE: Endocrine Physiology, 3rd ed.
New York: McGraw-Hill Medical, 2010.) Lumen-collecting duct
CHAPTER 61 The Hypothalamus and Posterior Pituitary Gland 619

TABLE 61–3 Key features of aquaporins.


Aquaporin Features

AQP1 Constitutively expressed in apical and basolateral membranes of epithelial cells of proximal tubules and descending limb of
Henle’s loop. Involved in 90% of water reabsorption

AQP2 Exclusively expressed in the collecting ducts. The only aquaporin directly regulated by ADH. Binding to the V2 ADH (AVP) receptor
stimulates insertion into the luminal membrane

AQP3, AQP4 Constitutively expressed in the basolateral membranes of epithelial cells in the collecting ducts. Enhance water reabsorption
following AQP2 insertion into the luminal membrane
AQP, aquaporin; ADH, antidiuretic hormone; AVP, arginine vasopressin.

to its movement from cytoplasmic pools and its insertion in hormone is known as vasopressin because of these vasocon-
the luminal (apical) epithelial cell membrane of the collecting strictor effects.
duct cells. The result is an increase in the number of functional
water channels in the luminal membrane, making it more per-
meable to water. AQP2, one of the several members of the Control of ADH Release
aquaporin family, is exclusively expressed in the collecting ADH is released into the circulation following either an
ducts of the kidney (Table 61–3). It is the only aquaporin that increase in plasma osmolality or a decrease in blood volume
is directly regulated by ADH via the V2 ADH receptor. AQP3 (Figure 61–6). Under physiologic conditions, the most impor-
and AQP4 are constitutively expressed in the basolateral mem- tant stimulus for ADH release is the “effective” plasma osmo-
branes of the collecting ducts and contribute to the enhanced lality detected by special osmoreceptor neurons located in the
water reabsorption following AQP2 insertion into the luminal hypothalamus and in three structures associated with the lam-
membrane. Water that enters the epithelial cell through AQP2 ina terminalis: the subfornical organ, the median preoptic
on the apical membrane leaves the cell through AQP3 and nucleus, and the organum vasculosum lamina terminalis.
AQP4, located in the basolateral membranes of these cells, The sensitivity of this system is quite high. That is, very small
eventually entering the vasculature. ADH also binds the V1 changes in plasma osmolality (as little as 1% change) above the
receptor, found in the vascular smooth muscle, producing osmotic threshold of 280–284 mOsm/kg water produce
contraction and increasing peripheral vascular resistance. The significant increases in ADH release.

Fluid loss
Hemorrhage, vomiting, diarrhea

10
MABP
Plasma ADH (pg/ml)

8
10% blood loss
FIGURE 61–6 Integration of signals
6
Baroreceptor stretch and firing that trigger ADH release. Release of ADH is
4 stimulated by an increase in plasma osmolality
9th and 10th cranial
and a decrease in blood volume. Small changes
2
Thirst nerve afferents in plasma osmolality above a threshold of
0 280–284 mOsm/kg produce an increase in
270 280 290 300 310 ADH release before the stimulation of thirst.
Plasma osmolarity (mOsm/kg)
A decrease in blood volume sensitizes the
Plasma osmolarity system and increases the responsiveness to
small changes in plasma osmolality. Blood
loss and a decrease in mean arterial blood
Shrinkage of Sympathetic
tone pressure (MABP) greater than 10% signal
osmoreceptors
the hypothalamus to increase the release
of ADH. The afferent signals are transmitted
by the 9th and 10th cranial nerves. These
ADH release
Restore fluid signals increase sympathetic tone, therefore
balance decreasing magnocellular neuron inhibition
and stimulating ADH release. (Modified with
Magnocellular permission from Molina PE: Endocrine Physiology, 3rd ed.
H2O reabsorption
neuron inhibition New York: McGraw-Hill Medical, 2010.)
620 SECTION IX Endocrine and Metabolic Physiology

ADH secretion is also stimulated by a decrease in blood inappropriate ADH secretion, may be the result of tumor pro-
pressure of greater than 10%. Factors that reduce cardiac out- duction of ADH. The tumor can be located in the brain, but ma-
put, such as a decrease in blood volume greater than 8%, lignancies of other organs such as the lung have also been shown
orthostatic hypotension, and positive pressure breathing, are to produce high levels of ADH. The excess production of ADH
all stimuli for ADH release. ADH secretion is far more sensitive results in the production of very small volumes of concentrated
to small changes in plasma osmolality than to changes in blood urine. Retention of water may lead to decreased plasma sodium
volume. The volume-induced sensitization of ADH release (hyponatremia). Management of this condition entails fluid re-
results in a more accentuated ADH response to changes in striction and in some cases the use of saline solutions to restore
plasma osmolality. ADH is barely detectable below a certain adequate plasma sodium levels.
plasma osmolality (287 mOsm/kg) threshold. Above this
threshold, the plasma AVP concentration rises steeply in direct
proportion to plasma osmolality.
CLINICAL CORRELATION
Disorders of ADH Production A head trauma patient on his third day in the surgical
Either excess or deficiency of ADH can result in clinical dis- intensive care unit is reported to have an excessive urine
ease. The concentrations of ADH may be altered in various output of 20 L in the past 24 hours. Laboratory findings
chronic pathophysiologic conditions, including congestive show hypernatremia (high serum Na) and hypotonic
heart failure, liver cirrhosis, and nephrotic syndrome. A urine. Withholding drinking water did not decrease urine
decrease in ADH release or action results in diabetes insipi- output or increase urine osmolarity. Urine osmolarity
dus, a clinical syndrome in which the ability to form concen- increased in response to exogenous antidiuretic hormone
trated urine is reduced. (vasopressin) administration. He is diagnosed as having
posttraumatic diabetes insipidus.
Diabetes insipidus Diabetes insipidus is characterized by Neurogenic diabetes insipidus is the result of decreased
the excretion of abnormally large volumes (up to 30 mL/kg of release of ADH characterized by polyuria (increased
body weight per day for adult subjects) of dilute (<250 mmol/kg) urine output) and hypernatremia. This can be the result
of destruction of neurons that produce and release ADH
urine and excessive thirst. Three basic defects have been identi-
resulting from inflammation, neoplasia, vascular abnor-
fied in the etiology (only the first two pertain to alterations re-
malities, or traumatic injury. Patients are treated with
lated to components of the ADH system itself):
desmopressin, a synthetic analog of ADH, which binds
• Decreased ADH release: Neurogenic (central or hypotha- to V2 receptors, increasing water reabsorption in the
lamic) diabetes insipidus is due to a decrease in ADH release kidney, decreasing urine output, and restoring serum
from the posterior pituitary, resulting from diseases affect- osmolarity.
ing the hypothalamic-neurohypophysial axis. Three causes
can be identified: traumatic, inflammatory or infectious,
and cancer related.
• Decreased renal responsiveness to ADH: Renal (nephro- CHAPTER SUMMARY
genic) diabetes insipidus results from renal insensitivity to ■ The parvocellular neurons of the hypothalamus produce
the antidiuretic effect of ADH. ADH production and release neuropeptides that are released at the median eminence and
are transported to the anterior pituitary, where they regulate
are not affected, but responsiveness at the distal tubule is
the release of anterior pituitary hormones.
impaired. Nephrogenic diabetes insipidus can be inherited
■ The hypothalamus integrates information from various brain
or acquired and is characterized by an inability to concen-
regions, the environment, and peripheral organs and mediates
trate urine despite normal or elevated plasma concentra- systemic responses that help maintain homeostasis.
tions of ADH. About 90% of inherited cases are males with
■ The magnocellular neurons of the hypothalamus produce
the X-linked recessive form of the disease who have muta- oxytocin and ADH, hormones that are released from the
tions in the ADH type 2 receptor gene. A small number of posterior pituitary into the systemic circulation.
cases of inherited nephrogenic diabetes insipidus are due to ■ Prohormone posttranslational modification and processing of
mutations in the AQP2 water channel gene. Acquired neph- oxytocin and ADH occur inside the secretory granules during
rogenic diabetes insipidus can result from lithium treat- axonal transport.
ment, hypokalemia, and postobstructive polyuria. ■ ADH binds to the V2 receptor and increases water reabsorption
• Excess water intake: Finally, the third possible cause of dia- by stimulating the insertion of aquaporin 2 into the apical
betes insipidus is excess water intake. This cause does not (luminal) membrane of tubular collecting duct epithelial cells.
involve dysfunction of the ADH system. ■ The release of ADH is more sensitive to small changes in
plasma osmolality than to small changes in blood volume.
Syndrome of inappropriate ADH secretion An increase ■ Deficiency of ADH results in the production of large quantities
or excess in the release of ADH, also known as the syndrome of of dilute urine.
CHAPTER 61 The Hypothalamus and Posterior Pituitary Gland 621

STUDY QUESTIONS 4. Oxytocin


A) is released from the anterior pituitary during parturition
1. The renal effects of ADH (AVP) are mediated by B) is synthesized in the posterior pituitary and released during
A) binding to V2 G protein–coupled receptor, protein parturition
phosphorylation, and insertion of aquaporin 2 into luminal C) is produced in the magnocellular neurons of the
cell membrane in collecting duct hypothalamus and released from the posterior pituitary
B) binding to nuclear receptors, protein phosphorylation, and during parturition
insertion of aquaporin 2 into basolateral membrane of D) is produced in the magnocellular neurons of the
collecting duct hypothalamus and released from the anterior pituitary
C) binding to V1 G protein–coupled receptor, protein during parturition
phosphorylation, and insertion of aquaporin 1 into luminal 5. Uterine muscle responsiveness to oxytocin
cell membrane of collecting duct
A) is not altered throughout pregnancy
D) binding to nuclear receptors, protein phosphorylation, and
B) is prevented by high prostaglandin levels during pregnancy
insertion of aquaporin 2 into luminal cell membrane of
C) is decreased by lower gap junction formation during the
collecting duct
third trimester
2. On a hot summer day after profuse sweating, the release of D) is enhanced by increased oxytocin receptor density
ADH occurs
A) when plasma osmolality is 270 mOsm/kg
B) immediately following stimulation of thirst
C) only after more than a 10% decrease in blood volume
D) before stimulation of thirst
3. Which of the following can be manifestations of the syndrome of
inappropriate ADH secretion?
A) hypernatremia with high urine osmolality
B) hyponatremia with high urine osmolality
C) hypophosphatemia with low urine osmolality
D) hyponatremia with low urine osmolality
This page intentionally left blank
62
C H A P T E R

Anterior Pituitary Gland


Patricia E. Molina

O B J E C T I V E S

■ Identify the three families of anterior pituitary hormones and their main
structural differences.
■ Understand the mechanisms that regulate anterior pituitary hormone
production and describe the actions of tropic hormones on target organs.
■ Diagram the short- and long-loop negative feedback control of anterior
pituitary hormone secretion.
■ Predict the changes in secretory rates of hypothalamic anterior pituitary and
target gland hormones caused by oversecretion or undersecretion of any of
these hormones or receptor deficit for any of these hormones.
■ Explain the importance of pulsatile and diurnal hormone secretion.

The anterior pituitary plays a central role in the regulation of hormone (TSH), growth hormone (GH), prolactin, and the
endocrine function through the production and release of gonadotropins [luteinizing hormone (LH) and follicle-
tropic hormones (Figure 62–1). The function of the anterior stimulating hormone (FSH)] (Figure 62–1). All of these hor-
pituitary, and thereby the production of tropic hormones, is mones are released into the systemic circulation.
under hypothalamic regulation by the hypophysiotropic neu-
ropeptides released in the median eminence, summarized in
Table 62–1. The tropic hormones produced by the anterior HYPOTHALAMIC CONTROL
pituitary bind to specific receptors at their target organs to
produce a physiologic response, most frequently involving the
OF ANTERIOR PITUITARY
release of a hormone (Figure 62–2). The hormones produced HORMONE RELEASE
by the target organs affect anterior pituitary function as well as
the release of hypophysiotropic neuropeptides, maintaining The production of pituitary tropic hormones is under direct
an integrated feedback control system of endocrine function. regulation by the hypothalamic neurohormones released from
neuronal terminals in the median eminence.
The responsiveness of the anterior pituitary to the inhibitory
FUNCTIONAL ANATOMY or stimulatory effects of hypophysiotropic neurohormones can
be modified by several factors, including hormone levels, nega-
The pituitary, or hypophysis, consists of an anterior and a tive feedback inhibition, and circadian rhythms The release of
posterior lobe that differ from one another in their embryo- anterior pituitary hormones is cyclic in nature, and this cyclic
logic origin, mode of development, and structure. The ante- pattern of hormone release is governed by the nervous system.
rior lobe, also known as the adenohypophysis, is the remnant Most daily (circadian) rhythms are driven by an internal bio-
of Rathke’s pouch; it is a highly vascularized structure consist- logic clock located in the hypothalamic suprachiasmatic
ing of epithelial cells derived from the ectodermal lining of the nucleus; this clock is synchronized by external signals such as
roof of the mouth. The pituitary cells that line the capillaries light and dark periods. Both sleep and circadian effects interact
produce the tropic (also known as trophic) hormones: adre- to produce the overall rhythmic pattern of pituitary hormone
nocorticotropic hormone (ACTH), thyroid-stimulating release and the associated responses. Some of the 24-hour

623

Ch62_623-632.indd 623 11/26/10 4:43:25 PM


624 SECTION IX Endocrine and Metabolic Physiology

Artery
Artery
Anterior pituitary
Posterior pituitary
gland
gland

GH
TSH ACTH

Thyroid hormone Cortisol,


aldosterone, &
androgens

H2O & Na
balance
FIGURE 62–1 Anterior pituitary hormones, target Growth &
Inflammation Insulin-like
organs, and physiologic effects. Thyroid-stimulating differentiation
& metabolism growth factor
Energy balance
hormone (TSH) stimulates the thyroid gland to produce
and release thyroid hormones that regulate growth, LH &
differentiation, and energy balance. Luteinizing hormone FSH Growth & differentiation
(LH) and follicle-stimulating hormone (FSH) stimulate
gonadal production of sex steroids, which mediate Prl
reproductive function and behavior. Adrenocorticotropic
hormone (ACTH) stimulates the adrenal glands to produce
steroid hormones, which regulate water and sodium
balance, inflammation, and metabolism. Prolactin (Prl)
stimulates breast development and milk production.
Growth hormone (GH) exerts direct effects on tissue
growth and differentiation and indirect effects through Estrogen, progesterone,
the stimulation of insulin-like growth factor I production, & testosterone
which mediates some of the growth and differentiation
effects of GH. (Modified with permission from Molina PE: Endocrine Reproductive Breast development
Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.) function & behavior Milk production

hormonal rhythms depend on the circadian clock (i.e., ACTH, administration produces effects that differ from the hor-
cortisol, and melatonin) and some are sleep related (i.e., prolac- mone’s natural physiologic effects. These observations have
tin, GH, and TSH). For example, GH secretion is influenced by highlighted the importance of trying to simulate as much as
the first slow-wave sleep episode at the beginning of the night. possible the endogenous cyclic patterns of hormone release
Pulses of prolactin and GH are positively linked to increases in when giving hormone replacement therapy to a patient.
delta-wave activity, present during the deepest phases of sleep
and occurring primarily during the first third of the night. Pulses
of TSH and cortisol are related to superficial phases of sleep. HORMONES OF THE
Although the regulation of the patterns of hormone release
is not well understood, it is clear that the respective patterns
ANTERIOR PITUITARY
of anterior pituitary hormone release play a crucial role in The hormones of the anterior pituitary can be classified into
achieving their physiologic effects and, thus, in maintaining three families: the glycoproteins, those derived from proopi-
homeostasis. The importance of this regulation has become omelanocortin (POMC), and those belonging to the GH and
evident because constant or continuous exogenous hormone prolactin family.
CHAPTER 62 Anterior Pituitary Gland 625

TABLE 62–1 Anterior pituitary cell type, regulatory GLYCOPROTEINS


hypothalamic factor, and hormone product.
Glycoprotein hormones are among the largest hormones
Anterior Hypothalamic Pituitary Hormone
Pituitary Cells Factor Produced
known to date. They include TSH, FSH, LH, and human chor-
ionic gonadotropin produced by the placenta. These hor-
Lactotrophs Dopamine (−) Prolactin mones are heterodimeric glycoproteins consisting of a
Corticotrophs CRH (+) POMC: ACTH, β-LPH, common α-subunit and a unique β-subunit, which confers the
α-MSH, β-endorphin biologic specificity of each hormone.
Thyrotrophs TRH (+) Thyroid-stimulating
hormone
Thyroid-stimulating Hormone
Gonadotrophs GnRH (+) LH and FSH TSH is a glycoprotein synthesized and secreted from thyrotro-
Somatotrophs GHRH (+) and SST (−) Growth hormone phs of the anterior pituitary gland. Thyrotrophs consti-
(+), stimulatory factor; (−), inhibitory factor; CRH, corticotropin-releasing tute approximately 5% of all adenohypophysial cells. They
hormone; POMC, proopiomelanocortin; ACTH, adrenocorticotropic hormone; synthesize and release TSH in response to stimulation by
LPH, lipotropin; MSH, melanocyte-stimulating hormone; TRH, thyrotropin-
releasing hormone; GnRH, gonadotropin-releasing hormone; LH, luteinizing thyrotropinreleasing hormone (TRH), which is synthesized
hormone; FSH, follicle-stimulating hormone; GHRH, growth hormone– in the hypothalamus and released from nerve terminals in
releasing hormone; SST, somatostatin.
the median eminence. TSH binds to a Gs protein–coupled

Hypothalamic Peptides
TRH GnRH CRH Somatostatin GHRH Dopamine

Median eminence

G protein−coupled receptors in anterior pituitary

Thyrotroph Gonadotroph Corticotroph Somatotroph Lactotroph

PLC PLC AC AC AC AC, K+ , Ca2+

TSH LH & FSH ACTH GH Prl

G protein−coupled receptors Class 1 cytokine receptor

AC AC AC Kinase activity Kinase activity

Target organ physiologic response

FIGURE 62–2 Cellular signaling pathways involved in hypothalamo-pituitary hormone-mediated effects. All hypothalamic
releasing and inhibiting factors mediate their effects predominantly via G protein–coupled receptors. Anterior pituitary hormones bind
to either G protein–coupled receptors (thyroid-stimulating hormone [TSH], luteinizing hormone [LH], follicle-stimulating hormone [FSH],
adrenocorticotropic hormone [ACTH]) or class 1 cytokine receptors (growth hormone [GH] and prolactin [Prl]). Most of the cellular responses
elicited by anterior pituitary hormones that bind to G protein–coupled receptors are mediated by modulation of adenylate cyclase activity.
The cellular responses evoked by anterior pituitary binding to class 1 cytokine receptors are mediated through protein kinase activation.
TRH, thyrotropin-releasing hormone; GnRH, gonadotropin-releasing hormone; CRH, corticotropin-releasing hormone; GHRH, GH-releasing
hormone; PLC, phospholipase C activity; AC, adenylate cyclase activity. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York:
McGraw-Hill Medical, 2010.)
626 SECTION IX Endocrine and Metabolic Physiology

receptor in the thyroid gland, and stimulates all the events has been implicated in feeding behavior and appetite regula-
involved in thyroid hormone synthesis and release. In addi- tion. The role of MC3R is not well defined.
tion, it acts as a growth factor for the thyroid gland. The release
of TSH from the anterior pituitary gland is under negative
feedback inhibition by thyroid hormones.
Adrenocorticotropic Hormone
The main hormone of interest produced by the cleavage of
POMC is ACTH. The release of ACTH is stimulated by psy-
Gonadotropins (FSH and LH) chological and physical stressors such as infection, hypoglyce-
The gonadotropic hormones LH and FSH are synthesized and mia, surgery, and trauma and is considered critical in mediating
secreted by gonadotrophs of the anterior pituitary in response the adaptive response of the individual to stress. ACTH is
to stimulation by gonadotropin-releasing hormone (GnRH). released in pulses, with the highest concentrations occurring
Gonadotrophs constitute about 5–10% of the pituitary cells. in the morning and the lowest concentrations around mid-
Most of the gonadotrophs (60%) produce both LH and FSH. night. ACTH released into the systemic circulation binds to a
The remainder of the gonadotroph population produces LH Gαs protein–coupled receptor at the adrenal cortex and stimu-
(18%) or FSH (22%) exclusively. GnRH is synthesized and lates the production and release of glucocorticoids (cortisol)
secreted by the hypothalamus in a pulsatile manner. and, to a lesser extent, mineralocorticoids (aldosterone). The
FSH and LH exert their physiologic effects on multiple cells release of cortisol follows the same diurnal rhythm as that of
of the reproductive system by binding to Gαs protein–coupled ACTH. The feedback inhibition of ACTH and of CRH release
receptors and activation of adenylate cyclase. Among the target by cortisol is mediated by glucocorticoid receptor binding in
cells for gonadotropins are ovarian granulosa cells, theca the hypothalamus and in the anterior pituitary.
interna cells, testicular Sertoli cells, and Leydig cells. The
physiologic responses produced by the gonadotropins include
stimulation of sex hormone synthesis (steroidogenesis), sper-
Melanocyte-stimulating Hormone
matogenesis, folliculogenesis, and ovulation. Therefore, their α-MSH is produced by the proteolytic cleavage of POMC,
central role is the control of reproductive function in both mainly in the pars intermedia of the pituitary gland
males and females. GnRH controls the synthesis and secretion (Figure 62–3). Only small amounts of α-MSH are produced in
of both FSH and LH by the pituitary gonadotroph cell. Gonad- the human pituitary under normal conditions. Melanocortin
otropin synthesis and release, as well as differential expression, peptides exert their effects through MC1R found in melano-
is under both positive and negative feedback control by cytes, which are key components of the skin’s pigmentary sys-
gonadal steroids and gonadal peptides. Gonadal hormones tem, endothelial cells, monocytes, and keratinocytes.
can decrease gonadotropin release both by decreasing the fre-
quency and amplitude of pulses of GnRH release from the β-Endorphin
hypothalamus and by affecting the ability of GnRH to stimu-
late gonadotropin secretion from the pituitary itself. β-Endorphin, the most abundant endogenous opioid peptide,
is another product of POMC processing in the pituitary
(Figure 62–3). The physiologic effects of this opioid peptide
are mediated by binding to opiate receptors, expressed in mul-
PROOPIOMELANOCORTIN- tiple cell types in the brain as well as in peripheral tissues. The
DERIVED HORMONES physiologic actions of endorphins are not well understood, but
may include analgesia, behavioral effects, and neuromodula-
Proopiomelanocortin (POMC) is a precursor prohormone tory functions.
produced by the corticotrophs of the anterior pituitary.
Corticotrophs account for 10% of the secretory cells of the
anterior pituitary. The production and secretion of POMC-
derived hormones from the anterior pituitary are regulated
GROWTH HORMONE &
predominantly by corticotropin-releasing hormone (CRH) PROLACTIN FAMILY
produced in the paraventricular nucleus of the hypothalamus
and released in the median eminence. CRH binds to Gs pro- Growth Hormone
tein–coupled CRH receptors. GH is a 191–amino acid peptide hormone, with a molecular
POMC is posttranslationally cleaved to ACTH; β-endorphin, weight of approximately 22 kd and structural similarity to pro-
an endogenous opioid peptide; and α-, β-, and γ-melanocyte- lactin and chorionic somatomammotropin (human placen-
stimulating hormones (MSH) (Figure 62–3). The biologic tal lactogen), a placental-derived hormone. GH is released
effects of POMC-derived peptides are largely mediated from the somatotrophs, an abundant (50%) cell type in the
through melanocortin receptors (MCRs), of which five have anterior pituitary. It is released in pulsatile bursts, with the
been described. MC1R, MC2R, and MC5R have defined roles majority of secretion occurring nocturnally in association
in the skin, adrenal steroid hormone production, and thermo- with slow-wave sleep (see Figure 60–7). The basis of the pulsa-
regulation, respectively. MC4R is expressed in the brain and tile release of GH and the function of this pattern are not fully
CHAPTER 62 Anterior Pituitary Gland 627

Pro-opiomelanocortin

Signal peptide

NH2 COOH

β-LPH

N-terminal
ACTH

γ-LPH β-end

ACTH
receptor β-MSH

γ-MSH α-MSH CLIP

Opiate
Melanocortin receptor
receptor
FIGURE 62–3 Proopiomelanocortin (POMC) processing. Corticotropin-releasing hormone stimulates the production, release, and
processing of POMC, a preprohormone synthesized in the anterior pituitary. POMC is posttranslationally cleaved to adrenocorticotropic hormone
(ACTH); β-endorphin, an endogenous opioid peptide; and α-, β-, and γ-melanocyte-stimulating hormones (MSH). The cellular effects of these
peptides are mediated via melanocortin (ACTH and MSH) or opiate (β-end) receptors. LPH, β-lipotropin; CLIP, corticotropin-like intermediate lobe
peptide. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

understood; however, nutritional, metabolic, and age-related The overall contribution of ghrelin to regulation of GH release
sex steroid mechanisms, adrenal glucocorticoids, thyroid in humans is still not fully elucidated.
hormones, and renal and hepatic functions are all thought
to contribute to the pulsatile release of GH and appear to Other regulators
be essential in achieving optimal biologic potency of the In addition to regulation by GHRH and SST, GH is regulated
hormone. by other hypothalamic peptides and neurotransmitters, which
act by regulation of GHRH and SST release, as summarized in
Regulation of GH release Table 62–2. Catecholamines, dopamine, and excitatory amino
The two principal hypothalamic regulators of GH (soma- acids increase GHRH and decrease SST release, resulting in an
totropin) release from the anterior pituitary are growth increase in GH release. Hormones such as cortisol, estrogen,
hormone-releasing hormone (GHRH) and somatostatin androgens, and thyroid hormone can also affect somatotroph
(SST), which exert stimulatory and inhibitory influences, responsiveness to GHRH and SST and consequently GH
respectively, on the somatotrophs (Figure 62–4). GH release is release. Metabolic signals such as glucose and amino acids can
also inhibited by insulin-like growth factor-I (IGF-I), the affect GH release. Decreased blood glucose concentrations
hormone produced in the periphery, particularly in the liver, (hypoglycemia) stimulate GH secretion in humans. Glucose
in response to GH receptor stimulation, in a classic negative and nonesterified fatty acids decrease GH release, whereas
feedback mechanism of hormone control. More recently, amino acids, particularly arginine, increase GH release.
ghrelin has been identified as an additional GH secretagogue. Consequently, arginine administration is also an effective chal-
Ghrelin is a peptide released predominantly from stomach. lenge to elicit an increase in GH release in the clinical setting.
628 SECTION IX Endocrine and Metabolic Physiology

Sleep
Aging
Hypoglycemia (+) (–) Disease
Stress
Glucose

GHRH (+) (–)


(–) SST (–)

Adipose tissue
Glucose uptake
(–) Lipolysis

(–)
GH

Muscle
Glucose & AA
uptake
Protein
synthesis
Chondrocytes
AA uptake
Protein synthesis
DNA & RNA synthesis
Chondroitin sulfate IGF-I
Collagen
Cell size & number

FIGURE 62–4 Growth hormone release and effects. Growth hormone release from the anterior pituitary is modulated by several factors,
including stress, exercise, nutrition, sleep, and growth hormone itself. The primary controllers of GH release are growth hormone–releasing
hormone (GHRH) that stimulates both the synthesis and secretion of growth hormone and somatostatin (SST) that inhibits growth hormone
release in response to GHRH and to other stimulatory factors such as low blood glucose concentration. Growth hormone secretion is also part of
a negative feedback loop involving IGF-1. High blood levels of IGF-1 lead to decreased secretion of growth hormone not only by directly
suppressing the somatotroph, but also by stimulating release of somatostatin from the hypothalamus. Growth hormone also feeds back to
inhibit GHRH secretion and probably has a direct (autocrine) inhibitory effect on secretion from the somatotroph. Integration of all the factors
that affect growth hormone synthesis and secretion leads to a pulsatile pattern of release. GH effects in peripheral tissues are mediated by GH
binding to its receptor and through the synthesis of insulin-like growth factor 1 (IGF-1) by the liver and at the tissue level. The overall effects of
GH and IGF-I are anabolic. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

GH is released from the anterior pituitary into the systemic Physiologic effects of GH
circulation. The half-life of the hormone averages 6–20 minutes. GH can have direct effects on cellular responses, by binding to
GH is degraded in the lysosomes following binding to its recep- the GH receptor at target tissues, and, indirectly, by stimulat-
tor and internalization of the hormone–receptor complex. ing the production and release of IGF-I, a mediator of several
of growth hormone’s effects at target tissues. IGF-I is a small
peptide (about 7.5 kd) structurally related to proinsulin that
TABLE 62–2 Factors that regulate growth hormone
mediates several of the anabolic and mitogenic effects of GH
release.
in peripheral tissues. The most important physiologic effect of
Stimulation of GH Release Inhibition of GH Release GH is stimulation of postnatal longitudinal growth. GH also
plays a role in regulation of substrate metabolism, adipocyte
GHRH Somatostatin
differentiation; and maintenance and development of the
Dopamine IGF-I immune system.
Catecholamines Glucose

Excitatory amino acids FFA GH receptor


Thyroid hormone
In the peripheral tissues, GH binds to specific cell surface recep-
tors belonging to the class 1 cytokine receptor superfamily (see
GHRH, growth hormone–releasing hormone; IGF-I, insulin-like growth factor-I; FFA,
free fatty acids. Figure 60–5) that includes the receptors for prolactin, erythro-
CHAPTER 62 Anterior Pituitary Gland 629

poietin, leptin, interferons, granulocyte colony–stimulating hormone in linear growth is clearly demonstrated by the
factor, and interleukins. GH receptors are present in many severe growth failure in children with congenital IGF-I defi-
biologic tissues and cell types, including liver, bone, kidney, ciency. These small peptide hormones are members of a
adipose tissue, muscle, eye, brain, heart, and cells of the immune family of insulin-related peptides including relaxin, insulin,
system. The GH molecule exhibits two binding sites for the GH IGF-I, and IGF-II. The IGFs are synthesized primarily by
receptor, resulting in dimerization of the receptor, a step that is the liver and act as a mitogen, stimulating DNA, RNA, and
required for biologic activity of the hormone. protein synthesis. IGF-I circulates in blood either free (half-
life is about 15–20 minutes) or bound to one of the several
GH effects at target organs specific binding proteins that prolong the half-life of the
peptide.
• Bone: GH, both directly and indirectly through circulating
and localized production of IGF-I, stimulates longitudinal
IGF receptor IGF-I and IGF-II bind specifically to two
growth by increasing the formation of new bone and carti-
high-affinity membrane-associated receptors that are ty-
lage. The growth effects of GH are not critical during the
rosine kinases and belong to the same family of receptors as
gestational period, but begin gradually during the first and
insulin. The insulin and IGF-I receptors, although similar in
second years of life and peak at the time of puberty. Before
structure and function, play different physiologic roles in
the epiphyses in long bones have fused, GH and IGF-1 stim-
vivo. The insulin receptor is primarily involved in metabolic
ulate chondrogenesis and widening of the cartilaginous epi-
functions, whereas the IGF-I receptor mediates growth and
physial plates, followed by bone matrix deposition. In addi-
differentiation. The separation of these functions is con-
tion to its effects on linear growth stimulation, GH plays a
trolled by several factors, including the tissue distribution of
role in regulating the normal physiology of bone formation
the respective receptors, the binding with high affinity
in the adult by increasing bone turnover, with increases in
of each ligand to its respective receptor, and the binding of
bone formation and, to a lesser extent, bone resorption.
IGF to plasma insulin-like growth factor binding proteins
• Adipose tissue: GH stimulates release and oxidation of free
(IGFBPs).
fatty acids, particularly during fasting. These effects are
mediated by a reduction in the activity of lipoprotein lipase,
the enzyme involved in clearing triglyceride-rich chylomi- Prolactin
crons and very-low-density lipoprotein particles from the Prolactin is a polypeptide hormone synthesized and secreted
bloodstream. Thus, GH favors the availability of free fatty ac- by lactotrophs in the anterior pituitary gland. The lactotro-
ids for adipose tissue storage and skeletal muscle oxidation. phs account for approximately 15–20% of the cell population
• Skeletal muscle: GH and IGF-I have anabolic actions on of the anterior pituitary gland, and this increases dramati-
skeletal muscle tissue mediated through stimulation of ami- cally in response to elevated estrogen levels, particularly dur-
no acid uptake and incorporation into protein, cell prolif- ing pregnancy. Prolactin levels are higher in females than in
eration, and suppression of protein degradation. males, and the role of prolactin in male physiology is not
• Liver: GH stimulates hepatic IGF-I production and release. completely understood. Plasma concentrations of prolactin
It stimulates hepatic glucose production. are highest during sleep and lowest during the waking hours
Overall, GH counteracts the action of insulin on lipid and in humans.
glucose metabolism, by decreasing skeletal muscle glucose uti-
lization, increasing lipolysis, and stimulating hepatic glucose Regulation of prolactin release
production. Prolactin release is predominantly under tonic inhibition by
Key aspects of GH physiology can be summarized as follows: dopamine derived from dopaminergic (D2) neurons of the
hypothalamus. D2 inhibition of lactotroph release of prolactin
• GH is produced and stored in somatotrophs in the anterior
is mediated by D2 Gαi protein–coupled receptors.
pituitary.
Prolactin release is affected by a large variety of stimuli
• The production of GH is pulsatile, mainly nocturnal, and is
provided by the environment, and the internal milieu, the
controlled mainly by GHRH and SST.
most important being suckling, increased levels of ovarian
• Circulating levels of GH increase during childhood, peak
steroid hormones, primarily estrogen. The release of prolac-
during puberty, and decrease with aging.
tin in response to suckling is a classical neuroendocrine reflex
• GH stimulates lipolysis, amino acid transport into cells, and
also referred to as a stimulus-secretion reflex (Figure 62–5).
protein synthesis.
This surge in prolactin release in response to a suckling stim-
• GH stimulates the production of IGF-I, which is responsible
ulus is mediated by a decrease in the amount of dopamine
for many of the activities attributed to GH.
released at the median eminence, relieving the lactotroph
from tonic inhibition. Estrogen stimulates growth of the lac-
Insulin-like Growth Factors totrophs during pregnancy as well as prolactin gene expres-
Many of the growth and metabolic effects of GH are medi- sion and release. Several neuropeptides have been identified
ated by the IGFs, or somatomedins. The importance of this as potential prolactin-releasing factors. These include TRH,
630 SECTION IX Endocrine and Metabolic Physiology

DISEASES OF THE
Hypothalamus
ANTERIOR PITUITARY
Dopamine
Alterations in function of the anterior pituitary can be due to
Dopamine
excess or deficient production of pituitary hormones or to
release Pituitary
altered responsiveness to hormone effects at the target organ.

HORMONE-PRODUCING
Prolactin PITUITARY ADENOMAS
Breast differentiation The most common cause of excess production of pituitary
Duct proliferation and branching hormones is a hormone-producing pituitary adenoma, a usu-
Glandular tissue development ally benign neoplasm. Prolactinomas are the most common
Milk protein synthesis
Lactogenic enzyme synthesis
(40–45%) pituitary tumors, followed by somatotroph (20%),
corticotroph (10–12%), gonadotroph (15%), and very rarely
thyrotroph adenomas. Small pituitary adenomas can cause
manifestations of excess tropic hormone production, whereas
• Mammary gland development
• Milk production larger tumors can produce neurologic symptoms by mass
effect in the sellar area. Patients with a prolactinoma present
with elevated levels of prolactin (hyperprolactinemia), milk
secretion (galactorrhea), and reproductive dysfunction. In
FIGURE 62–5 Physiologic effects of prolactin. Prolactin plays males, prolactinomas may cause infertility by producing
an important role in the normal development of mammary tissue and hypogonadism. In most cases, dopamine agonists are extremely
in milk production. Prolactin release is predominantly under negative effective in lowering serum prolactin levels, restoring gonadal
control by hypothalamic dopamine. Suckling stimulates the release of
function, decreasing tumor size, and improving visual fields
prolactin. Prolactin inhibits its own release by stimulating dopamine
decreased because of tumor compression of the optic chiasm.
release from the hypothalamus. (Modified with permission from Molina PE:
Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
GH-secreting adenomas can be associated with acromegaly or
bone and soft tissue overgrowth in adults, and with gigantism
in children. ACTH-releasing adenomas are associated with
excess cortisol production or Cushing syndrome; patients
oxytocin (OT), vasoactive intestinal peptide (VIP), and neu- present with central obesity, proximal myopathy, hyperten-
rotensin (NT). sion, mood changes, dorsocervical fat pads, and hyperglyce-
Prolactin regulates its own secretion through a short-loop mia, among other clinical signs and symptoms. Gonadotroph
feedback mechanism by binding to prolactin receptors located pituitary adenomas are frequently inefficient in hormone pro-
in neuroendocrine D2 neurons; this binding leads to increased duction. Thyrotropin-secreting tumors are exceedingly rare
hypothalamic dopamine synthesis (Figure 62–5). When the and are frequently large when diagnosed.
concentration of dopamine in the hypothalamo-hypophysial
portal blood increases, the release of prolactin from the lac-
totrophs is suppressed. HYPOPITUITARISM
Physiologic effects of prolactin The physiologic effects Hypopituitarism, or deficiency of anterior pituitary hor-
of prolactin are mediated by the prolactin receptor found in the mones, can be congenital or acquired. Pituitary insufficiency
mammary gland and the ovary. The main physiologic effects of can result from trauma, such as that associated with surgery,
prolactin are stimulation of growth and development of the penetrating injury, or automobile accidents, particularly
mammary gland, synthesis of milk, and maintenance of milk se- involving head trauma. Severe blood loss and decreased
cretion (Figure 62–5). Prolactin stimulates glucose and amino blood flow (ischemia) of the pituitary can also lead to pitu-
acid uptake and synthesis of the milk proteins β-casein and itary insufficiency. Ischemic damage to the pituitary gland or
α-lactalbumin, the milk sugar lactose, and milk fats by the mam- hypothalamic-pituitary stalk during the peripartum period
mary epithelial cells. During pregnancy, prolactin prepares the leads to Sheehan syndrome, manifested as hypothyroidism,
breast for lactation. The production of milk is prevented during adrenal insufficiency, hypogonadism (failure to resume nor-
pregnancy by the high progesterone levels. Additional effects of mal menses), and GH deficiency.
prolactin include inhibition of GnRH release, progesterone bio- GH deficiency and retarded growth may result from impaired
synthesis, and luteal cell hypertrophy during pregnancy. Prolac- release of GH from the pituitary gland because of diseases of the
tin also modulates reproductive and parental behavior. hypothalamus or pituitary gland or genetic predisposition.
CHAPTER 62 Anterior Pituitary Gland 631

Alternatively, mutations in the gene for the GH receptor can


tion of dopamine agonists to reduce prolactin production.
cause insensitivity to GH and growth retardation with low serum
Tumors that do not shrink in size with medical treatment
IGF-I concentrations. GH insensitivity syndrome is also known
may require focused radiation therapy and/or surgical
as Laron syndrome and is characterized by failure to generate
removal.
IGF-I and IGFBP-3. The typical manifestation is short stature or
dwarfism, which can be prevented by IGF-I treatment.
CASE B
EVALUATION OF ANTERIOR A middle-aged male patient consults his family physician
PITUITARY FUNCTION because he has noticed that his hat and wedding ring are
tight and his shoe size has increased one size during the
Measurements of anterior pituitary hormone concentrations past couple of years. He complains of joint aches and pains.
and of the respective target gland hormone levels are used to He also states that he has noticed his voice getting deeper
assess the functional status of the system. For example, paired and his facial features being thicker and coarser when com-
measures of TSH and thyroid hormone, FSH and estradiol, pared to his pictures of 10 years ago. Laboratory values
and ACTH and cortisol are used to evaluate the integrity of the show increased growth hormone and IGF-I levels and
respective systems. In addition, stimulation and inhibition increased fasting plasma glucose. An intravenous infusion
tests can be used to assess the functional status of the pituitary of glucose fails to decrease growth hormone levels. Brain
gland. These tests are based on the normal physiologic feed- MRI reveals a tumor localized to the pituitary. The patient
back mechanisms that control tropic hormone release. For is diagnosed with acromegaly resulting from a growth hor-
example, administration of the amino acid arginine can be mone–producing tumor.
used to elicit an increase in GH release in patients with sus- Acromegaly occurs as a result of growth hormone pro-
pected GH deficiency. In contrast, suppression tests can be duction in middle-aged adults. The symptoms of acromeg-
used to diagnose Cushing syndrome, a clinical state resulting aly develop slowly over many years, resulting in a frequent
from prolonged, inappropriate exposure to excessive endoge- delay in diagnosis after the estimated onset of symptoms.
nous secretion of cortisol. The clinical manifestations result from soft tissue growth in
response to growth hormone stimulation. This is evident in
thickening of facial features, hands, and feet but is also
associated with organomegaly (enlargement of internal
CLINICAL CORRELATION organs). Because of growth hormone’s anti-insulin actions
in adipose tissue, patients present with increased fasting
CASE A plasma glucose levels or impaired glucose tolerance. Diag-
A mother of two school-age children still in her reproduc- nosis is made by measurement of growth hormone release
tive years consults her physician because she has had no during the 2-hour period following a 75-g oral glucose load
menstrual periods for the past 6 months. A pregnancy test (similar to that used for the glucose tolerance test), as well
is negative, and she is not taking any medications. She also as by measurement of peripheral IGF-I levels. Treatment
complains of problems with her peripheral (temporal) consists of administration of long-acting somatostatin ana-
vision, and has also noted milky discharge from her nip- logs such as octreotide and surgical removal of the tumor
ples. An magnetic resonance image (MRI) scan of the in cases that do not respond to medical treatment. There
brain reveals a pituitary mass. On physical examination, are also GH receptor antagonists currently available that
she is afebrile and has a normal blood pressure. Laboratory can be used to treat the symptoms of GH excess.
values are within normal ranges for serum glucose, Na+
and K+. Increased levels of prolactin are measured, with
normal levels of all other pituitary hormones. She is diag-
nosed as having a prolactinoma. CHAPTER SUMMARY
Prolactinomas account for 50% of functioning pitu- ■ Anterior pituitary function is under regulation by the hypo-
itary tumors and are more frequent in females than in thalamus.
males. Tumor size is correlated with prolactin levels. ■ Anterior pituitary hormone release is under feedback regula-
Women present with amenorrhea (lack of menses), tion by peripheral hormone levels.
galactorrhea (milk discharge from nipples), and infertil- ■ Pulsatile release of hypothalamic, pituitary, and target organ
ity. Large tumors extending caudal to the pituitary are hormones plays an important role in endocrine function.
associated with visual defects due to compression of the ■ Growth hormone exerts direct and indirect (IGF-I) effects on
optic chiasm. In males, presentation is impotence, loss of linear growth and metabolism.
libido, and infertility as well as headaches and alterations ■ With the exception of dopamine inhibition of prolactin and
in visual field. Initial treatment consists of administra- SST inhibition of GH, adenohypophysial hormones are under
stimulatory hypothalamic control.
632 SECTION IX Endocrine and Metabolic Physiology

STUDY QUESTIONS 3. A patient has been taking pharmacologic doses of a steroidal


anti-inflammatory drug for a prolonged period for his asthma
1. A female patient presents with milky discharge from both flare-ups. Which of the following is likely to occur to
nipples and menstrual irregularity. The patient has no history of hypothalamic CRH release and plasma cortisol?
headache or visual abnormalities. A pregnancy test is negative, A) low CRH and high cortisol
and her mammogram and breast exam are normal. She has a list of B) high CRH and high cortisol
medications that her psychiatrist has recently prescribed. Which C) low CRH and low cortisol
of the following could be responsible for the milky discharge? D) high CRH and low cortisol
A) levodopa 4. Growth hormone excess in an adult male can present with
B) dopamine antagonist
A) longitudinal growth of the extremities
C) D2 agonist
B) thickening of cartilaginous tissue
D) serotoninergic agonist
C) hypoglycemia
2. Damage to the axons passing through the pituitary stalk during D) low IGFBP-3
an automobile accident could be expected to result in any of the
following with the exception of
A) decreased prolactin release
B) decreased TSH release
C) decreased LH and FSH release
D) decreased GH release
63
C H A P T E R

Thyroid Gland
Patricia E. Molina

O B J E C T I V E S

■ Identify the steps and control factors of thyroid hormone biosynthesis,


storage, and release.
■ Describe the distribution of iodine and the metabolic pathway involved in
thyroid hormone synthesis.
■ Explain the importance of thyroid hormone binding in blood for free and
total thyroid hormone levels.
■ Understand the significance of the conversion of tetraiodothyronine (T4) to
triiodothyronine (T3) and reverse T3 (rT3) in extrathyroidal tissues.
■ Understand how thyroid hormones produce their cellular effects.
■ Describe their effects on development and metabolism.
■ Understand the causes and consequences of excess and deficiency of thyroid
hormones.

Thyroid hormones play important roles in maintaining energy • endothelial cells lining the capillaries that provide the blood
homeostasis and regulating energy expenditure. Their physio- supply to the follicles;
logic effects, mediated at multiple target organs, are primarily to • parafollicular or C cells, involved in the production of
stimulate cell metabolism and activity. The vital roles of these calcitonin, a hormone with a minor role in calcium ho-
hormones, particularly in development, differentiation, and mat- meostasis (discussed in Chapter 64);
uration, are underscored by the severe intellectual and develop- • fibroblasts, lymphocytes, and adipocytes.
mental delay observed in infants with deficient thyroid hormone
function during gestation. Thyroid hormones are derived from
the amino acid tyrosine and are produced by the thyroid gland in THYROID FOLLICLE
response to stimulation by thyroid-stimulating hormone (TSH)
produced by the anterior pituitary. TSH, in turn, is regulated by The main function of the thyroid gland is the synthesis and
negative feedback by thyroid hormone and by the hypophysio- storage of thyroid hormone. The functional unit of the thy-
tropic peptide thyrotropin-releasing hormone (TRH). roid gland is the thyroid follicle, a spherical structure con-
sisting of a layer of thyroid epithelial cells arranged around
a large central cavity filled with colloid. Colloid makes up
FUNCTIONAL ANATOMY about 30% of the thyroid gland mass and contains a protein
called thyroglobulin. Thyroglobulin plays a central role in
The thyroid gland is a highly vascular, ductless gland the synthesis and storage of thyroid hormone. The parafol-
located in the anterior neck in front of the trachea. The cel- licular cells are in the spaces between follicles. Most para-
lular composition of the thyroid gland is diverse, including follicular cells synthesize and secrete the hormone
the following: calcitonin, and therefore they are frequently referred to as
• follicular (epithelial) cells, involved in thyroid hormone C cells.
synthesis;

633

Ch63_633-642.indd 633 11/30/10 3:27:57 AM


634 SECTION IX Endocrine and Metabolic Physiology

REGULATION OF BIOSYNTHESIS, TSH receptor activation results in stimulation of all of the


steps involved in thyroid hormone synthesis, including iodine
STORAGE, AND SECRETION OF uptake and organification, production and release of thyroid
hormones from the gland, and promotion of thyroid growth.
THYROID HORMONES The biologic effects of TSH include stimulation of activity and
production of the Na+/I− symporter, thyroglobulin, thyroid
HYPOTHALAMIC REGULATION peroxidase, and T4 and T3. Both growth and function of the
OF TSH RELEASE thyroid are stimulated by TSH stimulation. Therefore, contin-
ued stimulation of the TSH receptor causes hyperthyroidism
Thyroid hormone synthesis and release is under negative feed-
and thyroid hyperplasia.
back regulation by the hypothalamic–pituitary–thyroid axis
(Figure 63–1). TRH is a tripeptide synthesized in the hypo-
thalamus, released from nerve terminals in the median emi- THYROID HORMONE SYNTHESIS
nence, and transported to the anterior pituitary where it
stimulates release of TSH into the systemic circulation. The Thyroglobulin is a glycoprotein synthesized in the thyroid
release of TSH is inhibited by the thyroid hormones tri- follicular epithelial cells and secreted into the follicular lumen,
iodothyronine (T3) and tetraiodothyronine (T4, thyroxine). where it is stored in the colloid. It undergoes major posttrans-
TSH is transported in the bloodstream to the thyroid gland, lational modification during the production of thyroid hor-
where it binds to the TSH receptor located on the thyroid folli- mones consisting of iodination of multiple tyrosine residues,
cular epithelial cells. The TSH receptor is an important antigenic followed by coupling of some of the iodotyrosine residues to
site involved in thyroid autoimmune disease. Autoantibodies to form T3 and T4. A small amount of noniodinated thyroglobulin
the receptor may act as agonists mimicking the actions of TSH is also secreted into the circulation, and it can be increased in
in Graves’ disease. diseases such as thyroiditis and Graves’ disease.

Hypothalamus

TRH

(-)

Dopamine (-)
Somatostatin
Glucocorticoids Pituitary
(-)

FIGURE 63–1 The hypothalamic–pituitary–thyroid axis. TSH


Thyrotropin-releasing hormone (TRH) is synthesized in parvocellular
neurons of the paraventricular nucleus of the hypothalamus and
released from nerve terminals in the median eminence, from where
it is transported via the portal capillary plexus to the anterior
pituitary. TRH binds to a G protein–coupled receptor in the anterior
pituitary, leading to an increase in intracellular Ca2+ concentration, T4
which in turn results in stimulation of exocytosis and release of
thyroid-stimulating hormone (TSH) into the systemic circulation. T3
TSH stimulates the thyroid gland to increase the synthesis and
secretion of tetraiodothyronine (T4) and triiodothyronine (T3) into
the circulation. T4 and T3 inhibit the secretion of thyrotropin (TSH) Thyroid
both directly and indirectly by inhibiting the secretion of TRH.
Additional factors that inhibit TSH release are glucocorticoids,
somatostatin, and dopamine. (Modified with permission from Molina PE:
Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
CHAPTER 63 Thyroid Gland 635

The iodide required for thyroid hormone synthesis is read- ease and toxic nodules, and to ablate thyroid tissue in the
ily absorbed from dietary sources, primarily from iodized salt, treatment of thyroid cancer.
but also from seafood and plants grown in soil that is rich in The iodination of thyroglobulin residues is a process that
iodine. Iodide is removed from the blood primarily by the thy- occurs at the apical membrane of the thyroid follicular cell.
roid and the kidneys. Thus, once inside the cell, iodide must leave the follicular cell
through apical efflux via a chloride–iodide transporting
protein (iodide channel). The uptake, concentration, and
REGULATION OF IODINE METABOLISM efflux of iodide are all functions of TSH-stimulated transepi-
IN THE THYROID FOLLICULAR CELL thelial transport of iodide.
The transport of iodine through the Na+/I− symporter can
Iodide is concentrated in thyroid epithelial cells by a sodium– be substituted by other substances including perchlorate and
iodide (Na+/I−) symporter (Figure 63–2). This ability of the pertechnetate. Radiolabeled pertechnetate (Tc99m-pertechne-
thyroid gland to accumulate iodine allows therapeutic admin- tate) can be used for imaging of the thyroid gland that reflects
istration of radioactive iodine for the treatment of Graves’ dis- the ability of specific regions of the thyroid to take up the iso-

Follicle lumen
(colloid)
Apical
membrane

Follicular
Thyroid cell
gland

Iodide

2Na+/I –
symporter
I–
Na+

3Na+/2K+
ATPase
FIGURE 63–2 Mechanism of iodide concentration
Na +

by the thyroid gland. Iodide is transported into the cytosol


K+

of the follicular cell by active transport against a chemical


and electrical gradient. The energy is derived from the
Basolateral
K + ATP

electrochemical gradient of sodium. Two sodium ions are


membrane
transported inside the thyroid follicular cell with each iodide
P
Na +

AD

molecule. Sodium moves down its concentration gradient,


which is maintained by the Na+/K+-ATPase that constantly
pumps sodium out of the cytoplasm of the thyroid follicular
Low epithelial cell, maintaining the low intracytoplasmic
intracellular concentration of sodium. Iodide must reach the colloid space,
Na where it is used for organification of thyroglobulin. This process
is achieved by efflux through the iodide channel. One of the
early effects of TSH binding to its receptor is the opening of
these channels, which facilitate the leakage of iodide into the
extracellular space. This transcellular transport of iodide relies
on the functional and morphologic polarization of the thyroid
Iodide channel follicular epithelial cell. ATP, adenosine triphosphate. (Modified
Apical
with permission from Molina PE: Endocrine Physiology, 3rd ed. New York:
membrane
McGraw-Hill Medical, 2010.)
Colloid
636 SECTION IX Endocrine and Metabolic Physiology

1 BLOOD 90% T4 10% T3 roglobulin yields monoiodinated tyrosine (MIT) and diiodi-
Iodide Na+ I– 5 nated tyrosine (DIT) residues that are enzymatically coupled
trapping to form T3) or T4 by the enzyme thyroid peroxidase. Because
Hormone
secretion not all of the iodinated tyrosine residues undergo coupling,
2 T3 thyroglobulin stored in the follicular lumen contains MIT and
T4
Tg synthesis DIT residues as well as formed T3 and T4.
and secretion
Iodine metabolism within the thyroid can also be regulated
independently of TSH, particularly when plasma iodide levels
are increased via an autoregulatory phenomenon known as
I– I– MIT
the Wolff–Chaikoff effect. This effect lasts for a few days and
pool pool DIT is followed by the so-called “escape” phenomenon, at which
MIT point the organification of intrathyroidal iodine resumes and
Tg DIT
T3 T4 the normal synthesis of T4 and T3 returns.

Tg
REGULATION OF THYROID
HORMONE RELEASE
I–
Tg TSH stimulates release of thyroid hormones from the thyroid
4 gland. This process involves endocytosis of vesicles containing
Colloid uptake thyroglobulin from the apical surface of the follicular cell, lys-
Tg by endocytosis
I– I° osomal fusion of the vesicles, and proteolytic cleavage of thy-
iodine Thyroid iodine
peroxidase 3 MIT roglobulin. As a result, the products of proteolytic cleavage
Iodination include the thyroid hormones T4 and T3, which are released
+ conjugation Tg DIT
T3 T4
into the circulation, and the iodinated tyrosine residues (MIT
and DIT) that are deiodinated intracellularly. With normal
THYROID COLLOID
iodine intake, greater amounts of T4 than T3 are released
FIGURE 63–3 Overview of thyroid hormone synthesis in the (plasma concentrations of T4 are 40-fold higher than those of
thyroid follicular epithelial cell. Thyroid hormone synthesis T3). Most of the T4 released is converted to T3 (the more active
involves concentration of iodide by the Na+/I− symporter (1) and form of the hormone) in peripheral tissues by the removal of
transport of iodide through the epithelial cell and into the
iodine from carbon 5 on the outer ring of T4. Key features of
extracellular compartment of the follicular cells, where it is oxidized
thyroid regulation and function are listed in Table 63–1.
to iodine by thyroid peroxidase (3) and is then used in the iodination
of thyroglobulin (Tg) synthesized within the cell and released into the
colloid (2). Iodine organification is an extracellular process that
takes place inside the thyroid follicles at the apical membrane
surface facing the follicular lumen. Iodine is bound to either carbon
TABLE 63–1 Key features of thyroid regulation
3 or carbon 5 of the tyrosyl residues of thyroglobulin to form and function.
monoiodinated tyrosine (MIT) and diiodinated tyrosine (DIT). An MIT
Key Features
and a DIT, or two DITs are conjugated to form triiodothyronine (T3)
and tetraiodothyronine (T4), respectively. Secretion of the hormone TSH Binds to Gαs protein–coupled receptor
involves endocytosis of colloid containing thyroglobulin (4), followed on thyroid follicular cells. Main second-
by degradation of thyroglobulin and release of T4 and T3 (5). Some of messenger system is cAMP. Stimulates
the T4 produced is deiodinated in the thyroid follicle to T3, which is all steps involved in thyroid hormone
then released into the bloodstream. In addition, intracellular synthesis: iodine uptake and
organification, production and release
deiodination provides a mechanism for recycling iodide to participate
of thyroid hormone, and promotion of
in the synthesis of new thyroid hormone at the apical cell surface. A thyroid growth
small fraction of thyroglobulin is released from the follicular epithelial
cell into the circulation. (Reproduced with permission from Kibble J, Halsey CR: Thyroid gland Can store 2–3-month supply of thyroid
hormones in the thyroglobulin pool
The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)
(colloid). Produces more T4 than T3

Thyroid hormone Synthesis and release are under negative


tope and therefore to function normally. In the follicular feedback regulation by the hypothalamic–
lumen, tyrosine residues of thyroglobulin are iodinated by pituitary–thyroid axis. T4 is converted to T3
iodine (Io; formed by oxidation of I− by thyroid peroxidase) in peripheral tissues. Biologic activity of T3
is greater than that of T4. Binds to nuclear
(Figure 63–3). This reaction requires hydrogen peroxide.
receptors and modulates gene
Iodine bonds to carbon 3 or carbon 5 of the tyrosine residues transcription
on thyroglobulin in a process referred to as the organification TSH, thyroid-stimulating hormone; cAMP, cyclic adenosine monophosphate; T4,
of iodine. The iodination of specific tyrosines located on thy- tetraiodothyronine; T3, triiodothyronine.
CHAPTER 63 Thyroid Gland 637

TRANSPORT AND TISSUE DELIVERY from carbon 5 in the inner ring to yield reverse T3 (rT3). rT3 has
OF THYROID HORMONES little or no biologic activity. Following conversion of T4 to T3 or
rT3, these are converted to T2, a biologically inactive hormone.
Once thyroid hormones are released into the circulation, most of This extrathyroidal progressive deiodination of thyroid hor-
them circulate bound to plasma proteins (thyroid-binding glob- mones catalyzed by deiodinases plays a significant role in thy-
ulin [TBG], transthyretin, and albumin). This high percentage roid hormone metabolism and involves three deiodinases
of protein-bound hormone significantly prolongs thyroid hor- (type I, II, and II) that differ in their tissue distribution, cata-
mone half-life. A small fraction of each hormone circulates in the lytic profiles, substrate specificities, physiologic functions, and
free form that is bioavailable and can enter the cell to bind to regulation outlined in Figure 63–4.
the thyroid receptor. Of the two thyroid hormones, T4 binds more
tightly to binding proteins than T3 and thus has a lower metabolic BIOLOGIC EFFECTS OF
clearance rate and a longer half-life (7 days) than T3 (1 day). THYROID HORMONES
THYROID HORMONE METABOLISM Thyroid hormones are essential for normal growth and devel-
opment; they control the rate of metabolism and hence the
Thyroid hormone peripheral metabolism is a sequential deio- function of practically every organ in the body. Thyroid hor-
dination process catalyzed by tissue deiodinases, leading first mone receptors are expressed in most tissues and affect mul-
to a more active form of thyroid hormone (T3) and finally to tiple cellular events. The following are some examples of the
complete inactivation of the hormone (Figure 63–4). Approxi- specific effects of thyroid hormone.
mately 40% of T4 is deiodinated at carbon 5 in the outer ring to Their effects are mediated primarily by the transcriptional
yield the more active T3. In about 33% of T4, iodine is removed regulation of target genes, and are thus known as genomic

Glucuronidation
(T4G)
Tetraiodothyronine (T4, thyroxine).
NH2
Sulfation HO O CH2 CH COOH
(T4S)

Deiodinase I & II Deiodinase I & III

FIGURE 63–4 Thyroid hormone


Outer ring Inner ring metabolism. Peripheral metabolism of thyroid
deiodination deiodination hormones involves the sequential removal of
(T3) (rT3)
iodine molecules, converting T4 into the more
active T3 and inactivating thyroid hormones
before their excretion. In addition, thyroid
O O hormones can undergo conjugation in the liver,
=

which increases their solubility and facilitates


=

3' 3 CH2 CH C OH HO CH2 CH C OH


HO O O their biliary excretion. The type I iodothyronine
5
NH2 NH2 deiodinase is expressed predominantly in liver,
3,5,3' – Triiodothyronine
kidney, and thyroid. It catalyzes both outer- and
Reverse T3 (rT3)
(T3) inner-ring deiodination of thyroid hormones. It
is the primary site for clearance of plasma rT3
and a major source of circulating T3. Type II
deiodinase is expressed primarily in the human
Deiodinase I & III Deiodinase I & II brain, anterior pituitary, and thyroid. It has only
outer-ring deiodination activity and plays an
important role in the local production of T3 in
tissues that express this enzyme. The type III
deiodinase is located predominantly in human
O
brain, placenta, and fetal tissues. It has only
=

HO O CH2 CH C OH inner-ring activity and catalyzes the inactivation


of T3 more effectively than that of T4, thereby
NH2 regulating intracellular T3 levels. (Modified with
permission from Molina PE: Endocrine Physiology, 3rd ed.
Diiodothyronine (T2) New York: McGraw-Hill Medical, 2010.)
638 SECTION IX Endocrine and Metabolic Physiology

effects. Thyroid hormones enter cells by a carrier-mediated Liver


energy-, temperature-, and Na+-dependent process. The cel-
Thyroid hormone regulates triglyceride and cholesterol
lular actions of thyroid hormones are mediated by nuclear
metabolism, as well as lipoprotein homeostasis. It also modu-
receptors. They bind thyroid hormones with high affinity
lates cell proliferation and mitochondrial respiration.
and specificity, particularly for T3. Thyroid hormone recep-
tors are DNA-binding transcription factors that function
as molecular switches in response to hormone binding. The Pituitary
hormone receptor can activate or repress gene transcription, Thyroid hormone regulates the synthesis of pituitary hormones,
depending on the promoter context and ligand-binding sta- facilitates growth hormone production, and inhibits TSH.
tus. The cellular events mediated by thyroid hormones are:
• transcription of cell membrane Na+/K+-ATPase, leading to Brain
an increase in oxygen consumption;
Thyroid hormone controls expression of genes involved in
• transcription of uncoupling proteins, enhancing fatty acid
myelination, cell differentiation, migration, and signaling. It is
oxidation and heat generation without production of ATP;
necessary for axonal growth and development.
• protein synthesis and degradation, contributing to growth
and differentiation;
• epinephrine-induced glycogenolysis and gluconeogenesis,
affecting insulin-induced glycogen synthesis and glucose
DISEASES OF THYROID
utilization; HORMONE OVERPRODUCTION
• cholesterol synthesis and low-density lipoprotein receptor
regulation. AND UNDERSECRETION
The widespread distribution of thyroid hormone receptors
ORGAN-SPECIFIC EFFECTS OF and the multitude of physiologic effects that they exert are
highlighted in cases of abnormal thyroid function (Table 63–2).
THYROID HORMONE Dysfunction can result from three factors: (1) alterations in
the circulating levels of thyroid hormones, (2) altered metabo-
Bone lism of thyroid hormones in the periphery, and (3) resistance
Thyroid hormone is important for bone growth and develop- to thyroid hormone actions at the tissue level.
ment through activation of osteoclast and osteoblast activities. An individual whose thyroid function is normal is said to
Deficiency during childhood affects growth. In adults, excess be in a euthyroid state. The clinical state resulting from an
thyroid hormone levels are associated with increased risk of alteration in thyroid function is classified as either hypothy-
osteoporosis. roidism (low thyroid function) or hyperthyroidism (exces-
sive thyroid function). Autoimmunity plays an important role
Cardiovascular System in thyroid disease. Abnormal immune responses directed to
thyroid-related proteins result in two opposite pathogenic
Thyroid hormone has cardiac inotropic and chronotropic
processes: thyroid enlargement (hyperplasia) in Graves’
effects, increases cardiac output and blood volume, and
disease and thyroid inflammation and/or destruction in
decreases systemic vascular resistance. These responses are
Hashimoto’s thyroiditis.
mediated through thyroid hormone changes in gene transcrip-
The most common presentations of thyroid hormone abnor-
tion of several proteins including Ca2+-ATPase, phospholam-
malities are summarized as follows.
ban, myosin, β-adrenergic receptors, adenylate cyclase,
guanine nucleotide–binding proteins, Na+/Ca2+ exchanger,
Na+/K+-ATPase, and voltage-gated potassium channels. The HYPOTHYROIDISM
effect on β-adrenergic receptor expression accounts for the
sympathomimetic effects of high levels of thyroid hormone. Hypothyroidism is the condition resulting from insufficient
thyroid hormone action. It has an incidence of 2% in adult
women and is less common in men. Two main types are dis-
Fat tinguished, primary and secondary hypothyroidism, although
Thyroid hormone induces white adipose tissue differentiation, the former is more common.
lipogenic enzymes, and intracellular lipid accumulation; stim-
ulates adipocyte cell proliferation; stimulates uncoupling pro-
teins; and uncouples oxidative phosphorylation. The induction Primary Hypothyroidism
of catecholamine-mediated lipolysis by thyroid hormones When the decrease in thyroid function occurs in utero, the
results from an increased β-adrenoceptor (AR) number and a result is severe intellectual and developmental delay or cretin-
decrease in phosphodiesterase (PDE) activity resulting in an ism, underscoring the vital role that thyroid hormone plays in
increase in cAMP level and hormone-sensitive lipase (HSL) development and growth. Hypothyroidism may be associated
activity. with thyroid enlargement, resulting from inflammation as in
CHAPTER 63 Thyroid Gland 639

TABLE 63–2 Clinical and laboratory manifestations of excess or deficient thyroid hormone function.
Hypothyroidism Hyperthyroidism

Clinical Presentation Laboratory Values Clinical Presentation Laboratory Values

In utero: Primary: Palpitations, exercise impairment, Primary:


Cretinism, mental and growth retardation, short limbs Low free T4; low widened pulse pressure, tachycardia at Low TSH; high T4
Adult onset: or sometimes rest and during exercise, increase in (2-fold) and T3
Tiredness, lethargy, constipation, decreased appetite, normal T3; high blood volume, palpable enlarged (3–4-fold). In Graves’
cold intolerance, abnormal menstrual flow, hair loss, TSH thyroid gland, infiltrative disease, elevated
brittle nails, dry coarse skin, hoarse voice Secondary: ophthalmopathy, nervousness, anti-TSH receptor
Chronic: Low T4 and T3, irritability, hyperactivity, emotional antibody titers
Myxedema, thickened features, periorbital edema, normal or instability, tenseness, pounding of the Secondary:
swelling of hands and feet without indentation, inappropriately heart, heat intolerance, weight loss High TSH, T4, and T3
delayed muscle contraction and relaxation, delayed low TSH despite increased food intake, decreased
tendon reflexes, reduced stroke volume and heart or absent menstrual flow, increased
rate, decreased cardiac output, enlarged heart, number of bowel movements, warm
pericardial effusion, pleural and peritoneal fluid moist skin or velvety texture, proximal
accumulation, slowing of mental function, impaired muscle weakness, fine hair, fine tremor,
memory, slow speech, decreased initiative, excessive sweating
somnolence, hypothermia
T4, thyroxine; T3, triiodothyronine; TSH, thyroid-stimulating hormone.

Hashimoto’s thyroiditis and increases in TSH due to dietary TSH-secreting adenomas represent a very small fraction
iodine deficiency. (<1%) of all pituitary adenomas and result in a syndrome of
excess secretion of TSH. The hormonal profile is characterized
Secondary Hypothyroidism by the inability to suppress TSH despite increased levels of free
Secondary hypothyroidism is characterized by decreased thyroid hormones (T3 and T4).
TSH secretion and subsequently decreased thyroid hormone
release, and is usually due to hypopituitarism (decreased ABNORMALITIES IN IODINE
anterior pituitary function).
METABOLISM
HYPERTHYROIDISM Iodine is an essential component of thyroid hormone, but both
low and high iodine intake may lead to disease. Several drugs
Hyperthyroidism is excessive functional activity of the thyroid can interfere with the ability of the thyroid gland to concen-
gland, characterized by increased basal metabolism and distur- trate iodide. Perchlorate is a contaminant that can be found in
bances in the activity of the autonomic nervous system as a result drinking and groundwater, and occasionally in cow’s milk.
of excess thyroid hormone production. The incidence is higher in Ingestion of high levels, as with consumption of well water,
women than in men. Several conditions can lead to hyperthy- can lead to inhibition of iodine transport into the follicular
roidism, but the most common cause in adults is Graves’ disease. cells. Methimazole, the active form of carbimazole, inhibits
iodide uptake and organification of iodine. Thiouracil and
Graves’ Disease propylthiouracil inhibit iodine organification.
Abnormalities in the metabolism or supply of iodine have
Graves’ disease is an autoimmune condition leading to auton-
particular importance in fetal development. Severe iodine
omous thyroid hormone secretion resulting from the stimula-
deficiency of the mother may lead to insufficient thyroid hor-
tion of TSH receptor by TSH-like antibodies called
mone synthesis in both mother and fetus, resulting in develop-
thyroid-stimulating immunoglobulins (TSI). Clinically, about
mental brain injury. Iodine deficiency is the leading cause of
40–50% of patients with hyperthyroidism present with pro-
preventable mental retardation worldwide, although it is rare
truding eyes (exophthalmos or proptosis) as a result of lym-
in the USA where table salt is supplemented with iodine. On
phocyte and fibroblast infiltration of the extraocular tissues
the other hand, excess iodine given to the mother (e.g., use of
and muscles, and accumulation of hyaluronate, a glycosamin-
an antiarrythmic iodine-containing drug called amiodarone
oglycan produced by fibroblasts in the tissues and muscles.
or excess iodine supplementation) may inhibit fetal thyroid
The majority (90%) of patients with thyroid ophthalmopathy
function, leading to hypothyroidism and goiter, or may pre-
have Graves’ hyperthyroidism.
cipitate hyperthyroidism (iodine toxicity).
TSH-secreting Adenomas
Secondary hyperthyroidism is due to increased thyroid hor- Goiter
mone release by the thyroid gland in response to increased Goiter is defined as an overall enlargement of the thyroid
TSH levels derived from TSH-secreting pituitary adenomas. gland. It can be associated with either decreased (iodine
640 SECTION IX Endocrine and Metabolic Physiology

deficiency or Hashimoto’s) or increased (Graves’ disease)


population, and is more frequent in females than in
thyroid function. Enlargement of the gland can be the result
males. The excess production of thyroid hormone in
of increased TSH receptor stimulation leading to thyroid
Graves’ disease is the result of circulating IgG antibodies
hyperplasia or of localized inflammatory and/or autoimmune
that bind to the TSH receptor on the thyroid gland, acti-
processes.
vating the G protein–coupled receptor. This excess stim-
ulation of the receptor leads to follicular hypertrophy and
Thyroiditis hyperplasia, causing thyroid enlargement, as well as
Thyroiditis, or inflammation of the thyroid gland, may lead increases in thyroid hormone production. As a result of
to abnormalities in thyroid hormone state. This condition excess thyroid hormone production, the release of TSH is
may be acute, subacute, or chronic. Chronic thyroiditis markedly suppressed via negative feedback. Clinical
(Hashimoto’s thyroiditis, chronic lymphocytic thyroiditis, manifestations are the result of excess thyroid hormone
autoimmune thyroiditis) is an autoimmune disease of the thy- activity. Treatment for Graves’ disease includes antithy-
roid gland characterized by lymphocyte infiltration and circu- roid drugs (inhibitors of thyroid hormone synthesis),
lating autoimmune antibodies. These antibodies inhibit the radioiodine ablation of the thyroid tissue, and surgical
Na+/I− symporter, preventing iodide uptake and consequently removal of the thyroid gland.
thyroid hormone synthesis. Hashimoto’s thyroiditis is the most
common cause of adult hypothyroidism; it is more prevalent
in women than in men, and peaks at the age of 30–50 years.
CHAPTER SUMMARY
■ The thyroid gland is regulated by the hypothalamic–pituitary–
EVALUATION OF THE thyroid axis.
■ Dietary iodine is required for thyroid hormone synthesis.
HYPOTHALAMIC–PITUITARY– ■ The thyroid gland produces thyroid hormones by a process of
THYROID AXIS concentration of iodine in the thyroid, iodination of tyrosine
residues of thyroglobulin in the colloid space of the follicle, and
TSH AND THYROID HORMONE LEVELS endocytosis of colloid followed by proteolytic release of thyroid
hormones (T4 and T3).
TSH levels are useful in evaluation of the patient because small ■ Thyroid hormones undergo metabolism in peripheral tissues,
changes in free thyroid hormone levels lead to greater changes leading to the production of the more active T3 and deactiva-
in TSH levels. Serum TSH concentrations are considered the tion of thyroid hormones.
single most reliable test to screen for all common forms of ■ The presence of deiodinases and their substrate specificity play
hypothyroidism and hyperthyroidism, particularly in the a central role in thyroid hormone function in target tissues.
ambulatory setting. Overt primary hyperthyroidism is accom- ■ Thyroid hormone receptors are located intracellularly, bound
panied by low serum TSH concentrations. Interpretation of to DNA, and alter gene transcription on binding by thyroid
abnormal TSH levels is best done with simultaneous measures hormone.
of thyroid hormone levels. ■ Thyroid hormone actions are systemic and vital for develop-
ment, growth, and metabolism.

CLINICAL CORRELATION STUDY QUESTIONS


1. Which of the following blood laboratory values would be
A premenopausal woman presents with palpitations and a compatible with hyperthyroidism due to Graves’ disease?
history of weight loss over the past 6 months. In addition, A) low TSH and T4 levels
she complains of increased irritability and anxiety, inability B) high TSH but low T4 levels
to sleep, and heat intolerance. Physical examination reveals C) low TSH and high T4 levels
tachycardia (pulse of 120 beats/min), hypertension (a D) high T4 and low T3 levels
blood pressure of 139/80 mm Hg), and a diffusely enlarged 2. The following laboratory results are available before physical
nontender thyroid gland. In addition, she has infrequent examination of a new patient: very low TSH levels, and increased
blinking and characteristic stare, sweaty hands, and tremor. T4 and T3. Which of the following signs and symptoms would you
Her serum TSH level is low, and the level of free thyroxine expect to find?
is increased. She is diagnosed with Graves’ disease. A) heart rate of 45 beats/min, diarrhea, and weight gain
Graves’ disease accounts for 50–80% of cases of B) sweaty hands, 40-g goiter, and constipation
hyperthyroidism, affects approximately 0.5% of the C) heat intolerance, irritability, and weight loss
D) insomnia, delayed muscle relaxation, and diarrhea
CHAPTER 63 Thyroid Gland 641

3. Blood laboratory values that would be compatible with endemic 4. A radioactive iodine scan revealed greater concentration of
goiter due to iodine deficiency include radioiodine when compared to that of other asymptomatic
A) increased TSH, and decreased T4 and T3 levels individuals. Subsequent laboratory values came back positive for
B) increased free T4 levels and thyroid-stimulating Ig positive serum titers of autoantibodies to the TSH receptor. The
C) low TSH, T4, and T3 levels pathophysiology of disease in this patient involves
D) normal free T4 and low TSH levels A) increased deiodination of T4 to T3 in the liver
B) decreased thyroid hormone–binding levels
C) increased cAMP formation in the thyroid follicular cell
D) downregulation of the Na+/I– symporter
This page intentionally left blank
64
C H A P T E R

Parathyroid Gland and


Calcium and Phosphate
Regulation
Patricia E. Molina

O B J E C T I V E S

■ Identify the origin, target organs and cell types, and physiologic effects of
parathyroid hormone.
■ Describe the functions of osteoblasts and osteoclasts in bone remodeling
and the factors that regulate their activities.
■ Describe the regulation of parathyroid hormone secretion, the role of the
calcium-sensing receptor, and the negative feedback relationship between
parathyroid hormone and the biologically active form of vitamin D.
■ Identify the sources, biosynthetic pathway, and physiologic effects of vitamin
D and its active metabolites.
■ Describe the causes and consequences of excess or deficiency of parathyroid
hormone and vitamin D.
■ Describe the regulation of calcitonin release and the cell of origin and target
organs for calcitonin action.
■ Explain the hormonal regulation of plasma calcium concentration through
bone resorption, renal excretion, and intestinal absorption.
■ Explain the hormonal regulation of plasma phosphate concentration through
exchange with bone, renal excretion, and dietary intake and absorption.

The regulation of plasma calcium levels is critical for normal


cell function, neural transmission, membrane stability, bone
FUNCTIONAL ANATOMY
structure, blood coagulation, and intracellular signaling. This The parathyroid glands are pea-sized glands located at the top
regulation relies on the interactions between parathyroid and bottom posterior borders of the lateral lobes of the thyroid
hormone (PTH) from the parathyroid glands and dietary and gland. The chief cells synthesize and secrete the polypeptide
endogenous vitamin D (Figure 64–1). PTH stimulates bone PTH, which plays a major role in bone remodeling, calcium
resorption and the release of calcium (and phosphate) into homeostasis, renal excretion of phosphate, and activation of
the circulation. In the kidney, PTH promotes Ca2+ reabsorp- vitamin D.
tion and inorganic phosphate excretion in the urine. In addi-
tion, PTH stimulates the hydroxylation of 25-hydroxyvitamin
D (25(OH)D) at the 1-position, leading to the formation of
1,25-dihydroxyvitamin D (1,25(OH)2D), the active form of
REGULATION OF PTH RELEASE
vitamin D. 1,25(OH)2D exerts its main effects through PTH is synthesized as a prepropeptide that is posttranslation-
increased intestinal absorption of dietary calcium. In addition, ally modified to yield PTH, an 84–amino acid polypeptide.
and to a lesser degree, it contributes to renal reabsorption of PTH release is controlled in a tight feedback system by small
filtered calcium and bone resorption. The overall result of the changes in plasma calcium levels detected by the parathyroid
interactions between PTH and vitamin D is the maintenance calcium-sensing receptor, a G protein–coupled receptor
of normal plasma calcium concentrations. located on the plasma membrane of the parathyroid chief cells;

643

Ch64_643-654.indd 643 11/30/10 3:30:58 AM


644 SECTION IX Endocrine and Metabolic Physiology

1,25(OH)2D

FIGURE 64–1 Regulation of parathyroid Kidney:


hormone (PTH) release. A decrease in calcium ↑Vitamin D activation
Ca2+ ↑Ca2+ reabsorption
(Ca2+) stimulates the release of PTH from the
↓Pi reabsorption
parathyroid gland. PTH increases the activity of
1α-hydroxylase in the kidney, leading to ↑Ca2+ ↓PTH release
increased production of 1,25(OH)2D. In addition,
PTH increases the reabsorption of calcium and
decreases the reabsorption of inorganic
phosphate (Pi). In bone, PTH stimulates bone ↑PTH release
resorption, increasing the plasma calcium levels.
↑Pi
The increases in 1,25(OH)2D and plasma calcium
levels exert negative feedback inhibition of ↓ Ca2+
PTH release. Increases in plasma phosphate (Pi)
levels stimulate the release of PTH. (Modified with Bone:
permission from Molina PE: Endocrine Physiology, 3rd ed. ↑Bone
Ca2+ resorption
New York: McGraw-Hill Medical, 2010.)

it is also found in kidney tubule cells and thyroid C cells PTH-mediated bone resorption. Table 64–1 lists the factors
(Figure 64–2). An acute decrease in circulating calcium levels that regulate PTH release.
(hypocalcemia) triggers PTH release within seconds. In addi-
tion, increases in plasma phosphate levels increase PTH
secretion.
PTH release can be stimulated by a decrease in plasma Mg2+. PTH TARGET ORGANS AND
The balance of magnesium is closely linked to that of calcium; PHYSIOLOGIC EFFECTS
magnesium depletion or deficiency is frequently associated
with hypocalcemia. A combined decrease in Mg2+ and calcium The primary target organs for the physiologic effects of PTH
leads to impairment in the individual’s ability to secrete PTH. are kidney and bone. The main physiologic response elicited by
Moreover, severe hypomagnesemia impairs not only the PTH is to increase plasma calcium levels by increasing renal
release of PTH from the parathyroid gland in response to calcium reabsorption, bone resorption, and intestinal cal-
hypocalcemia, but also prevents the responsiveness of bone to cium absorption indirectly (via 1,25(OH)2D, the active form

↓ [Ca++]
↑ [Ca++]
Activates 1,25(OH)2D Relaxed
Ca++ sensor Ca++ sensor

FIGURE 64–2 Parathyroid Ca2+-


sensing receptor and regulation of PTH ↓ PTH release ↑ PTH release
release. PTH release is decreased by
increased serum calcium (Ca2+) Nucleus
concentrations and by increased
1,25(OH)2D. PTH is increased by decreased
calcium, decreased magnesium, and ↓ PTH synthesis
increased phosphate levels. Changes in
calcium concentrations are sensed by a
G protein–coupled receptor on the
parathyroid chief cells. (Modified with permission
from Molina PE: Endocrine Physiology, 3rd ed. New York:
McGraw-Hill Medical, 2010.) ↓ Magnesium ↑ Phosphate
CHAPTER 64 Parathyroid Gland and Calcium and Phosphate Regulation 645

TABLE 64–1. Regulation of parathyroid hormone peptide product of a different gene from that which encodes
release. for PTH, and is expressed in multiple tissues. It binds to the
PTHR1 in bone and kidney, resulting in elevated plasma cal-
PTH Release is Increased by PTH Release is Decreased by cium levels. PTHrP released from cancerous tissue is respon-
sible for the hypercalcemia of malignancy. Therefore, PTHR1
Hypocalcemia Hypercalcemia
not only mediates the physiologic effects of PTH, but also plays
Hyperphosphatemia 1,25(OH)2D an important role in the pathophysiologic effects of PTHrP.
Catecholamines Severe hypomagnesemia
PTH, parathyroid hormone.
CELLULAR EFFECTS OF PTH
In the kidney, PTH directly stimulates calcium reabsorption,
of vitamin D). PTH also increases renal 1α-hydroxylase activ- decreases the reabsorption of phosphate causing an increase in
ity and renal phosphate excretion. The effects of PTH are medi- phosphate excretion, and stimulates the activity of
ated through binding to G protein–coupled PTH receptors. At 1α-hydroxylase, the enzyme responsible for formation of
least three receptors have been identified, but the important 1,25(OH)2D. PTH regulation of calcium reabsorption is medi-
physiologic effects of PTH are mediated by PTHR1. ated in the distal tubules (Figure 64–3). PTH stimulates the
PTHR1 is expressed in bone osteoblasts and kidney, where it insertion and opening of the apical calcium channel, facilitat-
binds PTH and PTH-related protein (PTHrP). PTHrP is a ing calcium reabsorption as discussed in Chapter 48. It

Ca2+
Distal tubule
PTH
+
Ca2+ channel Luminal membrane

Calbindin-D28K

+
1,25(OH)2D

+
ATP ADP 3 Na+

Basolateral membrane
Ca2+/Na+
Ca2+ ATPase exchanger

Ca2+
Ca2+
Interstitial space
FIGURE 64–3 Parathyroid hormone (PTH) increases renal calcium reabsorption. The transcellular reabsorption of calcium (Ca2+) by
the distal tubule is regulated by PTH and 1,25(OH)2D. PTH increases the insertion of calcium channels in the apical membrane and facilitates the
entry of calcium. Inside the cell, calcium binds to calbindin-D28K, a vitamin D–dependent calcium-binding protein that facilitates the cytosolic
diffusion of calcium from the apical influx to the basolateral efflux sites. Calcium transport out of the cell through the basolateral membrane into
the interstitial space is mediated by a Na+/Ca2+ exchanger and a Ca2+-ATPase. 1,25(OH)2D contributes to the enhanced calcium reabsorption by
stimulating the synthesis of calbindin and the activity of Ca2+-ATPase. ATP, adenosine triphosphate. (Modified with permission from Molina PE: Endocrine
Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
646 SECTION IX Endocrine and Metabolic Physiology

Proximal tubule
Pi 3Na+

PTH
Luminal Type I Type IIa
membrane

Type IIa PTH


+
• Internalization
• Destruction

Type IIa Lysosomes


A–

Basolateral
Type III
membrane

Pi Na+
Pi

Pi

Interstitial space
FIGURE 64–4 Parathyroid hormone (PTH) decreases renal inorganic phosphate (Pi) reabsorption. Renal reabsorption of phosphate
occurs through apical sodium (Na+)/Pi cotransport. Three different Na/Pi cotransporters have been identified: types I, II, and III. Types I and II
cotransporters are located in the apical membrane. Type II cotransporters are expressed in the renal proximal tubule (type IIa) and in the small
intestine (type IIb). Type IIa cotransporters are the major target for PTH regulation and contribute to most (up to 70%) of proximal tubular Pi
reabsorption. PTH acutely stimulates internalization of the type IIa cotransporters, directing them to the lysosomes for destruction, resulting in
a decrease in Pi reabsorption as indicated by the dotted line. Type III cotransporters are most likely located at the basolateral membrane and
play a general “housekeeping” role in ensuring basolateral Pi influx if apical Pi entry is insufficient to satisfy cellular requirements. Basolateral
exit, which is necessary to complete transcellular Pi reabsorption, is not well defined. Several Pi transport pathways have been suggested
including Pi cotransport, anion (A−) exchange, and even an “nonspecific” Pi channel. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed.
New York: McGraw-Hill Medical, 2010.)

decreases the renal (and intestinal) reabsorption of phosphate these minerals. Bone remodeling involves the continuous
by decreasing the expression of the type II Na+/phosphate removal of bone (bone resorption) followed by synthesis of
cotransporters by stimulating their internalization via coated new bone matrix and subsequent mineralization (bone forma-
vesicles and subsequent lysosomal degradation (Figure 64–4). tion). Osteoclastic activity induced by PTH is indirectly medi-
In the bone, PTH binds to receptors found in osteoblasts stim- ated through osteoblast activation. Osteoclastic bone resorption
ulating the activity of several proteins, including osteoclast- involves several steps, including recruitment and differentia-
differentiating factor (ODF), also known as receptor activator tion of osteoclast precursors into mononuclear osteoclasts
of nuclear factor-κβ ligand (RANKL) or osteoprotegerin (preosteoclasts) and fusion of preosteoclasts to form multinu-
ligand (Figure 64–5). In addition, PTH stimulates osteoblast cleated functional osteoclasts (Figure 64–6). PTH stimulation
expression of genes involved in degradation of the extracellu- of osteoblast synthesis of ODF (RANKL or osteoprotegerin
lar matrix and bone remodeling (collagenase-3), production of ligand) facilitates recruitment and binding of osteoclast precur-
growth factors (insulin-like growth factor I), and stimulation sors expressing the ODF receptor (RANK). This interaction
and recruitment of osteoclasts (RANKL and interleukin-6). initiates the differentiation and activation of osteoclasts
(Figures 64–5 and 64–6). Another protein that participates in
this sequence of events is osteoprotegerin, a member of the
PTH MOBILIZATION OF BONE CALCIUM TNF receptor superfamily, secreted by osteoblasts. Osteoprote-
gerin acts as a natural antagonist of RANKL, decreasing
The inorganic phase of bone matrix is composed mainly of RANK–RANKL interaction and, as a result, bone resorption.
hydroxyapatite, which functions as a reservoir of calcium and Osteoclastic bone resorption involves the attachment of osteo-
phosphate ions and plays a major role in the homeostasis of clasts to the bone surface, generating an isolated extracellular
CHAPTER 64 Parathyroid Gland and Calcium and Phosphate Regulation 647

Osteoclast
Osteoprotegerin
precursor

RANKL
+ RANK
PTH

Osteoblast
Mature
Osteoclast
Osteoblast

Ca2+, Pi, alkaline phosphatase


HCO3 –

Osteoclast Cl–
Osteoblast

Cl–

H+

Cl–
H+ Hydroxyapatite
Bone

FIGURE 64–5 PTH-mediated osteoclast differentiation. Upper Panel: PTH binds to PTHR1 in osteoblasts and stimulates the expression
of receptor activator of nuclear factor-κβ ligand (RANKL) expression on the cell surface. RANKL binds to RANK, a cell surface protein on osteoclast
precursors. Binding of RANKL to RANK activates osteoclast gene transcription and the differentiation into a mature osteoclast, characterized by
the ruffled membrane under which bone resorption occurs. Lower Panel: Osteoclasts attach to the bone surface through β-integrins generating
an isolated extracellular microenvironment. Hydrogen ions generated are delivered across the plasma membrane by H+-ATPases at the cell’s
ruffled membrane. The acidification of this microenvironment to ~pH 4 favors the dissolving of hydroxyapatite and provides optimal conditions
for the action of the lysosomal proteases including collagenase and cathepsins. The products of bone degradation (ionized Ca2+, inorganic
phosphate [H2PO4−], and alkaline phosphatases) are endocytosed by the osteoclast and transported to and released at the cell’s antiresorptive
surface. Osteoprotegerin, a soluble protein secreted by osteoblasts that serves as a decoy ligand for RANKL, prevents binding of RANKL to RANK,
thereby inhibiting the process of osteoclastic bone resorption. As a result, there is a decreased differentiation of precursor cells into oscteoclasts
and decreased bone resorption. Production of osteoprotegerin is increased by estrogen and decreased by glucocorticoids and PTH. Cl−, chloride;
HCO3−, bicarbonate; H+, hydrogen ion. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

microenvironment between osteoclasts and the bone surface calcium and an increase in “free” or ionized calcium in the
that in effect functions as a lysosome in which bone resorption plasma. Alkalosis results in increased calcium binding and a
takes place. The products of bone degradation (including cal- decrease in ionized calcium in the plasma. A smaller fraction
cium and phosphate), as well as intracellular enzymes such as (10–20%) of protein-bound calcium is bound to globulins.
alkaline phosphatase, are also released into the circulation. Calcium is a key intracellular messenger, a cofactor for vari-
ous enzymes, and has diverse extracellular functions (e.g., in
the clotting of blood, maintenance of skeletal integrity, and
CALCIUM HOMEOSTASIS modulation of neuromuscular excitability). Therefore, stable
calcium levels are critical for normal physiologic function. For
The human body contains approximately 1,100 g of calcium, example, Na+ channel voltage gating is dependent on the extra-
99% of which is deposited in bones and teeth. The small cellular calcium concentration. Decreased plasma calcium
amount found in plasma is divided into three fractions: concentrations (hypocalcemia) reduce the voltage threshold
ionized calcium (50%), protein-bound calcium (40%), and for the action potential firing, resulting in neuromuscular
calcium complexed to citrate and phosphate forming soluble hyperexcitability. This can result in muscle cramps or numb-
complexes (10%). The complexed and ionized calcium frac- ness and tingling of fingertips, toes, and the perioral region.
tions (about 60% of total plasma calcium) can cross the plasma Clinically, neuromuscular irritability can be demonstrated by
membrane. The majority (80–90%) of protein-bound calcium mechanical stimulation of the hyperexcitable nerve leading to
is bound to albumin, and this interaction is sensitive to changes tetanic-like muscle contraction by eliciting Chvostek’s sign
in blood pH. Acidosis leads to a decrease in protein binding of (ipsilateral contraction of facial muscles elicited by tapping the
648 SECTION IX Endocrine and Metabolic Physiology

Bone remodeling Growth hormone

Osteoblast Stromal
vessels
Increase in insulin-like
Stroma
growth factor I
Trabeculae

Circulating
osteoblasts

Osteogenic Chondrocytes
cells in bone
marrow
Endochondral
Systemic bone formation
circulation

Periosteum
Osteoblast
Osteoclast

Periosteal appositional
bone formation

Blood vessel

PTH New lining


Local factors
cells
Lining cells
Osteoblasts

Osteoclasts Osteoid

New
osteocytes
Microcrack New bone
Osteocyte
apoptosis
Cement line

Old bone

FIGURE 64–6 Bone remodeling involves bone formation by osteoblasts and bone resorption by osteoclasts. PTH stimulates both
aspects of the process. Growth hormone, acting through insulin-like growth factor, also stimulates bone formation particularly during the linear
growth phase in children. Bone remodeling ensures bone repair and is necessary to maintain calcium homeostasis. (Reproduced with permission from
Canalis E, Giustina A, Bilezikian JP. Mechanisms of anabolic therapies for osteoporosis. NEJM. 2007;357:905-916. Copyright Massachusetts Medical Society. All rights reserved.)

skin over the facial nerve) or Trousseau’s sign (carpal spasm Bone
induced by inflation of the blood pressure cuff to 20 mm Hg
Calcium in bone is distributed in a readily exchangeable
above the patient’s systolic blood pressure for 3–5 minutes).
pool and a stable pool. The readily exchangeable pool is
involved in maintaining plasma calcium levels by the daily
INTERACTION OF BONE, KIDNEY, exchange of approximately 550 mg of calcium between the
AND INTESTINE IN MAINTAINING bone and extracellular fluid. The stable calcium pool is
involved in bone remodeling.
CALCIUM HOMEOSTASIS
Plasma concentrations of calcium are mainly regulated by the Kidney
actions of PTH and 1,25(OH)2D and three tissues: bone, kid- In the kidney, virtually all filtered calcium is reabsorbed, of
ney, and intestine. which about 40% is under hormonal regulation by PTH.
CHAPTER 64 Parathyroid Gland and Calcium and Phosphate Regulation 649

Intestine with a resulting increase in the release of calcium into the cir-
culation. The increase in calcium levels decreases PTH release
The availability of dietary calcium is a critical determinant of cal-
from the parathyroid gland, decreases activation of 25(OH)D
cium homeostasis. Dietary intake of calcium averages 1,000 mg
in the kidney, and stimulates release of calcitonin from the
per day, of which only 30% is absorbed in the intestinal tract.
parafollicular cells of the thyroid gland. At high pharmaco-
This percentage of dietary calcium that is absorbed is signifi-
logical concentrations, calcitonin can inhibit osteoclast activ-
cantly enhanced by 1,25(OH)2D.
ity and increase renal calcium excretion. Overall, PTH and
1,25(OH)2D are the critical hormones that work together to
HORMONAL REGULATION maintain plasma calcium levels within a normal range.
OF CALCIUM HOMEOSTASIS
A slight decrease in the calcium level results in an increased
ROLE OF VITAMIN D
release of PTH. In bone, PTH increases resorption and the IN CALCIUM HOMEOSTASIS
release of calcium and phosphate into the circulation. In the
kidney, PTH promotes calcium reabsorption and phosphate Synthesis and Activation of Vitamin D
excretion in urine. In addition, PTH stimulates the formation Vitamin D is a lipid-soluble vitamin synthesized either from
of 1,25(OH)2D. 1,25(OH)2D increases intestinal absorption of dietary plant- and animal-derived precursors or through the
dietary calcium and, to a lesser extent, renal reabsorption of action of sunlight on cholesterol-derived precursors found in the
filtered calcium. In bone, 1,25(OH)2D increases bone turnover, skin (Figure 64–7). Active vitamin D (calcitriol; 1,25(OH)2D)

UV light Liver

Cholecalciferol

Provitamin D 25(OH)Vitamin D
7-dehydrocholesterol Ergocalciferol
in skin (Vitamin D2) in diet Kidney
24α-Hydroxylase
Ca
PTH
1α-Hydroxylase 24, 25 (OH)2 Vitamin D
(Inactive form)

1, 25 (OH)2 Vitamin D
(Active form)

Bone Ca Ca PTH
resorption absorption reabsorption synthesis
Plasma Ca2+ concentrations

FIGURE 64–7 Vitamin D metabolism and physiologic effects at target organs. Provitamin D (7-dehydrocholesterol) in the skin is
converted to cholecalciferol by ultraviolet (UV) light. Cholecalciferol and ergocalciferol (from plants) are transported to the liver, where they
undergo the first step in bioactivation, the hydroxylation at C-25 to 25-hydroxyvitamin D (25(OH)D), the major circulating form of vitamin D.
The second hydroxylation step, at C-1, occurs in the kidney and results in the hormonally active 1,25(OH)2D. This activation step, mediated by
1α-hydroxylase, is under tight regulation by parathyroid hormone (PTH), calcium levels, and 1,25(OH)2D. The activity of 1α-hydroxylase is
stimulated by PTH and inhibited by calcium and 1,25(OH)2D. Decreased activity of 1α-hydroxylase favors C-24 hydroxylation and formation of
the less active 24,25(OH)2D. 1,25(OH)2D increases bone resorption, increases calcium absorption from the intestine (the major effect), increases
renal calcium reabsorption, and decreases the production of PTH by the parathyroid glands. The overall effect of 1,25(OH)2D is to increase plasma
calcium concentrations. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
650 SECTION IX Endocrine and Metabolic Physiology

is the product of two consecutive hydroxylation steps. The first deficiency can result in bone deformities (rickets) when it occurs
hydroxylation of the precursors cholecalciferol (D3, derived in children and decreased bone mass (osteomalacia) in adults.
from skin or diet) and ergocalciferol (D2, derived from diet) Vitamin D deficiency is associated with weakness, bowing of the
takes place in the liver. Cholecalciferol is produced in the skin weight-bearing bones, dental defects, and hypocalcemia.
by ultraviolet radiation acting on 7-dehydrocholesterol, an inert
precursor. Vitamin D circulates to the liver bound to vitamin
D–binding protein. The precursors are hydroxylated at C-25 to ROLE OF CALCITONIN
the prehormone 25(OH)D. 25(OH)D is the major circulating IN CALCIUM HOMEOSTASIS
and storage form of vitamin D, and circulates bound to vitamin
D–binding protein. In the kidney, it is hydroxylated by Calcitonin is a 32–amino acid peptide hormone produced by
1α-hydroxylase resulting in 1,25(OH)2D. This second hydroxy- the parafollicular or C cells in the thyroid gland in response to
lation step is a tightly regulated process enhanced by PTH and total plasma concentrations greater than 9 mg/dL. The two
under negative feedback regulation by plasma calcium levels. target organs for calcitonin’s physiologic effects are bone and
An increase in plasma calcium levels inhibits the hydroxylation kidney. Calcitonin inhibits bone resorption, and increases uri-
at C-1 and favors hydroxylation at C-24, leading to the synthesis nary calcium excretion. The cellular effects of calcitonin are
of an inactive metabolite of vitamin D (24,25(OH)2D). mediated through G protein–coupled receptors from the same
receptor family as the PTH, PTHrP receptor superfamily. Cal-
Cellular Effects of Vitamin D citonin does not appear to be critical for the regulation of cal-
cium homeostasis in humans; in fact, total removal of the
1,25(OH)2D mediates its effects through binding to a steroid thyroid does not produce major alterations in calcium homeo-
receptor located in the intestines, bone, kidney, and parathy- stasis. However, calcitonin has been used therapeutically for
roid gland, where it stimulates intestinal calcium absorption, the prevention of bone loss and for the short-term treatment
regulates bone turnover, increases renal calcium reabsorption, of hypercalcemia of malignancy.
and suppresses the synthesis of PTH (Figure 64–7).

Abnormal Vitamin D Levels ADDITIONAL REGULATORS OF


Vitamin D belongs to the class of vitamins that are lipid soluble CALCIUM AND BONE METABOLISM
(i.e., A, D, E, and K), and can be stored in tissues. Extremely
high levels of vitamin D (vitamin D toxicity) may lead to prob- PTH and vitamin D play central roles in the regulation of bone
lems such as calcinosis (calcification of soft tissues), deposi- metabolism. However, additional hormones participate in this
tion of calcium and phosphate in the kidney, and increased process (Table 64–2). Sex steroids (androgens and estrogens)
plasma calcium levels, resulting in cardiac arrhythmia. decrease bone resorption through increased osteoprotegerin
Deficiency of vitamin D is extremely common and can be synthesis, osteoblast proliferation, expression of type I collagen
the result of inadequate dietary intake or absorption, or lack of and alkaline phosphatase, and modulation of the effects of
sunlight, resulting in decreased conversion of inactive precursors growth hormone, vitamin D, progesterone, and PTH. Growth
to the substrates used in the synthesis of 25(OH)D. Vitamin D hormone and insulin-like growth factor-I stimulate proliferation

TABLE 64–2 Factors involved in the regulation of calcium and bone metabolism.
Regulator Action

PTH Increases bone resorption and plasma calcium

1,25(OH)2D Increases intestinal calcium absorption, bone resorption, and facilitates renal calcium reabsorption

Calcitonin Decreases bone resorption and plasma calcium

Sex steroids (androgens and estrogens) Increase 1α-hydroxylase activity


Increase osteoprotegerin synthesis
Net decrease in bone loss

Growth hormone and insulin-like growth factor Stimulate bone synthesis and growth

Thyroid hormone Increases bone resorption

Prolactin Increases renal calcium reabsorption and 1α-hydroxylase activity

Glucocorticoids Increase bone resorption, decrease bone synthesis

Inflammatory cytokines Increase bone resorption


PTH, parathyroid hormone.
CHAPTER 64 Parathyroid Gland and Calcium and Phosphate Regulation 651

and differentiation of osteoblasts, bone protein synthesis, and of the bone architecture and systemic preservation of calcium
growth, and promote synthesis of type I collagen. Normal thy- homeostasis. In early life, a careful balance exists between bone
roid function is required for physiologic bone remodeling; yet, formation by osteoblasts and bone resorption by osteoclasts.
excess thyroid hormone levels result in increased bone resorp- With aging, the process of coupled bone formation–resorption
tion. Glucocorticoids increase bone resorption, decrease bone (turnover) is affected by the reductions in osteoblast differen-
synthesis, and inhibit osteoprotegerin synthesis, together lead- tiation, activity, and life span, which are further potentiated in
ing to decreased bone mass. The proinflammatory cytokines are the perimenopausal years by hormone deprivation (estrogen
potent stimulators of bone resorption in vitro and in vivo. The and dehydroepiandrosterone) and an increase in osteoclast
overall interaction of these various factors during health and dis- activity.
ease plays an important role in maintaining bone mass. Decreased calcium intake below obligatory calcium loss
(through the urine, feces, and skin) mobilizes calcium from the
skeleton to maintain the ionized calcium concentration in the
REGULATION OF PHOSPHATE ECF, resulting in bone destruction. Vitamin D deficiency
decreases the concentration of ionized calcium in the ECF
BALANCE (from loss of the calcemic action of 1,25(OH)2D on bone),
resulting in stimulation of PTH release (secondary hyperpara-
Phosphorus in the form of phosphate accounts for more than
thyroidism), increased phosphate excretion (hypophos-
50% of bone mineral mass in the form of calcium phosphate.
phatemia), and failure to mineralize new bone as it is being
It plays numerous vital roles in cell function. Most food prod-
formed. Simple calcium deficiency is associated with compen-
ucts, whether plant or animal, contain relatively abundant
satory increases in PTH and calcitriol, which together cause loss
quantities of phosphorus. The skeleton contains 85% of the
of whole bone, whereas true vitamin D deficiency reduces the
body’s phosphorus; the rest is distributed in the ECF and ICF.
mineral content of the bony tissue itself, leading to abnormal
Total extracellular phosphorus is found in an ionized form
bone composition. However, these two nutritional deficiencies
and a nonionized form (organic and inorganic forms). The
cannot be completely separated because calcium malabsorption
inorganic form may be found ionized or free in the form of
is the first manifestation of vitamin D deficiency.
phosphate (50%); complexed with calcium, Mg2+, and Na+
(35%); or bound to protein (15%).
Phosphate homeostasis is maintained by intestinal absorp- CHILDHOOD–ADULT
tion, renal excretion, balance of phosphate exchange in and
out of the cells, and hormonal regulation. Most phosphate Bone mass increases throughout childhood and adolescence.
absorbed from the diet undergoes urinary excretion. Extracel- In girls, the rate of increase in bone mass decreases after
lular phosphate concentration is tightly regulated principally menarche, whereas in boys, gains in bone mass persist up to
through urinary excretion. Alterations in extracellular phos- 17 years of age and are closely linked to pubertal stage and
phate concentrations lead to rapid adjustments in renal phos- androgen status. By age 17–23, the majority of peak bone mass
phate excretion and slower and less regulated adjustments in has already been achieved in both sexes. Skeletal growth is
intestinal absorption. Phosphate excretion by the kidney is achieved through the interaction between osteoblasts and
stimulated by PTH (Figure 64–4). 1,25(OH)2D stimulates osteoclasts, which work cooperatively under the influence of
phosphate intestinal absorption by stimulating the brush- the mechanical strain placed on bone by skeletal muscle force
border membrane Na+–HPO42− cotransport in the upper small such as that exerted during exercise. Decreased mechanical
intestine. Thus, PTH promotes phosphate excretion, whereas strain (such as that associated with prolonged bed rest or
1,25(OH)2D and insulin promote phosphate renal reabsorp- immobilization) leads to bone loss, whereas increased mechan-
tion and intestinal absorption. Vitamin D deficiency leads to ical strain (weight-bearing exercise) stimulates osteoblastic
increased phosphate renal excretion and decreased intestinal activity and bone formation. Peak bone mass is attained in the
phosphate and calcium absorption, resulting in a severe loss of third decade of life and is maintained until the fifth decade,
both calcium and phosphate from bone (the major site of both when age-related bone loss begins in both men and women.
of these mineral stores) because of enhanced PTH activity,
resulting in loss of bone mineral and osteomalacia. This is in
contrast to osteoporosis induced by calcium deficiency. PREGNANCY AND LACTATION
Overall requirements for calcium are significantly increased
during pregnancy and lactation. The uptake and release of cal-
HORMONAL REGULATION OF cium from the skeleton are increased during pregnancy, and
BONE METABOLISM the rate of calcium mobilization continues to be increased
during the early months of lactation, returning to prepreg-
Bone remodeling results from the interactions of multiple ele- nancy rates during or after weaning. Calcium absorption, uri-
ments, including osteoblasts, osteoclasts, hormones, growth nary calcium excretion, and bone resorption are higher during
factors, and cytokines, the result being a dynamic maintenance pregnancy than before conception or after delivery.
652 SECTION IX Endocrine and Metabolic Physiology

TABLE 64–3 Measurements used frequently for the clinical evaluation of abnormalities in parathyroid hormone
function or calcium homeostasis.

Normal Range Abnormality

Plasma calcium 8.5–10.5 mg/dL Increased with ↑ PTH, vitamin D intoxication, ↑ bone resorption

Plasma phosphate 3–4.5 mg/dL Decreased in hyperparathyroidism, vitamin D deficiency; increased in renal
failure, hypoparathyroidism, vitamin D intoxication

Intact plasma PTH levels 10–65 pg/mL Increased in hyperparathyroidism; decreased in hypoparathyroidism

Alkaline phosphatase 30–120 U/L High levels indicate increased osteoblastic activity (bone turnover)

Bone-specific alkaline phosphatase 17–48 U/L High bone turnover, useful marker of active bone formation

Osteocalcin (intact) <1–23 ng/mL Marker for skeletal bone turnover


PTH, parathyroid hormone.

MENOPAUSE densitometry), or bone biopsy. DEXA uses x-rays to measure


bone density and provides two measures of how dense bone is:
The acute loss of bone that accompanies menopause involves the T score and the Z score. The T score compares the person’s
most of the skeleton but particularly affects the trabecular com- bone density with the average bone density of young healthy
ponent. The associated biochemical changes include increases adults of the same sex, a time when bone density is at its peak.
in the complexed fraction of plasma calcium (bicarbonate), The Z score compares a person’s bone density with that of peo-
increases in plasma alkaline phosphatase and urinary hydroxy- ple of the same age, sex, and weight, and is less valuable in
proline (representing increased bone resorption followed by a making predictions of risk of fracture or in making decisions
compensatory increase in bone formation), increased obliga- about treatment.
tory calcium loss in the urine, and a small but significant
decline in calcium absorption (Table 64–3). These changes are
ameliorated by hormone treatment with estrogen or selective
Prevention of Osteoporosis
estrogen receptor modulators (SERMs), calcium supplementa- The principal current approaches include:
tion, thiazide diuretic administration (which reduces calcium • Estrogen replacement therapy: Estrogen decreases bone loss
excretion), and restriction of salt intake, which reduces obliga- in postmenopausal women by inhibiting bone resorption, re-
tory calcium loss. In some cases of osteoporosis, calcium sulting in a 5–10% increase in BMD over 1–3 years. Calcium
absorption is low, and high bone resorption can be suppressed supplements enhance the effect of estrogen on BMD.
by treatment with vitamin D that in turn leads to improvement • Bisphosphonates: Bisphosphonates have a strong affinity
in calcium absorption. The process of bone loss is progressive, for bone apatite and are potent inhibitors of bone resorption.
starting at about the age of 50 in men and at menopause in These agents reduce the recruitment and activity of osteo-
women. Bone loss is faster in women than in men and affects clasts and increase their apoptosis.
some bones more than others; the consequences include • Calcitonin: Calcitonin reduces bone resorption by direct
decreased bone mineral density (BMD) and increased risk of inhibition of osteoclast activity. Intranasal calcitonin pro-
fractures. Estrogen deficiency is a major pathogenic factor in duces significant effects on BMD. Calcitonin is less effective
the bone loss associated with menopause and the subsequent in prevention of cortical bone loss than cancellous bone loss
development of postmenopausal osteoporosis. In males, bone in postmenopausal women.
loss is not associated with a rise in bone resorption markers. • PTH: Intermittent administration of human recombinant
Instead, bone loss in men is linked to an age-related decline in PTH restores bone strength by stimulating new bone forma-
gonadal function and is due to a decrease in bone formation, tion at the periosteal (outer) and endosteal (inner) bone sur-
not so much an increase in bone resorption. faces, thickening the cortices and existing trabeculae of the
skeleton, and perhaps increasing trabecular numbers and
their connectivity.
Bone Density • SERMs: SERMs are compounds that exert estrogenic ef-
Bone density determines the degree of osteoporosis and the fects in specific tissues and antiestrogenic effects in oth-
fracture risk. The main determinants of peak bone density are ers. Raloxifene competitively inhibits the action of estro-
genetics, calcium intake, and exercise. The most common test gen in the breast and the endometrium and acts as an
for measuring bone density is dual-energy x-ray absorptiom- estrogen agonist on bone and lipid metabolism. In early
etry (DEXA) scanning. Additional approaches include com- postmenopausal women, raloxifene prevents postmeno-
puted tomography, radiologic techniques (morphometry or pausal bone loss at all skeletal sites, reduces markers of
CHAPTER 64 Parathyroid Gland and Calcium and Phosphate Regulation 653

bone turnover to premenopausal concentrations, and re- roid glands. Because of the important role of PTH in the acute
duces the serum cholesterol concentration and its low- regulation of plasma calcium levels, an early manifestation of
density lipoprotein fraction without stimulating the surgical removal of the parathyroid glands is hypocalcemic
endometrium. tetany. The classic clinical sign is known as the Chvostek’s
• Vitamin D analogs: Vitamin D analogs induce a small in- sign, which is twitching or contraction of the facial muscles in
crease in BMD that seems to be limited to the spine. response to tapping the facial nerve at a point anterior to the
• Exercise: Physical activity early in life contributes to high ear and above the zygomatic bone.
peak bone mass. Walking, weight training, and high-impact
exercises induce a small (1–2%) increase in BMD at some
skeletal sites. These effects disappear if the exercise program PSEUDOHYPOPARATHYROIDISM
is stopped. Load-bearing exercise is more effective for in-
creasing bone mass than are other types of exercise. Pseudohypoparathyroidism—PTH resistance—is the result
of a decreased response to PTH because of a congenital defect
in the G protein associated with the PTHR1.
DISEASES OF PTH PRODUCTION
PRIMARY HYPERPARATHYROIDISM
CLINICAL CORRELATION
Excess PTH production is often due to parathyroid gland hyper-
plasia, adenoma, or carcinoma. The manifestations include A postmenopausal patient is referred for asymptomatic
increased PTH levels, increased plasma calcium levels (hyper- hypercalcemia and history of repeated episodes of uro-
calcemia), increased urinary calcium excretion (hypercalciu- lithiasis (kidney stones). Blood laboratory values reveal
ria) with increased formation of kidney stones (urolithiasis), increases in intact PTH, 1,25(OH)2D, and markers of bone
and decreased plasma phosphate levels due to the large increase resorption. Neck ultrasound revealed a mass below the
in urinary excretion. The increase in PTH results in increased right lobe of the thyroid gland. Surgical removal and patho-
bone resorption and further increases in extracellular calcium logical examination of the excised gland led to the diagno-
concentrations leading to increased filtered load of calcium in sis of parathyroid adenoma.
the kidney that exceeds the reabsorptive transport capacity, as Parathyroid adenomas cause the majority (90%) of cases
discussed in Chapter 48. of primary hyperparathyroidism. Excess PTH release leads
to increased bone resorption, increased hydroxylation of
25(OH)D, and increased intestinal calcium absorption.
SECONDARY HYPERPARATHYROIDISM Urolithiasis results from increased concentrations of cal-
cium in the glomerular filtrate. Increased bone resorption
Secondary hyperparathyroidism and hyperplasia of the para-
leads to increased release of bone proteins that are markers
thyroid glands are complications that occur in patients with
of bone resorption, such as osteocalcin and bone alkaline
chronic renal failure. In early renal failure, a reduction in
phosphatase. With measurement of total plasma calcium
plasma 1,25(OH)2D and moderate decreases in ionized cal-
during routine laboratory testing, hypercalcemia in the
cium contribute to greater synthesis and secretion of PTH.
absence of clinical manifestations is now a more frequent
As renal disease progresses, parathyroid expression of recep-
presentation of these adenomas.
tors for vitamin D and calcium is reduced, making the para-
thyroid gland more resistant to both the 1,25(OH)2D and
calcium negative feedback regulation of PTH release. Thus,
for any increase in plasma calcium, the inhibition of PTH
secretion is less efficient. As a result, for any particular plasma
CHAPTER SUMMARY
calcium concentration, secretion of PTH is enhanced, result- ■ PTH release is under negative feedback regulation by calcium
ing in a shift in the calcium–PTH set point toward secondary and vitamin D.
hyperparathyroidism. Hyperphosphatemia independent of ■ The main physiologic effects of PTH are mediated by the
calcium and 1,25(OH)2D levels further enhances uremia- PTHR1 expressed in bone and kidney.
induced parathyroid gland hyperplasia and PTH synthesis ■ PTHR1 binds PTH and PTHrP, a peptide responsible for the
and secretion, the latter by posttranscriptional mechanisms. pathophysiologic elevation of plasma calcium in some
malignancies.
■ In the kidney, PTH increases renal calcium reabsorption,
HYPOPARATHYROIDISM increases the activity of 1α-hydroxylase (which mediates the
final activation step in the synthesis of vitamin D), and
Hypoparathyroidism, resulting from impaired production of decreases phosphate reabsorption.
PTH, can be associated with other endocrine disorders and ■ In bone, PTH increases osteoclast-mediated bone resorption
neoplasias or may result from surgical removal of the parathy- indirectly through stimulation of osteoblast activity.
654 SECTION IX Endocrine and Metabolic Physiology

■ Calcitonin decreases bone resorption and lowers plasma 3. A 73-year-old woman is admitted to the hospital following a bout
calcium levels. of severe vomiting and generalized weakness. Initial laboratory
■ Synthesis of the active form of vitamin D (1,25(OH)2D; calcitriol) values reveal increased plasma calcium levels. The referring
involves hydroxylation in the liver (C-25) and kidney (C-1). physician tells you that she has breast cancer and her bone scan
■ 1,25(OH)2D increases bone resorption, renal calcium reabsorp- indicates metastasis to bone. Which of the following blood
tion, and intestinal calcium absorption. laboratory values would be compatible with this clinical scenario?
■ Plasma calcium levels are tightly regulated through hormone- A) low PTH and phosphate, and high alkaline phosphatase
mediated effects on bone. B) high PTH and phosphate, and low alkaline phosphatase
C) low PTH and phosphate, and low alkaline phosphatase
■ Bone mineral density and mass are under nutritional and
D) low PTH and phosphate, and normal alkaline phosphatase
hormonal control.
4. The most likely cause of hypercalcemia in the patient described
■ Phosphate is regulated principally through effects on renal
in Question 3 is
excretion.
A) increased PTH production
B) increased responsiveness of the PTH receptor 1
C) increased PTHrP production
STUDY QUESTIONS D) increased calcitonin release
1. A 43-year-old male is admitted to the emergency room for severe 5. Hyperventilation usually leads to muscle cramping (tetanic
pain in his left flank, radiating to the groin. The pain is contractions). What is the physiologic concept that explains what
intermittent and initiated after running a marathon on a hot happens in that situation?
summer day. The patient is asked for a urine specimen and blood A) hypercalcemia secondary to PTH-mediated bone resorption
is detected in the urine. He is hydrated and additional diagnostic B) increased dissociation of protein-bound calcium
procedures are done. Laboratory values show an increased C) decreased ionized plasma calcium levels
plasma calcium of 12 mg/dL, and increased plasma intact PTH D) increased renal calcium excretion
values of 130 pg/mL. Which of the following findings would be
predictable in this patient?
A) increased plasma phosphate
B) increased serum alkaline phosphatase
C) increased intestinal calcium loss
D) decreased urinary calcium excretion
2. In the patient described above, the mechanism underlying the
abnormalities observed is
A) increased calcitonin release
B) decreased hepatic 25-hydroxylase activity
C) increased osteoclast apoptosis
D) increased bone resorption
65
C H A P T E R

Adrenal Gland
Patricia E. Molina

O B J E C T I V E S

■ Identify the functional anatomy and zones of the adrenal glands and the
principal hormones secreted from each zone.
■ Describe and contrast the regulation of synthesis and release of the adrenal
steroid hormones (glucocorticoids, mineralocorticoids, and androgens) and
the consequences of abnormalities in their biosynthetic pathways.
■ Understand the cellular mechanism of action of adrenal cortical hormones
and identify their major physiologic actions, particularly during injury and
stress.
■ Identify the major mineralocorticoids, their biologic actions, and their target
organs or tissues.
■ Describe the regulation of mineralocorticoid secretion and relate this to the
regulation of sodium and potassium excretion.
■ Identify the causes and consequences of overproduction and underproduction
of glucocorticoids, mineralocorticoids, and adrenal androgens.
■ Identify the chemical nature of catecholamines and their biosynthesis and
metabolic fate.
■ Describe the biologic consequences of sympathoadrenal medulla activation
and identify the target organs or tissues for catecholamine effects along with
the receptor types that mediate their actions.
■ Describe and integrate the interactions of adrenal medullary and cortical
hormones in response to stress.
■ Identify diseases caused by oversecretion of adrenal catecholamines.

The adrenal glands contribute significantly to maintaining


homeostasis through their role in the regulation of the adap-
FUNCTIONAL ANATOMY
tive response to stress, in the maintenance of body water, AND ZONATION
sodium, and potassium balance, and in the control of blood
pressure. The main hormones produced by the human adre- The adrenal glands are located above the kidneys and consist
nal glands belong to two different families based on their of two different components: the outer cortex derived from
structure; these are the steroid hormones including the glu- mesodermal tissue and the inner medulla derived from a sub-
cocorticoids, mineralocorticoids, and androgens; and the population of neural crest cells (Figure 65–1). The cortex
catecholamines norepinephrine and epinephrine. makes up the majority of the gland and synthesizes the adrenal

655

Ch65_655-670.indd 655 11/30/10 3:32:28 AM


656 SECTION IX Endocrine and Metabolic Physiology

Zona Aldosterone
glomerulosa

Zona
Cortex

fasciculata Cortisol
and
androgens

Zona
reticularis
Medulla

Epinephrine
and
norepinephrine

Cortex

Medulla

FIGURE 65–1 Adrenal glands. The adrenal glands are composed of a cortex and a medulla, each derived from a different embryologic
origin. The cortex is divided into three zones: reticularis, fasciculata, and glomerulosa. The cells that make up the three zones have distinct
enzymatic capacities, leading to a relative specificity in the products of each of the adrenal cortex zones. The adrenal medulla is made of cells
derived from the neural crest; the adrenal cortex is made of cells derived from mesodermal tissue. (Reproduced with permission from Widmaier EP, Raff H,
Strang KT [editors]: Vander’s Human Physiology: The Mechanisms of Body Function, 11th ed. McGraw-Hill, 2007.)

steroid hormones. The medulla synthesizes the catecholamines step in their biosynthesis (steroidogenesis), which is the
epinephrine and norepinephrine. conversion of cholesterol to pregnenolone (Figure 65–2).
This step involves the release of cholesterol by the enzyme
HORMONES OF THE ADRENAL CORTEX cholesterol esterase, the transfer of cholesterol from
the outer mitochondrial membrane to the inner mitochon-
The adrenal cortex consists of three zones that vary in both drial membrane by steroidogenic acute regulatory
their morphologic and functional features and, thus, the ste- (StAR) protein (the rate-limiting step), and the conversion
roid hormones they produce (Figure 65–1): of cholesterol to pregnenolone by cytochrome P450 side-
chain cleavage (SCC) enzyme (P450scc; or cholesterol SCC
• The zona glomerulosa is the unique source of the mineral-
desmolase).
ocorticoid aldosterone.
Defects in the activity of specific enzymes involved in the
• The zona fasciculata produces the glucocorticoids, cortisol
pathway leading to production of steroid hormones can result in
and corticosterone, and the androgens, DHEA and DHEA
disease with various degrees of severity depending on the enzy-
sulfate (DHEAS).
matic step that is compromised. The key enzymes involved in
• The zona reticularis (which develops postnatally) produces
steroid hormone synthesis and the consequences of their defi-
glucocorticoids and androgens.
ciency are described in Table 65–1. The severity of the manifes-
The steroid hormones produced by the adrenal cortex are tations ranges from death in utero as in the case of congenital
classified into three general categories: glucocorticoids, deficiency of cholesterol SCC enzyme (P450scc) to abnormalities
mineralocorticoids, and androgens. They share an initial that become evident in adult life and that are not life threaten-
CHAPTER 65 Adrenal Gland 657

StAR

Cholesterol

Cholesterol side
chain cleavage
17α-hydroxylase/ 17α-hydroxylase/
17,20 lyase 17,20 lyase
Pregnenolone 17-OH-Pregnenolone DHEA

3β-hydroxysteroid 3β-hydroxysteroid
dehydrogenase dehydrogenase

Progesterone 17-OH-Progesterone Androstenedione

21-hydroxylase 21-hydroxylase
Androgens

DOC 11-Deoxycortisol

Aldosterone
11β-hydroxylase
synthase

Corticosterone Cortisol

Aldosterone
synthase Glucocorticoid

Aldosterone

Mineralocorticoid

FIGURE 65–2 Overview of the zone-specific adrenal steroid hormone synthetic pathway. Steroidogenic acute regulatory (StAR) protein
mediates cholesterol transfer into the mitochondria where cholesterol is converted to pregnenolone by cholesterol side-chain cleavage enzyme.
Pregnenolone is converted to progesterone by 3β-hydroxysteroid dehydrogenase. These two initial steps are shared in the synthetic pathway of
mineralocorticoids, glucocorticoids, and androgens. Activity of 17α-hydroxylase/17,20 lyase expressed in the zona fasciculata and zona reticularis
produces 17-OH-pregnenolone and 17-OH-progesterone necessary to produce androgens and to produce precursors for the glucocorticoid
pathway. Cortisol is produced by the activity of 11β-hydroxylase in the zona fasciculata and zona reticularis. Aldosterone is produced by
processing deoxycorticosterone (DOC) to corticosterone, and then to aldosterone by the enzyme aldosterone synthase unique to the zona
glomerulosa. (Modified with permission from Kronenberg HM, Melmed S, Polonsky KS, Larsen PR. Williams Textbook of Endocrinology. Philadelphia, Saunders Elsevier, 2008).

ing. An enzymatic defect of 21-hydroxylase accounts for 95% of ciency is the loss of sodium as a result of mineralocorticoid
the genetic abnormalities in adrenal steroid hormone synthesis deficiency. In contrast, patients with 11β-hydroxylase deficiency
(Figure 65–3). The second most frequent abnormality in gluco- produce excess 11-deoxycortisol and 11-deoxycorticosterone,
corticoid synthesis is deficiency of the enzyme 11β-hydroxylase. which have active mineralocorticoid activity. Because of the
Deficiencies in these enzymes result in impaired cortisol synthe- resulting excess in mineralocorticoid-like activity, patients with
sis, lack of negative feedback inhibition of the release of adreno- this deficiency retain salt and water and may present with high
corticotropic hormone (ACTH), high ACTH levels, and greater blood pressure (hypertension).
stimulation of cholesterol conversion to pregnenolone. Because
of the lack of negative feedback inhibition of ACTH release (due
to insufficient cortisol production) and the resulting high ACTH Glucocorticoid Synthesis and Release
levels and greater stimulation of steroidogenesis (resulting from The release of cortisol is under direct stimulation by ACTH
the higher ACTH-mediated stimulation of the initial steps in released from the anterior pituitary. The release of cortisol
steroid hormone synthesis), the intermediate metabolites (before follows a circadian rhythm that is exquisitely sensitive to
the enzymatic step that is deficient) continue to be synthesized, light, sleep, stress, and disease (see Figure 60–7). Release of
and their buildup leads to a shunting to the alternate enzymatic cortisol is greatest during the early waking hours; levels
pathways. Thus, more pregnenolone is shunted to the DHEA– decline as the afternoon progresses and are lowest at around
androstenedione pathway and more intermediate metabolites midnight.
are converted to androgens, resulting in virilization (presence Cortisol inhibits the biosynthesis and secretion of hypotha-
of masculine traits). One consequence of 21-hydroxylase defi- lamic corticotropin-releasing factor (CRH) and pituitary
658 SECTION IX Endocrine and Metabolic Physiology

TABLE 65–1 Key enzymes involved in steroid hormone synthesis and metabolism.
Enzyme and Relevance Physiologic Function Consequence of Deficiency

21-Hydroxylase

Accounts for 95% of genetic Converts progesterone to Decreased cortisol and aldosterone. Loss of sodium
abnormalities in adrenal steroid 11-deoxycorticosterone and because of mineralocorticoid deficiency. Virilization
hormone synthesis 17α-hydroxyprogesterone to because of excess androgen production
11-deoxycortisol
11β-Hydroxylase

Second most frequent abnormality in Converts 11-deoxycorticosterone to Excess 11-deoxycortisol and


adrenal steroid hormone synthesis corticosterone; 11-deoxycortisol to cortisol 11-deoxycorticosterone. Excess mineralocorticoid
activity. Salt and water retention
11β-Hydroxysteroid dehydrogenase type II

Inhibited by glycyrrhetinic acid, a Converts cortisol into cortisone that has less Decrease in glucocorticoid inactivation in
compound in authentic licorice affinity for the mineralocorticoid receptor mineralocorticoid-sensitive cells, leading to excess
mineralocorticoid activity

adrenocorticotropin (ACTH) in a classic example of negative glucocorticoid action within the cell, whereas increased
feedback regulation by hormones. This closely regulated 11β-hydroxysteroid dehydrogenase type II activity decreases
circuit is referred to as the hypothalamic–pituitary–adrenal glucocorticoid action.
(HPA) axis (Figure 65–4).
Mineralocorticoid Synthesis and Release
Metabolism of Glucocorticoids Aldosterone synthesis and release in the adrenal zona glomeru-
Free cortisol accounts for 5–8% of total cortisol in the circula- losa are predominantly regulated by angiotensin II and extracel-
tion. Most (>90%) of cortisol circulates in a conjugated form lular K+ and, to a lesser extent, by ACTH (Figure 65–5).
(e.g., as sulfate or glucuronide derivatives) or bound to pro- Aldosterone is part of the renin–angiotensin–aldosterone sys-
teins (noncovalent, reversible binding). Most of cortisol tem, which is responsible for preserving circulatory homeostasis
released into the blood is bound to glucocorticoid-binding in response to a loss of salt and water (please review Chapter 45).
α2-globulin (transcortin or cortisol-binding globulin A decrease in the effective intravascular blood volume leads to
[CBG]), a specific carrier of cortisol. The hepatic synthesis of decreased renal perfusion pressure, which is sensed by the juxta-
transcortin is stimulated by estrogen and decreased by liver glomerular apparatus (baroreceptor) and triggers the release of
disease (cirrhosis). The liver and kidney are the two major renin. Renin (pronounced REE-nin) release is also regulated by
sites of hormone inactivation and elimination, or catabolism. NaCl concentration in the macula densa, plasma electrolyte con-
Inactive hormones are mainly eliminated as urinary (mostly centrations, angiotensin II levels, and sympathetic tone. Renin is
conjugated) metabolites. Inactivation of cortisol to cortisone an enzyme synthesized in the juxtaglomerular cells of the kid-
and to tetrahydrocortisol and tetrahydrocortisone is followed ney that cleaves angiotensinogen (a protein produced by the
by conjugation and renal excretion. liver) to angiotensin I, which is later converted by angiotensin-
Localized tissue metabolism by the isoforms of the enzyme converting enzyme to angiotensin II. This renin–angiotensin
11β-hydroxysteroid dehydrogenase contributes to modula- system is part of an extremely powerful feedback system for long-
tion of the physiologic effects of glucocorticoids. Corticos- term control of blood pressure and volume homeostasis. Together,
teroid 11β-hydroxysteroid dehydrogenase type I is a angiotensin II, aldosterone, and antidiuretic hormone (ADH;
low-affinity NADPH-dependent reductase that converts cor- arginine vasopressin) produce vasoconstriction, and renal reten-
tisone back to its active form cortisol. This enzyme is tion of Na+ and water. The increase in circulating angiotensin II
expressed in liver, adipose tissue, lung, skeletal muscle, vas- binds to the angiotensin II receptor in the adrenocortical cells of
cular smooth muscle, gonads, and the central nervous sys- the zona glomerulosa, stimulating phospholipase C. This results
tem. The conversion of cortisol to cortisone, its less active in an increase in intracellular Ca2+, leading to stimulation of
metabolite, is mediated by the enzyme 11β-hydroxysteroid aldosterone synthesis and release (Figure 65–5).
dehydrogenase type II. This high-affinity NAD-dependent Potassium is also a major physiologic stimulus for aldoster-
dehydrogenase is expressed primarily in the kidney, where one production, illustrating a classic example of hormone
it converts cortisol to the inactive metabolite corti- regulation by the ion it controls (please review Chapter 46).
sone. This conversion is critical in preventing excess miner- Aldosterone increases potassium excretion in urine, feces,
alocorticoid activity resulting from cortisol binding to the sweat, and saliva, preventing hyperkalemia during periods of
mineralocorticoid receptor. Increased expression and activ- high potassium intake or after potassium release from skeletal
ity of 11β-hydroxysteroid dehydrogenase type I amplifies muscle during strenuous exercise. In turn, increases in circu-
CHAPTER 65 Adrenal Gland 659

A. 21 Hydroxylase deficiency Cholesterol

Pregnenolone

Progesterone 17-Hydroxypregnenolone

Cortisol Aldosterone 17-Hydroxyprogesterone Dehydroepiandrosterone

Deoxycortisol

B. 11 β-hydroxylase deficiency Cholesterol

Pregnenolone

Progesterone 17-Hydroxypregnenolone

Deoxycorticosterone 17-Hydroxyprogesterone 17-Hydroxyprogesterone

11-Deoxycortisol
Corticosterone 11-Deoxycortisol Androstenedione

Cortisol
Cortisol

Dehydroepiandrosterone

FIGURE 65–3 Alterations in steroid hormone synthesis in 21-hydroxylase and 11β-hydroxylase enzymatic deficiency. A)
21-Hydroxylase deficiency accounts for 95% of genetic abnormalities in adrenal steroid hormone synthesis. 21-Hydroxylase converts progesterone
to deoxycorticosterone and 17-hydroxyprogesterone to 11-deoxycortisol, the precursor metabolites for the synthesis of cortisol and aldosterone.
Thus, more pregnenolone is shunted to the DHEA–androstenedione pathway (more androgen synthesis), resulting in virilization (presence of
masculine traits). In addition, aldosterone deficiency leads to sodium wasting. B) The second most frequent abnormality in glucocorticoid synthesis
is 11β-hydroxylase deficiency, the enzyme that converts 11-deoxycorticosterone to corticosterone and deoxycortisol to cortisol. 11β-Hydroxylase
deficiency results in excess deoxycorticosterone and 11-deoxycortisol production. Both metabolites have active mineralocorticoid activity. The
resulting excess in mineralocorticoid-like activity leads to salt and water retention and may lead to hypertension. Because of the loss in negative
feedback inhibition of ACTH release by cortisol, excess adrenal stimulation leads to increased synthesis of adrenal androgens (androstenedione
and dehydroepiandrosterone). (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

lating potassium concentrations stimulate the release of androgens, including DHEA and DHEAS (Figure 65–2).
aldosterone from the adrenal cortex. DHEA is the most abundant circulating hormone in the body
The total amount of aldosterone released and the prevailing and is readily conjugated to its sulfate ester DHEAS. Regula-
plasma concentrations are markedly less than those of gluco- tion of DHEA production is not completely understood, but
corticoids. In addition, binding of aldosterone to plasma pro- it is, in part, controlled by ACTH. The adrenal androgens are
teins is minimal, resulting in a short plasma half-life. converted into androstenedione and then into potent andro-
gens or estrogens in the peripheral tissues. Dihydrotestoster-
one and 17β-estradiol, the most potent androgen and
Adrenal Androgen Synthesis and Release estrogen, are synthesized from DHEA. The importance of the
The third class of steroid hormones produced by the zona fas- adrenal-derived androgens to the overall production of sex
ciculata and reticularis of the adrenal glands is the adrenal steroid hormones is highlighted by the fact that approximately
660 SECTION IX Endocrine and Metabolic Physiology

Stress

Hypothalamus

CRH

Anterior
pituitary
gland
ACTH

Negative feedback

Adrenal gland

Cortisol

Bloodstream

FIGURE 65–4 Hypothalamic–pituitary–adrenal axis. Corticotropin-releasing hormone (CRH), produced by the hypothalamus and
released in the median eminence, stimulates the synthesis and processing of proopiomelanocortin, with resulting release of proopiomelanocortin
peptides that include adrenocorticotropic hormone (ACTH) from the anterior pituitary. ACTH binds to the melanocortin-2 receptor in the adrenal
gland and stimulates the cholesterol-derived synthesis of adrenal steroid hormones. Glucocorticoids released into the systemic circulation exert
negative feedback inhibition of CRH and ACTH release from the hypothalamus and pituitary, respectively, in a classic example of negative
feedback hormone regulation. This closely regulated circuit is referred to as the HPA axis. (Modified with permission from Molina PE: Endocrine Physiology,
3rd ed. New York: McGraw-Hill Medical, 2010.)

50% of total androgens in the prostate of adult men are DNA-binding domain. They are classified into types I and II.
derived from adrenal steroid precursors. Adrenal secretion of Type I receptors are specific for mineralocorticoids but have a
DHEA and DHEAS increases in children at the age of high affinity for glucocorticoids. Type II receptors are specific
6–8 years, and values of circulating DHEAS peak between the for glucocorticoids and are expressed in virtually all cells.
ages of 20 and 30 years. Thereafter, serum levels of DHEA The higher concentration of glucocorticoids, and the high
and DHEAS decrease. affinity of the mineralocorticoid receptor for glucocorticoids,
raises the issue of ligand–receptor specificity and resulting
physiologic action. Several factors are in place to enhance the
STEROID HORMONE TARGET specificity of the mineralocorticoid receptor for aldosterone.
ORGAN CELLULAR EFFECTS First, plasma glucocorticoids bind to CBG and albumin. This
plasma protein binding allows only a small amount (< 10%) of
Most of the physiologic effects of glucocorticoid and mineral- the unbound hormone to freely cross cell membranes. Second,
ocorticoid hormones are mediated through binding to intrac- aldosterone target cells possess enzymatic activity of
ellular receptors that belong to a superfamily of steroid, 11β-hydroxysteroid dehydrogenase type II, the enzyme that
thyroid, retinoid, and orphan receptors and that operate as inactivates cortisol to cortisone (Figure 65–6). Third, the min-
ligand-activated transcription factors to regulate gene expres- eralocorticoid receptor discriminates between aldosterone
sion. Mineralocorticoid and glucocorticoid receptors are and glucocorticoids. Aldosterone dissociates from the miner-
closely related, and share similarities in their ligand- and alocorticoid receptor five times more slowly than do the
CHAPTER 65 Adrenal Gland 661

Stimuli to renin

Liver

Kidney
Angiotensinogen
(453 aa)

Renin (enzyme)

Angiotensin I
(10 aa)

Angiotensin I
Angiotensin-converting Angiotensin-converting
enzyme enzyme
(endothelium) (endothelium)
Angiotensin II

Angiotensin II
(8 aa)

Adrenal
Cardiovascular cortex
system Aldosterone

Kidney

Vasoconstriction Salt and H2O


retention

Blood pressure

FIGURE 65–5 Regulation of aldosterone release by the renin–angiotensin–aldosterone system. A decrease in the effective circulating
blood volume triggers the release of renin from the juxtaglomerular apparatus in the kidney. Renin cleaves angiotensinogen, the hepatic
precursor of angiotensin peptides, to form angiotensin I. Angiotensin I is converted to angiotensin II by angiotensin-converting enzyme (ACE),
which is bound to the membrane of endothelial cells. Angiotensin II is a potent vasoconstrictor and stimulates the production of aldosterone in
the zona glomerulosa of the adrenal cortex. Aldosterone production is also stimulated by potassium and ACTH; aa = amino acids. (Reproduced with
permission from Widmaier EP, Raff H, Strang KT [editors]: Vander’s Human Physiology: The Mechanisms of Body Function, 11th ed. McGraw-Hill, 2007.)

glucocorticoids, despite their similar affinity constants. In SPECIFIC EFFECTS OF ADRENAL


other words, aldosterone is less easily displaced from the min-
eralocorticoid receptor than is cortisol. Together, these mecha-
CORTEX HORMONES
nisms ensure that under normal conditions, mineralocorticoid
action is restricted to aldosterone. However, when production Glucocorticoids
and release of glucocorticoids is excessive, or when the conver- Cortisol binds to the glucocorticoid receptor (type II glucocorti-
sion of cortisol to its inactive metabolite cortisone is impaired, coid receptor) (Figure 65–6). The hormone–receptor complex
the higher circulating and tissue cortisol levels may lead to translocates to the nucleus, where it binds to specific DNA
binding and stimulation of mineralocorticoid receptors. sequences (glucocorticoid response elements) and exerts its
662 SECTION IX Endocrine and Metabolic Physiology

GC MC

Cell surface
11β-HSD 2

GC GC CS MC
GR MR MR MR

GRE MRE MRE

FIGURE 65–6 Steroid hormone receptors and mineralocorticoid specificity. Mineralocorticoids (MC; aldosterone) and glucocorticoid
(GC; cortisol) hormones bind to intracellular receptors that share 57% homology in the ligand-binding domain and 94% homology in the DNA-binding
domain. Cortisol binds the mineralocorticoid (MR) receptor with high affinity. Because more cortisol is produced than aldosterone, this could
complicate the regulation of aldosterone-specific effects. Nonspecificity is prevented by the presence of an enzyme: 11-hydroxysteroid dehydrogenase
type 2 (11β-HSD2) in mineralocorticoid target cells. This enzyme converts cortisol into its less active form cortisone (CS) that has less affinity for the
mineralocorticoid receptor (MR). Another factor that contributes to ensure that mineralocorticoid effects are kept under regulation is the fact that
aldosterone dissociates from the mineralocorticoid receptor more slowly than cortisol despite their similar affinity constants. In other words,
aldosterone is less easily displaced from the mineralocorticoid receptor than is cortisol. Thus, glucocorticoids and mineralocorticoids bind to
intracellular receptors (GR and MR, respectively), which dimerize prior to binding to glucocorticoid- or mineralocorticoid-responsive elements (GRE
and MRE, respectively) in the nucleus, thus modulating (increasing or suppressing) transcription of specific genes. Cortisol, because of its high-affinity
binding to the MR, can produce mineralocorticoid-like effects (sodium retention). Conversion to cortisone (CS) decreases the affinity for the receptor
shown by the ill-fit of CS with the MR. Decreased activity of the 11β-HSD2 leads to decreased conversion of cortisol to cortisone and increased
mineralocorticoid activity. (Modified with permission from Molina, PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010)

physiologic effects by altering transcription of genes. Because corticoid deficiency and manifests as hypotension and decreased
virtually all cells express glucocorticoid receptors, the physio- sensitivity to vasoconstrictor administration. In the central ner-
logic effects are multisystemic. Glucocorticoids affect interme- vous system, glucocorticoids modulate perception and emotion
diary metabolism, stimulate proteolysis and gluconeogenesis, and may produce marked changes in behavior. Some of the main
inhibit muscle protein synthesis, and increase fatty acid mobili- physiologic effects of glucocorticoids are listed in Table 65–2. It
zation. Their hallmark effect is to increase blood glucose con- is important to note that some of these become evident only at
centrations, hence the name “glucocorticoids.” In the liver, high circulating levels of cortisol.
glucocorticoids increase the expression of gluconeogenic
enzymes. In muscle, glucocorticoids interfere with GLUT4
translocation to the plasma membrane, causing insulin resis- Mineralocorticoids
tance. In bone and cartilage, glucocorticoids decrease the The principal physiologic function of aldosterone is to regu-
insulin-like growth factor I, insulin-like growth factor–binding late renal sodium reabsorption and potassium excretion, hence
protein 1, and growth hormone expression and action, and affect the name “mineralocorticoid.” Aldosterone binds to the min-
thyroid hormone interactions. At high circulating levels, gluco- eralocorticoid receptor in the principal cells of the distal tubule
corticoids are catabolic and result in loss of lean body mass and the collecting duct of the nephron, producing an increase
including bone and skeletal muscle. Glucocorticoids modulate in sodium reabsorption and potassium excretion (Figure 65–7).
the immune response by increasing anti-inflammatory cytokine Aldosterone increases sodium entry at the apical membrane of
synthesis and decreasing proinflammatory cytokine synthesis, the cells of the distal nephron through the amiloride-sensitive
exerting an overall anti-inflammatory effect. Their anti-inflam- epithelial Na+ channel (ENaC). The Na+/K+-adenosine
matory effects have been exploited by the use of synthetic ana- triphosphatase (ATPase), located in the basolateral mem-
logs of glucocorticoids, such as prednisone, for the treatment of brane of the cells, maintains the intracellular sodium concen-
chronic inflammatory diseases. The development of potent tration by extruding the reabsorbed sodium toward the
inhaled steroids has been a major advance in the treatment of extracellular and blood compartments.
asthma. In the vasculature, glucocorticoids affect reactivity to The specific effects of aldosterone are to increase the syn-
vasoactive substances, such as angiotensin II and norepineph- thesis of Na+ channels in the apical membrane, increase the
rine. This interaction becomes evident in patients with gluco- synthesis and activity of Na+/K+-ATPase in the basolateral
CHAPTER 65 Adrenal Gland 663

TABLE 65–2 Physiologic effects of cortisol*.


System Effects

Metabolism Degrades muscle protein and increases nitrogen excretion


Increases gluconeogenesis and plasma glucose levels
Increases hepatic glycogen synthesis
Decreases glucose utilization (anti-insulin action)
Decreases amino acid utilization
Increases fat mobilization
Redistributes fat
Permissive effects on glucagon and catecholamine effect

Hemodynamic Maintains vascular integrity and reactivity


Maintains responsiveness to catecholamine pressor effects
Maintains fluid volume

Immune function Increases anti-inflammatory cytokine production


Decreases proinflammatory cytokine production
Decreases inflammation by inhibiting prostaglandin and leukotriene production
Inhibits bradykinin and serotonin inflammatory effects
Decreases circulating eosinophil, basophil, and lymphocyte counts (redistribution effect)
Impairs cell-mediated immunity
Increases neutrophil, platelet, and red blood cell counts

Central nervous system Modulates perception and emotion


Decreases CRH and ACTH release
CRH, corticotropin-releasing hormone; ACTH, adrenocorticotropic hormone. *Some of the effects listed (particularly in Immune Function) occur only with very high cortisol levels.

Angiotensin II Principal cells

Extracellular
Aldosterone
fluid [K+]
Cortical collecting-
duct cells Interstitial Lumen
K+ space
Na+
Interstitium
Nucleus Apical
membrane
Na+/K+ -ATPase 3Na+
Na+
Na+
K+ 2K+
ENaC
Na+ Basolateral
membrane
Na+
K+
Na+
Lumen

FIGURE 65–7 Renal physiologic effects of aldosterone. Aldosterone diffuses across the plasma membrane and binds to its cytosolic
receptor. The receptor–hormone complex is translocated to the nucleus, where it interacts with the promoter region of target genes, activating
or repressing their transcriptional activity and thereby increasing transepithelial Na+ transport. Aldosterone increases Na+ entry at the apical
membrane of the cells of the distal nephron through the amiloride-sensitive epithelial Na+ channel (ENaC). Aldosterone promotes potassium
excretion through its effects on Na+/K+-ATPase and epithelial Na+ and K+ channels in collecting duct cells. An increase in the extracellular fluid K+
concentration stimulates the secretion of aldosterone, and a decrease in K+ inhibits aldosterone secretion. Angiotensin II has a synergistic effect
on the stimulation of aldosterone production induced by hyperkalemia. ATP, adenosine triphosphate. (Reproduced with permission from Gennari JF.
Current concepts: Hypokalemia. NEJM. 1998;339:451. Copyright Massachusetts Medical Society. All Rights reserved.)
664 SECTION IX Endocrine and Metabolic Physiology

membrane (which pulls cytosolic Na+ to the interstitium in Glucocorticoid Deficiency


exchange for K+ transport into the cell), and increase the
Glucocorticoid deficiency can result from lack of ACTH stimu-
expression of H+-ATPase in the apical membrane and the Cl−/
lation of adrenal glucocorticoid production (secondary defi-
HCO3– exchanger in the basolateral membrane of intercalated
ciency) or from adrenal dysfunction (primary deficiency).
cells, described in Chapters 44 and 45. These cells express car-
Exogenous administration of synthetic analogs of glucocorti-
bonic anhydrase and contribute to the acidification of urine
coids in the chronic treatment of some diseases will also suppress
and alkalinization of plasma.
CRH and ACTH, thereby leading to adrenal atrophy. Therefore,
Mineralocorticoids act on fewer cell types than do glucocor-
the sudden discontinuation of treatment may be manifested as
ticoids. Their receptors are not as widely expressed as those for
an acute case of adrenal insufficiency, a medical emergency.
glucocorticoids. Classic aldosterone-sensitive tissues include
Most cases of ACTH deficiency involve deficiencies of other
epithelia in the distal parts of the nephron, the surface epithe-
pituitary hormones. Because aldosterone is mainly under the
lium of the distal colon, and the salivary and sweat gland ducts.
regulation of angiotensin II and K+, individuals may not neces-
Additional effects of aldosterone include increased sodium
sarily manifest with simultaneous mineralocorticoid deficiency
reabsorption in salivary and sweat glands, and increased K+
when impaired ACTH release is the causative factor. Glucocor-
excretion from the colon.
ticoid deficiency due to primary adrenal insufficiency is also
known as Addison’s disease, which can be the result of autoim-
Androgens mune or infectious destruction of the adrenal gland.
The physiologic effects of DHEA and DHEAS are not com-
pletely understood. Current knowledge indicates that low levels DISEASES OF OVERPRODUCTION
of DHEA are associated with cardiovascular disease in men and
with an increased risk of premenopausal breast and ovarian can-
AND UNDERSECRETION OF
cer in women. In contrast, high levels of DHEA might increase MINERALOCORTICOIDS
the risk of postmenopausal breast cancer. Exogenous adminis-
tration of DHEA to the elderly increases several hormone levels, Aldosterone Excess
including insulinlike growth factor-1, testosterone, dihydrotes- Primary hyperaldosteronism, also known as Conn’s syndrome,
tosterone, and estradiol. The specific mechanisms through is a condition in which autonomous benign tumors of the
which DHEA exerts its actions are not completely understood. adrenal glands hypersecrete aldosterone. The excess aldoster-
one leads to hypertension because of Na+ and H2O retention
DISEASES OF OVERPRODUCTION and hypokalemia because of excess K+ secretion. The release
of renin is suppressed.
AND UNDERSECRETION OF Secondary hyperaldosteronism is a renin-dependent phe-
GLUCOCORTICOIDS nomenon. A decrease in the effective arterial blood volume,
associated with ascites or heart failure, leads to continuous
Glucocorticoid Excess stimulation of the renin–angiotensin II–aldosterone system.
Glucocorticoid excess can be due to overproduction by an
adrenal tumor, overstimulation of adrenal glucocorticoid syn- Aldosterone Deficiency
thesis by ACTH produced by a pituitary tumor or an ectopic
tumor, or the iatrogenic administration of excess synthetic Primary hypoaldosteronism is most often due to primary
glucocorticoids. The clinical manifestation of glucocorticoid adrenal insufficiency as described above. Plasma renin levels
excess, known as Cushing’s syndrome, can be separated into are elevated, so this condition is also referred to as hyper-
two categories depending on its etiology. reninemic hypoaldosteronism.
ACTH-independent Cushing’s syndrome is usually due to Secondary hypoaldosteronism may be due to inadequate
an adrenal neoplasm autonomously releasing cortisol despite stimulation of aldosterone secretion (hyporeninemic hypoal-
suppressed ACTH (due to cortisol negative feedback). Excess dosteronism) despite normal adrenal function. This condition
exogenous glucocorticoid therapy is also a form of ACTH- is usually associated with renal insufficiency.
independent Cushing’s syndrome.
ACTH-dependent Cushing’s syndrome has two causes: DISEASES OF OVERPRODUCTION AND
Cushing’s disease is reserved for Cushing’s syndrome caused
by excess secretion of ACTH by pituitary corticotroph tumors
UNDERSECRETION OF ADRENAL
and is the most common endogenous form of the syndrome. ANDROGENS
ACTH production may also be ectopic (derived from extrapi-
tuitary tissue), most frequently because of small cell lung car- Adrenal Androgen Excess
cinoma. ACTH-dependent Cushing’s syndrome is characterized Congenital adrenal hyperplasia is an autosomal recessive disor-
by increased glucocorticoid levels due to excess stimulation by der due to 21-hydroxylase deficiency in approximately 90% of
ACTH leading to bilateral hyperplasia of the adrenal cortex. cases. In this condition, impaired cortisol production leads to a
CHAPTER 65 Adrenal Gland 665

lack of negative glucocorticoid feedback resulting in increased Catecholamines can undergo reuptake by extraneuronal sites,
ACTH release. Increased ACTH stimulation of adrenal hor- degradation at target cells by catechol-O-methyltransferase
mone synthesis leads to a buildup of cortisol precursors that (COMT) or monoamino oxidase (MAO), or direct filtration
due to the lack of 21-hydroxylase activity get shunted to the into the urine. The joint action of MAO and COMT on norepi-
androgen synthetic pathway producing an androgen excess. nephrine and epinephrine, especially in the liver, produces the
Steroid 21-hydroxylase (a cytochrome P450 enzyme) converts metabolite vanillylmandelic acid (VMA), which is then
17-hydroxyprogesterone to 11-deoxycortisol, and progester- excreted in the urine; dopamine metabolized through this path-
one to 11-deoxycorticosterone. Both 11-deoxycortisol and way yields homovanillic acid. Because these metabolites are
11-deoxycorticosterone are precursors for cortisol and aldoster- water soluble and have high levels of urinary excretion, they can
one, respectively. Total loss of 21-hydroxylase activity results in play an important role in the clinical detection of tumors that
cortisol and aldosterone deficiencies. If not detected and treated produce excess catecholamines. In humans, VMA is the major
in time, it can cause death in early infancy owing to shock, end product of norepinephrine and epinephrine metabolism.
hyponatremia, and hyperkalemia. Deficiency of 21-hydroxylase
leads to accumulation of steroid hormone precursors, and these
can be directed to the androgen hormone synthetic pathway. TARGET ORGAN CELLULAR EFFECTS
Increased androgen production can lead to virilization in affected
The physiologic effects of catecholamines are mediated by
girls and signs of postnatal androgen excess in both sexes, includ-
binding to G protein–coupled adrenergic receptors distrib-
ing rapid linear growth and accelerated skeletal maturation.
uted widely throughout the body (Table 65–3). Catecholamines
released from the adrenal medulla exert their effects almost
exclusively in peripheral tissues and not in the brain, because
HORMONES OF THE catecholamines do not readily cross the blood–brain barrier.
ADRENAL MEDULLA
Alpha-adrenergic Receptors
The adrenal medulla consists of cells that synthesize and
secrete the catecholamines epinephrine (in greater amounts) Alpha-adrenergic receptors have greater affinity for epineph-
and norepinephrine. rine than for norepinephrine or for isoproterenol, a synthetic
agonist. They are subdivided into α1- and α2-receptors.
α1-Adrenergic receptors are further subdivided into α1A,
CHEMISTRY AND BIOSYNTHESIS α1B, and α1D. α1-Adrenergic receptors play important roles in
the regulation of several physiologic processes, including myo-
Catecholamines are tyrosine-derived hormones (Figure 65–8). cardial contractility and chronotropy and hepatic glucose
The transporters involved in packaging epinephrine into metabolism (Table 65–4).
secretory vesicles are the vesicular monoamine transporters, α2-Adrenergic receptors are also subdivided into three
which are expressed exclusively in neuroendocrine cells. groups, including α2A, α2B, and α2C (Table 65–3). Some of the
Because of the expression of these transporters in sympatho- physiologic effects mediated by this subtype of receptor involve
medullary tissues, their function can be used diagnostically actions at two counteracting α2-receptor subtypes. For example,
for radioimaging and localization of catecholamine-producing stimulation of α2A-receptors decreases sympathetic outflow and
tumors (pheochromocytomas). The synthesis of cate- blood pressure, whereas stimulation of α2B-receptors increases
cholamines can be regulated by changes in the activity of blood pressure by direct vasoconstriction. α2-Adrenergic recep-
tyrosine hydroxylase by release from end-product inhibition tors are implicated in diverse physiologic functions, particularly
or by an increase in enzyme synthesis. in the cardiovascular system and the central nervous system.

CATECHOLAMINE RELEASE, Beta-adrenergic Receptors


TRANSPORT, AND METABOLISM Beta-adrenergic receptors have been subclassified as β1-, β2-,
and β3-receptors. They have greater affinity for isoproterenol
The release of catecholamines is a direct response to sympa- than for epinephrine or norepinephrine (Table 65–3). The
thetic nerve stimulation of the adrenal medulla (please β1-adrenergic receptor plays an important role in regulating
review Chapter 19). Acetylcholine released from the pre- contraction and relaxation of cardiac myocytes (Table 65–4).
ganglionic sympathetic nerve terminals binds to nicotinic The β2-adrenergic receptor mediates several physiologic
cholinergic receptors in the plasma membrane of the chro- responses, including vasodilatation, bronchial smooth mus-
maffin cells, leading to the exocytosis of secretory granules, cle relaxation, and lipolysis, in various tissues. Abnormalities
which release catecholamines into the interstitial space, from in the function of this adrenergic receptor may lead to hyper-
where they are transported in the circulation to their target tension. The β3-adrenergic receptor plays an important role
organs. Catecholamines have a short half-life and for the in mediating catecholamine-stimulated thermogenesis and
most part circulate bound to albumin. lipolysis.
666 SECTION IX Endocrine and Metabolic Physiology

HO

Tyrosine

COOH NH2

Tyrosine
hydroxylase

HO
HO
Dihydroxyphenylalanine (DOPA)

COOH NH2
DOPA
decarboxylase
HO
HO

Dopamine

Dopamine-β NH2
hydroxylase
HO
HO
Norepinephrine
OH

NH2
Phenylethanolamine
N-methyltransferase
HO
HO
Epinephrine
OH
N
CH3 H

FIGURE 65–8 Catecholamine synthetic pathway. Catecholamine synthesis from the precursor L-tyrosine involves four enzymatic
reactions that take place in the cytosol of chromaffin cells. These are the following: (1) hydroxylation of tyrosine to L-dihydrophenylalanine
(L-Dopa) by the enzyme tyrosine hydroxylase. This enzyme is found in the cytosol of catecholamine-producing cells and is the main control
point for catecholamine synthesis. The activity of this enzyme is inhibited by norepinephrine, providing feedback control of catecholamine
synthesis. (2) Decarboxylation of L-Dopa to dopamine by the enzyme dopa decarboxylase in a reaction that requires pyridoxal phosphate as
a cofactor. This end product is packaged into secretory vesicles. (3) Hydroxylation of dopamine to norepinephrine by the enzyme dopamine
β-hydroxylase, a membrane-bound enzyme found in synaptic vesicles that uses vitamin C as a cofactor. This reaction occurs inside the
secretory vesicles. (4) Methylation of norepinephrine to epinephrine by the enzyme phenylethanolamine N-methyltransferase. The activity of
this adrenal medullary enzyme, found in the cytosol of the chromaffin cell, is modulated by adjacent adrenal steroid production, underscoring
the importance of radial arterial flow from the cortex to the medulla. The latter enzymatic reaction occurs in the cytoplasm and thus requires
that norepinephrine leave the secretory granules by a passive transport mechanism. The epinephrine produced in the cytoplasm must reenter
the secretory vesicles through ATP-driven active transport. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill
Medical, 2010.)
CHAPTER 65 Adrenal Gland 667

TABLE 65–3 Adrenergic receptors and signaling pathways.


Adrenergic Receptor G Protein Second Messenger

β-Adrenergic receptors β1, β2, β3 Gαs G protein Activate adenylate cyclase

α1-Adrenergic receptors α1A, α1B, α1D Mostly Gαq/11 family of G proteins Usually activate PLCα (thereby activating PKC via
DAG and increasing intracellular Ca2+ via IP3) or PLA2

α2-Adrenergic receptors α2A, α2B, α2C Mostly varied Gαi and Gα0 proteins May decrease the activity of adenylate cyclase
(opposing the effects of β-adrenergic receptors).
Activate K channels. Inhibit Ca2+ channels and
activate PLCβ or PLA2 (an effect similar to that of
α1-adrenergic receptors)
PL, phospholipase; PKC, protein kinase C; DAG, diacylglycerol; IP3, inositol 1,4,5-trisphosphate.

CATECHOLAMINE REGULATION OF
PHYSIOLOGIC EFFECTS ADRENERGIC RECEPTORS
Catecholamines are released from the adrenal medulla in Chronic elevation of catecholamine levels leads to sustained
response to sympathetic stimulation and are central to the stimulation of adrenergic receptors, which can alter tissue
stress response to a physical or psychological insult such as responsiveness. For example, chronic exposure to β-agonists,
severe blood loss, decrease in blood glucose concentration, as in asthmatic patients treated with isoproterenol, promotes
traumatic injury, surgical intervention, or a fearful experience. receptor desensitization. In contrast, treatment with
Because catecholamines are part of the “fight–or-flight” α-agonists, as found in some nasal decongestants, results in
response, their physiologic effects include arousal, alerting, tachyphylaxis. Persistent exposure to an agonist of the adren-
papillary dilation, piloerection, sweating, bronchial dilation, ergic receptor can also result in an actual loss of receptors
tachycardia, inhibition of smooth muscle activity in the gas- because of degradation or receptor desensitization. Adrener-
trointestinal tract, constriction of the sphincters, and relax- gic receptors can also undergo upregulation because of
ation of the uterine muscles (Table 65–4). Catecholamines increased transcription of the gene for the receptor. Two hor-
ensure substrate mobilization from the liver, muscle, and fat mones are known to produce this effect: glucocorticoids and
by stimulating the breakdown of glycogen (glycogenolysis) thyroid hormone. In addition, glucocorticoids and thyroid
and fat (lipolysis). Thus, an increase in circulating cate- hormone can regulate the expression of several types of adren-
cholamines is associated with elevations in plasma glucose ergic receptors through posttranscriptional events.
and free fatty acid levels. Some of the most important effects of
catecholamines are exerted in the cardiovascular system,
where they increase heart rate (tachycardia), produce periph-
DISEASES OF OVERPRODUCTION
eral vasoconstriction, and elevate vascular resistance. OF ADRENAL CATECHOLAMINES
Endocrine cells of the sympathoadrenal system are named
TABLE 65–4 Catecholamine physiologic effects. chromaffin cells, and the tumors arising from these cells are
called pheochromocytomas. Pheochromocytomas produce
α-Adrenergic Mediated β-Adrenergic Mediated catecholamines, and patients present with signs of excess cat-
Vasoconstriction Vasodilation echolamine effects, such as sustained or paroxysmal hyper-
tension associated with headache, sweating, or palpitations.
Iris dilation Cardioacceleration

Intestinal relaxation Increased myocardial strength

Intestinal sphincter contraction Intestinal and bladder wall


relaxation CLINICAL CORRELATION
Pilomotor contraction Uterus relaxation
CASE A
Bladder sphincter contraction Bronchodilation
A term newborn infant presents with abnormal-appear-
Bronchoconstriction Calorigenesis
ing ambiguous genitalia that look more male than
Uterine smooth muscle Glycogenolysis female, including an enlarged clitoris. Blood tests reveal
contraction
hyponatremia, high 17-hydroxyprogesterone, and low
Cardiac contractility Lipolysis cortisol levels. CT scan reveals enlarged adrenal glands.
Hepatic glucose production Chromosomal and gonadal sex is assigned as female
668 SECTION IX Endocrine and Metabolic Physiology

■ Steroid hormone receptors undergo conformational changes


gender. The diagnosis of congenital adrenal hyperplasia on hormone binding that allow them to bind to DNA and
is made. stimulate gene transcription.
Congenital adrenal hyperplasia is most frequently ■ Specificity of the mineralocorticoid receptor is conferred by
(>90% of cases) the result of steroid 21-hydroxylase defi- conversion of cortisol to cortisone by 11β-hydroxysteroid
ciency due to a mutation. This is associated with ambig- dehydrogenase.
uous external genitalia in females or virilization during ■ Glucocorticoids facilitate fuel mobilization, decrease glucose
the first 2–3 years of life in both girls and boys. Adrenal utilization, and produce immunosuppression.
insufficiency (cortisol deficiency) may present with or ■ Aldosterone regulates body sodium balance and stimulates
without salt wasting (due to decreased aldosterone excretion of potassium.
synthesis). The lack of negative feedback inhibition of ■ Catecholamine release is under sympathetic neural control.
ACTH release results in excess ACTH production and ■ The response to stress by the host relies on close interaction
stimulation of the early events in adrenal steroid hor- between the steroid hormones and catecholamines to ensure
mone synthesis and adrenal growth (hyperplasia). The adequate fuel mobilization and hemodynamic control.
partial block in the synthetic pathway of cortisol and
aldosterone leads to shunting of pregnenolone to the
androgen hormone synthetic pathway, resulting in the STUDY QUESTIONS
excess in progesterone causing increased androgen 1. Regulation of steroid hormone production by the adrenals
synthesis. Treatment consists of replacement of gluco- involves which of the following steps?
corticoids and, if desired or possible, surgical correction A) ACTH binding to a G protein–coupled receptor, stimulation
of the external genitalia as soon as possible. of cholesterol transfer to inner mitochondrial membrane,
and formation of pregnenolone
B) ACTH binding to a nuclear receptor, stimulation of
CASE B pregnenolone transfer to inner mitochondrial membrane,
and formation of DHEA
A young adult male patient is admitted to the hospital C) hydrolysis of cholesterol ester, stimulation of pregnenolone
because of severe headache and hypertension (blood pres- transfer to inner mitochondrial membrane, and formation
sure 220/100 mm Hg). The patient’s history includes com- of androstenedione
plaints of episodic mild headaches that resolved D) ACTH binding to a G protein–coupled receptor, increased
spontaneously over the past 3 years. These have progres- cholesterol ester hydrolysis, and pregnenolone transfer to
sively increased in severity and frequency and are accom- inner mitochondrial membrane
panied by sweating, dizziness, palpitations, and pallor. 2. A deficiency in 21-hydroxylase activity would be associated with
Measurements of catecholamines and their metabolites in which of the following?
a 24-hour urine specimen were increased, leading to the A) central obesity, increased cortisol levels, and decreased
diagnosis of a pheochromocytoma. An MRI examination androstenedione
of the abdominal revealed a large, unilateral adrenal mass. B) virilization, increased ACTH, and decreased
Surgical resection of the adrenal was performed, reveal- cortisol levels
C) decreased ACTH levels and decreased cortisol levels
ing a large pheochromocytoma.
D) hypertension, decreased ACTH, and increased cortisol
Pheochromocytomas are rare tumors that produce cat-
levels
echolamines. They usually arise from the adrenal medulla, E) virilization, hypertension, increased ACTH, and increased
but about 10% can arise in extra-adrenal chromaffin tis- cortisol levels
sue. The classic clinical presentation is the triad of epi- 3. Regarding cortisol production and transport, which of the
sodic headache, sweating, and palpitations as a result of following statements is true?
the release of stored catecholamines from the tumor. An A) Most cortisol circulates unbound to protein.
unrecognized pheochromocytoma may lead to death as B) Peak plasma levels occur at noon.
the result of a hypertensive crisis, arrhythmia, or myocar- C) Most of the plasma cortisol is bound to transcortin.
dial infarction. D) More than 90% is excreted intact in the urine.
4. Physiologic effects of cortisol include
A) hypoglycemia, increased fatty acid mobilization, and
decreased central fat deposition
B) increased amino acid–derived gluconeogenesis,
CHAPTER SUMMARY increased glucose utilization, and hypoglycemia
■ Cortisol (adrenal glucocorticoid) production and release is C) hyperglycemia, decreased fatty acid mobilization, and
under ACTH regulation. decreased central fat deposition
■ Aldosterone (adrenal mineralocorticoid) release is under D) decreased glucose utilization, hyperglycemia,
angiotensin II and K+ regulation. and lipolysis
CHAPTER 65 Adrenal Gland 669

5. Regarding the production and release of aldosterone from the 6. Which of the following statements is true regarding
adrenals, which of the following statements is correct? catecholamine synthesis and release from the adrenals?
A) Aldosterone production in the zona glomerulosa is mainly A) Epinephrine accounts for 20% of total adrenal
under ACTH control. catecholamine release.
B) Aldosterone production in the zona glomerulosa is mainly B) Norepinephrine is derived from epinephrine through the
under angiotensin II control. action of the enzyme phenylethanolamine
C) Aldosterone production in the adrenal medulla is mainly N-methyltransferase.
under angiotensin II control. C) Catecholamine synthesis is regulated by tyrosine
D) Aldosterone production in the zona glomerulosa is mainly hydroxylase.
under K+ control. D) 35% of catecholamines released are excreted intact in the
urine.
This page intentionally left blank
66
C H A P T E R

Endocrine Pancreas
Patricia E. Molina

O B J E C T I V E S

■ Identify the principal hormones secreted from the endocrine pancreas,


their cells of origin, and their chemical nature.
■ Understand the nutrient, neural, and hormonal mechanisms that regulate
pancreatic hormone release.
■ List the principal target organs for insulin and glucagon and their major
physiologic effects.
■ Identify the disease states caused by overproduction, underproduction, or
decreased sensitivity to insulin, and describe the principal manifestations
of each.

The pancreas is a mixed exocrine and endocrine gland that mass of endocrine cells, and their principal secretory product
plays a central role in digestion and in the metabolism, utiliza- is insulin. The α-cells account for about 18–20% of the endo-
tion, and storage of energy substrates. Normal pancreatic crine cells and are responsible for glucagon secretion. A small
function is essential for the physiologic control of glucose number of δ-cells (4–6%) secrete somatostatin, and an even
homeostasis, which, in turn involves interaction of several tis- smaller number of cells (1%) secrete pancreatic polypeptide.
sues and hormones in the regulated balance between hepatic The rich vascularization by fenestrated capillaries allows
glucose release (from glycogen breakdown and gluconeogen- ready access to the circulation for the hormones secreted by
esis), dietary glucose absorption, and glucose uptake and dis- the islet cells. Venous blood from the pancreas drains into the
posal from skeletal muscle and adipose tissue. The pancreatic hepatic portal vein. Therefore, the liver, a principal target
hormones insulin and glucagon play central roles in regulating organ for the physiologic effects of pancreatic hormones, is
each of these processes; their overall effects are, in part, modi- exposed to the highest concentrations of pancreatic hormones.
fied by other hormones such as growth hormone, cortisol, and Following first-pass hepatic metabolism, the pancreatic endo-
epinephrine. crine hormones are distributed to the systemic circulation.
Parasympathetic, sympathetic, and sensory nerves richly
innervate the pancreatic islets, and the respective neurotrans-
FUNCTIONAL ANATOMY mitters and neuropeptides released from their nerve termi-
nals exert important regulatory effects on pancreatic endocrine
The pancreas is a retroperitoneal gland located near the duo- hormone release. Acetylcholine, released from the parasym-
denum, composed of exocrine cells that are clustered in acini pathetic nerve terminals, stimulates the secretion of insulin,
(see Chapter 51). Embedded within the acini are richly vascu- glucagon, somatostatin, and pancreatic polypeptide. Norepi-
larized, small clusters of endocrine cells called the islets of nephrine released from sympathetic nerve terminals inhibits
Langerhans, in which two endocrine cell types (β and α) pre- basal and glucose-stimulated insulin secretion and stimulates
dominate. The β-cells constitute about 73–75% of the total glucagon and pancreatic polypeptide secretion.

671

Ch66_671-682.indd 671 11/26/10 4:51:48 PM


672 SECTION IX Endocrine and Metabolic Physiology

PANCREATIC HORMONES its first pass, which extracts about 40–80% of insulin delivered.
Additional degradation of insulin occurs in the kidneys as well
INSULIN as at target tissues by insulin proteases following endocytosis
of the receptor-bound hormone. C-peptide is not readily
Insulin Synthesis degraded in the liver. Thus, the relatively long half-life of
C-peptide (35 minutes) allows its release to be used as an index
Insulin is a polypeptide hormone with a highly conserved of the secretory capacity of the endocrine pancreas.
amino acid sequence. It is produced from proinsulin cleavage
of the connecting C-peptide from the amino-terminal β-chain
and the carboxy-terminal α-chain (Figure 66–1A). C-peptide Regulation of Insulin Release
links the α- and β-chains, allowing proper folding of the mol- The release of insulin throughout the day is pulsatile and
ecule and the formation of disulfide bonds between the two rhythmic in nature (Figure 66–1C). The pulsatile release of
chains. Removal of the C-peptide exposes the end of the insu- insulin appears to be critical in the suppression of liver glucose
lin chain that interacts with the insulin receptor. Both insulin production and in insulin-mediated glucose disposal by adi-
and the free C-peptide are packaged into secretory granules of pose tissue. Insulin release increases after a meal in response
which about 5% are in a readily releasable pool and the rest to the increases in plasma levels of glucose and amino acids.
(>95%) in a reserve pool (Figure 66–1B). Insulin release from An increase in plasma glucose concentration is followed by a
granules from different pools leads to a biphasic pattern transient stimulation of insulin secretion known as first-phase
response to glucose stimulation of the β-cell. Only a small pro- secretion, which consists of a rapid burst of release to a
portion of the cellular stores of insulin is released even under high peak and then a steep decline to a low secretion rate
maximal stimulatory conditions. (Figure 66–1B). This is followed by second-phase secretion,
Stimulation of the pancreatic β-cell leads to release of equal which consists of a gradually increasing rate of secretion to a
amounts of insulin and C-peptide into the portal circula- plateau level. This biphasic response to glucose is a major
tion. Insulin circulates in its free form and has a half-life of characteristic of glucose-stimulated insulin secretion. The first
3–8 minutes. It is degraded predominantly by the liver during phase occurs over a period of minutes, the second over an

A Proinsulin Insulin C Pulsatile insulin release


C peptide
900
Portal vein INS.

S 750
conc. (pM)

α-chain S
N-terminal S S
S S 450
S S S
S S 300
S
C-terminal 150
β-chain
0
B Biphasic insulin release 0 20 40 60 80 100 120
Time (minutes)
Immature glucose
Insulin release per beta cell

granules 1st phase


15
(granules/min)

10
2nd phase
5

0
Readily releasable 0 5 10 15 20
granules t (min)

FIGURE 66–1 Principal feature of insulin synthesis and release. A) Insulin synthesis starts with the translation of insulin mRNA into
an inactive protein called preproinsulin. Preproinsulin undergoes posttranslational modification in the endoplasmic reticulum (ER) to form
proinsulin. The active form of insulin is produced by modification of proinsulin by cleavage of the C-peptide structure linking the α- and
β-chains. Insulin is composed of two amino acid chains. The chains are held together by two disulfide (S–S) bonds. A third disulfide bond is
present within the α-chain. Both insulin and the cleaved C-peptide are packaged in secretory granules that accumulate in the β-cell cytosol
and are coreleased in response to glucose stimulation. B) Insulin release occurs in a biphasic mode from secretory granules that are immediately
available for release (<5%) and from granules that must undergo a series of preparatory reactions including mobilization to the plasma
membrane (>95%). These granule preparatory or maturation processes are modulated by intracellular levels of ATP, ADP, and Ca2+. C) Insulin
release in response to a meal is characterized by increased frequency and amplitude of pulsatile release. Shown are portal insulin concentrations
during basal state (left) and after ingestion of a mixed meal (right) in normal patients. (Modified with permission from Porksen N et al: Human insulin release
processes measured by intraportal sampling. Am J Physiol Endocrinol Metab 2002;282(3):E695–E702.)
CHAPTER 66 Endocrine Pancreas 673

Glucose
1
GLUT2 carrier

Mitochondrion ATP

2
ATP-sensitive K+ channel
Endoplasmic reticulum K+

Ca2+ store
Depolarization

4 Ca2+ Ca2+
3
Secretory
granules
Voltage-gated Ca2+ channel
Ca2+

Exocytosis

Insulin
β CELL
FIGURE 66–2 Regulation of insulin release. Glucose is the principal stimulus for insulin release from the pancreatic β-cell. (1) It enters the β-cell
by a specific glucose-transporter protein (GLUT-2) and is immediately phosphorylated by glucokinase (not shown). The increased concentrations of
ATP and resulting greater ATP/ADP ratio lead to inhibition and (2) closure of the ATP-sensitive K+ channels (the target of sulfonylurea drugs), resulting
in depolarization of the plasma membrane and (3) opening of the voltage-dependent Ca2+ channels. As a result, there is an increased influx of
extracellular Ca2+ as well as (4) mobilization of Ca2+ from intracellular stores leading to the fusion of insulin-containing secretory granules with the
plasma membrane and the release of insulin (and C-peptide) into the circulation. PLC, phospholipase C; AC, adenylate cyclase; CCK, cholecystokinin;
GLP-1, glucagon-like peptide-1. (Reproduced with permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)

hour or more. Secretion is the result of a combination of an Regulation of K+ channels by ATP is mediated by the sulfonyl-
increase in the total amount of insulin released in each secre- urea receptor, and is the basis for the therapeutic use of sulfo-
tory burst and an increased pulse frequency of a similar mag- nylurea drugs in the treatment of type 2 diabetes mellitus.
nitude (Figure 66–1C). The β-cell Ca2+ concentrations can also be increased by amino
The pancreatic β-cell functions as a fuel sensor that responds acids through their metabolism and ATP generation, or by
to changes in plasma levels of energy substrates (glucose direct depolarization of the plasma membrane. Other factors
and amino acids), hormones (glucagon-like peptide 1 [GLP-1] that amplify the glucose-induced release of insulin from the
and epinephrine), and neurotransmitters (norepinephrine and β-cell include acetylcholine, cholecystokinin, gastrointestinal
acetylcholine) (Figure 66–2). Glucose is the principal stimulus peptide, and GLP-1. Catecholamines and somatostatin inhibit
for insulin release from the pancreatic β-cells. insulin.
The glucose-induced stimulation of insulin release is the
result of glucose metabolism by the β-cell and an increase in
the adenosine triphosphate (ATP)/ADP ratio in the cytosol. Physiologic Effects of Insulin
Glucose enters the β-cell through a membrane-bound glucose Insulin produces a wide variety of effects that range from
transporter (GLUT 2) (Figure 66–2). The production of ATP immediate (within seconds), such as the modulation of ion
from glucose oxidation results in an increase in intracellular (K+) and glucose transport into the cell; early (within minutes),
ATP/ADP that inhibits (closes) the ATP-sensitive K+ channels such as the regulation of metabolic enzyme activity; moderate
(KATP) in the β-cell, reducing the efflux of K+. This process (within minutes to hours), such as the modulation of enzyme
results in membrane depolarization, activation (opening) of synthesis; to delayed (within hours to days), such as the effects
voltage-dependent Ca2+ channels, and increased Ca2+ influx. on growth and cellular differentiation. Overall, the actions of
The increase in intracellular Ca2+ concentrations triggers the insulin at target organs are anabolic and promote the synthesis
exocytosis of insulin secretory granules and the release of of carbohydrate, fat, and protein, and these effects are medi-
insulin into the extracellular space and into the circulation. ated through binding to the insulin receptor (Table 66–1).
674 SECTION IX Endocrine and Metabolic Physiology

TABLE 66–1 Insulin effects on carbohydrate, fat, and protein metabolism.


Metabolic Effects Insulin Stimulates Insulin Inhibits

Carbohydrate Glucose transport in adipose tissue and muscle Glycogen breakdown in muscle and liver
metabolism Rate of glycolysis in muscle and adipose tissue Gluconeogenesis in the liver
Glycogen synthesis in adipose tissue, muscle,
and liver

Lipid metabolism Fatty acid and triacylglycerol synthesis in tissues Lipolysis in adipose tissue, lowering the plasma fatty
Uptake of triglycerides from the blood into adipose acid level
tissue and muscle Fatty acid oxidation in muscle and liver
Rate of cholesterol synthesis in the liver Ketogenesis

Protein metabolism Amino acid transport into tissues Protein degradation in muscle
Protein synthesis in muscle, adipose tissue, liver, Urea formation
and other tissues

Insulin Receptor pling of insulin receptor activation to signaling, mainly


the phosphatidylinositol 3-kinase (PI 3-kinase) and the
The insulin receptor belongs to the same family of the insu-
mitogen-activated protein kinase (MAPK) pathways
linlike growth factor receptor, and the insulin-related recep-
(Figure 66–3).
tor, all of which are involved in cell division, metabolism,
Activation of the PI 3-kinase pathway leads to activation of
and development (Figure 66–3). Insulin binding to the
enzymes that catalyze the cellular effects of insulin, particu-
receptor triggers receptor autophosphorylation on tyrosine
larly the metabolic effects of the hormone, including glucose
residues in the cytoplasmic domain (β-chain). The activated
transport, glycolysis, and glycogen synthesis, and regulation of
receptor phosphorylates tyrosine residues of several pro-
protein synthesis. Moreover, this pathway is involved in cell
teins known as insulin receptor substrates (IRS-1, -2, -3,
growth and transmits a strong antiapoptotic signal, promoting
-4). These IRS proteins facilitate the interaction of the insu-
cell survival. The other main signaling pathway that is acti-
lin receptor with intracellular substrates by serving as a scaf-
vated by insulin binding to its receptor is the MAPK pathway,
fold for recruitment of proteins involved in signal
which is involved in mediating the proliferative and differen-
transduction to downstream pathways. The result is cou-
tiation effects elicited by insulin.
The number of available insulin receptors is modulated by
Insulin exercise, diet, insulin, and other hormones. Chronic exposure
Insulin to high insulin levels, obesity, and excess growth hormone all
receptor
lead to a downregulation of insulin receptors. In contrast,
exercise and starvation upregulate the number of receptors.
The affinity of the receptor for insulin is increased following a
period of decreased insulin levels.
ATP

Insulin Effects at Target Organs


Early effects
The effects of insulin on skeletal muscle glucose utilization
P IRS-1 P dominate insulin action. Insulin mediates about 40% of glu-
cose disposal by the body, the great majority of which occurs
P P in skeletal muscle. The movement of glucose into the cell is
mediated by a family of carrier proteins, or glucose transport-
ers (GLUT), with their own unique tissue distribution. The
Other effector Various
main transporters and their predominant tissue distributions
systems and effects are summarized in Table 66–2.
secondary mediators
Insulin-stimulated glucose transport is mediated through
FIGURE 66–3 Intracellular responses triggered by insulin GLUT-4. Approximately 90% of GLUT-4 is sequestered intra-
binding to the insulin receptor. Red balls and balls labeled P cellularly in the absence of insulin or other stimuli such as
represent phosphate groups. IRS-1, insulin receptor substrate-1. exercise. Insulin binding to its receptor results in recruitment
(Reproduced with permission from Barrett KE, Barman SM, Boitano S, Brooks H: of GLUT-4 from cytosolic vesicular compartments to the
Ganong’s Review of Medical Physiology, 23rd ed. McGraw-Hill Medical, 2009.) plasma membrane.
CHAPTER 66 Endocrine Pancreas 675

TABLE 66–2 Main features of glucose transporters.


Transporter Expression Function

GLUT-1 Ubiquitous, with particularly high levels in human erythrocytes Glucose uptake by skeletal muscle and fat under
and in the endothelial cells lining the blood vessels of the brain. basal conditions
Expressed in skeletal muscle and fat

GLUT-2 Low-affinity glucose transporter present in pancreatic β-cells, Functions in the glucose sensor system and
liver, intestine, and kidney ensures that glucose uptake by pancreatic β-cells
and hepatocytes occurs only when circulating
glucose levels are high

GLUT-3 Primarily in neurons Together, GLUT-1 and GLUT-3 are crucial in


allowing glucose to cross the blood–brain barrier
and enter neurons

GLUT-4 Predominantly in striated muscle and adipose tissue. In contrast to The major insulin-responsive transporter
the other GLUT isoforms, which are primarily localized on the cell
membrane, GLUT-4 transporter proteins are sequestered in
specialized storage vesicles that remain within the cell’s interior
under basal conditions
GLUT-5 Spermatozoa and small intestine Predominantly a fructose transporter

Intermediate effects and protein loss, resulting in negative nitrogen balance and
The intermediate effects of insulin are mediated by modula- muscle wasting.
tion of protein phosphorylation of enzymes involved in meta-
bolic processes in muscle, fat, and liver (Table 66–1). In fat, Long-term effects Sustained insulin stimulation enhances
insulin inhibits lipolysis and ketogenesis by triggering the the synthesis of lipogenic enzymes and the repression of gluco-
dephosphorylation of hormone-sensitive lipase and stimulates neogenic enzymes. The growth-promoting and mitogenic ef-
lipogenesis by activating acetyl-CoA carboxylase. In the fects of insulin are long-term responses mediated through the
adipocytes, dephosphorylation of hormone-sensitive lipase MAPK pathway.
inhibits the breakdown of triglycerides to fatty acids and glyc-
erol, the rate-limiting step in the release of free fatty acids
mediated by lipolysis. This process thereby reduces the amount GLUCAGON
of substrate that is available for ketogenesis. Insulin antago-
nizes catecholamine-induced lipolysis through the phospho- Glucagon Synthesis
rylation and activation of phosphodiesterase, leading to a Glucagon is a 29–amino acid polypeptide hormone secreted
decrease in intracellular cAMP levels and a concomitant by the α-cells of the islets of Langerhans, which antagonizes
decrease in protein kinase A activity. insulin’s action. The primary sequence of glucagon is highly
In the liver, insulin stimulates the gene expression of conserved among vertebrates. Glucagon is synthesized as
enzymes involved in glucose utilization (e.g., glucokinase, proglucagon and then proteolytically processed to yield
pyruvate kinase, and lipogenic enzymes) and inhibits the glucagon. The prohormone proglucagon is expressed in the
gene expression of enzymes involved in glucose production pancreas, and also in other tissues, such as enteroendocrine
(e.g., phosphoenolpyruvate carboxykinase and glucose-6- cells in the intestinal tract and the brain. However, the
phosphatase) (Figure 66–4). Insulin stimulates glycogen processing of the prohormone differs among tissues. The
synthesis by increasing phosphatase activity, leading to the two main products of proglucagon processing are glucagon
dephosphorylation of glycogen phosphorylase and glycogen in the α-cells of the pancreas and GLP-1 in the intestinal
synthase. In addition, insulin-mediated dephosphorylation of cells. GLP-1 is produced in the intestine in response to a
inhibitory sites on hepatic acetyl-CoA carboxylase increases high concentration of glucose in the intestinal lumen. It is
the production of malonyl-CoA and simultaneously reduces known as an incretin, a mediator that amplifies insulin
the rate at which fatty acids can enter hepatic mitochondria for release from the β-cell in response to a glucose load. Gluca-
oxidation and ketone body production. gon has a short half-life (5–10 minutes) and is degraded
In muscle, insulin stimulates glucose uptake and favors mostly in the liver.
protein synthesis though phosphorylation of a serine/threo-
nine protein kinase known as mammalian target of rapamy-
cin (mTOR). In addition, insulin favors lipid storage in Regulation of Glucagon Release
muscle as well as in adipose tissue. Insulin deficiency leads to Glucagon release is inhibited by hyperglycemia (high blood
glucose accumulation in blood, a decrease in lipid storage, glucose levels) and stimulated by hypoglycemia (low blood
676 SECTION IX Endocrine and Metabolic Physiology

Glycogenolysis Glycolysis

Glycogen Glucose

G Glycogen I Glucokinase G
Glycogen
I synthase phosphorylase G Glucose-6-phosphate

Glucose-1-P
Fructose-6-phosphate
ATP Phospho-
ADP fructokinase G
Gluconeogenesis
Fructose-1,6-bisphosphate
(2) Pyruvate
Dihydroxyacetone
phosphate
(2) Oxaloacetate
I PEP carboxykinase G
(2) PEP
Glyceraldehyde-3-phosphate

(2) 3-Phosphoglycerate
1,3-Bisphosphoglycerate
(2) 1,3-Bisphosphoglycerate
3-Phosphoglycerate
Fructose-1,6-bisphosphate
Fructose-1,6-bisphosphate G
2-Phosphoglycerate
Fructose-6-phosphate

Phosphoenolpyruvate
Glucose-6-phosphate
I Glucose-6-phosphatase G I Pyruvate
kinase G
Glucose
Pyruvate
FIGURE 66–4 Glucagon and insulin effects on hepatic glucose metabolism. Binding of glucagon and insulin to their respective
receptors stimulates a cascade of protein phosphorylation steps that activate (or inhibit) key enzymes involved in the regulation of
glycogenolysis, gluconeogenesis, and glycolysis. The principal target enzymes for insulin- and glucagon-mediated effects are presented.
The overall result is an increase in hepatic glucose output. G, glucagon; I, insulin; PEP, phosphoenolpyruvate; ATP, adenosine triphosphate;
ADP, adenosine diphosphate. (Reproduced with permission from Jiang G, Zhang BB: Glucagon and regulation of glucose metabolism. Am J Physiol Endocrinol
Metab 2003;284(4):E671–E678.)

glucose levels). A meal rich in carbohydrates suppresses gluca- 66–4 and 66–5). Glucagon is the principal hormone involved in
gon release and stimulates insulin release from the β-cells prevention and counterregulation of hypoglycemia. The key
through intestinal release of GLP-1. Somatostatin also inhib- enzymatic steps regulated by glucagon that mediate the stimula-
its glucagon release. High amino acid levels following an tion of hepatic glucose output are summarized in Table 66–3.
amino acid–rich meal stimulate glucagon release. Epinephrine Glucagon can exert effects on adipose tissue as well.
stimulates release of glucagon through a β2-adrenergic mecha- However, these are important primarily during periods of pro-
nism (while it suppresses insulin release from β-cells through longed stress or food deprivation, particularly when insulin
an α2-adrenergic mechanism). Vagal (parasympathetic) stim- release is suppressed. In the adipocyte, glucagon activates hor-
ulation increases glucagon release. mone-sensitive lipase, the enzyme that breaks down triglycer-
ides (stored fat) into diacylglycerol and free fatty acids,
releasing them into the circulation. Glycerol released into the
Physiologic Effects of Glucagon circulation can be utilized in the liver for gluconeogenesis or
Glucagon mediates its effects by binding to the glucagon Gαs for reesterification. Free fatty acids are used as fuel by most
protein–coupled receptor (Figure 66–5). The principal target tissues, predominantly skeletal muscle and liver. In the liver,
tissue for glucagon is the liver. Glucagon’s main physiologic free fatty acids are used for reesterification or they can
effect is to increase plasma glucose concentrations by stimulat- undergo β-oxidation and conversion into ketone bodies.
ing de novo hepatic glucose production through gluconeogen- Thus, ketogenesis is regulated by the balance between the
esis and glycogen breakdown and decreasing glycolysis (Figures effects of glucagon and insulin at their target organs.
CHAPTER 66 Endocrine Pancreas 677

Glucagon

Adenylate
Phospholipase cyclase
C Gq

(PiP2) Receptor Gαs γ


β
Inositol 1.4.5 GDP
triphosphate Cytoplasm
(IP3) GTP cAMP

Ca2+

PKA Phosphorylase
PGC-1
PEPCK Kinase
G-6-Pase
a b
Phosphorylase

Glycolysis Glycogenesis Gluconeogenesis Glycogenolysis

Glucose
FIGURE 66–5 Glucagon receptor-mediated cellular effects. Glucagon binds to GPCR on target cells. The principal effects of glucagon
are mediated in hepatocytes where glucagon, through activation of adenylate cyclase and elevation in cAMP, leads to increased protein kinase
A (PKA) activity resulting in phosphorylation of enzymes responsible for control of glucose metabolism. Phosphorylase b is the inactive form of
the enzyme; phosphorylase a is the active form. Furthermore, there is a change in phospholipase C activity leading to a change in intracellular
Ca2+ release). The ultimate result is an increase in hepatic glucose production through increased gluconeogenesis and glycogenolysis. Additional
abbreviations: PGC-1, peroxisome proliferator-activated receptor coactivator-1; PEPCK, phosphoenolpyruvate carboxykinase; G-6-Pase, glucose-
6-phosphatase; PIP2, phosphatidylinositol 4,5-biphosphate. (Modified with permission from Cooper GM: The Cell: A Molecular Approach, 2nd ed. Sinauer, 2000.)

SOMATOSTATIN PANCREATIC POLYPEPTIDE


Somatostatin is a 14–amino acid peptide hormone produced Pancreatic polypeptide is a 36–amino acid peptide hormone
by the δ-cells of the pancreas. Its release is stimulated by high- that belongs to a peptide family including neuropeptide Y
fat, high-carbohydrate, and particularly protein-rich meals, and peptide YY. It is produced in the endocrine type F cells
and is inhibited by insulin. Somatostatin has a generalized located in the periphery of pancreatic islets and is released into
inhibitory effect on virtually all gastrointestinal and pancreatic the circulation after ingestion of food, exercise, and vagal
exocrine and endocrine functions. stimulation. The effects of pancreatic polypeptide include

TABLE 66–3 Effects of glucagon on hepatic glucose metabolism.


Effect on Target Enzyme Metabolic Response

Increased expression of glucose-6-phosphatase Frees glucose to enter the circulation

Suppression of glucokinase Decreases glucose entry into the glycolytic cascade

Phosphorylation (activation) of glycogen phosphorylase Stimulates glycogenolysis

Inhibition of glycogen synthase Inhibits glycogen synthesis

Stimulation of phosphoenolpyruvate carboxykinase expression Stimulates gluconeogenesis

Inactivation of phosphofructokinase-2 and activation of fructose-6-phosphatase. Inhibits glycolysis


PFK-2 is the kinase activity and fructose-2,6-bisphosphatase is the phosphatase Stimulates gluconeogenesis
activity of the bifunctional regulatory enzyme, phosphofructokinase-2/fructose-2,6-
bisphosphatase
Suppression of activity of the pyruvate kinase Decreases glycolysis
678 SECTION IX Endocrine and Metabolic Physiology

inhibition of pancreatic exocrine secretion, gallbladder con- absence of insulin therapy. T2DM results from impaired sensi-
traction, modulation of gastric acid secretion, and gastrointes- tivity to insulin (insulin resistance) and a subsequent relative
tinal motility. Pancreatic polypeptide crosses the blood–brain loss of normal regulation of insulin secretion and accounts for
barrier and has been postulated to play a role in regulating more than 90% of diabetes cases. It is usually associated with
feeding behavior. obesity in adults and is characterized by mild hyperglycemia.
It rarely leads to ketoacidosis.

AMYLIN
Type 1 Diabetes Mellitus
Amylin, or islet amyloid polypeptide, is a 37–amino acid The pathophysiology of T1DM involves a complete lack of
peptide hormone that belongs to the calcitonin family, insulin secretion due to autoimmune destruction of the islet
which includes calcitonin, calcitonin gene-related peptide, cells. This results in impaired entry of glucose into the cells
and adrenomedullin. Amylin is synthesized as a small precur- and accumulation of glucose in the blood. This leads to
sor, undergoes posttranslational modification (amidation), is increased plasma osmolarity and urinary loss of glucose,
stored in β-cell granules, and is released along with insulin accompanied by excess loss of water and sodium (polyuria).
and C-peptide. Plasma amylin concentrations increase after a The resulting dehydration triggers compensatory mecha-
meal or glucose infusion. Amylin appears to work with insulin nisms such as thirst (polydipsia). The inability of the cells to
to regulate plasma glucose concentrations in the bloodstream, utilize glucose resembles a state of cellular starvation, stimu-
suppressing the postprandial secretion of glucagon and slow- lating hunger (polyphagia) and triggering the activation of
ing gastric emptying. It is the main component of pancreatic compensatory responses to increase the release and availabil-
islet amyloid, found in the vast majority of patients with type 2 ity of fuel substrates though activation of lipolysis and prote-
diabetes mellitus, and is thought to contribute to destruction olysis.
of the pancreatic β-cell. Diabetic ketoacidosis is an acute pathologic event charac-
terized by elevated blood glucose levels and ketone bodies and
metabolic acidosis, resulting directly from decreased insulin
DISEASES ASSOCIATED WITH availability and simultaneous elevations of the counterregula-
tory hormones glucagon, catecholamines, cortisol, and growth
PANCREATIC HORMONES hormone. Diabetic ketoacidosis is precipitated by infections,
discontinuation of or inadequate insulin use, new-onset
HORMONE-PRODUCING TUMORS (untreated) diabetes, and other events such as the stress asso-
ciated with surgery.
Excess pancreatic hormone production and release are usually
In diabetic ketoacidosis, gluconeogenesis in the liver pro-
due to hormone-producing tumors, with insulinoma being
ceeds unopposed by the physiologic presence of insulin. The
the most frequent. Insulinomas produce excessive amounts of
excess blood glucose increases osmolarity, which, if severe,
insulin, and patients present with episodes of hypoglycemia,
can result in diabetic coma. Low insulin levels and the high
confusion, aggressiveness, palpitations, sweating, convulsions,
levels of counterregulatory hormones glucagon, epinephrine,
and even unconsciousness. These symptoms are mostly
and cortisol combine to increase the activity of hormone-sen-
observed before breakfast and following physical exercise. The
sitive lipase, increase the release of free fatty acids, and
compensatory or counterregulatory response of the body
decrease the activity of acetyl-CoA carboxylase, thereby
includes the release of catecholamines, glucagon, cortisol, and
impairing the reesterification of free fatty acids and promot-
growth hormone.
ing fatty acid conversion into ketone bodies (Figure 66–6).
Glucagonomas are unusual tumors that may produce symp-
The steps involved in ketogenesis are β-oxidation of fatty
toms of diabetes. Excessive glucagon production by the tumor
acids to acetyl-CoA, formation of acetoacetyl-CoA, and con-
may also result in an overall catabolic effect on fat and muscle,
version of acetoacetyl-CoA to 3-hydroxy-3-methylglutaryl-
leading to severe weight loss and anorexia.
CoA and then to acetoacetate, which is then reduced to
3-hydroxybutyrate. The enzymes involved in ketogenesis are
DIABETES MELLITUS summarized in Table 66–4. Acetoacetate can be spontane-
ously decarboxylated to acetone, a highly fat-soluble com-
The most common disease resulting from impaired pancreatic pound that is excreted slowly by the lungs and is responsible
hormone release or sensitivity is diabetes mellitus. The two for the fruity odor of the breath of individuals with diabetic
forms of diabetes mellitus, type 1 (T1DM) and type 2 (T2DM), ketoacidosis.
are characterized by impaired insulin release, but for different Ketone bodies released into the blood can freely diffuse
reasons. T1DM is the result of β-cell destruction. It accounts across cell membranes and serve as an energy source for extra-
for 2–5% of cases, and it occurs more frequently in younger hepatic tissues including the brain, skeletal muscle, and
people, hence its archaic name, juvenile-onset diabetes. T1DM kidneys. Ketone bodies are filtered and reabsorbed in the kid-
is characterized by the development of ketoacidosis in the ney. At physiologic pH, ketone bodies, with the exception of
CHAPTER 66 Endocrine Pancreas 679

Acetyl CoA
carboxylase
Adipocyte Acetyl CoA Malonyl CoA
(FA synthesis)
Tryglycerides
β-oxidation Acetyl CoA
+ HSL
Acetoacetyl CoA
Free fatty acids
HMG CoA
Mitochondria
Acetoacetate

Hepatocyte
H
HO C CH2 O=C–CH3
CH2 CH2
COO- COO-
Acetoacetate
3-β -Hydroxybutyrate
CH3
O=C
CH3
Acetone
FIGURE 66–6 Process of ketogenesis in insulin deficiency. Insulin deficiency and high levels of counterregulatory hormones
glucagon, epinephrine, and cortisol combine to increase the activity of hormone-sensitive lipase, increase the release of free fatty acids, and
decrease the activity of acetyl-coenzyme A (CoA) carboxylase, thereby impairing the reesterification of free fatty acids and promoting fatty
acid conversion into ketone bodies. The excess supply of fatty acetyl-CoA and the deficiency in oxaloacetate increase the oxidation to ketone
bodies, with the resulting release of ketone bodies into the blood. Plus signs (+) denote steps that are favored by insulin deficiency. HSL,
hormone-sensitive lipase; FA, fatty acid; HMG, 3-hydroxy-3-methylglutaryl. (Reproduced with permission from Molina PE: Endocrine Physiology, 3rd ed.
New York: McGraw-Hill Medical, 2010.)

acetone, dissociate completely. The resulting liberation of H+ excessive rise in plasma glucose, which in turn produces a
from ketone body metabolism exceeds the blood’s buffering compensatory and exaggerated second-phase hyperinsuline-
capacity, leading to metabolic acidosis with an increased mic response. This initial period of sustained hyperinsuline-
anion gap. If severe, this condition can lead to coma. mia downregulates the insulin receptors, decreasing
the sensitivity of tissues to insulin action and producing a state
of insulin resistance. The main pathologic defects in T2DM
Type 2 Diabetes Mellitus are excessive hepatic glucose production, defective β-cell
T2DM is the result of a decreased responsiveness of peripheral secretory function, and peripheral insulin resistance.
tissues to insulin action (insulin resistance) and inadequate
responsiveness of the β-cells to glucose, which later is followed
by a net reduction in β-cell mass. Patients with type 2 diabetes INSULIN RESISTANCE
secrete normal amounts of insulin during fasting, but
in response to a meal or glucose load, they secrete less insulin Insulin resistance is the inability of peripheral target tissues to
than nondiabetic patients. In addition to a relative reduction respond properly to normal circulating concentrations of
in insulin release, the pattern of insulin release is also altered insulin. To maintain euglycemia, the pancreas compensates by
following a meal, with pulses that are significantly smaller, secreting increased amounts of insulin. In patients with T2DM,
sluggish, and erratic, particularly after dinner. This abnormal- insulin resistance precedes the onset of the disease by several
ity results in significantly higher levels of fasting glucose in years. Compensating for insulin resistance by an increase
these patients. in insulin release is effective only temporarily. As insulin
Regardless of the etiology (e.g., abnormalities in glucose resistance increases, impaired glucose tolerance develops.
transport; abnormal insulin synthesis, processing, storage, or Ultimately, failure or exhaustion of the pancreatic β-cell results
secretion), the earliest physiologic indication of β-cell dys- in a relative decrease in insulin secretion. The combination of
function is a delay in the acute insulin response to glucose. insulin resistance and impaired β-cell function characterizes
The defect in the initial response to a glucose load leads to an clinical T2DM.
680 SECTION IX Endocrine and Metabolic Physiology

TABLE 66–4 Three principal enzymes involved in


tions, including retinopathy, nephropathy, and neuropa-
ketogenesis.
thy. Treatment aims at the control of blood sugar, and is
Enzyme Tissue Function monitored by prevailing levels of glycosylated hemoglo-
bin. Treatment can include sulfonylureas that increase
Hormone-sensitive Adipocytes Breaks down triglycerides,
pancreatic insulin secretion, biguanides that decrease
lipase releasing fatty acids into the
circulation hepatic glucose production, insulin sensitizers (glita-
zones) that attempt to restore insulin sensitivity, and drugs
Acetyl-CoA Liver Catalyzes the conversion of
carboxylase acetyl-CoA to malonyl-CoA,
that can decrease gastrointestinal absorption of glucose.
the primary substrate of Finally, insulin injections are often used because pharma-
fatty acid biosynthesis cological doses of insulin can increase glucose uptake
HMG-CoA synthase Liver Involved in the despite the relative insulin resistance. Newly available
conversion of acetyl-CoA analogs of GLP-1 can also be used to amplify the insulin
to acetoacetate response to glucose.
CoA, coenzyme A; HMG, 3-hydroxy-3-methylglutaryl.

Exercise has been demonstrated to increase glucose trans- CHAPTER SUMMARY


port in skeletal muscle and to decrease insulin resistance in ■ Insulin release is under nutrient, neural, and hormonal
patients with T2DM. The increase in glucose transport result- regulation.
ing from exercise is insulin-independent and involves the ■ The pancreatic β-cell functions as a glucose sensor in the
enzyme AMP-activated protein kinase. AMP-activated pro- process of insulin release.
tein kinase has been termed a master metabolic switch because ■ The PI 3-kinase pathway mediates most of insulin’s metabolic
it phosphorylates key target proteins that control flux through effects, and the MAPK pathway is mostly involved in mediating
metabolic pathways. Repeated physical activity improves insu- the proliferative responses.
lin sensitivity by increasing skeletal muscle expression and/or ■ The principal metabolic effects of insulin are to increase
activity of key signaling proteins involved in the regulation of glucose utilization in skeletal muscle, suppress hepatic glucose
glucose uptake and metabolism, lipid oxidation and/or turn- production, and inhibit lipolysis.
over, and oxidative capacity. ■ Glucagon antagonizes insulin’s effects by stimulating hepatic
glucose release.
■ T1DM results in hyperglycemia because of β-cell destruction
and loss of insulin.
CLINICAL CORRELATION ■ T2DM results in hyperglycemia because of tissue resistance
A middle-aged, overweight Hispanic man is referred for to insulin action and an inadequate β-cell response to
evaluation following detection of an increased fasting hyperglycemia.
plasma glucose of 150 mg/dL by his primary care physi- ■ A disruption in the balance of insulin and glucagon can lead to
cian. Attempts of lifestyle modifications including exer- ketogenesis and hyperosmolar coma.
cise and diet to reduce body weight over the past 7 months
have been unsuccessful. In addition, glycosylated hemo-
globin, an estimate of blood glucose control over the last STUDY QUESTIONS
month, remains elevated at 7%. The diagnosis of T2DM is 1. In a patient with severe hypoglycemia (38 mg/dL), the
established and treatment with oral hypoglycemic drugs differential diagnosis between self-administered insulin
is initiated. overdose and a tumor that produces excess insulin can be
T2DM is the result of deficient insulin action (insulin made by determining plasma levels of
resistance) leading to fasting hyperglycemia and the A) insulin
inability to have an adequate insulin-induced increase in B) somatostatin
glucose uptake from plasma after a glucose load. The inci- C) C-peptide
dence of T2DM has increased during the past several D) gastrin
decades due to an aging population, increased prevalence 2. Physiologic responses to insulin include
of obesity, and decreased physical activity, among other A) stimulation of glucose transport in skeletal muscle,
factors that include ethnicity, environmental, and genetic red blood cells and the brain
risk factors. Diabetes mellitus is associated increased risk B) inhibition of triglyceride synthesis in adipose tissue
factor for cardiovascular disease and long-term complica- C) stimulation of amino acid uptake by skeletal muscle
D) stimulation of glucose reabsorption in the kidney
CHAPTER 66 Endocrine Pancreas 681

3. A 57-year-old female patient is brought to the emergency room 5. When would plasma insulin levels be expected to be highest?
with a history of frequent urination, weight loss, and decreased A) following a carbohydrate-rich meal
oral intake. At presentation, the patient is lethargic, dehydrated, B) after intravenous administration of somatostatin
hypotensive, and tachycardic. Her caretaker reports that she C) during a surgical procedure
was recovering from a recent bout of pneumonia. She had been D) following vigorous exercise
diagnosed with T2DM five years prior to this incident. Which of
the following laboratory findings is most likely?
A) plasma glucose of 40 mg/dL
B) plasma osmolarity of >350 mOsm/L
C) low blood pH
D) high plasma ketones
4. A 21-year-old male patient with T1DM is brought to the
emergency room for abdominal pain, nausea, and vomiting of
16-hour duration. On examination, you notice that his insulin
pump has stopped functioning. Which of the following is likely
to be associated with his presentation?
A) high plasma insulin levels
B) increased plasma C-peptide levels
C) increased serum ketones
D) increased blood pH
E) decreased hepatic glycogen breakdown
This page intentionally left blank
67
C H A P T E R

Male Reproductive System


Patricia E. Molina

O B J E C T I V E S

■ Describe the physiologic functions of the principal components of the male


reproductive system.
■ Describe the endocrine regulation of testicular function by gonadotropin-
releasing hormone, follicle-stimulating hormone, luteinizing hormone,
testosterone, and inhibin.
■ Identify the cell of origin for testosterone, its biosynthesis, and transport
within the blood, metabolism, and clearance. List other physiologically
produced androgens.
■ List the target organs or cell types, the cellular mechanism of action, and the
physiologic effects of testosterone.
■ Describe spermatogenesis and the role of different cell types in this process.
■ Understand the neural, vascular, and endocrine factors involved in the
erection and ejaculation response.
■ Compare and contrast the actions of testosterone, dihydrotestosterone,
estradiol, and müllerian inhibitory factor in the process of sexual
differentiation.
■ Identify the causes and consequences of androgen overproduction and
underproduction in prepubertal and postpubescent adult males.

In utero sexual differentiation, maturation, spermatogenesis, numerous lobules made of convoluted tubes (seminiferous
and ultimately reproduction are all functions of the male repro- tubules) supported by loose connective tissue. The seminiferous
ductive system. The two principal functions of the testes, the tubules represent approximately 80–85% of the testicular mass.
male sex organs, are the production of sperm and the synthesis The Leydig cells, embedded in the connective tissue, are the
of testosterone. These processes ensure fertility and maintain endocrine cells responsible for the production of the most impor-
male sexual characteristics. Testicular function is regulated by tant circulating androgen, testosterone, a steroid hormone. The
the gonadotropins, follicle-stimulating hormone (FSH) and tubules consist of a basement layer lined with epithelial cells
luteinizing hormone (LH) as well as paracrine, neural, and forming the walls of the seminiferous tubules. These walls are
other endocrine factors. lined with germ cells (spermatogonia) and Sertoli cells.
Sertoli cells form tight junctions that functionally divide the
seminiferous tubules into two compartments or environments
FUNCTIONAL ANATOMY for the development of spermatozoa. The basal compartment
below the tight junctions has contact with the circulatory sys-
The male reproductive organs include the testes (the principal tem and is the space in which spermatogonia develop to pri-
male sex organs), the vas deferens, the ejaculatory ducts, the mary spermatocytes. The tight junctions open at specific
penis, and the accessory glands, which include the prostate and times and allow progression of spermatocytes to the adluminal
bulbourethral glands (Figure 67–1). The testes consist of compartment, where meiosis is completed. In the adluminal

683

Ch67_683-694.indd 683 11/30/10 4:02:03 AM


684 SECTION IX Endocrine and Metabolic Physiology

Vas Ejaculatory Seminal


deferens duct vesicle
Urinary Ureter
bladder

Pubic
bone
Efferent ductules
Epididymis
Seminiferous
tubule
Prostate
gland
Bulbourethral
Epididymis gland
Penis Rete
Urethra Testis testis

Vas
deferens

FIGURE 67–1 Functional anatomy of the male reproductive system. The male reproductive organs include the testes, the vas
deferens, the ejaculatory ducts, the penis, and the accessory glands, which include the prostate and bulbourethral glands. The testes consist of
numerous lobules made of tubuli seminiferi supported by loose connective tissue. The tubuli seminiferi are united to form larger ducts called
the tubuli recti. These larger tubules form a close anastomosing network of tubes called the rete testis, terminating in the ductuli efferentes.
The tubular network carries the seminal fluid from the testis to the epididymis, from where spermatozoa enter the vas deferens and then enter
the urethra through the ejaculatory ducts. The penis is composed of two functional compartments: the paired corpora cavernosa and the
corpus spongiosum. The corpora cavernosa form the greater part of the substance of the penis and consist of bundles of smooth muscle fibers
intertwined to form trabeculae, and containing numerous arteries and nerves. (Modified with permission from Widmaier EP, Raff H, Strang KT [editors]: Vander’s
Human Physiology: The Mechanisms of Body Function, 11th ed. McGraw-Hill, 2007.)

compartment, spermatocytes are protected by a blood–testis


barrier formed by tight junctions between the Sertoli cells. TABLE 67–1 Contribution of the genitourinary
The principal functions of Sertoli cells are as follows: system and accessory organs to sperm production.
• provide support for germ cells, forming an environment in Organ Function
which they develop and mature;
Genitourinary system
• provide the signals that initiate spermatogenesis and sustain
spermatid development; Efferent ductules, vas deferens, Movement of spermatozoa
ejaculatory duct, urethra Fluid reabsorption
• modulate pituitary gland function and, in turn, control of
spermatogenesis. Epididymis H+ secretion and acidification of
luminal fluid
The seminiferous tubules are united to form larger ducts that Capacitation of spermatozoa;
form a close anastomosing network of tubes (Figure 67–1). This glycoconjugation
tubular network carries the seminal fluid containing sperm Reservoir for mature
spermatozoa
from the testis to the epididymis; from here, spermatozoa enter Phagocytosis of aging
the vas deferens and then the ejaculatory ducts. The ejacula- spermatozoa
tory ducts move semen (sperm-containing fluid) into the ure- Accessory glands
thra. In addition to serving as a route of transport, the tubular
network or excretory system and the accessory organs play Seminal vesicle Secretion and storage of
fructose-rich product (preferred
important roles in the production of sperm-containing semen energy substrate for sperm),
through absorptive and secretory processes. As summarized in prostaglandins, ascorbic acid,
Table 67–1, these absorptive and secretory processes contribute fibrinogen- and thrombin-like
to the final composition of sperm-containing semen. Sperm proteins
account for approximately 10% of the volume of the ejaculated Prostate Secretion and storage of fluid
semen that is composed of testicular and epididymal fluid rich in acid phosphatase and
protease (prostate-specific
together with the secretory products of male accessory glands. antigen)
Most of the volume of the ejaculate is formed by the seminal
vesicles with the remainder consisting of epididymal fluids and Cowper’s glands Secretion of mucus into the
urethra on arousal
secretions from the prostate gland and bulbourethral glands.
CHAPTER 67 Male Reproductive System 685

The penis is composed of two functional compartments: the N


paired corpora cavernosa and the corpus spongiosum. The LH & FSH
corpora cavernosa form the greater part of the substance of
the penis and consist of bundles of smooth muscle fibers inter-
α
twined in a collagenous extracellular matrix. Interspersed GDP
β γ β γ
within this parenchyma is a complex network of endothelial C
cell–lined sinuses, arteries, and nerve terminals. The penis is
innervated by somatic and autonomic (both sympathetic and αi
parasympathetic) nerve fibers. The somatic innervation sup- GTP
plies the penis with sensory fibers and supplies the perineal
skeletal muscles with motor fibers. Autonomic nerves mediate
vascular dilation leading to penile erection, stimulate prostatic Adenylate cyclase Increase in translation
secretions, and control smooth muscle contraction of the vas of mRNAs for proteins
deferens during ejaculation. involved in cell growth and
differentiation, androgen
cAMP binding protein synthesis
(Sertoli cells), and
GONADOTROPIN REGULATION testosterone synthesis
(Leydig cells)
OF GONADAL FUNCTION PKA

The primary functions of the Leydig and Sertoli cells are to Gene expression
Protein regulation
produce the hormones involved in the regulation of reproduc- P
phosphorylation P
tive function and virilization and to produce spermatozoa.
These functions are regulated by the pituitary gonadotropins
Transcription Nucleus
FSH and LH. factors
The gonadotropins produce their physiologic responses by
binding to cell membrane G protein–coupled receptors
located in the Leydig and Sertoli cells (Figure 67–2). LH is the
principal regulator of testosterone production by the Leydig
cells. FSH plays an important role in the development of the Leydig & Sertoli Cells
immature testis, particularly by controlling Sertoli cell prolif-
eration and seminiferous tube growth. Because the tubules FIGURE 67–2 Receptor-mediated effects of gonadotropins
account for about 80% of the volume of the testis, FSH is of at target tissues. Model of signal transductional pathways of the
major importance in determining testicular size. FSH is gonadotropin receptor. On binding of FSH to the FSH receptor, the
Gαs subunit dissociates. Together with GTP, this complex directly
important in the initiation of spermatogenesis during puberty,
activates adenylyl cyclase, thereby leading to cAMP synthesis. cAMP
and is necessary for the production of androgen-binding activates protein kinase A (PKA), producing the dissociation of the
protein (ABP) by Sertoli cells and for the development of the catalytic subunit from the regulatory subunit. The active catalytic site
blood–testis barrier. of PKA can activate proteins by phosphorylation, and in the nucleus it
can phosphorylate transcription factors and affect gene transcription.

CONTROL OF GONADOTROPIN As a result, LH and FSH mediate several biologic responses at their
target cells. LH is the principal regulator of testosterone production
SYNTHESIS AND RELEASE by the Leydig cells. FSH plays an important role in the development
of the immature testis, particularly by controlling Sertoli cell
FSH and LH are glycoproteins consisting of a common proliferation and seminiferous tube growth. (Modified with permission
α-subunit, required for receptor binding, and a unique from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
β-subunit, which confers their biologic specificity. The synthe-
sis and release of the gonadotropins are regulated by the pulsa-
tile release of gonadotropin-releasing hormone (GnRH) ulation of gonadotropin release. Inhibins are heterodimer
from the hypothalamus, as well as by circulating hormones or glycoproteins consisting of an α- and a β-subunit (βA or βB).
their metabolites, as illustrated in Figure 67–3. Inhibin B is the physiologically important form in males.
LH stimulates testosterone production by the Leydig cell. Inhibin is produced and released from the Sertoli cells in
Testosterone released into the circulation inhibits LH release response to FSH stimulation, and its main function is to sup-
in a negative feedback loop. The negative feedback inhibition press FSH release in a classic negative feedback loop. Inhibin
of FSH release is mediated mainly by inhibin, a Sertoli cell– secretion appears to be dependent on Sertoli cell prolifera-
derived peptide. tion, maintenance, and spermatogenesis, all of these func-
In addition to the negative feedback inhibition produced by tions regulated by FSH. Inhibin B levels correlate with total
gonadal androgens, inhibin and activin contribute to the reg- sperm count and testicular volume and can be used as an
686 SECTION IX Endocrine and Metabolic Physiology

Hypothalamus
GnRH

Pituitary gland
LH
FSH

LH FSH

Negative
feedback

Negative
feedback
Sertoli cell

Inhibin

FIGURE 67–3 Negative feedback regulation of


gonadotropin synthesis and release. Gonadotropin Leydig cell
release from the anterior pituitary gland is under
control by hypothalamic GnRH release. LH stimulates
Leydig cell production and release of testosterone. FSH Testosterone
stimulates inhibin production and release from the
Sertoli cells. These two mediators regulate LH and FSH
release. The negative feedback inhibition of LH release
exerted by testosterone is mediated directly (through Blood vessels
androgen receptors) and indirectly (by local aromatase
conversion of testosterone to 17β-estradiol). Inhibin Testes
produces negative feedback inhibition of FSH release.
(Modified with permission from Molina PE: Endocrine Physiology,
3rd ed. New York: McGraw-Hill Medical, 2010.)

index of spermatogenesis. Activins belong to the same family either have a direct androgen receptor–mediated effect or be
of peptides as the inhibins and are homodimers or heterodi- metabolized to either 17β-estradiol through the action of aro-
mers of the β-subunit of the inhibins. The peptide and their matase or to 5α-dihydrotestosterone (DHT) through the
receptor are expressed in a wide variety of tissues. Activin action of 5α-reductase (Figure 67–4).
facilitates GnRH-mediated FSH release from the anterior Aromatase is expressed in the Sertoli cells as well as in
pituitary. extragonadal tissues. 17β-Estradiol produced by the testis
accounts for approximately 20% of the total circulating
estrogens in males. The majority of estradiol in males is
GONADAL FUNCTION produced in adipose tissue through the aromatization of tes-
tosterone and, to a smaller extent, of adrenal-derived andros-
The three principal hormones produced by the testis are tes- tenedione. Although some of the 17β-estradiol produced in
tosterone, estradiol, and inhibin. Testosterone, a steroid hor- peripheral tissues is released into the circulation, not all
mone produced by the Leydig cells, is the principal and most estrogens produced from testosterone are involved in medi-
important testicular and circulating androgen. Most of the ating endocrine responses. Some are involved in intracrine
testosterone released into the circulation is bound to plasma regulation of physiologic responses through estrogen recep-
proteins, primarily sex hormone–binding globulin (SHBG) tor stimulation (Figure 67–4). An example is testosterone-
and albumin. In the testes, testosterone is bound to androgen- induced negative feedback regulation of FSH release from
binding protein, a protein with great similarity to SHBG and a the anterior pituitary, which is mostly mediated by aromati-
product of the same gene. At its target cells, testosterone can zation of testosterone to estrogen. Another important exam-
CHAPTER 67 Male Reproductive System 687

Estradiol Testosterone PHYSIOLOGIC EFFECTS OF


DHEAS
ANDROGENS AT TARGET ORGANS
Aromatase 5α-
Sulfatase Reductase Testosterone and DHT affect sexual development, matura-
DHEA DHT tion, and function, and contribute to the maintenance of fertil-
Estradiol
AR ity and secondary sexual characteristics in the adult male. In
3β-OHD AR
addition, they exert anabolic effects in muscle and bone. Both
4A ER
17β-OHD
testosterone and DHT bind to the same androgen (nuclear)
receptor, on target cells (see Figure 67–4). DHT is the most
Testosterone potent activator of the androgen receptor. However, distinct
AR AR physiologic responses can be attributed to each hormone
Erβ/ERα
AR (Table 67–2), in part determined by the local conversion of
testosterone to DHT by 5α-reductase.
transcription

SEXUAL DEVELOPMENT
Testosterone target cells AND DIFFERENTIATION
FIGURE 67–4 Receptor-mediated effects of testosterone Sexual differentiation in humans is genetically and hormonally
at target tissues. Testosterone (a steroid hormone) enters the cell
controlled (Figure 67–6). Genes on the Y chromosome signal
by passive diffusion. It can be converted to dihydrotestosterone
(DHT) by 5α-reductase and bind to the androgen receptor (AR), or
primordial cells in the embryonic gonad ridge to differentiate
it can be converted to 17β-estradiol by aromatase. 17β-Estradiol into Sertoli cells and stimulate newly migrated germ cells to
can be released to act on a neighboring cell’s estrogen receptors differentiate as spermatogonia, developing into a testis. The
(ER) (paracrine mechanism); it can enter the circulation (endocrine cells of the embryonic testis secrete hormones that lead to
effects) or it can bind to either the estrogen receptor α or β the development of male secondary sexual characteristics. The
and subsequently bind to the nucleus and affect transcription. Sertoli cells secrete müllerian inhibitory factor or substance
Intracellular testosterone can arise from androstenedione (MIF or MIS) causing regression of the müllerian ducts.
(Δ4A), DHEA, or DHEAS. The desulfated DHEA is converted to The Leydig cells secrete testosterone causing differentiation
androstenedione by 3β-hydroxysteroid dehydrogenase (3β-OHD) and growth of the Wolffian duct structures. DHT causes
and androstenedione transformed into testosterone by growth of the prostate and penis and fusion of the labioscrotal
17β-hydroxysteroid dehydrogenase (17β-OHD). Testosterone,
folds (Figure 67–6).
DHT, and estradiol all bind to cytosolic steroid receptors. The
cytosolic androgen (and estrogen) receptor is complexed to
The key events involved in fetal development and sexual dif-
regulatory proteins (heat shock proteins). Hormone binding results ferentiation can be summarized as follows.
in the dissociation of the heat shock protein complex, dimerization
of the receptor, nuclear translocation, and DNA binding to Sex Determination
regulatory elements. The final result is the activation of gene
transcription. (Modified with permission from Molina PE: Endocrine Physiology, Mammalian sex determination leading to the development of
3rd ed. New York: McGraw-Hill Medical, 2010.) male or female phenotype involves three sequential processes:
• determination of the genetic sex of the embryo when an
X- or a Y-bearing sperm fertilizes the oocyte;
ple is the effect of testosterone on bones, where epiphysial
• determination of the fate of the bipotential or nondifferenti-
closure is mediated through osteoblast and chondroblast aro-
ated gonad and thus of gonadal sex;
matase conversion of testosterone to estradiol. The conver-
• differentiation of male or female internal and external geni-
sion of testosterone to DHT by 5α-reductase produces the
talia, or determination of phenotypic sex.
most potent natural androgen (Figure 67–4). Because andro-
gens stimulate the growth of prostate cancer cells and are also The determination of genetic sex is mediated through the
involved in benign prostatic hyperplasia, the inhibition of chromosomal set, which in the normal male is 46,XY. The sub-
enzymatic conversion of testosterone to DHT has been used sequent sexual differentiation is determined by genetic factors.
effectively as a target for pharmacologic interventions. Finas- One of the first genes involved in sexual differentiation is
teride and dutasteride, 5α-reductase inhibitors, are cur- on the Y chromosome and is called SRY (sex-regulating
rently used for the treatment of benign prostatic hyperplasia region of the Y). The product of the SRY gene is a protein that
and prostate cancer. stimulates the neutral gonadal tissues to differentiate into tes-
Testosterone, dehydroepiandrosterone (DHEA), and tes, thereby determining gonadal sex. SRY is necessary and
androstenedione are degraded primarily by the liver. sufficient to initiate the male development cascade. If the SRY
Biotransformation involves reduction to 17-ketosteroids con- gene is mutated or missing on the Y chromosome, the embryo
jugated (glucuronidation) prior to excretion (Figure 67–5). develops into a female.
688 SECTION IX Endocrine and Metabolic Physiology

LH

Mitochondria Cholesterol

StAR

Pregnenolone
P450 scc 3β-Hydroxysteroid dehydrogenase
Progesterone
17α-Hydroxylase
• Estrone
Androstenedione
• Epiandrosterone,
17β-HSD Androsterone,
Etiocholanolone
Testosterone

OH

FIGURE 67–5 Key steps in testosterone


biosynthesis and metabolism. Diagrammatic
representation of the typical biochemical 5α-Reductase
O OH
pathway and key enzymes involved in Leydig cell
steroidogenesis that facilitate the biosynthesis of Testosterone
testosterone from the precursor cholesterol. 3α-Hydroxysteroid
Testosterone finally diffuses out of the Leydig cell dehydrogenase
3α-Androstenediol
and reaches the interstitial space and the O
H (weak androgen)
peripheral circulation. In target cells, testosterone
can be converted to the most potent androgen, Dihydrotestosterone (DHT)
dihydrotestosterone (DHT), by 5α-reductase or to
17β-estradiol by aromatase. Testosterone,
dehydroepiandrosterone (DHEA), Aromatase
androstenedione, and 17β-estradiol are
degraded in the liver to 17-ketosteroids or polar
metabolites that are excreted in the urine. StAR, OH
steroidogenic acute regulatory protein; scc,
side-chain cleavage; HSD, hydroxysteroid
Estrone 16-OH-estrone Estriol
dehydrogenase; COMT, catechol-O-
methyltransferase. (Modified with permission from
Molina PE: Endocrine Physiology, 3rd ed. New York:
OH Catecholestrogen 2-Methoxyestrone
17β-Estradiol COMT
McGraw-Hill Medical, 2010.)

Sexual Differentiation gonads have differentiated into testes, the secretion of testicu-
lar hormones is sufficient to promote masculinization of the
Human male gonad differentiation begins in the sixth week of
embryo. The production of testosterone and MIF during a
gestation with the development of precursor Sertoli cells.
critical time in early gestation ensures male development. Ini-
These cells aggregate to form the seminiferous cords, which
tially, both the male (mesonephric or Wolffian) and female
are then infiltrated by primordial germ cells. By the end of the
(paramesonephric or müllerian) internal genital ducts are
ninth week, the mesenchyme that separates the seminiferous
present. In females, the mesonephric ducts regress and the
cords gives rise to the interstitial cells, which differentiate as
paramesonephric ducts develop into the uterine tubes, uterus,
steroid-secreting Leydig cells. Gonadotropic control of fetal
and upper vagina. In males, starting at the eighth week of ges-
testicular steroidogenesis is mediated initially by placenta-
tation, MIF mediates the regression of the paramesonephric
derived human chorionic gonadotropin (hCG) and later by
or müllerian ducts. The mesonephric duct system (Wolffian
fetal LH. The resulting increase in fetal testosterone produc-
ducts) remains and forms the vas deferens, epididymis, and
tion stimulates Leydig cell proliferation, increases the expres-
seminal vesicle. This process is dependent primarily on tes-
sion of steroidogenic enzymes, and increases the expression of
tosterone. In the female, in the absence of androgens, the
the androgen receptor in the target tissues.
Wolffian ducts regress and the müllerian ducts are spared
The postgonadal phase of sexual differentiation or external
from apoptosis, developing into the uterus, fallopian tubes,
genitalia differentiation is hormone dependent. Once the
CHAPTER 67 Male Reproductive System 689

TABLE 67–2 Specific actions of testosterone, dihydrotestosterone, and estradiol.


Testosterone DHT (5α-Reductase Activity) 17β-Estradiol (Aromatase Activity)

Embryonic development of Wolffian duct–derived structures Embryonic development of the prostate Epiphyseal closure

Postpubertal secretory activity Descent of the testes Prevention of osteoporosis

Pubertal growth of larynx and deepening of voice Phallic growth Feedback regulation of GnRH secretion

Anabolic effects on muscle and erythropoiesis Male pattern balding

Inhibition of breast development Development of pubic and underarm hair

Stimulation of spermatogenesis; libido Activity of sebaceous glands

Feedback inhibition of GnRH, LH, and FSH release


DHT, dihydrotestosterone; GnRH, gonadotropin-releasing hormone. The enzyme activities that metabolize testosterone to DHT or estradiol are in parentheses.

and vagina. Estrogens do not appear to be essential for nor- sexual differentiation and is completed during the late gesta-
mal sexual differentiation of either sex, as shown by normal tional period, and is mediated by testosterone and insulin-like
genital development in males with a mutant estrogen receptor peptide 3. In humans, failure of complete testicular descent
gene or with aromatase deficiency. The differentiation of into the scrotum (cryptorchidism) is one of the most common
external male genitalia is regulated particularly through the congenital abnormalities, involving approximately 3% of male
actions of DHT. births. Testicular descent into the scrotum is important because
Following müllerian duct regression and androgen- spermatogenesis requires lower temperatures (as in the scro-
dependent virilization of the urogenital system, the testes tum) than those found intra-abdominally. If untreated, cryp-
migrate from their site of origin next to the kidney into the torchidism can lead to infertility, and it has been associated
scrotum. This is the critical and final event involved in male with an increased risk of testicular tumors.

Epididymis,
Wolffian ducts seminal vesicles,
Sex vas deferens
determination Prostate & penis growth
SRY Testosterone Fusion of labioscrotal folds
Urogenital Leydig
ridge Bipotential Testis Insl3
cells Testicular descent
gonad
Sertoli MIF
cells
Uterus, fallopian
Mullerian ducts tubes, cervix,
vagina

Sertoli cell Leydig cell


activity activity

MIF Testosterone Testes descent


Insl3 Male external genital External genital
differentiation growth
Wolffian duct differentiation
Germ cell Mullerian duct
migration regression
4 5 6 7 8 9 10 11 12 13 14 15 40
Gestational period (weeks)

FIGURE 67–6 Male sexual differentiation. The bipotential gonad is differentiated into testes by the sex-determining region gene on the Y
chromosome (SRY). This period of sex determination is followed by gonad differentiation of the different cell types of the testis. The Sertoli cells of
the testis secrete müllerian-inhibiting factor (MIF). The Leydig cells produce testosterone and insulin-like growth factor 3. MIF produces regression
of the müllerian ducts. Testosterone stimulates the growth and differentiation of the Wolffian ducts, and growth of the penis and prostate. Insl3
participates in testicular descent, the final step in male sexual development. DHT produced from testosterone also participates in testis descent
and development of the prostate. (Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)
690 SECTION IX Endocrine and Metabolic Physiology

Sexual Maturation and Function gradual decline in circulating testosterone levels, followed at
age 50 by a reduction in sperm production. This period of
Puberty
gradual androgen decline is called the andropause and is
Puberty is the physiologic transition between childhood and characterized by diminished sexual desire and erectile capac-
adulthood and involves the development of fertility, secondary ity, decreased intellectual activity, fatigue, depression,
sexual characteristics and the pubertal growth spurt. The decreased lean body mass, skin alterations, decreased body
process takes place over a period of about 4 years. Puberty is hair, decreased bone mineral density resulting in osteoporosis,
triggered by increased pulsatile secretion of GnRH by the hypo- and increased visceral fat and obesity.
thalamus, leading to increases in serum gonadotropins and,
thus, to increases in gonadal secretion of sex steroids. The hypo-
thalamic–pituitary–gonadal system is active during the neona-
tal period but enters a dormant state in the juvenile, prepubertal FERTILITY AND SECONDARY SEXUAL
period. During the initial phase of puberty, plasma levels of LH CHARACTERISTICS
increase primarily during sleep. These sleep-associated surges
are later present throughout the day and mediate or result in an Spermatogenesis
increase in circulating testosterone levels. Spermatogenesis is the process of continuous germ cell dif-
Puberty is preceded by adrenarche, a period characterized by ferentiation to produce spermatozoa (Figure 67–7). Spermato-
increased adrenal production of DHEA and androstenedione at genesis is initiated at the time of puberty and is associated with
around 6–8 years of age. The peak concentrations of DHEA and the transition from a relatively hypogonadotropic state in the
androstenedione are reached during late puberty and early adult- prepubertal phase of development to the eugonadotropic state
hood. During this stage, there is some conversion of adrenal- in adulthood. It is compartmentalized within the blood–testis
derived androgens to testosterone, resulting in a small increase in barrier. It is under FSH regulation of the Sertoli cell and
circulating testosterone levels. The signal that triggers the increase requires testosterone production from the Leydig cells. Sper-
in the synthesis of DHEA and androstenedione is not known. matogenesis involves four basic processes:
The increase in pulsatile release of GnRH from the hypothal-
• Proliferation of spermatogonia (stem cells) giving rise to
amus into the hypophysial-portal blood is essential for the onset
spermatocytes (diploid cells)—Spermatogonia, derived
of puberty and is thought to be facilitated by leptin, a hormone
from primordial germ cells, line the seminiferous tubule near
secreted from adipose tissue. The increase in the amplitude of
the basement membrane. One or two divisions of sper-
GnRH pulses triggers a cascade of events including increases in
matogonia occur to maintain their population in a stem cell
the amplitude of FSH and LH pulses, followed by marked
pool, some cells stay in the “resting” pool, whereas others
increases in gonadal sex hormone production. As a result of the
proliferate several times and undergo 1–5 stages of division
increase in testosterone production during puberty, growth
and differentiation becoming spermatocytes (Figure 67–7).
hormone release is increased. Together, growth hormone and
The “resting” or stem cell spermatogonia remain dormant
gonadal steroids are responsible for normal pubertal growth
for a time and then join a new proliferation cycle of sper-
spurt during which the growth velocity increases from 4–6 cm
matogonia. These cycles of spermatogonial divisions occur
per year to as much as 10–15 cm per year. The physiologic
before the previous generation of cells has completed sper-
changes associated with puberty are summarized as follows:
matogenesis, so that multiple stages of the process are occur-
• Leydig cell maturation and initiation of spermatogenesis; ring simultaneously in the seminiferous tubules. This overlap
• testis enlargement, and reddening and wrinkling of scrotal ensures a residual population of spermatogonia that main-
skin; tain the ability of the testis to continuously produce sperm.
• pubic hair growth from base of the penis; • Meiosis of spermatocytes to yield spermatids (haploid
• penis enlargement; cells; 23 chromosomes): The primary spermatocytes un-
• prostate, seminal vesicle, and epididymis growth; dergo two divisions; the first meiotic division produces two
• facial (mustache and beard) and extremities hair growth, re- secondary spermatocytes. Division of the secondary sper-
gression of scalp line; matocytes completes meiosis and produces the spermatids.
• larynx enlargement, thickening of the vocal cords, and • Spermiogenesis, or maturation and development of sper-
deepening of the voice; matids into spermatozoa (sperm): This phase is character-
• enhanced linear growth; ized by nuclear and cytoplasmic changes that provide
• increased muscle mass and hematocrit; spermatozoa with key elements for their function, including
• increased libido and sexual potency. formation of the acrosome, as summarized in Table 67–3.
• Spermiation: This is the final process of release of mature
Maturity and senescence sperm from the Sertoli cells into the tubule lumen and in-
Sexual maturity in males is achieved at approximately age volves movement of the germ cells from the basal to the
16–18 years. During this stage, sperm production is optimal, adluminal region of the seminiferous tubule into the com-
plasma gonadotropins are normal, and most sexual anatomic partment protected by the blood–testis barrier. The mitotic
changes have been completed. Beginning at age 40, there is a phase occurs in the basal compartment, whereas the meiotic
CHAPTER 67 Male Reproductive System 691

Head
Tail sheath Nucleus
Spermatogonia Centrioles
Mitosis Mitochondria
Acrosome
Flaggellum
Primary spermatocyte Midpiece

First meiotic Tail


70 days

division
Secondary Spermatozoa
Second spermatocyte
meiotic Lumen of tubule
division
Spermatids

Spermatozoa
Spermatids Residual body

Secondary
Spermatocyte

Primary
Spermatocyte

Spermatogonia

FIGURE 67–7 Schematic representation of key events in spermatogenesis. The process of spermatogenesis involves proliferation
(mitosis) of spermatogonia, producing primary spermatocytes (diploid cells; 46 chromosomes). Spermatocytes undergo two meiotic divisions
to yield spermatids, or haploid cells (23 chromosomes). Spermatids undergo a process of maturation (spermiogenesis) and development into
spermatozoa. During this last phase, spermatozoa acquire the key elements for their function (Table 67–3). This continuous process takes about
70 days. At any given time, cells from all steps of spermatogenesis can be identified in the testes. (Modified with permission from Junqueira LC, Carneiro J:
Basic Histology: Text & Atlas, 11th ed. McGraw-Hill, 2005.)

and postmeiotic phases occur in the luminal compartment. The complete maturation of sperm and the development of
The overall results of spermatogenesis are the following: cell its capacity to fertilize the ovum are a function of additional
proliferation and maintenance of a reserve germ cell popu- processes occurring after sperm is released from the Sertoli
lation, reduction in chromosome number and genetic varia- cells. These processes are a function of the accessory glands and
tion through meiosis, and production of spermatozoa.

Regulation of Spermatogenesis TABLE 67–3 Key events in spermiogenesis and their


Spermatogenesis is dependent on gonadotropin stimulation functional importance in sperm function.
and testosterone production. FSH stimulates proliferation
Key Event Functional Importance
and secretory activity of the Sertoli cells, whereas LH stimu-
lates the production of testosterone. Testosterone in turn is a Nuclear chromatin condensation Haploid chromatin carries either
paracrine stimulator of spermatogenesis through receptor- X or Y chromosome
mediated events in the Sertoli cells. The LH-induced increase Acrosome development The acrosome is a large secretory
in intratesticular testosterone plays an essential role in the vesicle that overlies the nucleus
in the apical region of the sperm
initiation and maintenance of spermatogenesis by the Sertoli head and contains enzymes
cells. Androgens produced by the Leydig cells bind to needed for mucus penetration
androgen-binding globulin (ABG), a protein produced by and fertilization
the Sertoli cell in response to FSH stimulation and secreted Repositioning of spermatids; Microtubular structure provides
into the lumen of the seminiferous tubules. Testosterone is development and growth of motility, allowing sperm
produced by Leydig cells, which are interstitial cells that flagellum movement (3 mm/min) through
reside adjacent to the seminiferous tubules. Sertoli cells the genital tract
require the presence of testosterone for spermatogenesis, Formation of mitochondrial Provides energy (fructose-derived
underscoring the importance of paracrine mechanisms of sheath around flagellum ATP) for flagellar movement
hormone action. ATP, adenosine triphosphate.
692 SECTION IX Endocrine and Metabolic Physiology

the excretory system, summarized in Table 67–1. The develop- choline, stimulating the production of constitutive endothelial
ment and function of these organs are androgen dependent. nitric oxide. Nitric oxide produced locally in the smooth mus-
cle cell, or reaching it by diffusion from the adjacent endothe-
lial cells, is the major mediator of smooth muscle relaxation
ANABOLIC AND METABOLIC through activation of guanylate cyclase and increased
EFFECTS OF ANDROGENS production of cyclic guanosine monophosphate (cGMP). The
inhibition of cGMP phosphodiesterase (PDE5), the enzyme
In bone, the main physiologic effect of testosterone is to reduce that degrades cGMP, with drugs such as sildenafil preserves
bone resorption. This action is mediated both by direct effects smooth muscle relaxation and prolongs the erection period.
of testosterone on the androgen receptor and by localized aro- This is used commercially to treat erectile dysfunction.
matization of testosterone to estrogen. Testosterone increases The ejaculation phase of the sexual response consists of two
osteoblast lifespan and proliferation. Testosterone-derived sequential processes: emission and ejaculation. Emission is
estrogen is the critical sex hormone in the pubertal growth the deposition of seminal fluid into the posterior urethra and is
spurt, skeletal maturation, accrual of peak bone mass, and mediated by simultaneous contractions of the ampulla of the
maintenance of bone mass in the adult. It stimulates chondro- vas deferens, the seminal vesicles, and the smooth muscles of
genesis in the epiphysial growth plate, increasing pubertal linear the prostate. The second process is ejaculation, which results in
growth. At puberty, estrogen promotes the gradual, progressive expulsion of the seminal fluid from the posterior urethra
closure of the epiphysial growth plate and the termination of through the penile meatus. This process is controlled by sym-
chondrogenesis. In the adult, estrogen is important in main- pathetic innervation of the genital organs and occurs as a result
taining the constancy of bone mass through its effects on of a spinal cord reflex arc. Detumescence of the penis following
remodeling and bone turnover. Testosterone decreases osteo- ejaculation, and maintenance of the flaccid penis in the absence
blast and osteoclast apoptosis; stimulates osteoblast prolifera- of sexual arousal, is produced by sympathetic corporeal vaso-
tion, enhancing bone formation; and increases periosteal constriction and corporeal smooth muscle contraction by
apposition of bone. It increases protein synthesis and decreases noradrenergic, neuropeptide Y, and endothelin-1 fibers.
protein breakdown, having an overall anabolic effect in muscle.
Testosterone inhibits lipid uptake and lipoprotein lipase
activity in adipocytes, stimulates lipolysis by increasing the
number of lipolytic β-adrenergic receptors, and inhibits dif- DISEASES OF TESTOSTERONE
ferentiation of adipocyte precursor cells. DHEA stimulates
resting metabolic rate and lipid oxidation and enhances glu-
EXCESS OR DEFICIENCY
cose disposal by increasing the expression of glucose trans- Excess androgen activity in childhood leads to precocious
porters on the plasma membrane of adipocytes. puberty, defined in boys as the appearance of male secondary
sex characteristics before age 9 (Table 67–2). Hypothalamic
tumors, activating mutations of the LH receptor, congenital
NEUROENDOCRINE AND adrenal hyperplasia, and androgen-producing tumors are all
causes of premature virilization. Decreased testosterone pro-
VASCULAR CONTROL OF duction, or hypogonadism, can be caused by disorders at the
ERECTION AND EJACULATION hypothalamic/pituitary level (hypogonadotropic or secondary
hypogonadism) or by testicular dysfunction (hypergonadotro-
The physiologic process of human reproduction involves the pic or primary hypogonadism).
fertilization of a mature ovum through the deposition of Hypogonadotropic hypogonadism can be due to abnormali-
sperm-containing semen in the vagina of the female. This ties in hypothalamic GnRH secretion or impaired gonadotro-
event involves penile erection and ejaculation of the sperm- pin secretion by the anterior pituitary. This condition may result
containing semen at the time of copulation. Penile erection from genetic defects including Kallmann syndrome; abnormal-
results from smooth muscle relaxation mediated by a spinal ities of the GnRH receptor, LH, or FSH β-subunits; pituitary
reflex involving central nervous processing and integration of tumors (including prolactinomas); trauma; or surgery.
tactile, olfactory, auditory, and mental stimuli. Corporeal Abnormal testicular function in the presence of elevated
vasodilatation and corporeal smooth muscle relaxation allow gonadotropin levels (hypergonadotropic or primary hypogo-
increased blood flow into the corpus cavernosum. Corporeal nadism) is caused by testicular damage or impaired testicular
vasodilatation is mediated by the parasympathetic nervous development, which can be either congenital or acquired follow-
system. The concordant contraction of the perineal skeletal ing chemotherapy or radiation. Causes include cryptorchidism,
muscles leads to a temporary increase in corpus cavernosum gonadal dysgenesis, varicocele, enzyme defects in testosterone
blood pressure above mean systolic arterial pressure, helping biosynthesis, or LH receptor defects. Klinefelter syndrome is a
to increase penile firmness. sex chromosome disorder, in which affected males carry an
Parasympathetic fibers directly innervating the corporeal additional X chromosome. This genetic abnormality results in
smooth muscle and sinusoidal endothelial cells release acetyl- male hypogonadism, androgen deficiency, and impaired sper-
CHAPTER 67 Male Reproductive System 693

matogenesis. Klinefelter’s syndrome is the most common genetic CHAPTER SUMMARY


cause of human male infertility. Hyperprolactinemia from any
■ The Sertoli cells support spermatogenesis and form the
cause results in both reproductive and sexual dysfunction
blood–testis barrier.
because of prolactin inhibition of GnRH, FSH, and LH release,
■ Testicular function is under LH and FSH regulation.
resulting in hypogonadotropic hypogonadism.
■ LH and FSH production and release are under hypothalamic
GnRH stimulation and feedback inhibition by gonadal hormones.
CLINICAL PRESENTATION ■ The main functions of the testes are testosterone production
and spermatogenesis.
Excess testosterone in the prepubescent male is associated ■ The main hormones produced by the testis are testosterone,
with the appearance of all the changes of puberty at a very estradiol, and inhibin.
early age. These changes include enlargement of the penis and ■ Testosterone can be metabolized to DHT, a more potent
testicles; appearance of pubic, underarm, and facial hair; spon- androgen, or to 17β-estradiol, an estrogen.
taneous erections; production of sperm; development of acne; ■ Androgens exert their physiologic effects through modulation
and deepening of the voice. of gene transcription.
Low levels of testosterone lead to symptoms, which differ ■ Three essential testis-derived hormones regulate male sexual
according to the time of onset. Androgen deficiency during development: androgens, MIF, and insulin-like peptide 3.
puberty results in lack of pubertal growth spurt, lack of deepen-
ing of the voice, female distribution of secondary hair, anemia,
underdeveloped muscles, and genitalia with delayed or absent STUDY QUESTIONS
onset of spermatogenesis and sexual function. Hypogonadism, 1. A 20-year-old male patient presents to the doctor’s office
as well as aromatase deficiency and the inability to synthesize complaining about continuous growth, lack of facial hair
estradiol, result in lack of epiphyseal closure and continued development, and a smaller penis and testicles than his college
growth. Androgen deficiency in the adult after normal viriliza- friends. Laboratory values include low total testosterone and
tion has been completed leads to a decrease in bone mineral low LH. Thyroid-stimulating hormone and prolactin levels are
density (bone mass), decreased bone marrow activity resulting normal. He has no history of medications, drug use, or disease.
in anemia, alterations in body composition associated with Continued growth in this case is due to
muscle weakness and atrophy, changes in mood and cognitive A) increased estrogen production
function, and regression of sexual function and spermatogen- B) decreased inhibin release
esis. In the adult male, androgen deficiency decreases noctur- C) decreased testosterone production
nal erections and libido. D) decreased sensitivity to LH stimulation
2. In the patient described above, it was observed that in addition
to being much taller than young adults his age, his arms were
very long. The excessive height and arm length resulting from
delayed epiphyseal growth plate closure is due to
CLINICAL CORRELATION A) increased DHT formation
A 33-year-old father of two healthy children and his wife B) decreased adrenal DHEA production
are referred for evaluation more than a year after failure to C) decreased estradiol production
D) increased estriol production
conceive despite being sexually active and not taking any
contraceptive measures. On physical examination, the 3. A college football player purchases online natural hormone
analogs to increase his muscle mass. After a year of hormone
husband’s external genitalia are normal, but testes are
analog injections, his muscle mass increases significantly, and
smaller and softer than normal for his age. He is very mus-
he develops acne. Consultation with the team’s physician leads
cular and admits to being a body builder. Blood labora- to a complete physical exam that reveals small testicular size.
tory values show low testosterone levels, low LH and FSH, Being a newlywed, he wants to determine his fertility status and
and normal prolactin levels. Semen analysis shows low test results show low sperm count. Which of the following is the
sperm count (oligospermia). A diagnosis of anabolic ste- underlying mechanism for these manifestations?
roid-induced hypogonadotropic hypogonadism is made. A) increased conversion of DHT to testosterone.
Anabolic steroids are androgen hormone analogs that B) suppression of LH release from the pituitary
produce negative feedback inhibition of LH and FSH release. C) increased testicular testosterone concentrations
This in turn decreases testicular testosterone production D) increasing aromatase activity
and spermatogenesis. Some anabolic steroids that are 4. A 15-year-old male, underweight, high school student is brought to
abused by athletes are not aromatizable so that they are not the family physician due to what appears to be a delay in the onset
converted to estrogen and are different enough from native of puberty. The most likely mechanism underlying this defect is
testosterone to not be detected in the standard testosterone A) decreased aromatase activity
assays. Cessation of anabolic hormone utilization restores B) increased DHT production
hypothalamic–gonadal function in most of the cases. C) decreased leptin production
D) increased testosterone production
This page intentionally left blank
68
C H A P T E R

Female Reproductive
System
Patricia E. Molina

O B J E C T I V E S

■ Describe oogenesis, its relationship to follicular maturation, and the roles of


pituitary and ovarian factors in their regulation.
■ Describe the roles of pituitary hormones in regulation of ovarian function
including ovulation, formation and decline of the corpus luteum, and
estrogen and progesterone biosynthesis and secretion.
■ List the target organs and principal physiologic actions of estrogen and
progesterone and how they interact.
■ Identify the transport mechanisms and degradation pathways for estrogen
and progesterone.
■ Describe the cellular mechanisms of action for estrogen and progesterone.
■ Describe the endometrial changes (proliferative and secretory phases) that
occur throughout the menstrual cycle, and correlate them with the changes
in blood levels of pituitary and ovarian hormones.
■ Identify the pathways of sperm and egg transport required for fertilization
and for movement of the embryo to the uterus.
■ Describe the endocrine functions of the placenta, particularly the role of
placental hormones in rescue of the corpus luteum and maintenance of
pregnancy, and the fetal adrenal–placental interactions involved in estrogen
production.
■ Understand the roles of oxytocin, relaxin, and prostaglandins in the initiation
and maintenance of parturition.
■ Explain the hormonal regulation of mammary gland development during
puberty, pregnancy, and lactation, and explain the mechanisms that control
milk production and secretion.
■ Explain the physiologic basis for the effects of steroid hormone contraceptive
methods.
■ Describe the age-related changes in the female reproductive system,
including the mechanisms responsible for these changes, throughout life
from fetal development to senescence.

The principal functions of the female reproductive system are regulates the hypothalamic–pituitary–gonadal axis in a
to produce the ova for fertilization by sperm, and to provide classical negative feedback pattern. In addition, the ovarian
the appropriate conditions for embryo implantation, fetal cycle mediates the maturation and development of the repro-
growth and development, and birth. Endocrine regulation of ductive system throughout life. Throughout the cycle, a
the reproductive system is directed by the hypothalamic– selected ovarian follicle is rescued from its apoptotic fate to
pituitary–ovarian axis. The ovarian cycle, which involves undergo growth and development, culminating in ovulation.
changing patterns of hormone production and secretion, The remnant of the follicle undergoes transformation into the

695

Ch68_695-714.indd 695 11/30/10 4:06:12 AM


696 SECTION IX Endocrine and Metabolic Physiology

corpus luteum, a temporary endocrine structure that plays a under hormonal regulation. The ovaries are the principal
central role in preparation for, and maintenance of the initial female reproductive organs, and their functions are the stor-
stages of pregnancy. Parallel changes in endometrial morphol- age and release of the ovum and the production of the two
ogy and function (the menstrual cycle) occur throughout the principal female sex steroid hormones, estrogen and proges-
ovarian cycle in preparation for implantation of a fertilized terone. The ovaries consist of an outer cortex layer that
ovum. Ovarian and placental hormones maintain pregnancy contains different-sized follicles and their apoptotic remnants
and prepare the breast for lactation. embedded in connective tissue and an inner medulla
containing vascular connective tissue. The primordial follicle
contains a primary oocyte surrounded by epithelial pregranu-
FUNCTIONAL ANATOMY losa cells separated from the ovarian stroma by a basement
membrane. During follicular development, the epithelial cells
The female reproductive organs include the ovaries, the uterus differentiate into granulosa cells, and a layer of cells from the
and fallopian tubes, and the breasts or mammary glands ovarian stroma is transformed into theca cells. The larger,
(Figure 68–1). Their growth, development, and function are more mature follicles are filled with a transparent fluid and

A
Ampulla Fallopian
isthmus tube
Infundibulum Fundus of uterus
Fimbriae

Oviduct

Fimbriae
FIGURE 68–1. Functional anatomy of the Ovary
female reproductive tract. A) The female Ovarian ligament
reproductive organs include the ovaries, the Broad ligament Ovary
uterus and Fallopian tubes, and the breasts or Uterus
Perimetrium
mammary glands. The ovaries consist of an outer Layers of Myometrium Cervical canal
cortex layer that contains different-sized follicles uterus Endometrium
Vagina
and their remains undergoing apoptosis,
imbedded in connective tissue. The Fallopian tubes B
extend from each of the superior angles of the Primary
Mesovarium follicle Early antrum
uterus and consist of an isthmus, ampulla, and the Stroma formation
Primordial
infundibulum, which opens into the abdominal follicle Atretic
cavity and is surrounded by ovarian fimbria and follicle
attached to the ovary. The cilia of the epithelial
Graafian
lining of the Fallopian tubes contribute to the follicle
orientation of sperm movement, aiding in Blood
fertilization as well as facilitating the movement vessels
of the zygote (fertilized ovum) to the uterus for Corpus
implantation and fetal development. B) The albicans
ovary contains oocytes at different stages of Ovulation
development. Following ovulation, the follicle is Germinal
converted to the corpus luteum that contributes epithelium Ovary
to hormone production during the early pregnancy
period. C) The breast is organized in lobes made Mature corpus Early corpus
luteum Ductules or acini
of lobules, connected together by connective luteum Terminal duct
Terminal duct
lobular unit Lobule
tissue, blood vessels, and ducts. The lobules consist
of a cluster of rounded alveoli, which open into C
excretory lactiferous ducts and unite to form Breast Adipose tissue
larger ducts made of longitudinal and transverse Segmental duct
elastic fibers. These ducts converge toward the
Lactiferous duct
areola, beneath which they form ampular
Lactiferous sinus
dilatations, which serve as reservoirs for the milk.
Nipple
(A. Modified with permission from Molina PE: Endocrine
Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010. Fat
B. Modified with permission from Curtis O Byer, Louis W Lobule Alveoli
Shainberg, Grace Galliano, Louis Shainberg. Dimensions in unraveled Lactiferous Fat
Human Sexuality. 6th ed. McGraw-Hill; 2001. C. Modified Lobules Ampulla tubule
Duct
with permission from Gray H. Anatomy of the Human Body.
20th ed; 1918.)
CHAPTER 68 Female Reproductive System 697

consist of an external fibrovascular coat, connected with the follicular phase of the ovarian cycle.
surrounding stroma of the ovary by a network of blood vessels; • Luteal phase: The peak in LH immediately precedes ovula-
and an internal coat, consisting of several layers of nucleated tion and corpus luteum formation. The rise in LH results in
cells (granulosa cells) anchored in the zona pellucida, a progesterone and estrogen production by the corpus luteum
glycoprotein-rich eosinophilic material surrounding the during the early and midluteal phases of the menstrual
oocyte. The zona pellucida forms the corona radiata, which, cycle. Activation of the LH receptors in theca cells stimu-
close to the time of ovulation, is separated from the granulosa lates androstenedione production, providing the substrate
cells and expelled with the oocyte. Formation of the follicles for the enzymatic conversion to 17β-estradiol that is medi-
begins before birth, and their development and maturation ated by the enzyme aromatase in granulosa cells.
continue uninterrupted from puberty to the end of a woman’s
reproductive life, as discussed later.
The female genital tract is derived from the müllerian ducts OVARIAN HORMONE SYNTHESIS
and consists of the uterus, fallopian tubes, and vagina. The
fallopian tubes extend from each of the superior angles of the ESTROGEN
uterus and consist of the isthmus, the ampulla, and
the infundibulum, which opens into the abdominal cavity and The production of estrogen involves coordinated enzymatic
is surrounded by ovarian fimbriae and attached to the ovary activities between the theca and granulosa cells of the ovar-
(Figure 68–1). The epithelial lining of the fallopian tubes has ian follicle (Figure 68–2). During follicle growth, theca cells
secretory and ciliated cells that contribute to sperm movement, express the enzymes necessary to convert cholesterol to andro-
aiding in fertilization and facilitating the movement of the gens (mainly androstenedione, a weak androgen) but lack the
zygote (fertilized ovum) to the uterus for implantation and enzymes necessary to convert androgens to estradiol. Granu-
fetal development. These functions are also aided by the losa cells can convert androgens to estradiol and they can pro-
rhythmic contraction of the smooth muscle walls. duce progesterone, but they are unable to convert pregnenolone
The uterus is composed of a thick muscular coat and a or progesterone to androgens. Thus, theca cell–produced
mucous membrane, or endometrium, lined by columnar androgens diffuse into the granulosa cells where they are aro-
ciliated secretory epithelium. The endometrium undergoes matized to estradiol (Figure 68–2). More than 95% of circulat-
cycles of regeneration (proliferative phase), differentiation ing estradiol is directly secreted, with a smaller contribution
(secretory phase), and shedding (menstrual phase) from peripheral conversion of estrone to estradiol in premeno-
approximately every 28 days in preparation for embryo pausal women.
implantation.
The breast consists of glandular tissue organized in lobes ANDROGENS
connected by fibrous tissue, with fat deposits interspersed
between the lobes (Figure 68–1). The lobules open into excre- Female androgens are derived from the adrenal glands (dehy-
tory lactiferous ducts and unite to form larger ducts that con- droepiandrosterone and androstenedione), the ovaries
verge toward the areola, beneath which they form ampullar (androstenedione and testosterone), and peripheral conver-
dilatations, which serve as reservoirs for the milk. sion of androstenedione and dehydroepiandrosterone to tes-
tosterone. Ovarian androgen secretion parallels that of
estrogen throughout the menstrual cycle. Most of the circulat-
ing testosterone in females is derived from the peripheral con-
GONADOTROPIN REGULATION version of androstenedione; 30–40% of testosterone is directly
OF OVARIAN FUNCTION secreted. The conversion of testosterone to dihydrotestoster-
one in peripheral tissues is limited in females because of their
Pulsatile release of gonadotropin-releasing hormone (GnRH) higher levels of sex hormone–binding globulin (SHBG) than
from the hypothalamus stimulates pulsatile pituitary release of in males, as well as by the peripheral conversion to estrogen by
luteinizing hormone (LH) and follicle-stimulating hormone aromatase, protecting females from virilization by dihydrotes-
(FSH). Both FSH and LH stimulate ovarian production of tosterone.
estradiol and progesterone through binding to a transmem-
brane G protein–coupled receptor. Estradiol and progester-
one are the two principal ovarian steroid hormones involved PROGESTERONE
in the regulation of ovarian function and control of the repro-
The preovulatory LH surge results in luteinization of granu-
ductive cycle. The variations in pulsatile release of the gonado-
losa and theca cells, altering the steroidogenic pathway so that
tropins result in a cyclic response of ovarian function. Each
progesterone is the primary steroid hormone produced by
cycle lasts approximately 28 days and can be divided into two
each of these cell types after luteinization. The changes leading
phases (follicular and luteal) of approximately 14 days each:
to the ability to produce progesterone include increased
• Follicular phase: FSH is responsible for follicular recruit- expression of the enzymes involved in the conversion of cho-
ment and growth and for estrogen synthesis during the lesterol to progesterone (cholesterol side-chain cleavage
698 SECTION IX Endocrine and Metabolic Physiology

Theca Cell LH/CG receptor

PI-4, 5P2
AC Gs Gq PLCβ
ATP
βγ βγ

cAMP DAG IP3


Ca2+

PKA PKC

Protein
P Protein P Protein

FIGURE 68–2 Theca and granulosa cells


coordinate the production of estrogen. The
cAMP/PKA pathway DAG/PKA pathway
secretion of estradiol by the dominant follicle
requires cooperation between theca cells, which
synthesize androstenedione and testosterone;
Cholesterol Progesterone Androstenedione
and granulosa cells of mature follicles, which
P450scc P450C17α
convert androgens to estradiol and estrone.
Androgen synthesis in theca cells results from Androstenedione Systemic circulation
the activity of three enzymes: cholesterol
side-chain cleavage (P450scc), 17α-hydroxylase- Basal lamina

lyase (P450C17), and 3β-hydroxysteroid


dehydrogenase (3β-HSD). Luteinizing hormone Androstenedione 17β-Estradiol
(LH) stimulates the steroidogenic pathway
leading to formation of androstenedione in the Aromatase
theca cells. In granulosa cells, the enzyme
Protein P Protein
17β-hydroxysteroid dehydrogenase transforms
androstenedione into testosterone in follicles
from the primary stage. In mature follicles, PKA
follicle-stimulating hormone (FSH) stimulates
the activity of aromatase, which transforms
testosterone into 17β-estradiol. AC, adenylate cAMP
cyclase; PLCβ, phospholipase Cβ; PI-4,5P2,
phosphatidylinositol 4,5-bisphosphate; ATP, ATP
βγ
adenosine triphosphate; cAMP, cyclic adenosine AC
Gαs
monophosphate; DAG, diacylglycerol; IP3,
Follicular fluid
inositol 1,4,5-trisphosphate; PKA, protein kinase
A; PKC, protein kinase C. (Modified with permission Granulosa Cell FSH receptor
from Molina PE: Endocrine Physiology, 3rd ed. New York:
McGraw-Hill Medical, 2010.)

cytochrome P450 complex and 3β-hydroxysteroid dehydro- both inhibin B and inhibin A. Inhibin B is a good marker of
genase) and decreased expression of the enzymes that convert granulosa cell function under the control of FSH, whereas
progesterone to estrogens (17α-hydroxylase cytochrome P450 inhibin A is a marker of corpus luteum function under the
and aromatase cytochrome P450). control of LH. Inhibins contribute to the regulation of LH
and FSH release through endocrine feedback regulation at
INHIBINS, ACTIVINS, AND FOLLISTATIN the anterior pituitary.
Activin production by the granulosa cells changes during
Inhibin production by granulosa cells of mature follicles is folliculogenesis. Activin promotes granulosa cell prolifera-
regulated by FSH and LH, and by local factors such as growth tion, upregulates FSH receptor expression on granulosa
factors (epidermal, transforming, and insulin-like) and cells, and modulates steroidogenesis in both granulosa and
hormones (androstenedione, activin, and follistatin) in an theca cells. Activin is a physiologic antagonist to inhibin
autocrine and paracrine way. Preantral follicles secrete and specifically stimulates pituitary FSH synthesis and
inhibin B exclusively, whereas small antral follicles secrete secretion.
CHAPTER 68 Female Reproductive System 699

OVARIAN CYCLE Following ovulation, as the corpus luteum develops and


synthesis and circulating levels of progesterone increase,
During the ovarian cycle, throughout the reproductive life negative feedback inhibition of LH release is produced. With
span, a single mature oocyte is usually produced each month. the decline in LH levels comes the demise of the corpus luteum,
Most of the human oocytes (germ cells) present during pre- unless the corpus luteum is stimulated by human chorionic
natal development are destined to undergo atresia through gonadotropin (hCG), a placental hormone (described later) it
apoptosis, or programmed cell death. Only follicles that are regresses.
responsive to FSH stimulation (about 350) will enter the
final stage of development and progress to ovulation. The
ovarian cycle is divided into a follicular and a luteal phase. OOGENESIS AND FORMATION
The follicular phase begins on day 1 of the cycle, the first OF THE DOMINANT FOLLICLE
day of menses, and corresponds to the growth and develop-
ment of a dominant follicle. At midcycle (day 14), the increas- The fetal ovary begins germ cell development (oogenesis)
ing levels of estrogen stimulate a surge in LH release (positive early in fetal life. During early intrauterine development
feedback), which results in ovulation 36 hours later. (15 weeks), primordial germ cells (oogonia) proliferate and
After ovulation, during the luteal phase, the remnants of migrate to the genital ridge. On their arrival in the fetal ovary,
the ovulatory follicle form the corpus luteum, a transient some of the oogonia continue mitotic proliferation, while oth-
endocrine structure that produces estradiol, progesterone, ers begin to undergo apoptosis (Figure 68–5). Some of these
and inhibin A and is under LH regulation. The end of the oogonia begin the first meiotic division but arrest at the diplo-
luteal phase is marked by regression of the corpus luteum tene stage (end of prophase) at or near birth. The resulting
and associated decreases in estradiol, inhibin A, and proges- primary oocytes remain arrested in the first meiotic prophase
terone production. The hormonal changes that occur during until puberty when they are recruited to grow and mature and
the ovarian cycle produce differential regulation of gonado- undergo meiosis to produce an ovum.
tropin release. Primary oocytes are surrounded by a primordial follicle
consisting of a single layer of epithelial granulosa cells and a
less organized layer of mesenchymal theca cells. The pool of
OVARIAN REGULATION OF primordial follicles in the female ovary reaches its maximum
GONADOTROPIN RELEASE number at about 20 weeks of gestational age and then
decreases in a logarithmic fashion throughout life until com-
Gonadotropin release is under negative and positive feedback plete depletion occurs during menopause. When reproduc-
regulation by estradiol, progesterone, and inhibins A and B. tive life is initiated, less than 10% of the primordial follicles
The contributions of these ovarian hormones vary according remain.
to the stage of the ovarian cycle. Folliculogenesis, or formation of the dominant follicle,
consists of two stages: the gonadotropin-independent (pre-
antral) period and the gonadotropin-dependent (antral or
Follicular Phase Graafian) period. Primordial follicular growth up to the
During this phase, the dominant follicle produces high con- antral stage (up to 0.2 mm) occurs during fetal life and infancy
centrations of 17β-estradiol and inhibin B. As concentrations and is gonadotropin independent (Figure 68–5). Primary fol-
of estradiol and inhibin increase, they act on the hypothala- licles are formed when the flattened epithelial cells become
mus and pituitary to decrease FSH secretion. The increase in cuboidal and undergo mitosis. During the antral follicle
estradiol through positive feedback stimulates LH release dur- growth phase, there is further proliferation of the granulosa
ing midcycle (Figures 68–3 and 68–4). The midcycle LH surge cells, expression of FSH and steroid hormone receptors, and
resulting from positive feedback of estradiol is due to an association of the theca cells with the growing follicle and
increased responsiveness of gonadotropic cells to GnRH (fol- granulosa cells, leading to formation of the secondary folli-
lowing exposure to increasing estradiol), an increase in GnRH cles. Tertiary follicles are formed following further theca cell
receptor number, and a GnRH surge, triggered by the effect on hypertrophy and development. Their antrum is filled with
the hypothalamus of increasing estradiol. Progesterone also estrogen-rich fluid, and the theca interstitial cells start to
exerts positive feedback by enhancing pituitary sensitivity to express FSH and LH receptors.
GnRH and increasing GnRH release from the hypothalamus. The mechanisms that trigger initiation of follicular growth are
There is also a smaller pre-ovulatory FSH surge (Figure 68-4). thought to involve bidirectional communication between germ
This is because GnRH-stimulated FSH release is restrained by and somatic cells through gap junctions and paracrine factors,
the inhibitory effect of inhibin. including cytokines and growth factors (insulin-like growth
factor-I [IGF-I], epidermal and fibroblast growth factors, and
Luteal Phase interleukin-1β). When follicles reach a size of 2–5 mm, approxi-
The surge in LH levels induces ovulation and promotes mately 50% are rescued from apoptosis and enter the selection
the survival of the corpus luteum during the luteal phase. growth phase. This final developmental phase of follicular
700 SECTION IX Endocrine and Metabolic Physiology

NE β -endorphin
NPY + – IL-1
Leptin GABA
DA

GnRH

LH FSH
FSH

LH FSH

LH
FSH
+Follicular 80 200
development;
Inhibin A(pg/mL)

Inhibin B(pg/mL)
60 150
ovulation
40 100
Theca cells
20 50

Granulosa 0 0
cells

50 1500
Progesterone(nmo/L)

Estradiol(pg/mL)
40
1000
30
20
500
10
0 0

Ovary
FIGURE 68–3 Hypothalamic–pituitary–ovarian axis. Gonadotropin synthesis and release and differential expression are under
both positive and negative feedback control by ovarian steroid and peptide hormones. Ovarian hormones can decrease gonadotropin release
both by modulating gonadotropin-releasing hormone (GnRH) pulse frequency from the hypothalamus and by affecting the ability of GnRH to
stimulate gonadotropin secretion from the pituitary itself. Estradiol enhances luteinizing hormone (LH) and inhibits follicle-stimulating hormone
(FSH) release, whereas inhibins A and B (gonadal glycoprotein hormones) reduce FSH secretion. After ovulation, ovarian progesterone
production predominates. Progesterone increases hypothalamic opioid activity and slows GnRH pulse secretion, favoring FSH production and
decreasing LH release. Inhibin B peaks early in the follicular phase, whereas inhibin A peaks in the midluteal phase. The increasing inhibin B levels
in the midfollicular phase act at the pituitary gonadotroph to offset activin signaling and suppress FSH biosynthesis from early follicular phase
levels. It also prevents a larger FSH surge in response to estrogen-induced positive feedback on GnRH pulses. The decrease in inhibin A at the
end of the luteal phase creates an environment in which FSH levels can again increase. A variety other CNS and peripheral factors can alter GnRH
release. NE, norepinephrine; NPY, neuropeptide Y; IL-1, interleukin-1; GABA, γ-aminobutyric acid; DA, dopamine. (Modified with permission from Molina
PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

growth, in which antral follicles are protected from apoptosis, in circulating FSH stimulates antral follicle growth. One of
begins approximately 85 days before ovulation in the luteal the leading follicles grows faster than the rest and produces
phase of the cycle preceding ovulation (Figure 68–5). higher levels of estrogens and inhibins. While the reason for
During this gonadotropin-regulated growth phase, follicles why one follicle becomes dominant is unclear, this follicle is
grow exponentially, and FSH stimulates estrogen production likely to be more sensitive to FSH. In turn, the estrogens and
from granulosa cells through the induction of the aromatase inhibins produced by the largest follicle suppress pituitary
enzyme that catalyzes androgen conversion into estrogens, FSH released during the midfollicular phase. The decrease in
with stimulation of follicular fluid formation, cell prolifera- FSH then leads to apoptosis of the remaining growing antral
tion, and LH receptor expression in the dominant follicle. The follicles. Once a follicle is recruited for growth, the oocyte
average time between primary follicle development and ovula- enters a growth phase that leads to the completion of the first
tion is 10–14 days (Figure 68–5). meiotic division. The resumption of meiosis is mediated by
Follicles undergo cyclic recruitment and selection, leading the surge in LH. LH acts on mature follicles to terminate the
to the emergence of the preovulatory follicle(s). The increase program of gene expression associated with folliculogenesis.
CHAPTER 68 Female Reproductive System 701

DAY OF MENSTRUAL CYCLE


0 2 4 6 8 10 12 14 16 18 20 22 24 26 28–0

OVARY FOLLICULAR PHASE LUTEAL PHASE

Serum hormone concentrations

Progesterone

Estradiol

LH

FSH

Inhibin

UTERUS PROLIFERATIVE SECRETORY


PHASE PHASE

MENSTRUAL Spiral
PHASE artery
Gland Bleeding

Bleeding

Vein Basal artery


4 14 28
Time (days)
FIGURE 68–4 Hormonal events during the ovarian and endometrial cycles. Plasma concentrations of FSH, estrogen, LH, and progesterone,
during the human menstrual cycle and the corresponding proliferative and secretory changes in the endometrium and ovarian follicular development,
ovulation, and corpus luteum formation. (Reproduced with permission from Kibble J, Halsey CR: The Big Picture, Medical Physiology. New York: McGraw-Hill, 2009.)

The transcription of genes that control granulosa cell prolif- cycle, approximately 350 times during the female reproduc-
eration and those that encode steroidogenic enzymes is rap- tive life span.
idly turned off by LH. In addition, LH induces genes involved
in ovulation and luteinization. At this stage, mRNA transcrip-
tion virtually ceases and does not resume until 1–3 days after Ovulation
the egg has been fertilized, when the final phases of meiosis Ovulation involves rupture of the wall of the follicle at the
are completed. One preovulatory follicle is selected every surface of the ovary, releasing the oocyte and corona radiata
702 SECTION IX Endocrine and Metabolic Physiology

Basal lamina
Granulosa precursors 25 μ m primordial follicle
Oocyte
Birth to 6 months postpartum
Granulosa cells 60 μm primary follicle

Basal lamina
Theca cells Puberty to menopause
Granulosa cells 150 μm primary
Antral fluid follicle

Oocyte
Zona pellucida
Theca externa
Theca interna
Granulosa cells
5,000 μm antral (graafian) follicle
Antrum 2–4 weeks prior to ovulation
Cumulus oophorus
Zona pellucida
FIGURE 68–5 Follicle growth and Oocyte
development. Folliculogenesis, or formation of
the dominant follicle, consists of two stages: the
18
gonadotropin-independent (preantral) period and
16
the gonadotropin-dependent (antral or Graafian) PREOVULATORY

period. Primordial follicular growth up to the antral Follicle size (mm)


14

stage occurs during fetal life and infancy and is 12


DOMINANT
gonadotropin independent. The final developmental 10
phase of follicular growth, in which antral follicles
8
are protected from apoptosis, begins approximately
6 SELECTED
85 days before ovulation. One dominant follicle is
ATR ATR ATR
recruited in the luteal phase of the cycle preceding 4 – –
ATR ATR ATR ATR ATR ATR
ovulation. The larger, more mature follicles are filled 2

with a transparent albuminous fluid, consisting of RECRUITMENT SELECTION EARLY DOMINANCE LATE DOMINANCE
FINAL MATURATION
an external fibrovascular coat, connected with the Late luteal Early follicular Midfollicular Late follicular
surrounding stroma of the ovary by a network of
blood vessels; and an internal coat, consisting of
several layers of nucleated cells (granulosa cells)
Follicular diameter (mm)

anchored in the zona pellucida, a glycoprotein-rich


2-5
eosinophilic material surrounding the oocyte. The
zona pellucida forms the corona radiata, which
10 days
close to the time of ovulation is separated from the 1-2 Gonadotropin-independent
Gonadotropin-
granulosa cells and expelled with the oocyte during dependent
15 days
ovulation. Ovulation entails rupture of the wall of 0.5-0.9 Early antral
the follicle at the surface of the ovary, releasing the 0.2-0.4 20 days
25 days
oocyte and corona radiata into the peritoneal cavity. 0.15
ATR, atretic; LH, luteinizing hormone; FSH, follicle- Preantral LH
stimulating hormone; Ovul, ovulation; M, menstrual Ovul Ovul Ovul FSH Ovul
phase. (Modified with permission from Molina PE: Endocrine M M M

Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.) First cycle Second cycle Third cycle

into the peritoneal cavity, close to the opening of the fallo- serve as substrates for the aromatase enzyme in the granu-
pian tubes. The ovum is enclosed by an outer layer of cumu- losa cells.
lus cells and an inner, thick extracellular glycoprotein coat, The sequence of temporal events that occurs during ovula-
the zona pellucida. Ciliary movement on the mucous mem- tion is initiated in the responsive preovulatory follicle by a
brane of the fimbria aids movement of the ovum into the surge of LH, which starts 34–36 hours before and peaks 12–24
fallopian tubes. hours before ovulation. This leads to follicular rupture and
Throughout the preovulatory stage, the oocyte, granu- promotes follicular remodeling to form a corpus luteum. In
losa cells, and theca cells acquire specific functional char- addition, the surge in LH stimulates resumption of meiosis up
acteristics. The oocyte becomes competent to undergo to the second meiotic metaphase following extrusion of the
meiosis, granulosa cells acquire the ability to produce estro- first polar body (remnants of one X chromosome) before ovu-
gens and respond to LH via the LH receptor, and theca cells lation. Meiosis is again arrested and then completed with the
begin to synthesize increasing amounts of androgens that release of the second polar body following fertilization.
CHAPTER 68 Female Reproductive System 703

Formation of the Corpus Luteum trial maturation in preparation for implantation of the embryo.
The preovulatory endometrial proliferation leads to relative
The surge in LH release produces reorganization of the follicle
hypertrophy of the uterine mucosa.
and formation of the corpus luteum, composed of small (theca
lutein) and large (granulosa-lutein) cells, fibroblasts, endothe-
lial cells, and immune cells. On rupture of the follicle (follow- SECRETORY PHASE
ing ovulation), small amounts of bleeding into the antral cavity
lead to the formation of the corpus hemorrhagicum and the This phase corresponds to the ovarian luteal phase and is
invasion by macrophages and mesenchymal cells, leading to characterized by progesterone-induced differentiation of
revascularization of the corpus luteum. The granulosa-lutein the endometrial epithelial cells into secretory cells. During
cells transform into the corpus luteum, a temporary endocrine the secretory phase, there is a short, well-defined period of
gland. The corpus luteum continues to autonomously produce uterine receptivity for embryo implantation. Toward the end
and secrete progesterone and estradiol, playing a key role in of the secretory phase, glandular expression of estrogen recep-
regulating the length of the ovarian cycle, maintaining gesta- tors is markedly decreased, reflecting the suppressive effect of
tion in its early stages, and suppressing LH and FSH release increasing progesterone levels.
through the inhibition of GnRH release.
During the initial gestational period, placental production MENSTRUAL PHASE
of hCG prevents the regression of the corpus luteum, trans-
forming the corpus luteum of the ovarian cycle into the cor- The menstrual period is characterized by shedding of the
pus luteum of pregnancy. hCG stimulates the granulosa-lutein endometrium, resulting from proteolysis and ischemia in its
cells to produce progesterone, 17-hydroxyprogesterone, estro- superficial layer. Proteolytic enzymes accumulate in mem-
gen, inhibin A, and relaxin, a polypeptide hormone from the brane-bound lysosomes during the first half of the postovula-
insulin/IGF hormone family. Relaxin regulates the synthesis tory period. The integrity of the lysosomal membrane is lost
and release of metalloproteinases, mediators of tissue (uterus, with the decline in estrogen and progesterone on day 25, result-
mammary gland, fetal membranes, birth canal) growth and ing in lysis of the glandular and stromal cells and the vascular
remodeling, in preparation for birth and lactation. endothelium. Ischemia due to vasoconstriction of endometrial
vessels during the early part of the menstrual period results in
Luteolysis rupture of the capillaries, leading to bleeding. In addition, a
Luteolysis is a two-phase process of lysis or regression of the cor- significant increase in prostaglandin F2α in the late secretory
pus luteum and marks the end of the female reproductive cycle. endometrium contributes by releasing acid hydrolases from
The process involves an initial decline in progesterone secretion lysosomes and enhancing myometrial contractions, aiding in
(functional luteolysis), followed by programmed luteal cell the expulsion of degenerated endometrium.
apoptosis leading to gradual corpus luteum involution (struc-
tural or morphologic luteolysis) to form a small scar of connec- FERTILIZATION
tive tissue known as the corpus albicans. This sequence of
events takes place if fertilization does not occur within 1–2 days Fertilization is the union of the two germ cells, the ovum and
of ovulation. If fertilization occurs, the corpus luteum continues the sperm, restoring chromosome number to 46 and initiat-
to grow and function for the first 2–3 months of pregnancy. ing the development of a new individual. The final steps of
Thereafter, it slowly regresses as the placenta assumes the role mammalian oogenesis (and of spermatogenesis) prepare eggs
of hormone biosynthesis for the maintenance of pregnancy. (and sperm) for fertilization. In preparation for ovulation,
fully grown oocytes undergo “meiotic maturation,” prepar-
ing them to interact with sperm. A very low proportion of the
ENDOMETRIAL CYCLE sperm deposited into the vagina migrates up the female
reproductive tract to the site of fertilization in the ampullary–
The ovarian cycle is accompanied by cyclic growth and shed- isthmic junction of the fallopian tubes (Figure 68–6). During
ding of the endometrium controlled by estrogen and proges- this journey, sperm undergo activation (capacitation), a
terone (Figure 68–4). Three distinct phases can be identified series of changes in the sperm plasma membrane that increase
in the endometrium throughout the menstrual cycle. its affinity for the zona pellucida, enabling the sperm to bind
to the ovum and undergo the acrosome reaction. In the fal-
lopian tubes, sperm bind to the zona pellucida, leading to
PROLIFERATIVE PHASE fusion of the ovum and sperm plasma membranes to form a
The proliferative phase corresponds to the ovarian follicular single “activated” cell, the zygote (Figure 68–6). This simple
phase (Figure 68–4). It is characterized by estrogen-induced process requires several events:
endometrial epithelial cell proliferation and upregulation of • Sperm acrosome reaction and penetration of the ovum’s
estradiol and progesterone receptor expression to reach a peak zona pellucida: Sperm binding to the zona pellucida primes
by the time of ovulation. This is the initial phase of endome- the sperm cell to initiate the acrosome reaction. The
704 SECTION IX Endocrine and Metabolic Physiology

Perivitelline
Sperm
3 space
Zona
pellucida
Cortical
Ovum granules

Zona
pellucida

Ovum
4
FIGURE 68–6 Fertilization and embryo
migration. Sperm binds to the zona pellucida
and undergoes the acrosome reaction, releasing 5
its enzymatic contents, which are necessary for
penetration of the zona pellucida. In addition,
cortical granules in the ovum release their
Inner cell
contents, preventing multiple sperm from mass of
fertilizing one ovum. Once the sperm penetrates blastocyst
the zona pellucida and begins entry into the
perivitelline space, the sperm repositions itself Day 3 Day 2
Day 4 Day 1
from its original orientation with binding at the
tip of the head to binding in an equatorial or
sideways position, leading to fusion with the egg Blastocyst 4 cells 2 cells
plasma membrane and formation of the zygote. Morula Zygote
Day 7
This leads to completion of the meiotic division Fertilization Day 0
and initiation of mitotic divisions while the Embryo
zygote is being propelled through the fallopian
tubes through both ciliary movements by the
epithelium and rhythmic contractions of the Blastocyst
implanting
smooth muscle walls. The embryo enters the Acrosomal Ovulated
uterine cavity (where implantation occurs) as a reaction egg
blastocyst on day 4 following fertilization.
(Modified with permission from Molina PE: Endocrine Muscle layer
Endometrium Uterine wall
Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

acrosome reaction involves fusion of the acrosome with the sperm and ovum pronuclei reconstitutes a diploid cell,
sperm plasma membrane and exocytosis of its enzymatic called the zygote.
contents (proteases and glycosidases) and is required for
During migration of the zygote through the fallopian tubes
sperm penetration. During or after the acrosome reaction,
toward the site of implantation in the uterine cavity, mitosis
the fertilizing sperm detaches from the zona pellucida. It
yields a morula and then a blastocyst. The outer cells of the
penetrates through the zona pellucida and fuses with the
blastocyst are the trophoblast cells, which participate in the
ovum plasma membrane.
implantation process and form the fetal components of the
• Cortical and zona reaction: Fusion of the sperm and the
placenta. If fertilization does not take place, both the ovum
ovum triggers the second meiotic division of the ovum,
and sperm degenerate relatively rapidly in the female
leading to the formation of the mature oocyte and the sec-
reproductive tract.
ond polar body. In addition, this fusion triggers mecha-
nisms that prevent fertilization of the ovum by multiple
sperm, such as exocytosis of cortical granules (cortical re- IMPLANTATION
action) from the oocyte, resulting in proteolysis of zona
pellucida glycoproteins, as well as cross-linking of pro- The human embryo (blastocyst) enters the uterus three days
teins forming the perivitelline barrier. The fusion of the before implantation. The window of implantation corresponds
CHAPTER 68 Female Reproductive System 705

to the short period of endometrial receptivity for the embryo, In premenopausal women, 17β-estradiol produced by the ova-
between days 20 and 24 of the menstrual cycle. Outside of this ries is the chief circulating estrogen. Serum estradiol concen-
period, implantation fails. Many of the physiologic events trations are low in preadolescent girls and increase at menarche,
crucial to successful implantation are due to cyclic changes in defined as the time of the first menstrual bleed. Estradiol pro-
ovarian hormone levels, leading to morphologic and func- duction varies cyclically throughout the menstrual cycle. The
tional maturation of the endometrium. highest rates of production and serum concentrations are in the
preovulatory phase and the lowest during the premenstrual
phase (Figure 68–4). Estradiol levels increase during pregnancy.
PHYSIOLOGIC EFFECTS OF After menopause, serum estradiol concentrations decrease to
values similar to or lower than those in men of a similar age.
OVARIAN HORMONES Most of the estradiol released into the blood circulates bound
to SHBG and to albumin, with only 2–3% circulating in the free
ESTROGEN form. Estradiol (as well as androstenedione) is converted to
estrone (a weak estrogen) in peripheral tissues (Figure 68–7).
Estrogen Synthesis, Transport,
and Metabolism
The primary source of estradiol in women is the granulosa cell
Estrogen Receptor–Mediated
of the ovaries. However, both granulosa and theca cells and (Genomic) Effects
both gonadotropins (FSH and LH) are required for the produc- The estrogen receptors are members of the superfamily of
tion of estrogen. The theca cells secrete androgens that diffuse steroid hormone receptors (see Figure 60–6). Two subtypes
to the granulosa cells to be aromatized to estrogens (Figure 68–2). of estrogen receptors have been identified that differ in

CH3 CH3 CH3

C O C O H C OH

O HO HO
H H
Progesterone Pregnenolone Pregnandiol
H
O

HO
Estradiol

FIGURE 68–7 Metabolic fate of


HO progesterone and estrogen. Progesterone
Estrone and estrogen are degraded primarily in the
H liver. Estradiol and androstenedione are
O O
OH converted to estrone (a weak estrogen) in
peripheral tissues. Estrone is converted to
HO
estriol, primarily in the liver. Estrogens are
HO HO metabolized by sulfation or glucuronidation,
2-Hydroxyestrone Estriol and the conjugates are excreted into the
O urine. Estrogen can also be metabolized
through hydroxylation and subsequent
CH2O methylation to form catecholestrogens and
methoxyestrogens (not shown). (Modified
HO with permission from Molina PE: Endocrine Physiology,
2-Methoxyestrone 3rd ed. New York: McGraw-Hill Medical, 2010.)
706 SECTION IX Endocrine and Metabolic Physiology

structure and tissue distribution and are encoded by differ- membrane forms of estrogen receptor. Although these recep-
ent genes. Estrogen receptor alpha (α) is considered the tors remain largely uncharacterized, they are thought to
classic estrogen receptor. It is found predominantly in endo- resemble their intracellular counterparts.
metrium, breast cancer cells, and ovarian stroma. Estrogen
receptor beta (β) is found predominantly in granulosa cells Physiologic Actions of Estrogen
and developing spermatids, as well as in several nonrepro- at Target Organs
ductive target tissues, including the kidney, intestinal
mucosa, lung parenchyma, bone marrow, bone, brain,
Reproductive system
endothelial cells, and prostate gland. Estrogen exerts multiple effects in reproductive organs
Estrogen receptors are mostly nuclear, but are also found in (Figure 68–8):
the cytoplasm. Similar to other steroid hormones, free estro- • Uterus: Estrogen promotes proliferation of the endometri-
gen diffuses into the cell and binds to the ligand-binding um by stimulating mitosis and angiogenesis. It sensitizes
domain of the receptor, which dissociates from its cytoplasmic uterine smooth muscle to the effects of oxytocin by
chaperone proteins. The estrogen receptor complex then increasing the expression of oxytocin receptors and contrac-
translocates into the cell nucleus, where it binds as homodim- tile proteins. Estrogen increases watery cervical mucus
ers or heterodimers to specific sequences of DNA called estro- production.
gen response elements, regulating gene transcription. The • Ovary: Estrogen exerts potent mitotic effects on granulosa
physiologic effects of estrogens that are mediated through cells and augments the FSH-mediated differentiation pro-
transcriptional activation take minutes or hours to occur. cess (an autocrine action).
• Breast: Estrogen stimulates growth and differentiation of
the ductal epithelium, induces mitotic activity of ductal
Nongenomic Effects of Estrogen cylindrical cells, and stimulates the growth of connective
Some rapid effects of estrogen cannot be explained by a tran- tissue (Figure 68–9). The density of estrogen receptors in
scriptional mechanism (nongenomic) and are the result of breast tissue is highest in the follicular phase of the men-
direct estrogenic action on cell membranes mediated by cell strual cycle and decreases after ovulation. Estrogen can also

Neuroprotection

Influence on mood
Reduction of
intraocular pressure

Slowing of
skin aging

FIGURE 68–8 Systemic effects Arterial vasodilation


of estrogen. In addition to its Maintenance of
reproductive organ effects, estrogen bone density
has neuroprotective effects and
Cardioprotection
reduces perimenopausal mood
fluctuations in women. Estrogen is
Growth and proliferation
cardioprotective and may protect of breast tissue: risk factor
against colon cancer, and it has for breast cancer
Increased production of
vasodilatory effects. In the liver, liver proteins such as
estrogen stimulates the uptake of coagulation factors and
serum lipoproteins and the production hepatic lipoprotein
of coagulation factors. Estrogen receptors Putative reduction
protects against bone loss. In the skin, in risk of colon cancer
it increases skin turgor and collagen
production and reduces the depth of
wrinkles. (Reproduced with permission from
Gruber CJ et al. Mechanisms of Disease:
production and actions of estrogens. NEJM. Growth and differentiation of, and
2002;346:340. Copyright Massachusetts Medical water retention in primary sex organs:
Society. All rights reserved. risk factor for endometrial cancer
CHAPTER 68 Female Reproductive System 707

Atrophic ducts
Estrogen
GH, IGF-I Puberty
EGF

FIGURE 68–9 Hormonal regulation of breast


development and lactogenesis. Mammary gland
development is initiated at puberty through the
actions of estradiol and growth factors and is
further regulated during pregnancy through the
effects of prolactin and human placental lactogen
Lactogenesis Ductal growth (hPL). Throughout pregnancy, progesterone inhibits
lactogenesis. This inhibitory effect is removed
following parturition, when prolactin levels act
unopposed to stimulate lactogenesis. Through
Progesterone, Sexual neuroendocrine reflexes, suckling stimulates the
Prolactin Estrogen, Cortisol, maturation &
Prolactin, Thyroid release of oxytocin from the posterior pituitary,
Pregnancy & IGF-I, hPL, pregnancy
hormone producing the milk “let-down” reflex. GH, growth
post-partum Cortisol,
lactation Insulin hormone; IGF-I, insulin-like growth factor I; EGF,
Oxytocin epidermal growth factor. (Modified with permission from
Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill
Lobuloalveolar growth Medical, 2010.)

indirectly affect mammary gland development by increas- PROGESTERONE


ing prolactin and progesterone levels and inducing proges-
terone receptors in mammary epithelium. The growth- Progesterone is produced by both theca and granulosa cells
promoting effects of estrogen have been implicated in breast and is the predominant ovarian hormone produced in the
and endometrial cancer. luteal phase, a period during which plasma concentrations
• Other body systems: increase 10-fold. Production of progesterone by the corpus
a) Liver: Estrogen affects the expression of apoprotein luteum is mainly under LH stimulation. The majority (80%) of
genes and increases lipoprotein receptor expression, progesterone secreted circulates bound to albumin. The prin-
resulting in a decrease in serum concentrations of total cipal targets of progesterone are the reproductive tract and the
cholesterol and low-density lipoprotein (LDL) choles- hypothalamic–pituitary axis. The degradation of progesterone
terol, increases in serum high-density lipoprotein is similar to that of androgens and estrogens and occurs
cholesterol and triglyceride concentrations, and decreas- primarily in the liver.
es in serum lipoprotein A concentrations. It regulates
the hepatic expression of genes involved in coagulation
and fibrinolysis. Estrogen decreases plasma concentra- Progesterone Receptor–Mediated Effects
tions of fibrinogen, antithrombin III and protein S, and Progesterone appears to exert most of its effects by directly
plasminogen activator inhibitor type 1. Elevated plasma regulating transcription of genes through two specific pro-
estrogen levels are associated with an overall increase in gesterone receptor proteins, termed A and B. These proges-
the potential for fibrinolysis. Estrogen stimulates the terone receptor proteins arise from a single gene and act as
synthesis of transport proteins (thyroxine-binding ligand-inducible transcription factors, regulating the expres-
globulin and transcortin). sion of genes by binding specific progesterone responsive
b) Central nervous system: Estrogen has neuroprotective elements on the DNA. The expression of progesterone recep-
actions, and its age-associated decline is associated with tors is upregulated by estrogen and downregulated by
a decline in cognitive function. progesterone in most target tissues. Previous exposure to
c) Bone: Overall, the effects of estrogen are antiresorptive. estrogen induces the production of receptors for progester-
Estrogen promotes bone maturation and closure of one and is required for progesterone to act on the reproduc-
epiphysial plates in long bones at the end of puberty. It tive tract.
conserves bone mass by suppressing bone turnover and Progesterone receptor expression in the uterus is increased
maintaining balanced rates of bone formation and bone during the first half of the menstrual cycle. During the sec-
resorption. Estrogen affects the generation, life span, ond half, as serum progesterone levels increase, total proges-
and functional activity of both osteoclasts and osteoblasts. terone receptor levels in the uterus decrease. In the mid and
It decreases osteoclast formation and activity and late luteal phases, the density of progesterone receptors in
increases osteoclast apoptosis. the luminal and glandular epithelium declines markedly to
708 SECTION IX Endocrine and Metabolic Physiology

undetectable levels; on the other hand, stromal and myome- tion and early pregnancy by increasing the collagen
trial cells continue to express high levels of progesterone framework and distensibility of the uterus.
receptor despite high circulating progesterone and absent
At the end of pregnancy, the decrease in progesterone levels
estrogen receptor.
is associated with increased prostaglandin synthase activity
In general, progesterone acts on the reproductive tract
and prostaglandin F2α production, enhancing uterine con-
to prepare it for initiation and maintenance of pregnancy.
tractility. The antiprogestin mifepristone antagonizes the
The major physiologic roles of progesterone are mediated in
actions of progesterone on prostaglandin synthesis and catab-
the uterus and ovary, where it stimulates the release of mature
olism and stimulates prostaglandin production, thereby pro-
oocytes, facilitates implantation, and maintains pregnancy
ducing its abortive effects.
through stimulation of uterine growth and differentiation and
suppression of myometrial contractility. In the brain, proges- • Effects on lactation: In the mammary gland, progesterone
terone modulates sexual behavior. Increased progesterone lev- stimulates lobular–alveolar development in preparation for
els during the luteal phase increase both core and skin milk secretion but suppresses milk protein synthesis before
temperatures. This results in a biphasic pattern of body core parturition. Progesterone antagonizes prolactin’s effects in
temperature throughout the menstrual cycle, with a higher mid to late pregnancy, preventing the synthesis of milk pro-
temperature in the luteal phase of the cycle. teins. The sudden decrease in circulating progesterone that
occurs with parturition is associated with a concurrent in-
crease in prolactin secretion and the onset of lactation.
Physiologic Actions of Progesterone • Antiestrogen actions: Progesterone antagonizes estrogen
at Target Organs induction of many of the known hormone-responsive genes.
This effect is mediated by downregulation of estrogen re-
• Effects on the uterus during early pregnancy: Progester- ceptor protein concentrations, decreasing the active estro-
one induces stromal differentiation; stimulates glandular gen concentration (and antagonizing the action of estrogen
secretions, changing the pattern of proteins secreted by receptor at the molecular level), particularly in the uterus.
endometrial cells; and modulates cyclic proliferation during
the menstrual cycle.
Progesterone induces uterine cell proliferation and differ- THE PLACENTA
entiation in early pregnancy through regulation of localized
growth factor synthesis and cell type–specific expression Structure and Physiologic Function
of their receptors, thereby regulating cellular sensitivity to The placenta is derived from two major cell types, which are the
the autocrine or paracrine effects of growth factors and source of the principal placental hormones. The outer cell mass
producing an environment that supports early embryonic of the blastocyst, the precursor to the trophoblast, is in contact
development. with the endometrium and undergoes proliferation and tissue
penetration during implantation. The trophoblast has two cell
• Promotion and maintenance of implantation: Progester-
populations: an inner cytotrophoblast and an outer invasive
one plays a major role in preparing the endometrium for
syncytiotrophoblast. The maternal side of the placenta contains
implantation of a fertilized ovum. It facilitates implantation
fetal chorionic villi that provide an extensive surface area for
by stimulating the synthesis of enzymes responsible for lysis
nutrient and gas exchange between the fetal and maternal circu-
of the zona pellucida. Finally, it promotes and maintains im-
lation. The villi are covered with multinucleated syncytiotro-
plantation through effects on both the maternal uterus and
phoblast and trophoblast stem cells, stromal cells, and blood
the developing blastocyst.
vessels. The villous cytotrophoblast cells are entirely secluded
• Effects on uterine contractility: Progesterone induces
from maternal elements, with the exception of any molecules
quiescence of the myometrium by increasing resting
that might be transported across the placenta by the syncytiotro-
membrane potential and preventing electrical coupling
phoblast. By contrast, the extravillous trophoblast cells are con-
between myometrial cells. It also prevents uterine contrac-
tinuously exposed to maternal tissues. The middle layer of the
tions by blocking the ability of estradiol to induce mem-
placenta consists of densely packed cytotrophoblast cell columns
brane expression of α-adrenergic receptors (α-adrenergic
and serves as structural support for the underlying villi.
activation causes contractions). Progesterone decreases
The physiologic functions of the placenta can be classified
prostaglandin synthesis and increases the rate of prosta-
as follows:
glandin inactivation through the stimulation of prosta-
glandin 15-dehydrogenase and opposes the stimulatory • supportive, transporting nutrients and oxygen necessary for
effects of estrogen on endometrial prostaglandin F2α fetal growth; removing of waste products;
expression in the luteal phase of the menstrual cycle. • immune, suppressing the local immune system to prevent
Progesterone maintains the levels of relaxin, inhibiting immunologic rejection of the fetus by the mother;
spontaneous or prostaglandin-induced myometrial con- • endocrine, including hormone synthesis, transport, and
traction, and contributes to the maintenance of implanta- metabolism to promote fetal growth and survival.
CHAPTER 68 Female Reproductive System 709

Inability of the placental unit to perform these functions leads LDL. As discussed earlier, progesterone plays an important
to multiple complications of human pregnancy, including abor- role in maintaining uterine quiescence during pregnancy,
tion, miscarriage, impaired fetal growth, and preeclampsia. inhibiting prostaglandin synthesis, and modulating the
immune response to preserve pregnancy.
• Estrogen: The main source of estrogen during the initial
Endocrine Function of the Placenta phase of pregnancy is the corpus luteum, being replaced
The placenta produces cytokines, hormones, and growth fac- later by placental production. The production of estrogen by
tors that are essential for regulation of the fetomaternal unit. the placenta requires coordinated interaction between fetal
In addition, the placenta expresses enzymes involved in hor- and maternal adrenal gland steroid hormone production
mone metabolism, playing an important role in protection of (fetoplacental unit of steroid biosynthesis). The placenta
the fetus from maternal adrenal-derived androgens through lacks 17α-hydroxylase activity and is thus unable to convert
aromatase activity and from glucocorticoids through the activ- progesterone to estrogen or to produce androgens. This lack
ity of 11β-hydroxysteroid dehydrogenase type II. The princi- of placental androgen production protects the female fetus
pal placental hormones are as follows: from masculinization; protection is also aided by the strong
• hCG: hCG is a heterodimeric glycoprotein from the same aromatase activity that converts maternal and fetal adrenal-
hormone family as LH, FSH, and TSH. It is produced by the derived androgens to estrogens. Therefore, maternal and
syncytiotrophoblast and released into the fetal and maternal fetal adrenal-derived androgens (dehydroepiandrosterone
circulation. It is known as the hormone of pregnancy and sulfate [DHEAS]) are required for placental 17β-estradiol
is the basis for the pregnancy test. hCG is detected in serum and estriol production. Estriol is synthesized through
at days 6–8 after implantation, and its levels peak at 60–90 the aromatization of 16α-hydroxyandrostenedione derived
days of gestation, declining thereafter. hCG has structural from 16α-hydroxyepiandrosterone sulfate produced by the
and functional similarity to LH, has a much longer half-life, fetal liver and desulfated in the placenta (Figure 68–10);
and exerts its physiologic effects primarily through binding 16α-hydroxyepiandrosterone sulfate is derived from
to the LH receptors. The main function of hCG is to main- DHEAS produced in the fetal adrenal gland. The enzymes
tain the corpus luteum to ensure the production of proges- involved are placental sulfatase (DHEAS deconjugation),
terone until placental production takes over. Maternal hCG 3β-hydroxysteroid dehydrogenase (pregnenolone to proges-
levels are a useful index of functional status of the tropho- terone conversion), and aromatase. The principal physio-
blast (placental health). logic effects of estrogen during pregnancy include stimula-
• Human placental lactogen and growth hormone: hPL is tion of uterine growth, prostaglandin synthesis, thickening
produced by the syncytiotrophoblast and is secreted into of the vaginal epithelium, sensitization to oxytocin effects,
both the maternal and fetal circulations after the sixth week growth and development of the mammary epithelium, and
of pregnancy. In the fetus, hPL modulates embryonic inhibition of milk production.
development, contributes to regulation of intermediary • Corticotropin-releasing hormone (CRH): CRH is pro-
metabolism, and stimulates the production of IGFs, insulin, duced by the syncytiotrophoblast and trophoblast cells of
adrenocortical hormones, and pulmonary surfactant. Dur- the placenta. Its structure and function are similar to those
ing pregnancy, hGH-V, a GH variant expressed by the pla- of hypothalamus-derived CRH. The CRH concentration in-
centa, becomes the predominant GH in the mother. This creases throughout pregnancy and peaks during labor. Pla-
hormone has structural and functional similarity to pitu- cental production of CRH has been linked to the length of
itary GH and is not released into the fetus. Starting from the gestation in humans. CRH is secreted into the maternal cir-
15th to the 20th week of gestation up to term of pregnancy, culation in large amounts during the third trimester of preg-
placental GH gradually replaces maternal pituitary GH, nancy and may play an important role in the onset of labor.
which becomes undetectable. hGH-V stimulates IGF-I pro-
duction and modulates maternal intermediary metabolism,
increasing the availability of glucose and amino acids to the
PREGNANCY AND LACTATION
fetus. Placental GH is not detectable in the fetal circulation,
and thus it does not appear to have a direct effect on fetal
Hormonal Control of Parturition
growth. Uterine contractility during pregnancy and parturition can be
• Progesterone: The major source of progesterone during the divided into at least four distinct phases:
initial phase of pregnancy is the corpus luteum under hCG • Phase 0: During pregnancy, the uterus is maintained in a
regulation. Starting from about week 8 of gestation, the pla- relatively quiescent state, mainly through the effects of pro-
centa (syncytiotrophoblast) becomes the principal source of gesterone. Additional factors involved in modulation of
progesterone, leading to increasing levels of maternal pro- uterine activity during this period are prostacyclin, relaxin,
gesterone throughout pregnancy. Because the placenta is parathyroid hormone-related peptide, and CRH. The initia-
unable to produce cholesterol from acetate, cholesterol for tion of parturition results from the transition from the qui-
placental progesterone synthesis is derived from circulating escent phase (phase 0) to a phase of activation (phase 1).
710 SECTION IX Endocrine and Metabolic Physiology

Mother Placenta Fetus

Pregnenolone
sulfate
Acetate Cholesterol LDL
cholesterol
Estriol
glucosiduronate

Pregnenolone
DHEA

Progesterone

16α -OHDEA

Urinary
excretion

16α -OHDEAS
Estriol

FIGURE 68–10 Fetoplacental unit hormone synthesis. The production of estrogen by the placenta requires the coordinated interaction
between fetal and maternal adrenal gland steroid hormone production. The placenta lacks 17α-hydroxylase and is thus unable to convert
progesterone to estrogen or to produce androgens. Maternal and fetal adrenal-derived androgens (dehydroepiandrosterone sulfate, DHEAS) are
required for 17β-estradiol and estrone production. Estriol is synthesized through the aromatization of 16α-hydroxyandrostenedione derived from
16α-hydroxyepiandrosterone sulfate (16α-OHDEAS) produced by the fetal liver and desulfated in the placenta; 16α-hydroxyepiandrosterone
sulfate in turn is derived from DHEAS produced in the fetal adrenal gland. LDL, low-density lipoprotein; 16α-OHDEA, 16α-hydroxyepiandrosterone.
(Modified with permission from Molina PE: Endocrine Physiology, 3rd ed. New York: McGraw-Hill Medical, 2010.)

• Phase 1: This phase of parturition is associated with activa- • Phase 3: This postpartum phase involves uterine involution
tion of uterine function and is characterized by release from after delivery of the fetus and placenta and is mainly due to
the inhibitory mechanisms maintaining uterine quiescence the effects of oxytocin.
throughout pregnancy and activation of factors promoting
uterine activity. These factors include uterine stretch and
tension caused by the fully grown fetus, activation of the fe- Mammary Gland Development
tal hypothalamic–pituitary–adrenal axis, and increased Mammary gland development involves cell proliferation, dif-
prostaglandin synthesis. Mechanical stretch or hormonal ferentiation, and morphogenesis. Most mammary gland devel-
priming leads to upregulation of gene expression of proteins opment occurs postnatally and involves branching and
that facilitate smooth muscle contraction, including con- extension of ductal growth points and secretory lobules into a
nexins (key components of gap junctions), prostaglandin fatty stroma. This process is regulated by the associated altera-
and oxytocin receptors, and ion channel proteins. tions in hormones and growth factors during the various
• Phase 2: This phase of parturition is a period of active uter- reproductive states (puberty and pregnancy) (Figure 68–9).
ine contraction and is stimulated by prostaglandins, oxyto- Ductal elongation is mediated by estrogen, GH, IGF-I, and
cin, and CRH. Prostaglandins, particularly those produced epidermal growth factor. Ductal branching and alveolar bud-
in the intrauterine tissues, play a central role in the initiation ding are regulated by progesterone, prolactin, and thyroid
and progression of labor. They induce myometrial contrac- hormone. Progesterone stimulates ductal side branching and
tility and help produce the changes associated with cervical alveolar development. Prolactin acts directly on mammary
softening at the onset of labor. epithelium to induce alveolar development. Both progesterone
CHAPTER 68 Female Reproductive System 711

and prolactin synergize to stimulate proliferation of ductal stage of puberty (at age 8–13) in girls involves breast develop-
epithelium. ment, accompanied by ovarian and follicular growth. This is
During pregnancy, prolactin, progesterone, and hPL act on followed by androgen- plus estrogen-induced pubic and axil-
duct lobular units to promote expansion and differentiation of lary hair growth and the onset of menses (approximately at
their secretory function. This stage of mammary differentia- age 13), indicating sufficient estrogen production to stimulate
tion into a secretory function is called stage I lactogenesis. endometrial proliferation. The first cycles are usually anovula-
The elevated levels of progesterone prevent milk production tory, becoming fully ovulatory after 2–3 years. In girls, serum
during this period. leptin concentrations increase as pubertal development pro-
The second stage (stage II lactogenesis) is initiated after gresses, and this increase in leptin levels parallels the increase
termination of pregnancy. The sudden decrease in circulating in body fat mass.
progesterone that accompanies parturition in association with
the concurrent increase in prolactin secretion marks the onset MENOPAUSE
of lactation. The decrease in progesterone levels results in
removal of the inhibition of synthesis of α-lactalbumin and Menopause is the permanent cessation of menstruation
β-casein. In the presence of prolactin, insulin, and glucocorti- resulting from loss of ovarian follicular activity. It is preceded
coids, the synthesis of milk proteins is established. Continuous by a perimenopausal period, starting when the first features
milk production is maintained by prolactin secretion from the of impending menopause begin (i.e., irregular menstrual
anterior pituitary throughout the period of lactation. The high bleeding and cycle frequency) and lasting until at least 1 year
prolactin levels are in part due to increased lactotroph synthe- after the final menstrual period. During the menopausal
sis resulting from the high estrogen levels during pregnancy. transition, gonadotropins, estradiol, and inhibin show a
Prolactin is the main regulator of milk protein synthesis marked degree of variability in their circulating levels. Within
through its effects on the prolactin receptor located on mam- 1–2 years after the final menstrual period or the onset of
mary epithelial cells. Prolactin release is under negative con- menopause, FSH levels are markedly increased, LH levels are
trol by dopamine; thus, pharmacologic analogs of dopamine moderately high, and estradiol and inhibin levels are low or
such as bromocriptine inhibit lactogenesis. Weaning, or cessa- undetectable. Postmenopausally, adrenal androstenedione is
tion of the lactation period, is followed by involution of the the major source of estrogen, and serum testosterone levels
terminal duct lobular units mediated by alveolar cell apoptosis fall moderately.
and gland remodeling, returning the breast to its mature qui- Starting from the mid-thirties, ovarian follicular apoptosis
escent state. accelerates, leading to a steady decline in ovarian estradiol
production (Figure 68–11). This loss of ovarian function
Hormonal Control of Milk Secretion results in a 90% loss of circulating estradiol. However, extrago-
and Ejection nadal estrogen synthesis increases as a function of age and
body weight, and most of the estradiol is formed by extrago-
The onset of adequate milk production during the postpartum
nadal conversion of testosterone. The predominant estrogen
period requires developed mammary epithelium, persistent
in menopausal women is the weak estrogen estrone, produced
elevation in plasma prolactin, and a decrease in circulating
through aromatase conversion of androstenedione.
levels of progesterone. Milk secretion from the mammary
The decline in ovarian function associated with the peri-
glands is triggered by stimulation of tactile receptors in the
menopausal period is also responsible for an early decline in
nipples by suckling. Sensory impulses are transmitted to the
the release of inhibin B leading to an increase in follicular
secretory oxytocinergic neurons in the hypothalamus, which
phase FSH. The decrease in serum inhibin B is believed to
in response release oxytocin into the systemic circulation.
reflect the age-related decrease in ovarian follicle reserve,
Oxytocin produces contraction of the myoepithelial cells of
which is the primary source of serum inhibin B. The later rise
the lactiferous ducts, sinuses, and breast tissue alveoli.
in serum LH during the menopausal transition is due to the
cessation of ovarian follicle development. Despite a 30%
decrease in GnRH pulse frequency with aging, there is an
AGE-RELATED CHANGES IN THE increase in the overall amount of GnRH secreted. FSH levels
FEMALE REPRODUCTIVE SYSTEM gradually increase with age in women who continue to cycle
regularly. The consequences of loss of ovarian function during
PUBERTY reproductive life may be severe. Symptoms include hot flushes,
night sweats, vaginal dryness and dyspareunia (painful
Female puberty is initiated by low-amplitude nocturnal pulses intercourse), loss of libido, loss of bone mass with subsequent
of gonadotropin release. The increased synthesis and secretion osteoporosis, and abnormalities of cardiovascular function,
of estrogen by the ovary cause the progressive skeletal matura- including a substantial increase in the risk of ischemic heart
tion that eventually leads to epiphysial fusion and the termina- disease. As already mentioned, estrogens (like androgens) have
tion of linear growth. The onset of puberty causes a rapid general metabolic roles that are not directly involved in repro-
increase in bone mass that correlates with bone age. The initial ductive processes. These include actions on vascular function,
712 SECTION IX Endocrine and Metabolic Physiology

Hormonal changes in menopause


Ovarian estradiol Adrenal estrone
Androgen: estrogen ratio
Progesterone levels

Negative feedback GnRH


FIGURE 68–11 Changes in
gonadotropin and ovarian
hormone production associated
with aging. Lower levels of inhibin
LH and FSH
B and estradiol result in impaired Aromatase
negative feedback regulation of
gonadotropin release, increasing
Testosterone Estradiol
FSH and LH. Production of Androstenedione Estrone
androstenedione and testosterone
during early menopause continues, Inhibin B Ovary
Adipose tissue
with some conversion to estradiol by Estradiol
aromatase activity in adipose tissue.
Adrenal-derived androstenedione is
converted to estrone, principally in Androstenedione
adipose tissue. (Reproduced with DHEA
permission from Gruber CJ et al. Production Progesterone
and actions of estrogens. NEJM. Adrenals
2002;346:340.)

lipid and carbohydrate metabolism, and bone mineralization


and epiphysial closure at sites such as breast, bone, vasculature,
CONTRACEPTION AND THE
and brain. Within these sites, aromatase action can generate FEMALE REPRODUCTIVE TRACT
high levels of estradiol locally without significantly affecting
circulating levels. Circulating C19 steroid precursors are essen- The multiple steps involved in the regulation of ovarian
tial substrates for extragonadal estrogen synthesis. The levels hormone production, the consequent modifications of the
of these androgenic precursors decline markedly with advanc- endometrium, and the regulation of uterine motility are all
ing age in women, possibly from the mid to late reproductive under tight control, ensuring ovulation, fertilization, implan-
years. This is thought to contribute to the greater risk of bone tation, and maintenance of pregnancy. Multiple approaches
mineral loss and fracture, and possibly the decline in cognitive have been implemented for contraception. Some of the princi-
function, in women as compared with men. pal interventions are summarized in Table 68–1.

TABLE 68–1 Principal contraceptive methods.


Method Mechanism Involved

Steroid contraceptives Suppression of LH surge preventing ovulation

Intrauterine devices Prevent blastocyst implantation by altering the endometrial lining


Some release progesterone, modifying the endometrial lining

Barrier methods: condoms, foam, and Prevent fertilization by either interfering with the access of sperm to the uterine cavity or
diaphragms destroying sperm in the vaginal cavity

Sterilization Surgically disrupts the continuity of the fallopian tubes, impairing access of the fertilized ovum to
the uterine cavity and implantation

Abortive Antiprogestin mifepristone produces an increase in prostaglandin F2α synthesis, leading to


expulsion of the embryo

Rhythm Relies on changes in mucus thickness and body temperature throughout the menstrual cycle,
indicating a “safe” period for intercourse
CHAPTER 68 Female Reproductive System 713

DISEASES OF OVERPRODUCTION for her age and according to height of her parents. Body mass
AND UNDERSECRETION OF index is 19%. Physical examination does not reveal abnormal-
ities in her clitoris or vagina. No physical problems are identi-
OVARIAN HORMONES fied. Laboratory values are negative for elevations in prolactin.
The diagnosis of “exercise”-induced amenorrhea is made.
Alterations in female reproductive endocrine function are of A frequent cause of amenorrhea in adolescents is hypo-
multiple etiologies and produce manifestations that range thalamic amenorrhea. Energy deficit results in suppression
from precocious puberty to infertility, depending on the age at of hypothalamic secretion of GnRH in anorexia nervosa,
presentation. The most frequent are abnormalities in the men- exercise-induced amenorrhea, and amenorrhea associated
strual cycle, consisting of either absent menstruation (amen- with chronic illness. This is in part mediated by leptin defi-
orrhea) or excess bleeding (metrorrhagia), and infertility. ciency due to decreased adipose tissue. Leptin is a hormone
Abnormalities of ovarian development and function are usu- secreted by adipose tissue and signals energy availability.
ally caused by defective development of the gonads and rarely Young athletes, particularly ballet dancers, who start train-
by defects in the synthesis of ovarian steroids. In general, ing at a young age, may present with primary amenorrhea,
increased ovarian hormone production can be due to increased and this is frequently associated with low BMI and body
gonadotropin release (hypergonadotropic hypergonadism) weight. Additional factors that can contribute to lack of
related to tumors, brain inflammatory diseases, and head menses are the decreased androgen aromatization to estro-
injury, among other causes, or it can result from excess hor- gen resulting from low fat mass.
mone production by ovarian tumors. Decreased ovarian
hormone production can be genetic (e.g., FSH and LH recep-
tor gene mutations, mutation of the β-subunit of FSH, enzy-
matic deficiencies) or acquired (e.g., radiation) despite CHAPTER SUMMARY
adequate gonadotropin release (hypergonadotropic hypogo-
■ Gonadotropin release is under negative and positive feedback
nadism). Decreased ovarian hormone production due to regulation by ovarian steroid and peptide hormones.
impaired gonadotropin release (hypogonadotropic hypogo-
■ Estrogen synthesis requires LH and FSH regulation of
nadism) is rare and may result from GnRH receptor gene coordinated metabolism by granulosa and theca cells of the
mutations, lesions in the hypothalamic area, and other causes. ovarian follicle.
■ LH and FSH rescue selected ovarian follicles from apoptosis
and stimulate their growth and maturation.
CLINICAL CORRELATION ■ The corpus luteum is a temporary endocrine organ that plays a
central role during the initial stages of pregnancy.
CASE A ■ The ovarian cycle produces cyclic changes in steroid hormone
production, which in parallel produce marked morphologic
A postmenopausal woman presents with a history of acute and functional changes in the endometrium, preparing it for
low back pain. She had menopause at 20 years prior to con- embryo implantation.
sultation and has never received hormone replacement ■ Estrogen has important systemic effects affecting the risk of
therapy. She reports a history of a wrist fracture four years cardiovascular disease, osteoporosis, and endometrial and
before this visit. Lumbar spine films reveal a new vertebral breast cancer.
fracture. Dual-energy x-ray absorptiometry of the hip ■ Progesterone ensures uterine quiescence and prevents
shows a low bone mineral density. The diagnosis of osteo- lactogenesis during pregnancy.
porosis is made. ■ Mammary gland morphologic development occurs during
Postmenopausal osteoporosis is a common disease with a puberty and is functionally modified during pregnancy by
spectrum ranging from asymptomatic bone loss to disabling prolactin and hPL, ensuring lactogenesis.
hip fracture. Osteoporosis is a disease of increased skeletal
fragility accompanied by low bone mineral density measured
by dual-energy x-ray absorptiometry. Fractures occur because STUDY QUESTIONS
of microarchitectural deterioration resulting from deteriora-
tion in the trabecular and cortical skeleton. Risk factors for 1. A 30-year-old female patient arrives at your office because of
missed menstrual periods for 2 months. Her history indicates
osteoporosis include low calcium and vitamin D intakes,
regular menstrual periods in the past. During physical
sedentary life, smoking, alcohol use, and low estrogen. examination, you suspect that she may be pregnant. Which of the
following laboratory values would be compatible with your
diagnosis?
CASE B A) low plasma progesterone and high LH
A 19-year-old female ballet dancer is brought to the clinic B) high prolactin, low LH, and low progesterone
C) high urinary estradiol and low progesterone
because she has never started menstruating. Height is normal
D) high urinary hCG and increased plasma progesterone
714 SECTION IX Endocrine and Metabolic Physiology

2. A 5-month pregnant woman is referred to your office with newly 4. The effects of progesterone on the myometrium during
diagnosed hypertension. You are concerned that the fetus and pregnancy include
placenta may be compromised. To assess fetal and placental A) preventing electrical coupling between myometrial cells.
health, which of the following hormone measurements would be B) increased estradiol-induced α-adrenergic receptor
most informative? expression
A) urinary estriol and serum hCG C) decreased prostaglandin inactivation
B) serum progesterone and prolactin D) increased prostaglandin synthase activity
C) serum LH and hPL 5. Systemic and hepatic effects of estrogen include
D) urinary estriol and serum progesterone A) increased serum concentrations of total cholesterol and
3. Increased fasting plasma glucose levels in pregnant women with LDL
no history of diabetes may be related to B) increased lipoprotein receptor expression
A) increased estrogen production C) increased plasma concentrations of fibrinogen and
B) decreased progesterone clearance antithrombin III
C) increased hPL/GH-V production D) decreased synthesis of thyroxine-binding globulin and
D) increased insulin degradation transcortin
69
C H A P T E R

Endocrine Integration
of Energy and Electrolyte
Balance
Patricia E. Molina

O B J E C T I V E S

■ Identify the normal range of plasma glucose concentrations and the


hormonal regulation of its metabolism, storage, and mobilization.
■ Identify the specific roles of insulin, glucagon, glucocorticoids,
catecholamines, growth hormone, and thyroid hormone in the regulation of
energy substrate utilization, storage, and mobilization.
■ Describe the hormonal regulation of energy substrate metabolism during the
fed and fasted states and understand the consequences of its dysregulation.
■ Identify the mechanisms involved in the maintenance of long-term
energy balance.
■ Identify the normal range of dietary sodium intake, its body distribution, and
routes of excretion. Explain the roles of antidiuretic hormone, aldosterone,
angiotensin, and atrial natriuretic hormone in the regulation of sodium balance.
■ Identify the normal range of dietary potassium intake, its body distribution,
and routes of excretion. Explain the hormonal regulation of plasma potassium
concentration, distribution, and balance in the acute and chronic settings.

NEUROENDOCRINE REGULATION energy metabolism to different extents, according to age, sex,


nutritional state, and metabolic demands of the individual.
OF ENERGY STORAGE, Because the autonomic nervous system also interacts with the
endocrine system in the modulation of glucose and fat metabo-
MOBILIZATION, AND UTILIZATION lism, the system is involved in neuroendocrine regulation. The
Two distinct phases directly related to the ingestion of a meal autonomic nervous system exerts its effects both directly and
alternate throughout the day in the regulation of energy metab- indirectly. For example, activation of the sympathetic nervous
olism. The fed state reflects overall anabolic metabolism, dur- system through norepinephrine release from nerve terminals
ing which energy is stored in the form of energy-rich compounds and epinephrine release from the adrenal medulla stimulates
(adenosine triphosphate [ATP], phosphocreatinine), glycogen, skeletal muscle glycogenolysis, glucagon release, and hepatic glu-
fat, and proteins. The fasted or catabolic phase is the period cose output while inhibiting the release of insulin (Figure 69–1).
during which endogenous energy sources are utilized.
The anabolic and catabolic phases alternate to preserve ade- NEUROENDOCRINE REGULATION
quate glucose supply to the brain as well as sufficient energy to OF ENERGY METABOLISM
maintain body functions, such as thermoregulation (maintain-
ing a constant core temperature), food digestion, and physical DURING THE FED STATE
activity. The two hormones at the core of maintaining this bal-
ance are insulin and glucagon; in particular, their ratio plays a Glucose
critical role in the dynamic regulation of substrate metabolism Blood glucose regulation occurs through interactions among
(summarized in Table 69–1). However, several other established hormonal, neural, and hepatic autoregulatory mechanisms. Fol-
and newly discovered hormones participate in the regulation of lowing a meal (postprandial state), in response to the increase

715

Ch69_715-728.indd 715 11/26/10 10:39:05 AM


716 SECTION IX Endocrine and Metabolic Physiology

TABLE 69–1 Regulation of metabolic processes acids are provided. GH and testosterone are of particular
by insulin/glucagon ratios. importance during growth and development, as well as during
adulthood and senescence.
Anabolic Metabolic Catabolic
(↑ I:G) Process (↓ I:G)
NEUROENDOCRINE REGULATION
↑ ↓
OF ENERGY METABOLISM
Glycogen synthesis
(liver and muscle)

↓ Glycogen breakdown ↑ DURING THE FASTED STATE


↓ Gluconeogenesis ↑ During the fasted postabsorptive state, catabolism of stored
↑ Fatty acid synthesis and ↓ energy sources provides the energy required for bodily func-
triglycerides (hepatocytes tions. The amount of energy expended by an awake, resting indi-
and adipose tissue) vidual, measured 12–14 hours following the last meal, and at
↑ Muscle protein synthesis ↓ normal (or thermoneutral) body temperature is called the basal
metabolic rate (BMR). The BMR is the amount of energy
↑ Lipogenesis and ↓
triglyceride formation required to maintain breathing, brain activity, enzymatic activity,
and other functions without any physical movement of the indi-
↓ Lipolysis ↑
vidual. Any deviation from the basal condition, such as changes
↓ Free fatty acid oxidation ↑ in body temperature (fever or hypothermia), the level of activity
↓ Ketone body formation ↑ of the individual (exercise or sleeping), or time from the last meal
(fed or fasted), will affect the metabolic rate. In addition, BMR
↓ Muscle proteolysis ↑
can be directly affected by hormone action, particularly thyroid
I, insulin; G, glucagon. hormones, which increase Na+/K+-ATPase activity and body
temperature, resulting in an increase in the BMR.

in pancreatic insulin release, glucose uptake is increased in


muscle, fat, and the liver; hepatic glucose output is suppressed;
Glucose
and glycogen synthesis is increased. In the resting postabsorptive state, release of glucose from the
Glucose disposal by insulin-sensitive tissues is regulated liver through glycogenolysis and gluconeogenesis is the key
initially by an increase in glucose transport and enzyme phos- regulated process. During fasting, hepatic glucose production is
phorylation leading to the activation of glycogen synthase, increased and peripheral glucose utilization is inhibited. Initially,
phosphofructokinase, and pyruvate dehydrogenase (see hepatic glucose output is derived from breakdown of hepatic gly-
Figure 66–5). The majority of insulin-stimulated glucose cogen stores through glycogenolysis. Following an overnight
taken up is stored as glycogen. fast, glycogenolysis provides approximately 50% of the overall
hepatic glucose output. As hepatic glycogen stores are depleted
during a period of prolonged fasting (approximately 60 hours),
Fat the contribution of glycogenolysis to hepatic glucose output
Most of the body’s energy reserve is stored in adipose tissue in becomes negligible, with hepatic gluconeogenesis predominat-
the form of triacylglycerol in the adipocytes. The principal hor- ing. Glycogenolysis depends on the relative activities of glycogen
mone involved in lipogenesis is insulin, through activation of synthase and phosphorylase (see Figure 66–4). Gluconeogenesis
lipogenic enzymes. Opposing the effects of insulin are growth is regulated by the activities of fructose-1,6-diphosphatase, phos-
hormone and leptin (described later), which inhibit lipogenesis. phoenolpyruvate carboxykinase, pyruvate kinase, and pyruvate
The balance between lipogenesis and lipolysis followed by fatty dehydrogenase, and by the availability of the principal gluconeo-
acid oxidation determines the overall accumulation of body fat. genic precursors, lactate, glycerol, glutamine, and alanine.
Both processes are under hormonal and cytokine regulation. A smaller, yet significant amount (approximately 25%) of sys-
temic glucose production in the postabsorptive state is derived
Protein from renal gluconeogenesis. Usually, the kidney is not a net pro-
ducer of glucose. The proximal tubule cells produce glucose at a
The balance between protein synthesis and degradation is rate similar to that of glucose utilization by the renal medulla.
regulated by interactions among hormonal, nutritional, neu-
ral, and inflammatory mediators. Hormonally, regulation of
protein metabolism is predominantly under the influence of Fat
insulin, hGH, and insulin-like growth factor-I (IGF-I). After an overnight fast, most of the resting energy requirement is
During the fed state, insulin acts primarily to inhibit proteoly- provided by oxidation of fatty acids derived from adipose tissue.
sis, and GH stimulates protein synthesis. IGF-I has antiprote- Lipolysis in adipose tissue is mostly dependent on the concentra-
olytic effects during the postabsorptive state that progress to tions of hormones (epinephrine stimulates lipolysis, and insulin
stimulation of protein synthesis in the fed state or when amino inhibits lipolysis). During a period of acute energy deprivation
CHAPTER 69 Endocrine Integration of Energy and Electrolyte Balance 717

Pancreas

Glucagon
Insulin

Paraventricular
glucose nucleus
CRH, ADH
Pituitary
Locus ceruleus
Cortisol GH (noradrenergic
system)
Glycogenolysis Norepinephrine
Gluconeogenesis Gluconeogenic
ACTH
substrates

Cortisol FIGURE 69–1 Neuroendocrine response


FFA to exercise. A. The principal pathways activated
by stress are the hypothalamic–pituitary–adrenal
Epinephrine Sympathetic
FFA Norepinephrine ganglion
axis and the sympathetic nervous system,
Lipolysis resulting in the release of corticotropin-releasing
Norepinephrine
Dorsal-root hormone (CRH), antidiuretic hormone (ADH),
Glycogenolysis Neuropeptides
Heart rate ganglion catecholamines, and growth hormone (GH). In
FA oxidation Neuropeptides
Protein synthesis Blood pressure the periphery, increased production and release
Peripheral of cortisol, glucagon, and catecholamines and
vasoconstriction suppressed release of insulin favor an overall
B catabolic response. Stimulation of hepatic
Protein Glycogen Fat glycogenolysis and gluconeogenesis, muscle
glycogenolysis, and adipose tissue lipolysis
Amino acids Glucose Glycerol Fatty acids
ensures the production and mobilization of
energy stores to meet the enhanced metabolic
ATP Glycolysis
demands of the individual. Reproductive and
NH2 growth functions are inhibited, conserving
Pyruvate
energy to sustain fundamental processes that
CO2
Urea ensure survival. B. Stimulation of hepatic
Acetyl coenzyme A glycogenolysis and gluconeogenesis, muscle
glycogenolysis and adipose tissue lipolysis
Krebs cycle CO2 ensure the production and mobilization of
ATP energy stores to sustain the enhanced metabolic
demands of the individual as shown in the
Oxidative metabolic pathway. ATP, adenosine triphosphate;
O2 H2O
phosphorylation FFA, free fatty acid; FA, fatty acid. (Modified with
permission from Molina PE: Endocrine Physiology, 3rd ed.
ATP New York: McGraw-Hill Medical, 2010.)

or prolonged starvation, lipolysis mobilizes triglycerides, provid- of acute glucose deficiency (insulin-induced hypoglycemia)
ing nonesterified fatty acids as energy substrates for tissues such or increased energy demand (as with strenuous exercise), cat-
as muscle, heart, and liver; and substrates for glucose (glycerol) echolamines play an important role in the stimulation of lipoly-
and lipoprotein (free fatty acids) synthesis to the liver. Unlike sis (Figure 69–1). The amount of energy stored as triglycerides
most other tissues, the brain cannot utilize fatty acids for energy in adipose tissue is substantial. For example, an adult with 15 kg
when blood glucose levels become compromised. In this case, of body fat has enough energy to support the whole body
ketone bodies provide the brain with an alternative source of energy requirements (8.37 MJ; 2,000 kcal) for about 2 months.
energy, providing close to two thirds of the brain’s energy needs
during periods of prolonged fasting and starvation.
The release of glycerol and free fatty acids from adipose tis- Protein
sue is inhibited by insulin and stimulated primarily by cate- Unlike excess fat and glucose, which are stored as fat and gly-
cholamines. During fasting, or more frequently during periods cogen in adipose tissue, liver, and muscle, there is no storage
718 SECTION IX Endocrine and Metabolic Physiology

pool for body protein. Therefore, under catabolic conditions, MAINTENANCE OF LONG-TERM
essential proteins are catabolized. Cortisol, epinephrine, and
glucagon together favor muscle protein breakdown and
ENERGY BALANCE AND FAT STORAGE
hepatic amino acid uptake, some of which can be utilized for The balanced transition from fed to fasted and the consump-
gluconeogenesis. tion of adequate energy commensurate with the level of physi-
cal activity ensure that adequate energy reserves are available
for short-term increases in metabolic demands, such as those
COUNTERREGULATION described for exercise. An imbalance in either energy intake or
TO ACUTE STRESS expenditure leads to one of the following two extremes: loss of
lean body mass or wasting syndrome and obesity. In the
During acute decreases in plasma glucose (hypoglycemia) or in absence of chronic physical or psychiatric illness, development
response to acute stress, the role of the counterregulatory hor- of a wasting syndrome is infrequent. Obesity is an important
mones glucagon, epinephrine, growth hormone, and cortisol health problem that increases the risk of several diseases.
becomes evident. Glucagon’s primary role is to stimulate hepatic Because of the rising incidence of obesity in our society, a brief
gluconeogenesis and glycogenolysis, resulting in an overall discussion follows on the endocrine physiologic responses
increase in hepatic glucose output. GH and cortisol facilitate implicated in the development of this condition.
glucose production and limit glucose utilization. Their effects
are not immediate; thus, they are mostly involved in defense
against prolonged hypoglycemia. Cortisol exerts permissive Obesity
effects on the lipolytic action of catecholamines and GH in adi- Obesity is defined as a significant increase above the ideal
pose tissue and on the glycogenolytic action of catecholamines weight. The increase in body mass index (BMI), an indicator
in skeletal muscle. In addition, cortisol induces hepatic enzy- of the adiposity or fatness that accompanies obesity, has
matic gene expression required for enhanced gluconeogenic become an important worldwide health problem. Life expec-
rates and exerts permissive effects on the stimulation of gluco- tancy is reduced when BMI is significantly increased. Obesity
neogenesis in the liver by glucagon and epinephrine. is associated with an increased risk of type 2 diabetes melli-
Epinephrine stimulates hepatic glycogenolysis and hepatic tus, dyslipidemia, hypertension, heart disease, and cancer.
and renal gluconeogenesis, largely by mobilizing gluconeo- Approximately 30% of the US population is considered obese,
genic precursors including lactate, alanine, glutamine, and according to the definition of the World Health Organization.
glycerol. It also limits glucose utilization by insulin-sensitive Body weight and the excess weight gain leading to obesity
tissues. The role of epinephrine is critical when glucagon are determined by interactions among genetic, environmental,
release is deficient. Together, glucagon and epinephrine act and psychosocial factors that affect the physiologic mediators
within minutes to raise plasma glucose concentrations. of energy intake and expenditure, several of which pertain to
The contribution of the activation of the autonomic nervous the endocrine system. Energy expended by the individual can
system is more easily understood when described in the context be in the form of work (physical activity) or heat production
of acute and severe hypoglycemia. The decrease in plasma glu- (thermogenesis), which can be affected by environmental
cose concentrations (hypoglycemia) within and below the phys- temperature, diet, and the neuroendocrine system (cate-
iologic postabsorptive concentration range of about 70–110 mg/ cholamines and thyroid hormone). The uncoupling of ATP
dL (3.9–6.1 mmol/L) triggers the activation of a counterregula- production from mitochondrial respiration dissipates heat
tory neuroendocrine response. The release of counterregula- and affects the efficiency with which the body utilizes energy
tory hormones glucagon, epinephrine, GH, and cortisol substrates. The expression of proteins involved in this process
contributes to the increase in hepatic glucose output and the (uncoupling protein-1 expressed in brown adipose tissue and
suppression of tissue glucose uptake, partly through an increase uncoupling protein-3 in skeletal muscle) is modulated by cat-
in tissue fatty acid oxidation. As plasma glucose levels are echolamines, thyroid hormones, and leptin.
restored, peripheral glucose sensors in the portal vein, small The role of genetics in the predisposition to obesity has been
intestine, and liver decrease firing. This afferent signal is trans- demonstrated convincingly. Susceptibility genes have been
mitted to the hypothalamus and to the nucleus solitarius in the identified that increase the risk of developing obesity, and their
medulla through the vagus nerve, conveying information on the relevance has been shown in studies in which pairs of twins
prevailing peripheral glucose levels. In the hypothalamus, glu- were exposed to periods of positive and negative energy bal-
cose sensors contribute to the central nervous system integra- ance. The differences in the rate of weight gain, the proportion
tion of these signals. This initiates an appropriate response of weight gained, and the sites of fat deposition showed greater
through the inhibition of hepatic and adrenal nerve activity, similarity within pairs than between pairs, indicating a close
with consequently decreased release of adrenomedullary cate- genetic relationship. Although a clear correlation between
cholamines. The decreased sympathetic activation allows hyper- energy expenditure and weight gain has not been demon-
glycemia to induce pancreatic insulin secretion. Thus, glucose strated, increasing physical activity, which represents
acts as a feedback signal contributing to integration of the neu- 20–50% of total energy expenditure, has been actively pro-
roendocrine mechanisms that regulate its homeostasis. moted as an approach to prevent obesity and improve insulin
CHAPTER 69 Endocrine Integration of Energy and Electrolyte Balance 719

responsiveness. Environmental factors are also thought to they modulate the expression of hypothalamic neuropeptides
unmask genetic tendencies toward obesity. known to regulate feeding behavior and body weight, resulting
The responsiveness to hormones that regulate lipolysis in inhibition of food intake and increase in energy expendi-
varies according to the distribution of fat depots. The lipoly- ture. While insulin release is directly correlated to meals, that
tic response to norepinephrine is greater in abdominal than of leptin does not correlate with food intake but reflects body
in gluteal or femoral adipose tissue in both men and women. fat mass (Figure 69–2).
The exaggerated release of free fatty acids from abdominal
adipocytes directly into the portal system, an increased
hepatic gluconeogenesis, and hepatic glucose release, and Hypothalamic Integration
hyperinsulinemia are hallmarks of patients with upper-body The hypothalamus receives innervation from several areas,
obesity. The endocrine properties of the different fat depots notably the nucleus tractus solitarius and area postrema in
may be more important than the anatomic location. The the brainstem. These areas relay many neural and hormonal
severity of medical complications is more closely related to signals from the gastrointestinal tract. Mechanical stretch
body fat distribution, being greater in individuals with receptors sense stretch of the stomach and other areas of the
abdominal (visceral) obesity than those with an excess total intestine. Gastrointestinal hormones such as cholecystokinin
body fat. Differential fat deposition leading to upper-body or (CCK), released following a meal in response to the presence
abdominal obesity is reflected in a high waist–hip ratio, an of lipids or protein in the intestinal lumen, are involved in
index used for predicting risks associated with fat accumula- afferent signaling to the brain regarding the intestinal nutri-
tion. The presence of visceral obesity, insulin resistance, dys- tional content. The nucleus tractus solitarius also relays taste
lipidemia, and hypertension is collectively termed the information to the hypothalamus and other centers. Other sig-
metabolic syndrome. nals regarding smell, sight, memory of food, and the social
Excess energy intake in relation to the energy expended by context under which it is ingested are also integrated and may
the organism leads to the accumulation of fat. The fat mass also influence energy intake by modulating output from the
itself is determined by the balance between breakdown hypothalamus. Integration of these signals results in the acti-
(lipolysis) and synthesis (lipogenesis). The sympathetic ner- vation of gene expression of mediators implicated in the regu-
vous system is the principal stimulator of lipolysis, particu- lation of satiety and development of obesity. These genes
larly when the energy demands of the individual are control thermogenesis (uncoupling proteins), hormone syn-
increased. When intake exceeds energy utilization, lipogen- thesis (ghrelin, leptin, and CCK and adiponectin), and neu-
esis occurs in liver and adipose tissue. Lipogenesis is influ- rotransmitter (neuropeptide Y) availability, as summarized in
enced by diet (increased by carbohydrate-rich diets) and Table 69–2.
hormones (principally GH, insulin, and leptin) through The relative contributions of these mediators to the regula-
modification of transcription factors (e.g., peroxisome pro- tion of caloric intake, energy expenditure, body weight, and fat
liferator-activated receptor-γ [PPAR-γ]). The transcription mass are not completely understood. However, important new
factor PPAR, the target for the insulin sensitizer thiazolidin- discoveries, such as the secretory function of adipose tissue,
edione drugs, affects gene transcription of several enzymes have provided new insight into potential factors contributing
involved in glucose and fat metabolism and is involved in to obesity. Adipose tissue is an endocrine tissue participating in
preadipocyte differentiation into mature fat cells. The main a complex network regulating energy homeostasis, glucose and
hormones involved in fat storage are insulin (which stimu- lipid metabolism, vascular homeostasis, immune response,
lates lipogenesis), and GH and leptin (which reduce lipogen- and even reproduction. Among the hormones identified that
esis). Other hormones involved in the regulation of body fat are produced by adipose tissue are leptin, cytokines (TNF-α,
stores include testosterone, dehydroepiandrosterone, and interleukin-6), adipsin and acylation-stimulating protein,
thyroid hormone. angiotensinogen, plasminogen activator inhibitor-1, adi-
ponectin, resistin, and steroid hormones (Table 69–2). Secre-
tion of almost all of these hormones and cytokines is
Regulation of Energy Intake dysregulated as a consequence of both excess and deficiency in
Regulation of energy intake is mediated by several factors. the mass of adipose tissue, suggesting that they are involved in
Central integration of peripheral signals, including those the pathophysiology of both obesity and cachexia. Of particu-
mediated by mechanoreceptors and chemoreceptors, signals lar interest are the contributions of the proinflammatory cytok-
the presence and energy density of food in the gastrointestinal ines, such as TNF-α, to the development of insulin resistance
tract. Hypothalamic glucose sensors monitor fluctuations in in obese individuals and the potential role of leptin as a regula-
circulating glucose concentrations. Hormones signal the cen- tor of fat mass.
tral release of peptides that regulate appetite and satiety. Two
hormones that have been identified as crucial in the long-term
regulation of energy balance are insulin and leptin, the prod- Leptin
uct of the ob gene (discussed later). Both hormones are Leptin is a peptide hormone (146 amino acids) thought to
released in proportion to body fat (Figure 69–2). In the brain, serve as an indicator of energy stores, as well as a modulator of
720 SECTION IX Endocrine and Metabolic Physiology

Arcuate
nucleus Nucleus
of the
solitary tract
Hypothalamus
Vagus
nerves
Spinal
nerves Neurons
PVN in other
hypothalamic
Orexigenic areas

Anorexigenic
Ghrelin ARC NPY/
AgRP
Leptin Insulin POMC/
CART

PYY CCK NPY-Y1R MC3 R Ghrelin R Hunger Thermogenic response


NPY-Y2R MC4 R Leptin R
NPY-Y4R Insulin R Meal size Energy stores
GLP-1R
Meal duration Substrate metabolism

Intake Expenditure

Stretch
Chemoreceptors
Nutrients

– + – +
– Insulin
Ghrelin + Leptin
PYY3-36

Stomach
Adipose tissue Pancreas
(or duodenum)

FIGURE 69–2 The brain integrates multiple peripheral and neural signals to control the regulation of energy homeostasis,
maintaining a balance between food intake and energy expenditure. The hypothalamus receives innervation from several areas, notably the
nucleus tractus solitarius and area postrema in the brainstem, that relay many neural and hormonal signals from the gastrointestinal tract, such
as mechanical signals indicating stretch of the stomach and other areas of the intestine; and hormonal signals indicating the presence of food in
the gut, such as CCK. Additional signals regarding smell, sight, memory of food, and the social context under which it is ingested are also
integrated and may also influence energy intake by modulating output from the hypothalamus. Collectively, these signals act on two subsets of
neurons that control food intake in the arcuate nucleus of the hypothalamus (ARC), which stimulate and inhibit energy intake. Orexigenic
(appetite-stimulating) neurotransmitters are agouti-related peptide (AgRP) and neuropeptide Y (NPY). Anorexigenic (appetite-suppressant)
neurotransmitters are cocaine- and amphetamine-regulated transcript (CART) and proopiomelanocortin (POMC) neurotransmitters. Both
neuronal populations innervate the paraventricular nucleus (PVN), which, in turn, sends signals to other areas of the brain. These include
hypothalamic areas such as the ventromedial nucleus, dorsomedial nucleus, and the lateral hypothalamic area, which modulate this control
system. Brain circuits integrate information from the NTS and multiple hypothalamic nuclei to regulate overall body homeostasis. Leptin and
insulin decrease appetite by inhibiting the production of neuropeptide Y (NPY) and AgRP, while stimulating melanocortin-producing neurons in
the arcuate nucleus region of the hypothalamus. NPY and AgRP stimulate eating, and melanocortins inhibit eating. Ghrelin stimulates appetite
by activating the NPY/AgRP-expressing neurons. PYY3-36, released from the colon, inhibits these neurons and transiently decreases appetite.
Integration of these signals results in regulation of energy intake, satiety, control of thermogenesis, and energy expenditure.

energy balance. The specific effects of leptin on fat metabo- • decrease in triglyceride content by increasing fatty acid
lism are as follows: oxidation;
• decrease in activity and expression of esterification and
• decrease in fat storage;
lipogenic enzymes;
• increase in sympathetic-mediated energy expenditure;
• decrease in lipogenic activity of insulin, favoring lipolysis.
• increase in expression of uncoupling proteins;
CHAPTER 69 Endocrine Integration of Energy and Electrolyte Balance 721

TABLE 69–2 Mediators implicated in regulation of energy intake.


Mediator Regulation and Target Effect

Gastrointestinal tract

Cholecystokinin Released in the duodenum during a meal. Stimulates the vagus nerve projecting
to the NTS and signals within the hypothalamus to induce satiety

Ghrelin Released from the GI tract before meals. Plasma levels are low in obese patients.
Stimulates growth hormone release, decreases fat oxidation, increases food
intake and adiposity. Overall has antileptin action

PYY3-36 Member of the neuropeptide Y family, released in the distal small intestine
and colon in response to food. Blood levels remain elevated between meals.
Decreases food intake at the arcuate nucleus in the hypothalamus

Adipose tissue

Adiponectin (adipocyte complement-related Produced by adipose tissue (decreased in obese patients; plasma levels correlate negatively
protein of 30 kDa, AdipoQ) with triglycerides). Increases insulin sensitivity and tissue fat oxidation, resulting in reduced
circulating fatty acid levels and reduced intramyocellular and liver triglyceride content

Acylation-stimulating protein Produced by adipose tissue. Paracrine signal increases efficiency of triacylglycerol
synthesis in adipocytes, resulting in more rapid postprandial lipid clearance

Leptin Produced in adipose tissue. Acts on neuropeptide Y, AgRP-containing neurons, and


α-MSH neurons in the arcuate nucleus of the hypothalamus to decrease food intake

Resistin Peptide hormone induced during adipogenesis. It antagonizes insulin action

Hypothalamus

Neuropeptide Y (NPY) Produced by hypothalamic neurons that express AgRP. Release is under leptin, insulin,
and cortisol regulation. Stimulates food intake via the NPY5 receptor

α-MSH Product of POMC in hypothalamic neuronal subset under leptin regulation.


Decreases food intake through melanocortin-4 receptors in the hypothalamus

Cocaine- and amphetamine-regulated Leptin and amphetamines stimulate production of this peptide
transcript (CART) by hypothalamic POMC-expressing neurons. Reduces food intake

Agouti-related peptide (AgRP) Released from hypothalamic NPY-expressing neurons. Inhibits


neuronal melanocortin-4 receptors and increases food intake

Orexins (A and B) Produced by neurons in the lateral hypothalamus perifornical area. Regulated
by glucose, leptin, neuropeptide Y, and POMC neurons. They stimulate food intake
NTS, nucleus tractus solitarius; GI, gastrointestinal; PYY, polypeptide YY; AgRP, agouti-related peptide; MSH, melanocyte-stimulating hormone; POMC, proopiomelanocortin.

Leptin produced by white adipose tissue functions as a sig- and amphetamine-regulated transcript (CART). Thus, lep-
nal that provides information about the level of energy stores tin-induced inhibition of food intake results from both the
(adipose tissue mass). The signal is integrated by hypotha- suppression of orexigenic and the induction of anorexigenic
lamic neurons, and an effector response, most likely involving neuropeptides (Figure 69–2).
modulation of appetite centers and sympathetic nervous sys- The feedback regulatory loop for leptin’s effects has been
tem activity, regulates the two main determinants of energy well established in rodents; however, many unsolved questions
balance: intake and expenditure. Leptin secretion exhibits a remain about its applicability to body weight regulation in
circadian rhythm, with a nocturnal rise over daytime secre- humans. The role of leptin in humans appears to be mostly
tion. These changes in leptin plasma concentrations are not one of the adaptations to low energy intake rather than a brake
influenced by meal ingestion and meal-induced increases in on overconsumption and obesity. Leptin concentrations
the circulating insulin concentration. decrease during fasting and energy-restricted diets, indepen-
The effects of leptin are mediated through the leptin recep- dent of body fat changes, stimulating an increase in food intake
tor, a member of the gp130 family of cytokine receptors, which before body energy stores become depleted. Because leptin
activates a gene transcription factor on two populations of levels do not increase in response to individual meals, it is not
hypothalamic neurons. This process results in reduced expres- thought to serve as a meal-related satiety signal. Finally, it is
sion of two orexigenic (feeding-inducing) neuropep- notable that obese individuals have high plasma leptin con-
tides, neuropeptide Y and agouti-related peptide (AgRP); centrations that do not result in the expected reduction in
and enhanced expression of two anorexigenic peptides, food intake and increase in energy expenditure, suggesting
α-melanocyte-stimulating hormone (α-MSH) and cocaine- that obesity is related to leptin resistance.
722 SECTION IX Endocrine and Metabolic Physiology

Ghrelin stimulate the release of renin from juxtaglomerular cells in the


afferent and efferent arterioles.
Ghrelin is a hormone produced by the enteroendocrine cells
of the stomach, and to a lesser extent by the pituitary, and
hypothalamus. Circulating levels of ghrelin decrease during Antidiuretic hormone ADH directly controls water ex-
meals and are highest in the fasted state. Ghrelin levels are cretion by the kidneys. ADH secretion and compensatory
decreased in obese individuals and increased in individuals thirst are stimulated by hypothalamic osmoreceptors and by
consuming low calorie diets, chronic strenuous exercise, can- decreased stimulation of aortic and carotid stretch receptors.
cer anorexia, and anorexia nervosa. In humans, ghrelin has Release of ADH is inhibited by increased stretch of mechano-
been shown to be a potent growth hormone secretagogue and receptors (stretch receptors) in the atria of the heart. ADH
appetite stimulant. stimulates insertion of aquaporins into the cell membrane, in-
creases water reabsorption in the collecting ducts, and concen-
trates excreted urine.
ELECTROLYTE BALANCE
Angiotensin II Angiotensin II increases blood pressure by
REGULATION OF SODIUM BALANCE several mechanisms, including direct vasoconstriction, poten-
tiation of the activity of the sympathetic nervous system at both
Sodium is the primary electrolyte that regulates extracellular the central and peripheral levels, stimulation of aldosterone
fluid (ECF) levels and osmolarity in the body. Regulation of synthesis and release with consequent sodium reabsorption by
extracellular Na+ concentration controls the distribution of the kidney, stimulation of ADH release and increased water re-
water between the ECF and intracellular fluid (ICF) and main- tention, and intrarenal efferent arteriolar constriction.
tains cell volume, ensuring normal physiologic function.
Sodium is maintained in the ECF by the action of Na+/K+-
Aldosterone Aldosterone increases sodium reabsorption and
ATPase, whereas water crosses cell membranes through ubiq-
potassium excretion in the distal tubule and the collecting duct of
uitously expressed aquaporins (maintaining ICF and ECF
the nephron, playing a central role in determining total body Na+
isotonicity). The overall mass of Na+ is under aldosterone reg-
mass, and thus long-term blood pressure regulation. Aldosterone
ulation, whereas the Na+ concentration in plasma is under
release from the adrenals is stimulated by angiotensin II.
antidiuretic hormone (ADH) regulation. Thus, low Na+ con-
centrations do not necessarily mean that total Na+ mass is low.
In chronic heart failure, osmolarity can be low, yet Na+ mass Atrial natriuretic factor Atrial natriuretic factor is a pep-
can be high because of excess water and Na+ in the ECF, with tide hormone produced in atrial myocardial cells and released in
greater increases in total body water than in Na+ mass. response to increased stretch, usually resulting from increased
intravascular volume. It increases renal Na+ excretion and water
loss through an increase in glomerular filtration rate and a de-
Hormonal Regulation crease in Na+ reabsorption in the medullary collecting duct.
of Sodium and Water Balance
The system that controls total body water is a negative feed- Abnormalities in Sodium
back homeostatic mechanism, of which thirst and ADH are
the major effectors. Two stimuli regulate the system: tonicity and Water Balance
of the ECF through osmoreceptors and intravascular volume Abnormalities in sodium and water balance can be classified
through stretch or baroreceptors. into four categories. Excess Na+ is characterized by expansion
The system works primarily to maintain intravascular vol- of the ECF volume and frequently by low effective circulating
ume and to a lesser extent to maintain tonicity. Thirst is stimu- blood volume (i.e., heart failure, hypoalbuminemia, renal
lated by an increase in tonicity (1–2% changes are sufficient to insufficiency). Deficit in Na+ is characterized by reduced ECF
elicit thirst) and by reductions in the ECF volume. Water intake volume. Excess water is due to either excess intake or
is inhibited by hypotonicity and ECF volume expansion. enhanced ADH release and is manifested by hyponatremia
Sudden decreases in blood volume are sensed by mechano- and hypoosmolarity. Water deficit is due to lack of intake or
receptors in the left ventricle, carotid sinus, aortic arch, and excess loss (renal and nonrenal) and is manifested by hyper-
renal afferent arterioles (Figure 69–3). These mechanorecep- natremia and hyperosmolarity.
tors respond to decreased stretch resulting from decreases in
systemic arterial pressure, stroke volume, renal perfusion, or
peripheral vascular resistance by triggering an increase in REGULATION OF POTASSIUM BALANCE
sympathetic outflow from the central nervous system, activa-
tion of the renin–angiotensin–aldosterone system, and Potassium is the most abundant cation in the body and the
nonosmotic release of antidiuretic hormone, as well as stimu- main intracellular electrolyte. Most (98%) of the potassium in
lation of thirst. Low blood pressure results in decreased renal the body is sequestered within cells. The ratio of extracellular to
perfusion pressure and lower glomerular filtration rates, which intracellular potassium (1:10) is maintained by a sodium–
CHAPTER 69 Endocrine Integration of Energy and Electrolyte Balance 723

Cardioregulatory
center

Glossopharyngeal
& vagal afferents
from baroreceptors
Sympathetic trunk

Sympathetic ganglia

AVP
Sympathetic
nerves
Aldosterone FIGURE 69–3 Neuroendocrine control
of blood volume. Sudden decreases in blood
volume are sensed by mechanoreceptors in the
left ventricle, carotid sinus, aortic arch, and renal
afferent arterioles, triggering an increase in
Renin
Angiotensin II sympathetic outflow from the central nervous
production system, activation of the renin–angiotensin–
Peripheral
vasoconstriction aldosterone system, nonosmotic release of
arginine vasopressin (AVP), and stimulation of
thirst. The decreases in renal perfusion pressure
and glomerular filtration rates stimulate the
release of renin, the enzyme responsible for
the conversion of angiotensinogen to
angiotensin I (later converted by angiotensin-
converting enzyme to angiotensin II).
Angiotensin II, aldosterone, and ADH produce
vasoconstriction, venoconstriction, and renal
Water reabsorption retention of Na+ and water. (Modified with
Na reabsorption permission from Molina PE: Endocrine Physiology, 3rd ed.
K+ excretion New York: McGraw-Hill Medical, 2010.)

potassium pump and is the major determinant of resting mem- ment. Only a small fraction (10%) of potassium is excreted
brane potential. Small losses (1%, or 35 mmol) of total body through the gastrointestinal tract and the majority is excreted by
potassium content can seriously disturb the delicate balance the kidney. Thus, the kidney is responsible for long-term potas-
between intracellular and extracellular potassium and can result sium homeostasis, as well as for regulating the serum potassium
in profound physiologic changes. The tissues most severely concentration. On a short-term basis, serum potassium is also
affected by potassium imbalance are muscle and renal tubular regulated by the shift of potassium between the ICF and ECF.
cells. Manifestations of hypokalemia include generalized mus- This short-term regulation of serum potassium is principally
cle weakness, paralytic ileus, and cardiac arrhythmias. controlled by insulin and catecholamines through regulation of
the transcellular distribution of potassium. Dietary potassium,
which is rapidly absorbed by the gut, increases serum potassium
Intake, Distribution, and transiently. The release of insulin and catecholamines during a
Excretion of Potassium meal quickly shifts the potassium into the cells.
The daily intake of potassium in the Western diet is about The principal site for the regulation of K+ excretion is the
80–120 mmol, and this exceeds the minimum daily require- distal tubule, where secretion is indirectly but tightly coupled
724 SECTION IX Endocrine and Metabolic Physiology

to sodium reabsorption via the amiloride-sensitive sodium result is an increase in Na+ reabsorption and an increase in K+
channel, and under regulation by aldosterone. Because the excretion.
kidney is the major regulator of potassium homeostasis, renal
dysfunction results in abnormal levels of serum potassium. Insulin Insulin stimulates entry of K+ into the cell through
Potassium contributes to regulation of its balance by stimu- activation of the electroneutral Na+/H+ antiporter, leading to
lating aldosterone secretion by the zona glomerulosa cells of Na influx. The increase in intracellular Na+ produced by insu-
the adrenal cortex. Aldosterone enhances renal and colonic lin triggers the activation of the electrogenic Na+/K+-ATPase,
K+ secretion, promoting the loss of K+ in the urine and stool which extrudes Na+ from the cell in exchange for K+. The treat-
(Figure 69–4). Sustained hyperkalemia does not occur in ment of patients with diabetic ketoacidosis with high insulin
individuals with normal renal function despite marked doses produces a significant influx of K+ into the cells that may
increases in potassium intake because of an adaptive change in result in hypokalemia, manifested by changes in the electro-
distal tubular K+ secretion, such that intake is matched by cardiogram.
rapid and equivalent increases in K+ excretion. The mecha-
nisms involved in the chronic adaptation to increased levels of
K+ include changes in apical K+ and Na+ conductance and in Catecholamines Catecholamines (β-adrenergic receptor
basolateral Na+/K+-ATPase pump activity, an increase in api- stimulation) increase cellular potassium uptake by stimulating
cal Na+ delivery and reabsorption, and an increase in K+ excre- cell membrane Na+/K+-ATPase. Indirectly, catecholamines
tion per nephron to match K+ intake. stimulate glycogenolysis, resulting in a rise in plasma glucose
concentrations, release of insulin from the pancreas, and insu-
lin-mediated effects on K+ redistribution. Stimulation of the
Hormonal Regulation α-adrenergic receptor shifts K+ out of the cell and can also af-
of Potassium Balance fect K+ distribution through inhibition of pancreatic insulin
Total body stores of potassium and its cellular distribution in release.
the body are closely regulated by key hormones. Insulin and catecholamines are both stimulated by the inges-
tion of glucose- and potassium-rich foods, thereby maintain-
Aldosterone Aldosterone increases the synthesis and activity ing K+ homeostasis despite large dietary intake. These hormones
of Na+/K+-ATPase in the basolateral membrane of the distal tu- are essential in moving potassium primarily into the intracel-
bule, promoting the exchange of cytosolic Na+ for K+. The overall lular compartment of the liver and striated muscle cells.

β-Adrenergic Potassium concentration in extracellular fluid


catecholamines
Insulin Aldosterone
Cortical
Na+ collecting-
K+
duct cells K+
Na+
Na+
H+
Na+/K+ Na+/K+
-ATPase Na+ -ATPase
Na+
K+ K+

Na+

Na+
K+
Muscle Na+
cell
Lumen

A B
FIGURE 69–4 Key hormones involved in normal potassium homeostasis. A) Insulin stimulates entry of K+ into the cell through the
activation of the electroneutral Na+/H+ antiporter. The increase in intracellular Na+ produced by insulin triggers the activation of the electrogenic
Na+/K+-ATPase, which extrudes Na+ from the cell in exchange for K+. Catecholamines (β-adrenergic receptor stimulation) increase cellular
potassium uptake by stimulating cell membrane Na+/K+-ATPase. Stimulation of the α-adrenergic receptor shifts K+ out of the cell. B) Aldosterone
promotes potassium excretion through its effects on the Na+/K+-adenosine triphosphatase (ATPase) and epithelial sodium and potassium
channels in collecting duct cells. Angiotensin II has a synergistic effect on the stimulation of aldosterone production induced by hyperkalemia.
(Reproduced with permission from Gennari F. Current concepts: Hypokalemia. NEJM. 1998;339:451. Copyright Massachusetts Medical Society. All Rights reserved.)
CHAPTER 69 Endocrine Integration of Energy and Electrolyte Balance 725

Acid–Base and Osmolar Regulation dysregulated state are mediated by chronic activation of the
of Potassium Distribution hypothalamic–pituitary–adrenal (HPA) axis and the sympa-
thetic nervous system, producing marked alterations in endo-
Intracellular potassium homeostasis is also affected by changes crine function, such as the following:
in acid–base balance and osmolarity. Sudden changes in
plasma osmolarity redistribute water between the ICF and • Inhibition of reproduction function: Enhanced release of
ECF. This movement of water out of a cell creates a solvent corticotropin-releasing hormone (CRH) and β-endorphin
drag phenomenon, pulling K+ out of the cell and therefore suppresses GnRH release directly and indirectly through
increasing serum potassium. Similarly, metabolic acidosis the release of glucocorticoids. Glucocorticoids decrease the
caused by a loss of bicarbonate or a gain in hydrogen ion con- release of luteinizing hormone and produce gonadotropin
centration [H+] leads to a shift of K+ across cell membranes resistance at the gonads. This suppression in gonadal func-
and hyperkalemia. However, integrity of renal function and tion is evident in patients with anorexia nervosa, athletes,
stimulation of aldosterone release rapidly correct this imbal- and ballet dancers.
ance. In diabetic ketoacidosis, there is a net loss of K+ from the • Inhibition of the GH–IGF-I axis: Chronic activation of the
body because of osmotic diuresis, despite elevations in ECF K+ HPA axis suppresses GH release and inhibits the effects of
concentrations (hyperkalemia), because of insulin deficiency. IGF-I at target tissues.
Following aggressive insulin treatment, hypokalemia becomes • Suppression of thyroid function: CRH and cortisol sup-
apparent. Opposite effects are observed during alkalosis. In press the production of thyroid-stimulating hormone and
metabolic alkalosis, the excess bicarbonate causes H+ in the inhibit the activity of peripheral 5ʹ-deiodinase, leading to
ECF to fall, leading to entry of Na+ into the cell in exchange for the euthyroid sick syndrome.
H+. Na+ is pumped out of the cell by the Na+/K+-ATPase in • Dysregulation of energy substrate metabolism: An in-
exchange for K+ movement into the cell, creating a shift of K+ crease in catecholamines stimulates lipolysis and decreases
into the cells. triglyceride synthesis in white adipose tissue. In the liver,
Hypokalemia is a common electrolyte abnormality encoun- increased epinephrine levels stimulate hepatic glycogenoly-
tered in clinical practice. It is almost always the result of potas- sis and, together with high cortisol levels, increase hepatic
sium depletion induced by abnormal fluid losses (i.e., vomiting, glucose output. High cortisol levels resulting from activa-
colonic diarrhea, profuse sweating, diuretic use, or nasogastric tion of the HPA increase gluconeogenesis, produce insulin
suction). Patients present with muscle weakness and changes resistance in peripheral tissues, inhibit the lipolytic action of
in the electrocardiogram. More rarely, hypokalemia occurs GH, and inhibit bone osteoblastic activation (remodeling)
because of an abrupt shift of potassium from the ECF into by sex steroids. This leads to increases in visceral adiposity
cells, frequently as an effect of prescription drugs. Hyper- and loss of BMD and lean body mass. This aspect of the
kalemia, also a common electrolyte disorder, is caused by stress response may be of particular importance in the treat-
renal dysfunction, decreased aldosterone production by the ment of diabetic patients during stressful periods such as
adrenal gland, potassium shifting from the intracellular to the surgery or infection.
extracellular compartment, and some drugs. Patients can pres- • Alterations in the Immune Response: The significant in-
ent asymptomatic or have altered electrocardiogram. crease in circulating cortisol levels affects virtually all as-
pects of the immune response, including cytokine pro-
duction, leukocyte trafficking and recruitment, and
NEUROENDOCRINE REGULATION production of chemokines. Overall, glucocorticoids exert
an anti-inflammatory response. Activation of the auto-
OF THE STRESS RESPONSE nomic nervous system also affects the immune response
through effects on neutrophil demargination and cytokine
Alterations in the environment or in the host that require
production.
adaptation involve the synchronized interaction of virtually all
aspects of neuroendocrine function that have been described Short-term activation of these stress response mechanisms
(Figure 69–5). The adaptation to a biologic, psychosocial, or ensures that energy substrates are available to meet the increased
environmental insult to the host is referred to as the stress metabolic demands of the individual. However, prolonged
response; in the acute setting, it is also termed the fight-or- duration and increased magnitude of these activities lead to
flight response. It is now clear that this stress response can be erosion of lean body mass and tissue injury. Nevertheless,
chronic, with a significant cost to the health of the individual impaired activation or lack of responsiveness of the HPA and
(Figure 69–5). autonomic nervous system can also be deleterious, as in the
Chronic activation of the mechanisms that restore homeo- case of the critically ill patient. Thus, the overall regulation of
stasis results in excessive and, in some cases, inadequate the neuroendocrine responses that mediate the physiologic
responses that ultimately alter the function of virtually all functions involved in maintaining and restoring homeostasis is
organ systems (e.g., hypertension, autoimmune disorders, critically important in situations such as illness, trauma, sur-
metabolic syndrome) (Figure 69–5). Many of the effects of this gery, or fasting.
726 SECTION IX Endocrine and Metabolic Physiology

Stress
Developmental history
Nutrition
Genetic variation
Aging

CRH Stress system LC


ADH NE

Systemic sympathetic
HPA axis adrenomedullary systems

Cortisol NE, E
Target tissues
GH and/or
IGF-I Metabolic syndrome
(insulin resistance, visceral obesity, sarcopenia) Sleep apnea
LH, T, E2
TSH, T3
TG APR ABP
LDL Cytokines Coagulopathy
Osteopenia
Polycystic HDL
or
ovary syndrome
osteoporosis

Endothelial dysfunction
and inflammation

Atherosclerosis
Cardiovascular and
neurovascular disease

FIGURE 69–5 Neuroendocrine responses to chronic or severe stress. Chronic activation of the neuroendocrine response to restore
homeostasis influences virtually all organ systems. The short-term activation of these stress response mechanisms ensures that energy
substrates are available to meet the increased metabolic demands of the individual. However, prolonged duration and increased magnitude
of these activities lead to erosion of lean body mass and tissue injury. ABP, arterial blood pressure; ACTH, adrenocorticotropic hormone; APR,
acute-phase reactants; ADH, antidiuretic hormone; CRH, corticotropin-releasing hormone; iCRH, immune CRH; E, epinephrine; E2, estradiol; GH,
growth hormone; HPA, hypothalamic–pituitary–adrenal; IGF-I, insulinlike growth factor I; IL-6, interleukin 6; LC, locus ceruleus; LH, luteinizing
hormone; NE, norepinephrine; T, testosterone; TG, triglycerides. (Adapted with permission from Chrousos G. Stress and disorders of the stress system. Nat Rev
Endocrinol. 2009;5(7):347–381, 2009.)

CLINICAL CORRELATION night time, over time diminishing the autonomic responses
to hypoglycemic episodes. Hypoglycemia deprives the
A woman with type 1 diabetes mellitus on an insulin brain of its preferred substrate, glucose, leading to rapid
pump is brought to the emergency room after she fainted activation of neuroendocrine responses aimed at restoring
during her daily exercise class. First responders adminis- glycemia including increased release of glucagon, epi-
tered intravenous glucose solutions after obtaining a nephrine, and growth hormone. Together, these counter-
blood sample for analysis. On examination, she is tachy- regulatory hormones increase hepatic glucose output
cardic and agitated, and has sweaty palms. Laboratory (from gluconeogenesis and glycogenolysis) and peripheral
values showed high insulin and low plasma glucose levels. glycogenolysis and lipolysis. The activation of the sympa-
A diagnosis of insulin-induced hypoglycemia is made. thetic nervous system leads to increased heart rate. Insu-
Diabetic patients treated with insulin are at risk for lin-induced hypoglycemic episodes are more frequent in
developing insulin-induced hypoglycemia. These epi- patients treated with insulin than in those treated with
sodes are sometimes asymptomatic and occur during oral hypoglycemics.
CHAPTER 69 Endocrine Integration of Energy and Electrolyte Balance 727

CHAPTER SUMMARY 2. Which of the following processes takes place immediately after a
balanced meal?
■ Energy substrate mobilization, utilization, and storage are
A) suppression of pancreatic insulin release
under neuroendocrine regulation.
B) increased muscle and fat glucose uptake
■ Hepatic glycogen and adipose tissue triglycerides are the C) increased hepatic glycogenolysis
principal sites of energy storage. D) suppressed lipogenesis
■ The central nervous system integrates the counterregulatory 3. Activation of the renin–angiotensin–aldosterone system during
response to acute decreases in energy substrate availability. loss of effective intravascular volume results in all of the
■ Regulation of sodium balance determines blood volume and following except
blood pressure control. A) increased renal sodium and fluid retention
■ The kidney is responsible for long-term potassium homeostasis B) potentiation of the activity of the sympathetic nervous
and serum potassium concentration. system
■ Insulin and catecholamines regulate the cellular distribution C) peripheral venodilatation
of potassium. D) enhanced ADH release
4. Regulation of body potassium content and distribution can be
affected by all of the following except
STUDY QUESTIONS A) aldosterone-induced increase in K+ excretion
B) insulin stimulation of intracellular K+ efflux
1. Which of the following neuroendocrine responses contributes to C) β-adrenergic stimulation of cell membrane Na+/K+-ATPase
meeting the enhanced energy demands during exercise? D) sudden changes in plasma osmolarity
A) glucagon stimulation of hepatic glycogen synthesis
B) epinephrine stimulation of hepatic glycogenolysis
C) norepinephrine-induced stimulation of insulin release
D) cortisol inhibition of gluconeogenesis
This page intentionally left blank
SECTION X INTEGRATIVE PHYSIOLOGY

70
C H A P T E R

Control of Body
Temperature
Hershel Raff and Michael Levitzky

O B J E C T I V E S

■ Understand the mass balance characteristics of the control of internal body


temperature.
■ List and define the four mechanisms of heat transfer from the skin to the
environment.
■ Explain the feedback control of internal body temperature.
■ Understand the short-term response to cold (to increase heat production and
minimize heat loss) and heat (to decrease heat production and maximize
heat loss).
■ Describe the adaptations to cold and warm environments.
■ Understand the mechanisms of fever.

Like all mammals, humans are endotherms, meaning they gen- tem (changes in environmental temperature) can be very large
erate their own internal heat. Humans are also homeotherms— compared with the maintenance of a stable internal body tem-
they maintain body temperature within a narrow range despite perature. In fact, the gain of the body temperature control
large swings in environmental temperature. The maintenance system is 25–30—compare this with the gain of only 4 for the
of internal body temperature within a narrow range is therefore blood pressure restoration in response to moderate hemorrhage
one of the most important regulated variables in humans. This shown in Figure 1–5. Furthermore, there is usually a daily (cir-
is because enzymatic reactions, and optimal cell and organ cadian) rhythm in body temperature with a low point in the
function, occur in a fairly small range of temperatures. Despite early morning and a high point in the early evening. Body tem-
wide swings in environmental temperature, the core body tem- perature also varies depending on the level of muscular activity
perature—the internal temperature in the organs such as the and with the menstrual cycle in women.
liver (often estimated by rectal or tympanic membrane The core body temperature is the central compartment of a
temperature)—is usually maintained within ±0.6°C (±1.0°F). mass balance system (see Figure 1–4) composed of heat gained
The core temperature averages about 37°C (98.6°F) for humans, from the environment and produced in the body by cellular
although it varies from person to person. The maintenance of a metabolism, and the heat lost to the environment. Since body
stable body temperature involves a negative feedback control temperature is usually quite stable in the steady state, heat pro-
system with a very high gain since the perturbation to the sys- duction is approximately equal to heat loss.

729

Ch70_729-734.indd 729 11/26/10 10:39:16 AM


730 SECTION X Integrative Physiology

MECHANISMS EFFECTOR MECHANISMS


OF HEAT LOSS OR GAIN REGULATING TEMPERATURE
Most of the gain or loss of heat between the body and the envi- Body temperature can be increased when the body is cold by
ronment is through the skin. Heat is mainly transferred to the decreasing heat loss and/or by increasing heat production.
skin from the internal environment by the circulatory system. Decreasing heat loss is accomplished primarily by vasoconstric-
There are four general mechanisms of heat transfer between tion of the arterioles of the skin, minimizing heat transfer to the
the body and the environment. Radiation is the emission of environment. Another mechanism of heat conservation is the
heat to and from the skin by electromagnetic waves—the rate reduction of body surface area by, for example, curling up, and
of the temperature transfer by radiation is proportional to the behavioral responses such as putting on warm clothes or mov-
temperature difference between the body surface and the envi- ing to a warmer environment. As mentioned above, the short-
ronment. Conduction is intermolecular thermal heat transfer term increase in heat production is accomplished primarily by
and usually occurs between the skin and air. One loses heat an increase in voluntary movement and by shivering.
more rapidly when immersed in water because conduction Body temperature can be decreased when the body is hot by
between the skin and water is faster than that between skin and increasing heat loss and/or by decreasing heat production.
air. Convection is the loss or gain of heat by the movement of Increasing heat loss is accomplished primarily by vasodilation of
air or water over the body. Because heat rises, air carries heat arterioles in the skin, thereby increasing heat transfer from the
away from the body by convection. This is one reason why blood to the skin and then to the environment, and by sweating
having a fan circulating air in a room helps one to keep cool on to lose heat by increased evaporation. Sweat production is
a hot day. Finally, evaporation of water from the skin and the increased by increasing activity of autonomic nerves innervat-
respiratory tract can carry a large amount of heat generated by ing sweat glands in the skin. The principal mechanism to
the body because of the amount of heat required to transform decrease heat production is a decrease in voluntary movement.
water from the liquid to the gas phase. Air circulation also Now let us return to Figure 70–1 to summarize the control
improves the rate of evaporation of sweat from the skin. of body temperature. When first exposed to a cold environ-
ment, the thermal sensors in the skin signal the brain to
decrease blood flow to and sweat production from the skin,
FEEDBACK MECHANISMS thus minimizing heat loss. If the core temperature decreases,
shivering and increases in adrenal medullary epinephrine can
REGULATING BODY TEMPERATURE increase to increase heat production, although the latter is a
minor pathway in humans. When one is exposed to a hot envi-
As mentioned above, the control of the internal body tempera-
ronment, thermal sensors in the skin can increase blood flow
ture is a classic example of negative feedback control. If you
to the skin and sweat production, thus increasing heat loss. If
recall from Chapter 1, feedback control systems involve sen-
core temperature increases, sweat production can increase
sors that detect the regulated variable (in this case, body tem-
profusely to greatly increase heat loss by evaporation. How-
perature) with afferent input to the controller, a controller (in
ever, if evaporation cannot occur or is difficult because the air
this case, in the brain), and efferent input to the effectors that
is already saturated or nearly saturated with water vapor,
can alter the rate of heat gain or loss.
sweating is not a very effective means of heat loss.
Figure 70–1 summarizes the regulation of body temperature.
The sensors are located in the skin (peripheral thermorecep-
tors) and the brain (central thermoreceptors), primarily in
the hypothalamus. It is the central thermoreceptors that sense ADAPTATION TO A HOT
core temperature and provide input to control body tempera- OR COLD ENVIRONMENT
ture, whereas the peripheral thermoreceptors provide the brain
with information about changes in environmental tempera- Humans can adapt to a variety of environments. You will learn
ture. The main temperature control center is also located in the in the next chapter that humans can adapt when chronically
hypothalamus. The most important effectors are the sympa- exposed to low oxygen (hypoxia) at altitude. Humans also
thetic nerves to sweat glands, arterioles of the skin, and the have the ability to adapt to changes in environmental tempera-
adrenal medulla, as well as motor neurons to skeletal muscles. ture. Adaptation to a hot environment is much better under-
Heat production in humans is usually by metabolism. The stood than adaptation to a cold environment.
basal metabolic rate can be altered by circulating thyroid hor- Most people have difficulty when first exposed to a very hot
mone (see Chapter 63), and by shivering thermogenesis, environment and have difficulty exercising. The core tempera-
driven by innervation of skeletal muscle. Shivering is the ture can increase and a feeling of malaise and weakness can
rhythmic, involuntary contraction and relaxation of skeletal ensue. Heat stroke can occur if core temperature increases
muscles that generates heat due to increased metabolic rate. Of above 41°C and body functions begin to fail. After days to
course, one can voluntarily increase heat production from weeks of exposure to a hot environment, people do better—this
skeletal muscle with movement. process of acclimatization allows us to improve performance
CHAPTER 70 Control of Body Temperature 731

Voluntary motor responses

Begin
Begin
Cerebral cortex
Skin temperature Core temperature

Peripheral Central
thermoreceptors thermoreceptors

Hypothalamus
Involuntary motor responses
Via
sympathetic Via
nerves motor
nerves

Adrenal medulla Sweat glands Skin arterioles Skeletal muscles

Epinephrine

FIGURE 70–1 Summary of temperature-regulating mechanisms beginning with peripheral and central thermoreceptors. The
dashed arrow from the adrenal medulla indicates that this pathway is of minor importance in humans. (Reproduced with permission from Widmaier EP,
Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)

during hot weather. When the environment is warmer than


skin temperature, the only efficient way to lose heat is by
FEVER
increased evaporation from sweating. Over a few months in a Fever is an increase in body temperature that occurs usually
hot environment, the ability to sweat increases dramatically due to a pathophysiological process such as infection. It is due
(up to a 4-fold increase). Since sweat contains electrolytes, and to an increase in the hypothalamic set point for temperature
specifically sodium, an increase in blood aldosterone occurs, so that body temperature increases to a new plateau at which
which not only increases sodium reabsorption in the kidney body temperature is then regulated. Although infection is the
(see Chapter 44), but also increases sodium and chloride reab- most common cause of fever, there are other etiologies includ-
sorption from sweat in the sweat glands. The increase in aldos- ing a form of severe hyperthyroidism called thyroid storm (see
terone can lead to hypokalemia due to increased renal Chapter 63).
potassium excretion and loss of potassium in sweat, however, Figure 70–2 summarizes the development of fever due to
so increased dietary potassium intake is important during infection. Pyrogens are circulating factors that cause fever.
acclimatization to a hot environment. This is particularly true Although some bacteria release exogenous pyrogens such as
when working or exercising. You have probably observed mar- lipopolysaccharides (endotoxins), the unifying cause of fever
athon runners lose the ability to function when performing during infection is the release of small proteins called endog-
heroic exercise in a hot environment, particularly when they enous pyrogens from macrophages and other immune cells.
have not had time to acclimatize to hot weather. Macrophages activated during infection are an integral part of
The adaptation to a cold environment is not as well under- the immune response. Examples of endogenous pyrogens are
stood particularly since modern humans have the ability to cytokines such as the interleukins and tumor necrosis factor.
wear highly efficient warm clothing and to heat their environ- These endogenous pyrogens can activate vagal afferents to the
ments. It is known that constriction of the blood vessels in the hypothalamus, and can also circulate to the brain to directly
skin and the inhibition of sweat production does minimize heat alter the hypothalamic set point for temperature. Since these
lost to a cold environment. There may be the ability to increase pyrogens are peptides, they must be able to signal the hypo-
basal metabolic rate, although the role of changes in hormones thalamus despite the existence of the blood–brain barrier.
such as catecholamines and thyroid hormone in this response One theory to explain this is that the areas in the brain that can
is controversial. Probably the most common cause of lethal sense cytokines, such as the circumventricular organs near
hypothermia is immersion in cold water for an extended the hypothalamus, have a “leaky” blood–brain barrier (see
period of time. Wearing a wetsuit is one way to avoid this. Chapter 27). Endogenous pyrogens can be sensed and the
732 SECTION X Integrative Physiology

Infection
to curling up increase body temperature in the same way as
exposure to cold. Although it is not known exactly why an
increase in body temperature is beneficial during infection, it
is thought that some of the cells of the immune system operate
more efficiently at higher body temperature. When a fever
Liver Multiple organs
Macrophages Macrophages
“breaks” and the set point of the hypothalamus returns to nor-
mal, the body must dissipate heat to return body temperature
to normal. Therefore, profuse sweating can occur as one recov-
Secrete Secrete
endogenous pyrogens endogenous pyrogens
ers from the acute phase of an infection.
(IL–1, IL–6, ? others) (IL–1, IL–6, ? others) There are a variety of drugs that can be used to lower tem-
perature during a fever. Aspirin inhibits prostaglandin synthe-
Firing of neural sis throughout the body. In addition to the action of
receptors Plasma IL–1, IL–6, ? others prostaglandins in the resetting of the hypothalamic set point
Vagus Systemic
for temperature, local production of prostaglandins is thought
nerve circulation to be involved in the muscle and joint aches that occur with
fever, which is why aspirin is effective in treating those symp-
Hypothalamus toms. Acetaminophen is a potent inhibitor of prostaglandin
Temperature setpoint
synthesis within the brain, so it also is effective in treating
fever. Glucocorticoid therapy with drugs such as prednisone
can also inhibit fever due to their inhibition of the immune
response (see Table 65–2), as well as their inhibition of prosta-
glandin synthesis. If fever is long-lasting and excessive (>40°C
Skeletal muscles Skin arterioles
Curl up, Vasoconstriction [>104°F]), tissue damage and organ failure can ensue, so
put on clothes heroic measures may be instituted to lower body temperature.
Shivering and blankets An example of this is immersion in cold water to decrease
body temperature. Finally, high fever can be much more dan-
gerous in the elderly.
Heat production Heat loss

CLINICAL CORRELATION
A 32-year-old woman is brought into an emergency room
Heat production greater than heat loss by her husband. She is confused, and complaining of palpi-
tations, (rapid, strong heart beats) shortness of breath, and
Heat retention
excessive sweating. Her blood pressure and heart rate are
markedly increased and her body temperature is danger-
ously high at 40.3°C. The woman is placed on a cooling bed
Body temperature to try to lower her body temperature. The emergency room
doctor suspects an infection and orders the appropriate
FIGURE 70–2 Pathways by which infection causes fever. IL-1,
interleukin 1; IL-6, interleukin 6. (Reproduced with permission from Widmaier
blood testing. However, a medical student notices that the
EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
woman’s eyes are bulging out and immediately suspects
that the woman may have severe thyrotoxicosis (a patho-
logical increase in thyroid hormone secretion). When the
student interviews the patient and her husband, it is discov-
appropriate change in hypothalamic set point occurs, possibly ered that the woman always feels warm in a cold room and
due to release of prostaglandins in or near the hypothalamic often has palpitations. Blood tests are immediately ordered
controller that results in an alteration in the set point for body and show a suppressed serum thyroid-stimulating hor-
temperature. mone (TSH) and an increased serum thyroxine level indi-
Once the hypothalamic set point is increased, the body tem- cating primary hyperthyroidism.
perature is lower than the set point, so the hypothalamus Hyperthyroidism is a very common disease, particularly
“thinks” that the body is too cold. The brain then activates all in women. By far the most common cause is primary
of the mechanisms previously described to increase body tem- hyperthyroidism defined as an increase in thyroid gland
perature when cold. This is why one can feel cold just before function independent of TSH, the anterior pituitary hor-
and during the onset of fever despite the fact that body tem- mone that controls most aspects of thyroid hormone syn-
perature is normal or even increasing. The increase in heat thesis and gland function. The most common cause of
production due to shivering and the decrease in heat loss due
CHAPTER 70 Control of Body Temperature 733

■ Feedback control of body temperature involves sensors


primary hyperthyroidism is Graves disease (see Chapter (thermoreceptors) in the skin and hypothalamus, a central
63). It is caused by an autoimmune phenomenon in which controller in the hypothalamus, and effectors consisting of
antibodies that activate the TSH receptor on the thyroid autonomic (sympathetic) control of blood flow to the skin and
follicular cell are produced and stimulate thyroid hor- sweat production.
mone synthesis and secretion just as TSH normally would. ■ Adaptation to hot weather involves increasing the capacity to
The increase in thyroid hormone in the blood then sup- sweat to lose heat by evaporation.
presses pituitary TSH release due to negative feedback. ■ Fever due to infection results from pyrogens released from
The immune dysfunction can also result in a lymphocytic bacteria and from macrophages—these pyrogens increase the
infiltration of the muscles and fat behind the eyes. The set point of the hypothalamic controller such that heat
resultant swelling pushes the eyes forward (away from the production and body temperature increase.
eye socket) resulting in the eyes bulging out—this is called
proptosis or exophthalmos. To confirm the cause of thyro-
toxicosis is Graves disease, a radioactive iodine uptake test STUDY QUESTIONS
is performed by having the patient ingest a small amount 1. Which of the following will increase body temperature?
of radioactive iodine to measure the amount and distribu-
A) an increase in heat loss from the skin to the environment
tion of iodine that accumulates in the thyroid gland. Iodine B) a decrease in basal metabolic rate
is required for the synthesis of thyroid hormone and its C) a decrease in circulating thyroid hormone
uptake is stimulated by activation of the TSH receptor (see D) shivering
Table 63–1 and Figure 63–3). The iodine uptake is uni- 2. All of the following are mechanisms to increase heat transfer
formly increased in Graves disease and in our patient. The from the skin to the environment except
combination of the blood tests and uniformly increased A) vasoconstriction
thyroid uptake, plus other tests and physical findings such B) radiation
as proptosis, strongly suggests that the patient has Graves C) conduction
disease. In fact, her increase in body temperature, her D) convection
mental confusion, and other symptoms define her as the 3. Which of the following is false?
most extreme form of thyrotoxicosis called thyroid storm. A) Sweating increases due to an increase in sympathetic
The short-term goal in treating this patient is decreasing nerve input.
her body temperature and heart rate. This is accomplished B) Skin arterioles vasoconstrict primarily due to input from
by giving drugs that decrease thyroid hormone synthesis, motor nerves.
beta-adrenergic blocking drugs to lower her heart rate, C) The hypothalamus is the location of the central controller
stroke volume, and blood pressure, and glucocorticoid ther- of body temperature.
apy that is immunosuppressive and also decreases the con- D) The hypothalamus is the location of the central
thermoreceptors.
version of thyroxine to triiodothyronine the more potent
endogenous thyroid hormone. The long-term goal is nor- 4. All of the following will reduce fever except
malizing the thyroid function. This is usually accomplished A) aspirin-induced decrease in hypothalamic
by giving radioactive iodine to destroy the thyroid. Alterna- prostaglandin synthesis
B) acetaminophen-induced decrease in hypothalamic
tive therapies are drugs that inhibit thyroid hormone syn-
prostaglandin synthesis
thesis or, in an extreme case, a surgical thyroidectomy.
C) an increase in endotoxin in the blood
Why was this woman’s body temperature so elevated? D) cortisol-induced suppression of immune function
Thyroid hormone causes an increase in metabolic rate in
virtually all organs of the body. This results in an increase
in heat production. When severe enough, the ability to lose
heat to the environment by sweating cannot keep up with
the increase in heat produced by increased metabolism,
and the body temperature increases. This can be extremely
dangerous, and urgent treatment is usually required.

CHAPTER SUMMARY
■ Internal body temperature is the central compartment of a
mass balance system—heat loss usually equals heat gain.
■ Heat transfer between the skin and environment occurs by
radiation, conduction, convection, and evaporation.
This page intentionally left blank
71
C H A P T E R

Hypoxia and Hyperbaria


Michael Levitzky and Hershel Raff

O B J E C T I V E S

■ Discuss the physiologic responses to hypoxia and hyperbaria.


■ Identify the physiologic stresses involved in the ascent to altitude.
■ Predict the initial responses of the central nervous, cardiovascular, and
respiratory systems to the ascent to altitude.
■ Describe the acclimatization of the cardiovascular, respiratory, and renal
systems to residence at high altitudes.
■ Identify the physiologic stresses involved in diving.
■ Predict the responses of the respiratory, cardiovascular, and renal systems to
various types of diving.

HYPOXIA inspired air passes through the airways, it is warmed to body


temperature and completely humidified. Therefore, the partial
Hypoxia means low oxygen. As was discussed in Chapter 37, pressure exerted by the water vapor in the air entering the
tissue hypoxia has many causes that can be classified into four alveoli is 47 mm Hg.
main types (see Table 37–7). One of these is low arterial partial The alveolar Po2 can therefore be calculated by using the
pressure of oxygen (“hypoxic hypoxia”) that may result from alveolar air equation discussed in Chapter 33:
a low alveolar partial pressure of oxygen. Other causes of P
Pao2 = Pio2 – ____
Aco2
hypoxic hypoxia are described in Chapter 37. The most com- R
+ [F] (1)
mon reason for decreased inspired oxygen in a healthy indi-
The inspired Po2 is equal to 0.21 times the total barometric
vidual is ascent to altitude.
pressure (if ambient air is breathed) after the subtraction of the
water vapor pressure of 47 mm Hg:
ALTITUDE AND Pio2 = 0.21 × (PB – 47mm Hg) (2)

ACCLIMATIZATION The alveolar Pco2 decreases at greater altitudes because


hypoxic stimulation of the arterial chemoreceptors increases
The barometric pressure decreases at greater altitudes because alveolar ventilation (hyperventilation). As shown in
the total pressure at any altitude is proportional to the weight Figure 71–1, the decrease in alveolar Pco2 brings the alveolar
of the air above it. There is a greater change in barometric Po2 closer to the inspired Po2, as calculated by equation (1). For
pressure per change in altitude closer to the earth’s surface example, at an altitude of 15,000 ft, the total barometric pres-
than at very great altitudes because air, which is attracted to sure is about 429 mm Hg. The inspired Po2 is therefore 0.21 ×
the earth’s surface by gravity, is compressible. (429 – 47) mm Hg, or 80.2 mm Hg. The alveolar Pco2 is likely
The fractional concentration of oxygen in the atmosphere to be decreased to about 32 mm Hg, resulting in a Pao2 of about
does not change appreciably with altitude; it is therefore 21% 45 mm Hg. At 18,000 ft, the total barometric pressure is about
of the total pressure of dry ambient air at any altitude. As 380 mm Hg; at 20,000 ft, it is 349 mm Hg.

735

Ch71_735-744.indd 735 12/8/10 10:07:11 PM


736 SECTION X Integrative Physiology

Altitude (1000 feet) CARDIOVASCULAR SYSTEM


0 5 10 15 20 25 30
20
There is an increase in cardiac output, heart rate, and sys-

Highest permanent habitation


Denver
140
18 temic blood pressure at altitude. These effects are probably a
result of increased sympathetic stimulation of the cardiovas-
16 Inspired gas PO2 120 cular system secondary to arterial chemoreceptor stimula-

Partial pressure (mm Hg)


tion. There may also be a direct stimulatory effect of hypoxia
Mt. Blanc
Partial pressure (kPa)

14
100 on the myocardium. Alveolar hypoxia results in hypoxic

Silver hut
12 Alveolar pulmonary vasoconstriction (see Chapter 34). The
gas PO2 80 increased cardiac output, along with hypoxic pulmonary
10 vasoconstriction and sympathetic stimulation of larger pul-

Everest
60 monary vessels, results in an increase in mean pulmonary
8
artery pressure and tends to abolish any preexisting zone 1
6 Alveolar gas PCO2
(see Figure 34–7) by recruiting previously unperfused capil-
40
laries. Undesirable consequences of these effects include
4 vascular distention and engorgement of the lung secondary
20 to the pulmonary hypertension, which may lead to high-
2
altitude pulmonary edema and a greatly increased right
0 0 ventricular workload. People who are more susceptible to
0 1 2 3 4 5 6 7 8 9
high-altitude pulmonary edema also seem to have greater
Altitude (km)
hypoxic pulmonary vasoconstriction responses than less
FIGURE 71–1 Calculated inspired and alveolar partial susceptible individuals. Analysis of high-altitude pulmonary
pressures of oxygen and carbon dioxide at rest plotted versus edema fluid shows that it contains large-molecular-weight
increasing altitude. Note that as increasing arterial chemoreceptor proteins, which indicates that the edema is caused by
drive decreases alveolar PCO2, alveolar PO2 is closer to inspired PO2. increased capillary permeability as well as increased capil-
(Adapted with permission from Lumb AB: Nunn’s Applied Respiratory Physiology, 5th
lary hydrostatic pressure. The increased capillary permeabil-
ed. Oxford: Butterworth-Heinemann, 2000 [Figure 16.1, p. 359].)
ity may result from capillary stress failure caused by high
pulmonary artery pressure and blood flow and by altered
release of cytokines or other mediators.
ACUTE EFFECTS OF ALTITUDE The effects of ascent to high altitude on the cerebral circu-
lation are complex. Hypoxic stimulation of the arterial
Ascent to altitude causes significant alterations in the central chemoreceptors causes hypocapnia and respiratory alkalo-
nervous system, the cardiovascular system, the respiratory sis, as already discussed. Cerebral arterial hypocapnia not
system, and the regulation of body fluids by the renal system. only could cause constriction of the cerebral blood vessels,
Because low temperatures are commonly encountered at alti- but would also cause alkalosis of the cerebrospinal fluid.
tude, the temperature regulatory mechanisms discussed in Most of the central nervous system symptoms of acute moun-
Chapter 70 are also usually involved. tain sickness could be attributed to cerebral hypoperfusion,
If a healthy person were to ascend rapidly to altitudes greater alkalosis, or both. However, it now appears that in most cases
than 3,000–4,500 m (approximately 10,000–15,000 ft) above sea the symptoms of acute mountain sickness result from cere-
level, he or she would suffer a deterioration of nervous system bral hyperperfusion and edema. This hyperperfusion is
function. Similar problems could occur if cabin pressure is lost in mainly a result of vasodilation, which is the direct effect of
an airplane. The symptoms are mainly due to hypoxia; they may hypoxia on the cerebral blood vessels. As the cerebral arteri-
include sleepiness, laziness, a false sense of well-being, impaired oles dilate, the hydrostatic pressure in the cerebral capillaries
judgment, blunted pain perception, increasing errors on simple increases, increasing the tendency of fluid to leave the cere-
tasks, decreased visual acuity, clumsiness, and tremors. Severe bral capillaries and cause cerebral edema. The hyperperfusion
hypoxia may result in a loss of consciousness or even death. and cerebral edema increase intracranial pressure, compress-
If an unacclimatized person (a person who has not adapted ing and distorting intracranial structures. This may lead to a
to being at altitude) ascends to altitudes greater than general increase in sympathetic activity in the body, increas-
3,000–4,500 m above sea level, he or she may suffer from a ing the possibility of pulmonary edema and promoting renal
group of symptoms known collectively as acute mountain salt and water retention.
sickness. The symptoms include headache, dizziness, breath-
lessness at rest, weakness, malaise, nausea, anorexia, sweating,
palpitations, impaired vision, partial deafness, sleeplessness, RESPIRATORY SYSTEM
fluid retention, and dyspnea on exertion. These symptoms are
a result of hypoxia and hypocapnia, and alkalosis or cerebral The decreased arterial partial pressures of oxygen that occur at
edema, or both. altitude stimulate the arterial chemoreceptors and increase
CHAPTER 71 Hypoxia and Hyperbaria 737

alveolar ventilation; the central chemoreceptors are not Prevention and Treatment
responsive to hypoxia, as was discussed in Chapter 38. of Acute Mountain Sickness
Although the arterial chemoreceptors are not very sensitive to
changes in arterial Po2 above approximately 60 mm Hg (see Acetazolamide, a carbonic anhydrase inhibitor, taken for a
Figure 38–8), at an arterial Po2 of 45 mm Hg, minute ventila- few days before ascending to altitude can prevent the symp-
tion is approximately doubled. Because carbon dioxide pro- toms of acute mountain sickness in many people. The mecha-
duction is initially normal (it does increase with the increased nism by which it does this is unclear because acetazolamide
work of breathing caused by greater alveolar ventilation), alve- has several actions that may help prevent acute mountain sick-
olar and arterial Pco2 decrease to approximately 20 mm Hg, ness. It decreases reabsorption of bicarbonate by the proximal
causing respiratory alkalosis. Arterial hypocapnia also results tubule of the kidney. This may lead to a moderate metabolic
in “diffusion” of carbon dioxide from the cerebrospinal fluid acidosis that may partly offset the respiratory alkalosis and
into the blood (see Figure 38–6), causing an increase in the pH therefore also help stimulate ventilation. Acetazolamide is also
of the cerebrospinal fluid. Therefore, not only are the central a diuretic, so it may help prevent fluid retention and edema. It
chemoreceptors unresponsive to the hypoxia of altitude, but is likely that both proposed mechanisms are involved. Acetazo-
their activity is also depressed by the secondary hypocapnia lamide may also inhibit hypoxic pulmonary vasoconstriction.
and the alkalosis of the cerebrospinal fluid. By far the most important treatment of acute mountain
Increased tidal volumes and respiratory rates increase the sickness is increasing alveolar Po2. This can be performed at
elastic work of breathing. High ventilatory rates may be altitude by breathing increased concentrations of oxygen from
accompanied by active expiration, resulting in dynamic com- a gas cylinder. Descent to lower altitudes is most effective in
pression of airways. This airway compression, coupled with treating acute mountain sickness.
arterial chemoreceptor-mediated reflex parasympathetic
bronchoconstriction in response to the arterial hypoxemia,
results in increased resistance work of breathing. More tur- Acclimatization to Altitude
bulent airflow, which is likely to be encountered at higher Longer-term compensation to high altitude begins to occur
ventilatory rates, may also contribute to increased resistance after several hours of ascent and continues for days or even
work. Maximum airflow rates may increase because of weeks. The immediate responses to the ascent and the early
decreased gas density at altitude. The deeper inspirations and late adaptive responses are summarized in Table 71–1.
and expirations would likely result in a more uniform Renal compensation for respiratory alkalosis begins within
regional distribution of alveolar ventilation; previously col- a day: renal excretion of base is increased, and hydrogen ions
lapsed or poorly ventilated alveoli would be better ventilated. are conserved. A second major compensatory mechanism is
The increased pulmonary blood flow seen acutely at high erythropoiesis. Within 3–5 days, new red blood cells are pro-
altitude, coupled with the more uniform alveolar ventilation, duced, increasing the hematocrit and the oxygen-carrying
would be expected to improve regional ventilation–perfu- capacity. Much of this response is due to an increase in the
sion matching. Surprisingly, studies have not shown striking secretion of the hormone erythropoietin from the kidney due
differences in VA/Qc relationships at high altitude, although to local renal hypoxia; erythropoietin then stimulates red cell
they do appear to improve. production in bone marrow (see function 5 in Chapter 39).
At high altitude, the partial pressure gradient for oxygen (Commonly known as “Epo,” this protein is available for injec-
diffusion is decreased because the alveolar Po2 is decreased tion and has been used for “blood doping” in athletes.) Thus,
more than the mixed venous Po2. This decrease in the partial although the arterial Po2 is not improved, the arterial oxygen
pressure gradient is partly offset by effects of increases in car- content increases because of the increased blood hemoglobin
diac output and increased pulmonary artery pressure, which concentration. This is at the cost of a higher blood viscosity
increase the surface area available for diffusion by recruit- and ventricular workload. Increased concentrations of 2,3-BPG
ment of unperfused capillaries and decrease the time erythro- (DPG) may help release oxygen at the tissues (Figure 36–2).
cytes spend in pulmonary capillaries. Hypoxic stimulation of the arterial chemoreceptors persists
Oxygen loading in the lung may be compromised at alveolar and may be augmented during the acclimatization. A more
partial pressures of oxygen low enough to be below the flat part immediate finding is that the ventilatory response curve to car-
of the oxyhemoglobin dissociation curve (see Figure 36–1), bon dioxide shifts to the left (Figure 38–4). That is, for any given
causing a low arterial oxygen content. Hypocapnia may aid alveolar or arterial Pco2, the ventilatory response is greater after
somewhat in oxygen loading in the lung but will interfere with several days at high altitude. The current theory for the increase
oxygen unloading at the tissues (see Figure 36–2). The main in ventilation that occurs during acclimatization is that carotid
short-term compensatory mechanism for maintenance of oxy- body chemoreceptor sensitivity to hypoxia increases through
gen delivery is the increased cardiac output. The hemoglobin cellular mechanisms that are not well established. The increase in
concentration may also increase slightly within the first 2 days. alveolar ventilation results in an increase in alveolar, and hence
This is a result of hemoconcentration secondary to fluid shift- arterial, Po2. It is this adaptive response that allows people to
ing into the extravascular space, not an increase in erythrocyte live at altitude for long periods of time. The central nervous sys-
production. tem symptoms usually abate at the same time as the resolution of
738 SECTION X Integrative Physiology

TABLE 71–1 Physiologic responses to high altitude relative to sea-level control values.a
Immediate Early Adaptive (72 Hours) Late Adaptive (2–6 Weeks)

Minute ventilation ↑ ↑ ↑

Respiratory rate Variable Variable Variable

Tidal volume ↑ ↑ ↑

Arterial PO2 ↓ ↓ ↓

Arterial PCO2 ↓ ↓ ↓

Arterial pH ↑ ↑↔ ↑↔

Arterial HCO3− ↔ ↓ ↓

Evaluation of lung function

Vital capacity ↔ ↔ ↔

Maximum airflow rates ↑ ↑ ↑

Functional residual capacity ↔ ↔ ↔

Ventilatory response to inhaled CO2 ↔ ↑ ↑

Ventilatory response to hypoxia ↔ ↔ ↔

Pulmonary vascular resistance ↑ ↑ ↑

Oxygen transport

Hemoglobin ↔ ↑ ↑

Erythropoietin ↑ ↔ ↔

P50 ↓ ↑ ↑

2,3-BPG ↔ ↑ ↑

Cardiac output ↑ ↔ ↔↓

Central nervous system

Headaches, nausea, insomnia ↑ ↔ ↔

Perception, judgment ↓ ↔ ↔

Cerebral edema ↑ ↔ ↔
a
These values apply to native sea-level inhabitants. ↑, increased; ↓, decreased; ↔, no change.
Adapted with permission from Guenter CA. Pulmonary Medicine, 2nd ed. Philadelphia, PA: Lippincott; 1982.

the cerebral edema and higher intracranial pressure. This is likely


due to increased reabsorption of cerebrospinal fluid, autoregula-
HYPERBARIA
tion of cerebral blood flow, and a sympathetically mediated vaso- Hyperbaria means increased ambient pressure. Healthy peo-
constriction that for some reason takes several days to develop. It ple can experience hyperbaric conditions during underwater
is also possible that the cerebral vessels produce less nitric oxide, diving. Hyperbaric chambers are used clinically to increase
which may mediate the cerebral vasodilatation in response to ambient gas pressures as treatment for decompression illness
hypoxia (see Chapter 27). (discussed below) or to enhance wound healing (hyperbaric
The increased cardiac output, heart rate, and systemic blood oxygen therapy or HBOT).
pressure return to normal levels after a few days at high alti-
tude. This probably reflects a decrease in sympathetic activity
or downregulation of adrenergic receptors. Nevertheless, DIVING
hypoxic pulmonary vasoconstriction and pulmonary hyper-
tension persist (along with increased blood viscosity due to The major physiologic stresses involved in diving include
higher hematocrit), leading to right ventricular hypertrophy increased ambient pressure, decreased effects of gravity
and frequently chronic cor pulmonale (right ventricular fail- because of buoyancy, altered respiration, hypothermia, and
ure secondary to pulmonary hypertension). sensory impairment. The severity of the stress depends on the
CHAPTER 71 Hypoxia and Hyperbaria 739

depth attained, the length of the dive, and whether the breath An additional effect of neck-deep immersion is immersion
is held or breathing apparatus is used. The physiologic stresses diuresis. Within a few minutes of immersion, urine flow
of elevated ambient pressure, decreased effects of gravity, and increases 4–5-fold. These findings are consistent with stimula-
altered respiration are the focus of this discussion. tion of stretch receptors in the cardiac atria and elsewhere in
thoracic vessels by the increased thoracic blood volume. This,
PHYSICAL PRINCIPLES in turn, is believed to decrease the secretion of antidiuretic
hormone (ADH) by the posterior pituitary gland or to cause
The pressure at the bottom of a column of liquid is propor- the release of natriuretic hormones from the cardiac atria
tional to the height of the column (h), the density of the liquid (see Chapter 45).
(ρ), and the acceleration of gravity (g):
Respiratory Effects
Pressure = h × p × g (3)
The pressure outside the chest wall of a person standing or
For example, for each 10 m (33 ft) of seawater, ambient pres- seated in neck-deep water is greater than atmospheric,
sure increases by 1 atm. Thus, at a depth of 10 m of seawater, averaging about 20 cm H2O (14.7 mm Hg). This positive
total ambient pressure is equal to 1,520 mm Hg or twice the pressure outside the chest opposes the normal outward elas-
barometric pressure at sea level. tic recoil of the chest wall and decreases the functional
The tissues of the body are composed mainly of water and residual capacity by about 50%. This occurs at the expense
are therefore incompressible, but gases are compressible and of the expiratory reserve volume, which may be decreased
follow Boyle’s law. Thus, in a breath-hold dive, the volume of by as much as 70%. The intrapleural pressure is less nega-
gas in the lungs is inversely proportional to the depth attained. tive at the functional residual capacity because of decreased
At 10 m of depth (2 atm), lung volume is cut in half; at 20 m outward elastic recoil of the chest wall. The work that must
(3 atm), it is one third the original lung volume. As a gas is be done to bring air into the lungs is greatly increased
compressed, its density increases. because extra inspiratory work is necessary to overcome the
As the total pressure increases, the partial pressures of the positive pressure outside the chest. Immersion up to the
constituent gases also increase, according to Dalton’s law (see neck in water results in an increase in the work of breathing
Chapter 33). The biologic effects of gases are generally depen- by about 60%.
dent on their partial pressures rather than on their fractional The hydrostatic pressure effects of water outside the chest
concentrations. Also, as the partial pressures of a gas increases, prevent a submerged person, who is trying to breathe through
the amount dissolved in the tissues of the body increases, a tube that is communicating with the air above the surface of
according to Henry’s law (see Chapter 35). the water, from descending more than about 3 ft. This is
true even if the increased airway resistance offered by the tube
EFFECTS OF IMMERSION were negligible and if the person avoided increasing the effec-
UP TO THE NECK tive dead space by occluding the mouth end of the tube and
exhaling directly into the water (or by using a one-way valve).
Merely immersing oneself up to the neck in water causes pro- The reason is that the maximal inspiratory pressure that nor-
found alterations in the cardiovascular and pulmonary sys- mal individuals can generate is about 80–100 cm H2O (i.e.,
tems. These effects are mainly a result of an increase in pressure intrapleural pressures of –80 to –100 cm H2O). Because 100 cm
outside the thorax, abdomen, and limbs. is 1 m (39.37 in), the maximum depth a person can attain
while breathing through a tube in this manner is a little more
Cardiovascular and Renal Effects than 1 m (3 ft).
During immersion up to the neck, increased pressure outside
the limbs and abdomen results in less pooling of systemic BREATH-HOLD DIVING
venous blood in gravity-dependent regions of the body (see
Chapter 30). If the water temperature is below body temper- During a breath-hold dive, the total pressure of gases within
ature, a sympathetically mediated venoconstriction occurs, the lungs is approximately equal to the increased ambient
also augmenting venous return (see Chapter 70). The pressure. Therefore, the volume within the thorax must decrease
increased venous return increases the central blood volume proportionately and partial pressures of gases must increase.
by approximately 500 mL. Right atrial pressure increases
from about –2 to +16 mm Hg. As a result of this increase in The Diving Reflex
preload, the cardiac output and stroke volume increase by Many subjects demonstrate a profound vagally mediated
about 30%. The increases in pulmonary blood flow and pul- bradycardia (decreased heart rate) and increased systemic
monary blood volume result in increased mean pulmonary vascular resistance with face immersion (especially into
artery pressure, capillary recruitment, an increase in the dif- cold water), as well as apnea. This “diving reflex” is initiated
fusing capacity, and a somewhat improved matching of ven- by sensors in the face or nose with afferents to the brain via
tilation and perfusion. the trigeminal nerves. A similar response is seen when
740 SECTION X Integrative Physiology

Face immersion
FIGURE 71–2 The author’s (MGL) electrocardiographic response to face immersion in ice water (the diving reflex). The experiment
was performed in the prone position, and face immersion was performed without changing the position of the head to avoid the effects of
changes in baroreceptor activity. The heart rate decreased from about 75 to about 43 beats/min. (Time between the vertical ticks above or below
the ECG = 3sec.) (Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New York: McGraw-Hill Medical, 2007.)

aquatic mammals such as whales and seals dive. The reflex to alveolus is reversed during descent and results in signifi-
decreases the workload of the heart and severely limits per- cant retention of carbon dioxide in the blood.
fusion to all systemic vascular beds except for the strongest During ascent, the ambient pressure decreases rapidly, lung
autoregulators—namely, the heart and brain. The cardiovas- volume increases, and alveolar gas partial pressures decrease
cular effects of the diving reflex are similar to those produced accordingly. Alveolar Pco2 decreases, allowing CO2 to diffuse
by stimulation of the arterial chemoreceptors when no from pulmonary capillary blood into the alveoli. However,
increase in ventilation can occur, except that the diving reflex the rapid decrease in alveolar Po2 during ascent may result in a
also appears to cause the spleen to slowly contract, which decrease in arterial Po2 sufficient to cause the breath-hold diver
releases erythrocytes stored in the spleen into the venous to lose consciousness. This loss of consciousness can occur
blood. This increases the oxygen-carrying capacity of the rapidly and without warning and usually occurs as divers
blood and therefore the oxygen content of the blood at the ascend to a depth of approximately 5 m or less. It is therefore
same arterial Po2. The bradycardic component of the diving known as “shallow water blackout.”
reflex is demonstrated in Figure 71–2.

THE USE OF UNDERWATER


Gas Exchange BREATHING APPARATUS
Breath-hold divers usually hyperventilate before a dive so
that the typical alveolar Po2 and Pco2 might be 120 and Self-contained underwater breathing apparatus, or scuba
30 mm Hg, respectively. Indeed, breath-hold divers must gear, mainly consists of a tank of compressed gas that can be
take care not to hyperventilate so much that their Pco2 gets so delivered by a demand regulator to the diver when the diver’s
low that they do not reach their “breakpoint” until after they mouth pressure decreases during inspiration to slightly less
lose consciousness from arterial hypoxemia. (The break- than the ambient pressure. Expired gas is simply released into
point is the point at which an individual can no longer vol- the water as bubbles. Therefore, during a dive with scuba gear,
untarily hold his or her breath, which is mainly determined gas pressure within the lungs remains close to the ambient pres-
by Paco2.) During a breath-hold dive to a depth of 10 m sure at any particular depth. As a result, the physiologic stresses
(33 ft), lung volume decreases and gases are compressed. on the respiratory system during scuba diving are mainly a
Total gas pressure approximately doubles: thus, after consequence of elevated gas densities and partial pressures.
20 seconds at 10 m, the alveolar Po2 may be 160–180 mm Hg; During scuba diving, the inspiratory work of breathing is
even after 1 minute at 10 m, the alveolar Po2 is well above not a great problem at moderate depths because gas is deliv-
100 mm Hg. The alveolar Pco2 (that would be less than ered at ambient pressures. At very great depths, however,
40 mm Hg at the surface because of hyperventilation before increased gas density becomes a problem because it increases
the dive) also increases to above 40 mm Hg during descent, the airway resistance work of breathing during turbulent flow.
reversing the gradient for CO2 transfer. Carbon dioxide For example, in long-term experiments performed with sub-
therefore diffuses from the alveoli into the pulmonary capil- jects simulating dives of over 600 m (2,000 ft) inside hyper-
lary blood. The alveolar Pco2 therefore does not increase as baric chambers, all subjects reported that they could breathe
much as would be predicted by the compression of gases only through their mouths; the work of breathing through the
caused by the increased pressure as CO2 diffuses into the nose was too great. This is one reason for replacing nitrogen
blood. This is believed to result from the much greater solu- with helium for deep dives. Helium is only about one seventh
bility of CO2 than O2 in the blood. Thus, the transfer of oxy- as dense as nitrogen.
gen from alveolus to blood is undisturbed until ascent; The respiratory control system’s sensitivity to carbon diox-
however, the normal transfer of carbon dioxide from blood ide is decreased at great depths because of increased gas
CHAPTER 71 Hypoxia and Hyperbaria 741

densities and high arterial Po2 and because divers learn to sup- of gases, mainly carbon dioxide, and then capped. The total
press carbon dioxide stimuli to conserve compressed gas. pressure in the gas layer above the liquid remains greater than
atmospheric pressure. The partial pressures of gases dissolved
in the liquid phase are in equilibrium with the partial pres-
CLINICAL PROBLEMS sures in the gas phase. Gases dissolve in the liquid phase
ASSOCIATED WITH DIVING according to Henry’s law. When the bottle is uncapped, the
pressure in the gas phase suddenly decreases and the gas dis-
Clinical problems that may be encountered in diving, particu- solved in the liquid phase comes out of solution, forming bub-
larly to great depths, include barotrauma, decompression ill- bles.
ness, nitrogen narcosis, oxygen toxicity, and high-pressure The bubbles formed in decompression sickness may enter
nervous syndrome (HPNS). the venous blood or affect the joints of the extremities. Bub-
Barotrauma occurs when ambient pressure increases or bles that enter the venous blood are usually trapped in the
decreases, but the pressure in a closed unventilated area of the pulmonary circulation and rarely cause symptoms. The symp-
body that cannot equilibrate with ambient pressure does not. toms that do occasionally occur, which are known as “the
The barotrauma of descent is called “squeeze.” It can affect the chokes” by divers, include substernal chest pain, dyspnea, and
middle ear, if the Eustachian tube is clogged or edematous, so cough and may be accompanied by pulmonary hypertension,
that a person cannot equilibrate pressure in the middle ear; the pulmonary edema, and hypoxemia. This is obviously an
sinuses; the lungs, resulting in pulmonary congestion, edema, extremely dangerous form of decompression illness. Even
or hemorrhage; and even cavities in the teeth. The barotrauma more dangerous, of course, are bubbles in the circulation of
of ascent can occur if gases are trapped in areas of the body the central nervous system, which may result in brain damage
and begin to expand as the diver ascends. If a diver does not and paralysis. They may result from alveolar rupture and
exhale while ascending, expanding pulmonary gas may over- arterial gas embolism, as discussed previously, or be carried
distend and rupture the lung (“burst lung”). This may result in from the venous blood to the arterial side through a patent
hemorrhage, pneumothorax, or air embolism. Gases trapped foramen ovale (see Chapter 30) or an intrapulmonary shunt.
in the gastrointestinal tract may cause abdominal discomfort Bubbles that form in the joints of the limbs cause pain (“the
and eructation (belching) or flatus as they expand. Barotrauma bends”). Osteonecrosis of joints may also be caused by inad-
of the ears, sinuses, and teeth may also occur on rapid ascent equate decompression.
from great depths. The treatment for decompression illness is immediate
Decompression illness occurs when gas bubbles form in the recompression in a hyperbaric chamber that forces the gas in
blood and body tissues as the ambient pressure decreases. The bubbles back into solution, followed by slow decompression.
term “decompression illness” encompasses two different prob- Decompression illness may be prevented by slow ascents from
lems, both of which involve gas bubbles. Arterial gas embo- great depths (using decompression tables) and by substituting
lism is gas bubbles in the arterial blood. Because bubbles do helium for nitrogen in inspired gas mixtures. Helium is only
not seem to form in the arterial blood itself, arterial gas embo- about half as soluble as nitrogen in body tissues.
lism usually occurs when airway obstruction prevents expand- Gas bubbles, although they are sterile, are perceived by the
ing gas from being exhaled. As expanding alveoli rupture, gas body as foreign. They elicit inflammatory and other responses,
bubbles may enter pulmonary capillaries and be carried into including platelet activation, blood clotting, the release of
the arterial blood. Arterial gas embolism is a likely conse- cytokines and other mediators, leukocyte aggregation, free
quence of an ascending diver neglecting to exhale on rapid radical production, and endothelial damage. These responses
ascent. Bubbles resulting from arterial gas embolism may be are not reversed by recompression and may continue unless
carried to cerebral blood vessels where they may cause a stroke. additional treatment is initiated.
The second component of decompression illness, decompres- Divers who ascend from submersion with no immediate
sion sickness, occurs when bubbles form in tissues of the body. effects of decompression may subsequently suffer decompres-
During a dive, the increasing ambient pressure causes an ele- sion illness if they ride in an airplane within a few hours of the
vation of the partial pressure of nitrogen in the body. The high dive. Commercial airplanes normally maintain cabin pres-
partial pressure of nitrogen causes this normally poorly solu- sures well below 760 mm Hg, with cabin pressures similar to
ble gas to dissolve in the body tissues and fluids according to those at altitudes 5,000–8,000 ft above sea level. This is why
Henry’s law (discussed in Chapter 35). This is especially the people with pulmonary impairment may need to use supple-
case in body fat, which has a relatively high nitrogen solubility. mental oxygen during an airplane flight.
At great depths, body tissues become supersaturated with Very high partial pressures of nitrogen directly affect the
nitrogen. central nervous system, causing euphoria, loss of memory,
During a fast ascent, ambient pressure decreases rapidly and clumsiness, and irrational behavior. This nitrogen narcosis or
nitrogen comes out of solution, forming bubbles in body tis- “rapture of the deep” occurs at depths of 30 m (100 ft) or more
sues and fluids. The effect is the same as opening a bottle of a and at greater depths may result in numbness of the limbs, dis-
carbonated beverage. During the production of a carbonated orientation, motor impairment, and ultimately unconscious-
beverage, it is exposed to higher-than-atmospheric pressures ness. The mechanism of nitrogen narcosis is unknown.
742 SECTION X Integrative Physiology

Oxygen toxicity can be caused by inhalation of 100% oxy-


always lived at sea level because the increased pulmonary
gen at 760 mm Hg, or of lower oxygen concentrations at higher
artery pressure and increased blood viscosity have greatly
ambient pressures. It can cause central nervous system, visual
increased his right ventricular afterload. This has resulted
system, and alveolar damage, although pulmonary manifesta-
in right ventricular hypertrophy, which explains his right
tions are rare among divers. The mechanism of oxygen toxic-
axis deviation. This has resulted in right ventricular fail-
ity is controversial but probably involves the formation of
ure. As the right ventricle failed, it could no longer keep up
superoxide anions or other free radicals.
with venous return, leading to increased venous pressure,
Exposure to very high ambient pressures, such as those
which explains his distended jugular veins, and increased
encountered at very great depths (greater than 75 m), is associ-
capillary hydrostatic pressure, particularly in lower parts of
ated with tremors, decreased mental ability, nausea, vomiting,
the body. This caused his peripheral edema. His headache,
dizziness, and decreased manual dexterity. This high-pressure
sleep difficulties, and problems with his memory are a
nervous syndrome (HPNS) usually occurs when nitrogen has
result of insufficient oxygen delivery to his brain. The most
been replaced by helium to decrease gas density, prevent nitro-
effective treatment for chronic mountain sickness is to have
gen narcosis, and help avoid decompression sickness. Small
the patient move to a lower altitude, but this may not be
amounts of nitrogen added to the inspired gas mixture help
possible for psychosocial reasons. Other treatments may
counteract the problem. One hypothesis explaining HPNS is
include repetitive removal of red blood cells, and possibly
that the syndrome may result from alterations in cerebral neu-
acetazolamide.
rotransmission.
A person inside an intact submarine is not at risk for any of
the clinical problems associated with diving discussed above.
The rigid walls of a submarine allow its inhabitants to reside at
an ambient pressure similar to that at the surface. CHAPTER SUMMARY
■ The main physiologic stresses of altitude are hypoxia and
secondary hypocapnia and respiratory alkalosis; acclimatiza-
tion occurs mainly by renal compensation for the respiratory
alkalosis, erythropoiesis, elevated 2,3-BPG concentrations, and
CLINICAL CORRELATION resolution of cerebral edema.
■ The main physiologic stresses of diving are caused by
Physicians on a medical mission in the mountains of increased ambient pressures causing gas compression, leading
South America treat a 65-year-old man who has lived at to increased partial pressures and densities and viscosities
altitudes above 3,600 m (about 12,000 ft) since he was of gases.
born. He has a chronic headache, feels dizzy and tired,
has trouble remembering things, and does not sleep
well. He is cyanotic (his skin and mucosa appear to be STUDY QUESTIONS
blue; see Chapter 36), and has peripheral edema. His
jugular veins are distended even though he is sitting 1. Three hours after climbing from sea level to an altitude of
3,500 m, a healthy person breathing ambient air would likely
upright. An electrocardiogram indicates right axis
have a lower-than-normal
deviation (a frontal plane mean electrical axis greater
A) pulmonary artery pressure
than +105°).
B) alveolar ventilation
The patient has chronic mountain sickness (also known C) arterial pH
as Monge’s disease) with chronic cor pulmonale. Chronic D) arterial Pco
2
cor pulmonale is right ventricular failure secondary to E) work of breathing
pulmonary hypertension, in this case exacerbated by 2. Which of the following would be lower-than-normal sea-level
polycythemia (increased hematocrit resulting from values in a healthy person breathing ambient air after 5 days at
increased production of red blood cells). His pulmonary an altitude of 15,000 ft?
hypertension is mainly a result of the effects of chronic A) plasma bicarbonate concentration
hypoxic pulmonary vasoconstriction, which not only B) hematocrit
causes constriction of pulmonary arterioles, but also C) pulmonary vascular resistance
causes structural changes in the affected blood vessels D) 2,3-BPG (2,3-DPG)
over time. He has polycythemia because of chronic hypox- E) alveolar ventilation
emia leading to erythropoiesis. The increase in hemat- 3. Immersion in cold water up to the neck decreases
ocrit resulted in higher blood viscosity. His low arterial Po2 A) the inspiratory work of breathing
combined with his increased hematocrit (see Chapter 36) B) venous return
explains his cyanosis. The workload of his right ventricle C) the functional residual capacity
is much greater than that of a healthy individual who has D) the inspiratory reserve volume
E) right atrial pressure
CHAPTER 71 Hypoxia and Hyperbaria 743

4. During the descent phase of a 1-minute breath-hold dive to a 5. When a healthy person immerses his or her face in cold water
depth of 10 m
A) vagal tone to the cardiac pacemakers decreases
A) lung volume is increasing B) sympathetic stimulation of most of the systemic vascular
B) alveolar pressure is increasing beds, except for the coronary and cerebral beds, increases
C) the partial pressure gradient for diffusion of oxygen from the C) heart rate increases
alveoli into the pulmonary capillary blood is decreasing D) systemic vascular resistance decreases
D) the partial pressure gradient for carbon dioxide diffusion
from the pulmonary capillary blood into the alveoli is
increasing
This page intentionally left blank
72
C H A P T E R

Exercise
Michael Levitzky and Kathleen H. McDonough

O B J E C T I V E S

■ Identify the physiologic demands of exercise.


■ Predict the physiologic responses to acute exercise.
■ Describe the effects of long-term exercise programs (training) on the
physiologic response to exercise.

Exercise increases the metabolism of the working muscles. molecule of glucose (2 molecules when glucose is the substrate
Physical activity increases the requirement for oxygen and the and 3 when the glucose-6-phosphate produced by hydrolysis
production of carbon dioxide. Moderate to severe levels of of glycogen is the substrate), as opposed to the 32 molecules of
exercise also cause increased lactic acid production. The ATP produced per molecule of glucose by oxidative phospho-
respiratory and cardiovascular systems must increase the rylation. Therefore, anaerobic metabolism cannot be sus-
amount of oxygen supplied to the exercising tissues and tained for long periods of time. During strenuous exercise,
increase the removal of carbon dioxide and hydrogen ions glycolysis can maintain ATP production for muscular activity
from the body. Although the muscular and cardiopulmonary at least for a short time and, on completion of the exercise ses-
systems are the primary responders to exercise, there are also sion, the oxygen debt is then repaid by the increased oxygen
adjustments in intermediary metabolism and fluid and elec- consumption that continues in spite of the cessation of the
trolyte balance that allow the individual to maintain an higher rate of skeletal muscle contraction. Lactate produced
increase in physical activity. during the anaerobic phase can be converted to glucose by the
liver and glycogen and triacylglycerol stores are subsequently
restored.
MUSCLE METABOLISM
During exercise, increases in cardiac output (CO) and the ACUTE EFFECTS OF INCREASES
supply of substrates for energy are coordinated primarily by
the sympathetic nervous system. Epinephrine increases glu- IN PHYSICAL ACTIVITY
cose output from the liver and fatty acid output from adipose
The response to exercise in an untrained person is mainly a
tissue, providing more substrate for oxidative metabolism by
function of increased CO and alveolar ventilation to meet the
the exercising muscle. Muscle also has an intracellular supply
demand for increased oxygen delivery and carbon dioxide and
of glycogen that provides glucose-6-phosphate for energy.
hydrogen ion removal.
Oxidative phosphorylation of glucose supports the initial
few minutes of exercise and, during the intermediate phase,
both glucose and fatty acids are used in approximately equal CARDIOVASCULAR SYSTEM
proportions. More prolonged exercise is mainly supported by
fatty acid oxidation. With strenuous and prolonged exercise, The CO increases during exercise to provide more blood flow
oxygen delivery may be limiting and glycolysis and lactic acid to the exercising muscles. In most situations, blood flow to the
formation become more important sources of energy. Anaer- skin increases in order to help dissipate the heat generated by
obic metabolism provides only 2–3 molecules of ATP per muscle metabolism. CO increases nearly linearly with work

745

Ch72_745-752.indd 745 11/26/10 10:38:23 AM


746 SECTION X Integrative Physiology

rate up to 4–5-fold in a healthy untrained person during max-


Cardiac output (L/min)

25 imal exercise (Figure 72–1); it may increase even more in a


Trained trained athlete.
20
Untrained The CO increases immediately with the onset of exercise
or even in anticipation of exercise. The cerebral cortex initi-
ates the command to exercise, resulting in increased skeletal
muscle activity and metabolism (Figure 72–2). At the same
5 time, the cerebral cortex exerts a “central command” on the
medullary cardiovascular centers from corticohypothalamic
Work rate pathways that decreases parasympathetic activity to the heart
and increases sympathetic activity to the heart and blood
vessels. The set point of the arterial baroreceptor reflex is
Heart rate (beats/min)

200
Untrained Trained increased, which allows arterial pressure to be regulated to a
higher-than-resting level. It is also possible that chemorecep-
tor and mechanoreceptor afferents from active skeletal mus-
70 cles influence the medullary cardiovascular centers. Such
inputs would also contribute to the increases in sympathetic
activity and mean arterial pressure that accompany exercise
and may also contribute to increasing the baroreceptor reflex
Work rate
set point.
Heart rate and stroke volume increase to support the increase
in CO. Heart rate increases linearly with increasing work rate
Stroke volume (ml)

Trained up to about three times that at rest; stroke volume does not
125
increase linearly with workload, but plateaus at an increase of
Untrained
approximately 50%. The increase in heart rate is mediated by
70
reduced parasympathetic and augmented sympathetic activity
to the SA node; the increased stroke volume occurs primarily
by a sympathetically mediated increase in myocardial contrac-
O2 consumption tility and ejection fraction, with the Frank–Starling mecha-
FIGURE 72–1 Changes in cardiac output, heart rate, and nism (see Figure 24–4) playing a lesser role.
stroke volume with increasing workload in untrained and trained The distribution of the left ventricular output changes dur-
individuals. (Reproduced with permission from Widmaier EP, Raff H, Strang KT: ing exercise to support increased local oxygen consumption
Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.) (Figure 72–3). Blood flow to the exercising skeletal muscle

Begin
FIGURE 72–2 Control
of the cardiovascular Brain Exercising skeletal muscles
system during exercise. “Exercise centers” Contractions
“Exercise centers” in the brain
(mainly in the cerebral cortex
and hypothalamus) adjust Afferent Stimulate Local chemical
autonomic control to the input mechanoreceptors changes
Arterial baroreceptors Medullary in the muscles
heart and blood vessels and
Reset upward cardiovascular
increase the baroreceptor center
reflex set point via the Afferent
medullary cardiovascular input Stimulate Dilate
chemoreceptors arterioles
center. Afferent input from in the muscles in the muscle
Parasympathetic output to heart
mechanoreceptors and Sympathetic output to heart, veins,
chemoreceptors in the and arterioles in abdominal
exercising muscles also organs and kidneys Muscle blood flow
influences the medullary
cardiovascular center.
(Reproduced with permission from Cardiac output
Widmaier EP, Raff H, Strang KT: Vasoconstriction in
Vander’s Human Physiology, 11th
abdominal organs
and kidneys
ed. McGraw-Hill, 2008.)
CHAPTER 72 Exercise 747

Flow during Rest Exercise Recovery


strenuous exercise

Minute ventilation (L/min)


(ml/min)

750 (4%)
Flow
at rest (2)
(ml/min) 750 (4%)

Brain 650 (13%)


(1)
Heart 215 (4%) 12,500 (73%)

Skeletal muscle 1030 (20%)

Time
Skin 430 (9%)
FIGURE 72–4 Changes in ventilation during exercise.
Note the instantaneous increase at the onset of exercise (1) and the
Kidneys 950 (20%)
instantaneous decrease at the end of exercise (2). (Reproduced with
permission from Widmaier EP, Raff H, Strang KT. Van der’s Human Physiology. 11th ed.
Abdominal McGraw-Hill; 2008.)
1200 (24%)
organs 1900 (11%)

Other 525 (10%) vascular resistance (SVR) caused by dilation of blood vessels
Total 5000 600 (3%) supplying the exercising muscle:
600 (3%) MABP – RAP = CO × SVR (1)
400 (2%) Right atrial pressure (RAP) does not usually change sig-
17,500 nificantly during exercise; it may increase slightly during
FIGURE 72–3 Distribution of the left ventricular output at strenuous exercise because of the large increase in venous
rest and during strenuous exercise. Flows at rest are typical for a return (preload).
70-kg individual. The numbers in parentheses represent the percent The increased MABP is characterized by an increase in
of the total for each organ. (Reproduced with permission from Widmaier EP, pulse pressure without a significant change in diastolic pres-
Raff H, Strang KT. Van der’s Human Physiology. 11th ed. McGraw-Hill; 2008.) sure. The pulse pressure mainly increases because of the
greater stroke volumes noted above and because of more rapid
aortic pressure development (increased dP/dt) caused by
increased ventricular contractility. Furthermore, the set point
(including the respiratory muscles), heart, and skin increases for the baroreceptor reflex increases to allow the increased sys-
significantly, while blood flow to the kidneys, gastrointestinal temic blood pressure (see Figure 72–2).
tract, and other abdominal organs decreases significantly.
Blood flow to the brain is not significantly altered. The increase
in blood flow to the exercising muscles occurs primarily by RESPIRATORY SYSTEM
local metabolic regulation; the increased blood flow to the
skin occurs primarily by withdrawal of sympathetic tone. The Minute ventilation increases (hyperpnea) linearly with both
decreased blood flow to the kidneys and other abdominal oxygen consumption and carbon dioxide production up to a
organs occurs by sympathetically mediated vasoconstriction. level of about 60% of the maximal work capacity. Above that,
Venous return must increase in order for the CO to increase. minute ventilation increases faster than oxygen consumption but
Three mechanisms combine to increase venous return: continues to rise proportionally to the increase in carbon dioxide
increased activity in the exercising skeletal muscle compressing production. This increase in ventilation above oxygen consump-
veins and squeezing blood through one-way valves toward the tion at high work levels is caused in part by the increased lactic
heart (the skeletal muscle pump), increased tidal volume and acid production that occurs as a result of anaerobic metabolism.
breathing frequency increasing the abdominal–thoracic pres- The hydrogen ions liberated in this process directly stimulate the
sure difference so that intrapleural pressure is more negative arterial chemoreceptors. In addition, the buffering of hydrogen
and abdominal pressure more positive during inspiration (the ions by bicarbonate ions results in production of carbon dioxide
thoracoabdominal pump), and a sympathetically mediated in addition to that derived from aerobic metabolism.
increase in venous tone, which decreases venous capacitance. The ventilatory response to constant work rate exercise con-
The dilation of skeletal muscle arterioles also lowers resistance sists of three phases (Figure 72–4). At the beginning of exercise,
to blood flow from the arterial to the venous vessels. there is an immediate increase in ventilation, which may even
Mean arterial blood pressure (MABP) increases moder- occur in anticipation of physical activity. This is followed by a
ately with increasing exercise, indicating that the increase in phase of slowly increasing ventilation, ultimately increasing to a
CO must be slightly greater than the decrease in systemic final steady-state phase if the exercise is not too strenuous. The
748 SECTION X Integrative Physiology

initial immediate increase in ventilation may constitute as much TABLE 72–1 Ventilatory response to exercise.
as 50% of the total steady-state response. The increase in min-
ute ventilation is usually a result of increases in both tidal vol- Immediate response—“neural component”
ume and breathing frequency. Central command
The mechanisms by which exercise increases minute ventila- Learned or conditioned reflex
tion are summarized in Table 72–1. No single factor can fully Direct connections from motor cortex—collaterals from motor
account for the ventilatory response to exercise, and much of the neurons to muscles
response remains unexplained. The immediate increase in ven- Coordination in hypothalamus
tilation occurs too quickly to be due to changes in metabolism Proprioceptors or mechanoreceptors in limbs—probably not
or blood gases. This “neural component” consists in part of col- muscle spindles or Golgi tendon organs
lateral fibers to the respiratory muscles from the motor cortex Subsequent increase
neurons that primarily innervate the exercising skeletal muscles. During moderate exercise
It may also be partly accounted for by a conditioned reflex; that Arterial chemoreceptors
is, a learned response to exercise. Input to the respiratory centers PO2—usually no change in mean
from proprioceptors located in the joints and muscles of the
PCO2—usually no change in mean
exercising limbs may play a role. The ventilatory and cardiovas-
[H+]—usually no change unless cross anaerobic threshold
cular responses to exercise may be coordinated (and in part ini-
[K+]—released from exercising muscle cells could stimulate
tiated) in an “exercise center” in the hypothalamus. arterial chemoreceptors—minor role
The arterial chemoreceptors do not appear to play a role in Oscillations in arterial PO2 and PCO2
the initial ventilatory response to exercise. In mild or moder- Metaboreceptors
ate exercise, average arterial Pco2 and Po2 are not altered signifi-
Nociceptors—H+, K+, bradykinin, arachidonic acid released during
cantly (see Figure 72–5), even during the increasing ventilation exercise could stimulate pain receptors
phase (the “humoral component”). It is possible that the arte- Cardiac receptors
rial chemoreceptors are responding to greater oscillations in Venous chemoreceptors
the blood gases during exercise. Other potential afferent stim- Temperature receptors
uli to ventilation during moderate exercise are summarized in During exercise severe enough to exceed anaerobic threshold
Table 72–1.
Lactic acid buffered by HCO3− produces CO2
The work of breathing is increased during exercise. Larger
Arterial chemoreceptors
tidal volumes result in increased work necessary to overcome
↑ PCO2
the elastic recoil of the lungs and chest wall during inspiration
↑ [H+]
because the lungs are less compliant at higher lung volumes and
Central chemoreceptors
because the elastic recoil of the chest wall is inward at high tho-
↑ PCO2 → ↑ H+
racic volumes. The high airflow rates generated during exercise
result in a much greater airway resistance component of the Potentiation of chemoreceptor responses during exercise
work of breathing because of greater turbulence and dynamic Reproduced with permission from Levitzky MG: Pulmonary Physiology, 7th ed. New
York: McGraw-Hill Medical, 2007.
compression of airways secondary to active expiration. .
In normal adults, the resting minute ventilation (VE ) of 5–6
L/min can be increased to as much as 150 L/min during short
periods of maximal exercise. Maximal increases in CO during increase in pulmonary blood flow. This indicates a decrease in
exercise are only in the range of four to five times the resting pulmonary vascular resistance that occurs passively by
level in healthy adults compared with this 25-fold potential recruitment and distention of pulmonary vessels. It results in
increase in minute ventilation. Therefore, it is the cardiovas- more uniform distribution of pulmonary blood flow and more
cular system rather than the respiratory system that is the lim- uniform matching of ventilation and perfusion throughout
iting factor in exercise by healthy people. However, significant the lung. Because ventilation increases more than perfusion in
decreases in respiratory function that can occur with a variety moderate to strenuous
. . exercise, the whole lung ventilation–
of pulmonary diseases often limits exercise capacity. The perfusion ratio (VA /QC ) increases to a range of 2.0–4.0. Thus,
breathing frequency may increase to 40–50 breaths/min in the location of the perfusion is better matched to the location
healthy adults (and as high as 70 breaths/min in children) with of the ventilation during exercise, but the ventilation–perfusion
strenuous exercise. ratios increase in most alveolar–capillary units.
Arterial Pco2 is stable until anaerobic metabolism results in The diffusing capacities for oxygen and carbon dioxide
appreciable lactic acid generation (Figure 72–5). The hydro- normally increase substantially during exercise. The increase
gen ions liberated directly stimulate alveolar ventilation via in diffusing capacity during exercise is largely a result of the
the arterial chemoreceptors and may cause arterial Pco2 to fall increase in pulmonary blood flow. Recruitment of capillaries,
below the resting arterial Pco2. especially in upper regions of the lungs, increases the surface
As CO increases, mean pulmonary arterial and mean left area available for diffusion. Increased linear velocity of blood
atrial pressures increase, but the increase is not as great as the flow through pulmonary capillaries reduces the time that red
CHAPTER 72 Exercise 749

100 the arterial chemoreceptors (especially the carotid bodies)


80 and cause a further compensatory increase in alveolar ventila-
ventilation

tion, maintaining arterial pH near the normal level.


(L/min)
Minute

60

40

20 FLUID AND ELECTROLYTE BALANCE


0
Exercise causes a decrease in plasma volume, particularly if
the exercise is performed in a warm environment. The
Arterial PO2

110
increased metabolism generates heat. As was discussed in
(mmHg)

100 Chapter 70, evaporation of water in perspiration produced by


90 sweat glands is a primary mechanism of heat loss by the body.
A 70-kg person can lose more than 2 L/h of water via perspira-
Arterial PCO2

40 tion. A second cause of decreased plasma volume during exer-


(mmHg)

cise is increased filtration of plasma fluid from capillaries into


the interstitial space. Capillary hydrostatic pressure increases
30 in exercising muscles because of arteriolar vasodilation and to
a lesser extent because MABP increases. Also, the circulating
Arterial [H+]

60 blood volume decreases because as body temperature increases,


(nmol/L)

48 compliance of the veins of skin increases to help dissipate heat,


36
resulting in a greater amount of blood in the skin.
O2 consumption (ml/min) Perspiration is hypotonic to plasma, so relatively more water
Rest Maximal is lost than ions. The composition of sweat is variable from
exercise person to person and is also dependent on the rate of sweat
FIGURE 72–5 Effect of exercise on minute ventilation, arterial production. At higher flows, sweat contains increased concen-
oxygen and carbon dioxide partial pressures, and arterial trations of sodium and chloride, and less potassium although
hydrogen ion concentration with increasing exercise workloads. it remains hypotonic to plasma. Training (see the next section)
Note that the average arterial carbon dioxide levels are not altered and heat acclimatization lead to production of more dilute
significantly until exercise is sufficiently strenuous to produce increased
sweat, which helps conserve sodium and chloride and increase
levels of lactic acid. As the additional hydrogen ions stimulate the
exercise tolerance; this effect is probably mediated by aldoster-
arterial chemoreceptors, arterial carbon dioxide levels decrease; the
individual is therefore hyperventilating. (Reproduced with permission from
one release in response to sodium ion loss.
Widmaier EP, Raff H, Strang KT: Vander’s Human Physiology, 11th ed. McGraw-Hill, 2008.)
Ingesting sodium chloride with water improves recovery from
exercise and/or heat stress dehydration. Drinking water without
salt decreases extracellular fluid osmolality and decreases thirst,
blood cells spend in contact with the alveolar air, decreasing delaying rehydration. This is the basis of sports drinks that con-
the perfusion limitation of gas transfer. The alveolar–arterial tain sodium chloride and potassium; some also contain carbo-
oxygen difference usually increases during exercise. hydrates for energy. However, overhydration causing dangerous
. .This is
probably because of a number of factors, including VA /QC mis- hyponatremia during exercise is a significant problem particu-
match, diffusion limitation of gas transfer, a decreased mixed larly in those new to strenuous, sustained exercise.
venous Po2 , an increased alveolar Po2 , and alterations in the
oxyhemoglobin dissociation curve.
The loading of carbon dioxide into the blood and the unload- TRAINING EFFECTS
ing of oxygen from the blood are enhanced in exercising mus-
cles. Oxygen unloading is improved because the tissue Po2 in the The ability to perform physical exercise increases with train-
exercising muscle is decreased, resulting in an increased diffu- ing. Most of the changes that occur as a result of physical train-
sion gradient for oxygen. Oxygen unloading is also enhanced ing are a function of alterations in the cardiovascular system
by the rightward shift of the oxyhemoglobin dissociation curve and in muscle metabolism (Figure 72–1). The maximal oxy-
caused by the increased local Pco2 (the Bohr effect), hydrogen gen uptake increases with physical training mainly as a result
ion concentrations, and temperatures found in exercising mus- of a higher maximal CO. As stated earlier, the maximal CO is
cle. Low capillary Po2 also leads to improved carbon dioxide probably the limiting factor in exercise. Physical training
loading because lower oxyhemoglobin levels shift the carbon decreases the resting heart rate and increases the resting stroke
dioxide dissociation curve to the left (the Haldane effect). volume; an elite cross-country skier can have a resting heart
Exercise strenuous enough to cause a significant degree of rate as low as 40 beats/min. The maximal heart rate does not
anaerobic metabolism results in metabolic acidosis second- appear to be affected by physical training, but the heart rate of
ary to the increased lactic acid production. As discussed previ- a trained person is lower than that of an untrained person at
ously, the hydrogen ions generated by this process stimulate most levels of physical activity. Stroke volume is increased.
750 SECTION X Integrative Physiology

The arterial hemoglobin concentration and the hematocrit do


not appear to change with physical training at sea level, but the CLINICAL CORRELATION
arteriovenous oxygen content difference can increase with
physical training. This is probably a function of the greater A 58-year-old man, complaining of chest pain on exertion,
effects of local pH, Pco2 , and temperature in the exercising was referred to a cardiologist for a graded exercise stress
muscles, as well as an increased ability of the muscles to use test. The patient’s electrocardiogram, blood pressure, heart
oxygen. Blood volume is usually higher after an exercise– rate, tidal volume, respiratory rate, and arterial oxygen sat-
training program. uration by pulse oximetry were monitored during the test,
Physical training augments the oxidative capacity of skeletal which used a treadmill to provide an incremental work-
muscle by inducing mitochondrial proliferation and increasing load. At the stage of the test when the workload had dou-
the concentration of oxidative enzymes and the synthesis of gly- bled, the patient complained of chest pain and the stress
cogen and triglyceride. These alterations result in lower concen- test was stopped. During the test, his heart rate increased
trations of blood lactate in trained subjects than those found in from 75 to 135/min, and his blood pressure increased from
untrained people, reflecting increased aerobic energy produc- 150/90 to 180/95 mm Hg. His electrocardiogram was nor-
tion. Nevertheless, blood lactate levels during maximal exercise mal at rest, but showed significant ST segment depression
may be higher in trained athletes than in untrained people. at the highest exercise level (see Chapter 25). His oxygen
Maximal ventilation and resting ventilation do not appear to consumption increased from 300 mL/min to 1.5 L/min
be affected by physical training, but ventilation at submaximal during the test. Pulse oximetry did not show significant
loads is decreased, probably because of the lower lactic acid oxygen desaturation.
levels of the trained person during submaximal exercise. The Exercise stress tests are used to help diagnose cardiovas-
strength and endurance of the respiratory muscles appear to cular and pulmonary disease, as well as to determine the
improve with training. Total lung capacity is not affected by severity of the disease and the efficacy of treatment. Patients
training; vital capacity may be normal or elevated. Pulmonary may be asymptomatic at rest because of the physiologic
diffusing capacity is often elevated in athletes, probably as a reserves of the cardiovascular and respiratory systems; these
result of their increased blood volumes and maximal COs. reserves are often decreased in cardiopulmonary disease.
There are many changes in other physiologic systems that The data obtained during the increased workload of an
can occur with training. One of the most important is the exercise stress test can be useful in differentiating whether
female athlete triad that, by definition, occurs in women. The the problem is cardiovascular, respiratory, or both. In this
triad is amenorrhea (lack of menstrual cycles), osteoporosis case, the problem was cardiovascular, with coronary artery
(significant decreases in bone mineral density), and disor- disease the most likely diagnosis. The respiratory system
dered eating. This triad is found primarily in elite female ath- was able to meet the increased demand for alveolar ventila-
letes with low body energy stores. The amenorrhea (or tion in order to supply more oxygen and remove more car-
oligomenorrhea—irregular menses) is caused primarily by a bon dioxide. However, the increased cardiac work led to ST
decrease in luteinizing hormone (LH) release from the ante- segment depression, indicating an inability of the coronary
rior pituitary. Although the precise signal for this is not known, arteries to dilate sufficiently to provide additional blood
a decrease in the hormone leptin, released by adipose tissue, flow to meet the higher myocardial oxygen demand. The
resulting in a reduction of gonadotropin-releasing hormone cardiac workload, particularly the left ventricular work-
(GnRH) pulsatility has been implicated. Exercise by itself does load, is increased during exercise because of the increases
not cause amenorrhea—it contributes to the negative caloric in contractility and heart rate and stroke volume to increase
balance due to decreased caloric intake. The loss of bone mass the cardiac output, and because the increase in blood pres-
is primarily due to low estrogen levels because of decreased sure increases the afterload. Furthermore, left ventricular
LH (see Chapter 68). This greatly increases the risk of stress coronary perfusion mainly occurs during diastole, which is
fractures in female athletes. Unfortunately, some of this loss of much shorter at higher heart rates.
bone mass is not reversible even if normal menses and estro- Treatment of coronary artery disease is dependent on the
gen secretion occur with treatment. It is generally appreciated severity of the disease and may include low doses of aspirin
that eating disorders are more common in athletes. In women, to prevent blood clotting, drugs to decrease blood pressure
these are primarily anorexia nervosa (refusal to maintain and blood cholesterol, or interventions such as angioplasty
caloric input to maintain body weight) and bulimia nervosa (to open clogged arteries) with or without stents (to keep
(characterized by self-induced vomiting, as well as laxative them opened), or coronary artery bypass surgery.
and diuretic abuse). Although these extreme eating disorders
are not a requisite for the triad, caloric intake is usually sig-
nificantly lower than the increased energy expenditure in ath-
letes. The treatment of the female athlete triad generally CHAPTER SUMMARY
includes alterations in training type and intensity, increased ■ The main physiologic stresses of exercise are increased oxygen
caloric intake, and even administration of birth control pills or demand and removal of carbon dioxide and hydrogen ions, and
estrogen to try to restore bone mass. dissipation of heat produced by muscle metabolism.
CHAPTER 72 Exercise 751

■ The response to exercise is mainly an increase in cardiac output 2. Which of the following would be expected to have the largest
and alveolar ventilation. percent increase as a healthy person transitions from rest to
■ In a healthy young person, exercise is limited by the cardiovas- moderate exercise?
cular system, not the respiratory system. A) heart rate
■ Prolonged exercise decreases plasma volume, particularly in a B) stroke volume
warm environment. C) cardiac output
■ Training improves exercise performance mainly by improving D) mean arterial blood pressure
the cardiovascular response. E) diastolic arterial blood pressure
■ The female athlete triad is the combination of menstrual 3. Which of the following would be expected to decrease as a
disorders and low estrogen, decreases in bone mass, and healthy untrained person undergoes an exercise–training
decreased caloric intake despite increased energy expenditure program?
leading to a low body fat mass. A) resting stroke volume
B) resting cardiac output
C) resting heart rate
STUDY QUESTIONS D) maximal oxygen uptake
E) maximal alveolar ventilation
1. Which of the following would be expected to decrease as an
untrained person transitions from rest to moderate exercise?
A) pulmonary vascular resistance
B) pulmonary blood flow
C) alveolar ventilation
D) tidal volume
E) diffusing capacity
This page intentionally left blank
73
C H A P T E R

Aging
Hershel Raff

O B J E C T I V E S

■ Understand the changes in the nervous, muscular, cardiovascular, respiratory,


renal, gastrointestinal, and endocrine function that occur with aging.
■ Appreciate the pathophysiology of Alzheimer disease.

The life expectancy in most areas of the world has increased One of the important pathological causes of neurological
over the past few centuries. Much of this is due to the devel- decline associated with aging is dementia that is defined as a
opment of vaccines against infectious diseases as well as the significant loss of memory recall and other cognitive func-
development of antibiotics and antiviral therapy resulting in tions. Although there are many types of dementia, Alzheimer
decreased infant mortality. Furthermore, improvements in disease, a pathological deterioration of cognitive and behav-
the treatment of other chronic conditions such as cancer and ioral function associated with aging, is the most common type.
cardiovascular disorders, as well as improved sanitation, have This condition is associated with the development of amyloid
increased life expectancy. Along with this increase of lifespan, plaques and neurofibrillary tangles in the central nervous
there are many chronic diseases associated with aging such as system that leads to inflammation and neurotoxicity.
type 2 diabetes mellitus and osteoporosis that have increased
in frequency. Successful or healthy aging promotes the con-
cept that aging is not an illness, but a natural phenomenon MUSCULAR SYSTEM
that can be influenced by lifestyle, genetics, and environmen-
tal factors. In general, functional reserves such as exercise There is an overall loss of skeletal muscle mass (sarcopenia)
capacity decrease with age and, eventually, function itself with normal aging—this results in a decrease in lean body
declines and the susceptibility to disease increases. It is also mass. In addition, there is a loss of mitochondria, tendon
important to note that the effects of aging are quite variable strength, and elasticity. There is also loss of blood vessel den-
between males and females, ethnic groups, and populations. sity and cellular respiratory enzyme activity, leading to a
Table 73–1 summarizes some aging-related changes in the decrease in the ability to extract oxygen during exercise and
organ systems you have learned about in this book. decreased force production.

NEURAL SYSTEM CARDIOVASCULAR SYSTEMS


It is generally accepted that healthy individuals lose some cogni- One of the hallmarks of healthy aging is an increase in systolic
tive abilities with age. However, this is different from the devel- blood pressure with either no change or a decline in diastolic
opment of neurological diseases and syndromes associated with blood pressure (Figure 73–1). This increase in pulse pressure
aging. Reflexes can slow, resulting in an increased response time. indicates a loss of vascular compliance. Calcification of blood
Normal aging can often include a decline in visual, auditory, gus- vessels increases with age. When extreme, this “stiffening” of
tatory, and olfactory acuity, as well as deterioration in gait and the vascular bed is defined as arteriosclerosis. Table 73–2 lists
balance. This is why fall prevention is so critical in the elderly. other common cardiovascular changes with aging.

753

Ch73_753-756.indd 753 11/26/10 10:37:45 AM


754 SECTION X Integrative Physiology

TABLE 73–1 Aging-related changes. TABLE 73–2 Cardiovascular changes with aging.
Organ System Aging-related Changes Heart Decrease in maximal heart rate during exercise
Decrease in maximal cardiac output during
Nervous system Degenerative changes in neurons exercise
Loss of dendrites and synaptic connections Decrease in left ventricular compliance
Decreases in sensation, memory recall, and Increase in valve calcifications
communication
Increase in calcification of coronary arteries
Diminished visual, gustatory, auditory, and
olfactory senses Vascular Increase in total peripheral resistance
Muscular Decrease in number of skeletal muscle fibers and Increase in “stiffness”, decrease in aortic compliance
decrease in strength and capillary density (rarefaction)

Cardiovascular See Table 73–2 CV control Blunted baroreceptor reflex


Decrease in response to beta-adrenergic
Respiratory Loss of elasticity of lungs (increase in stimulation
compliance)
Decrease in the number of alveoli and surface Blood Decrease in total body water and blood volume
area for gas exchange CV hormones Decrease in plasma renin activity, angiotensin,
Reduced vital capacity and increased dead space and aldosterone
Reduced ability to clear airways by coughing

Renal/urinary Decrease in number of functional nephrons


Decrease in glomerular filtration rate
Decrease in tubular secretion and reabsorption
Increased risk of dehydration
PULMONARY SYSTEM
Increase in the frequency of incontinence Important changes in the structure and function of the pulmo-
Gastrointestinal Decreased capacity to secrete pancreatic nary system include loss of alveolar elastic recoil resulting in
enzymes increased lung compliance, decreased respiratory muscle
Decreased vitamin D and calcium absorption strength, changes in the rib cage and spinal column resulting
(contributes to loss of bone mineral density) in decreased chest wall compliance, and loss of alveolar sur-
Decrease in motility (contributes to constipation) face area and pulmonary capillary volume. The decreases in
Endocrine Decrease in some hormones in the blood lung elastic recoil can lead to an increase in functional
(e.g., GH, androgens, DHEA) residual and closing capacities.
Menopause and associated phenomena One of the hallmarks of aging is an increase in the difference
(e.g., loss of bone mineral density) between the partial pressure of alveolar oxygen (PAo2), which
Data from Shier D, Butler J, Lewis R. Hole’s Human Anatomy & Physiology 11th is calculated using the alveolar air equation and does not
Edition. McGraw Hill Higher Education. Boston, 2007 and Kane RL, Ouslander JG, change significantly with age, and arterial oxygen (Pao2). This
Abrass IB, Resnick B. Essentials of Clinical Geriatrics 6th Edition. McGraw Hill Medical,
New York, 2009. increase in the so-called A–a oxygen gradient reflects a nearly
linear decrease in Pao2 with age. This is primarily due to an
increase in ventilation–perfusion mismatching resulting
from changes in pulmonary blood flow and volume, increases
200 in pulmonary compliance leading to airway and alveolar col-
lapse, and a decrease in diffusing capacity due to a decreased
surface area for gas exchange. When evaluating blood gases in
150 an elderly patient, it is important to realize that a lower Pao2
Pressure (mmHg)

Systolic pressure may be a result of normal aging rather than due to a specific
disease that requires treatment.
Mean pressure
100

Diastolic pressure
50
RENAL/URINARY SYSTEM
After the age of about 30 years, the number of functional
glomeruli declines and, by age 80, may have decreased from a
0
0 20 40 60 80
peak of about 2.2 to about 1.2 million. In addition, there is a
Age (years) decrease in renal plasma flow due to changes in the small
FIGURE 73–1 Changes in arterial pressure with age in the renal arteries and the afferent arterioles. As a result, creatinine
US population. (Adapted with permission from National Institutes of Health clearance, an index of glomerular filtration rate, decreases
Publication #04-5230, August 2004.) by approximately 30%. There is also a decrease in urinary
CHAPTER 73 Aging 755

diluting and concentrating capacity. Despite this, basal water cal changes in autonomic function and behavior, as well as sys-
and electrolyte balance are usually not significantly altered temic consequences. There appears to be a higher incidence of
during healthy aging. The decrease in total body water is more cardiovascular dysfunction after menopause. The decline in
due to a loss of lean body mass rather than renal function. male reproductive function (the andropause) is much more
The risk of dehydration is increased during healthy aging. subtle. Although there can be a decrease in plasma free and
This is due to a decrease in the ability to maximally concentrate total testosterone levels and sperm production, it is well known
the urine due to a decline in tubular function. Thirst is also that many men can remain fertile well into their 80s.
compromised with aging. Furthermore, the antidiuretic hor- There is a subtle decline in insulin sensitivity such that fast-
mone (arginine vasopressin) response to an increase in plasma ing blood glucose tends to increase, as does the risk of the
osmolality can be altered, and the ability to defend total body development of type 2 diabetes mellitus. Finally, growth hor-
water and blood volume during dehydration is compromised. mone and insulin-like growth factor I (IGF-1) concentra-
Incontinence is involuntary urination with many causes, tions decline with aging. It has been hypothesized that these
and is a significant health and hygiene problem in the elderly. contribute to the loss of muscle mass described earlier, and
In men, the development of benign prostatic hyperplasia can some have advocated growth hormone replacement therapy in
lead to difficulty in voiding. In women after menopause, estro- the elderly with established growth hormone deficiency.
gen deficiency is known to be associated with structural
changes in the vagina, urethra, and bladder, all of which
increase the risk of incontinence.
CLINICAL CORRELATION
(Adapted from Toy E et al: CASE FILES; Pathology. McGraw-
GASTROINTESTINAL SYSTEM Hill, 2006):
An elderly man is found wandering around your campus
The changes in the gastrointestinal tract with aging are subtler looking for his way home. You notice a wristband that says
than the other systems described in this chapter. A loss of he is 88 years old and a resident of a local nursing home. He
swallowing coordination, combined with increased gastro- is not bleeding, has no obvious bruises, and is not limping.
esophageal reflux, can lead to an increased risk of aspiration In addition to being lost and disoriented, his sentences do
pneumonia. The colon tends to thicken, resulting in increased not make sense and his conversation is not coherent. He is
contraction strength that may increase the risk of diverticuli, alert and cooperative, but confused. You call the nursing
which are pouches that can form from the wall of the colon home and they immediately call for an ambulance that
that can become infected and even burst, releasing luminal arrives in 10 minutes and takes the man to the emergency
contents into the peritoneal cavity. The decline in vitamin D room of the local hospital. The emergency room physician
absorption and tendency to get less sunlight leads to vitamin makes a provisional diagnosis of dementia and examines
D insufficiency and a subsequent decrease in calcium absorp- the patient. He has no obvious injuries nor does he have
tion. This can exacerbate the decline in bone mineral density neurological signs that would suggest a brain tumor or
that occurs with age. The subtle decline in liver function can stroke. A representative of the nursing home arrives at the
alter drug metabolism sufficiently to increase the risk of drug emergency room with the patient’s medical records that
toxicity. Although decreases in motility of the intestine are not indicate the man has Alzheimer disease.
dramatic, there is an increase in the risk of constipation. Alzheimer disease is the most common cause of demen-
Finally, the maximal function of the exocrine pancreas can tia in the elderly. It usually starts with a slow loss of advanced
result in a decrease in enzyme secretion into the small intes- mental functions such as the ability to do mathematical
tine—this is usually not sufficient to alter the digestion and problems (e.g., pay bills, manage the check book or house-
absorption of proteins and fats. hold finances) and word recall. There is often a change in
mood and behavior early in the disease. As the disease pro-
gresses, higher brain functions involving the cerebral cor-
ENDOCRINE SYSTEM tex are lost, leading to disorientation (demonstrated in our
patient) and memory loss. In late stages, patients often do
There are many hormonal changes that occur with aging. The not speak or move.
most dramatic is the cessation of ovarian function in women Although there is a familial form of Alzheimer disease,
that results in menopause described in detail in Chapter 68. most cases in the elderly occur without a strong family his-
Briefly, loss of estrogen synthesis from the ovaries results in a tory. When the brain is examined post mortem, atrophy of
large increase in gonadotropin secretion from the anterior the cerebral cortex is a common finding. The most com-
pituitary due to loss of negative feedback. Low estrogen levels mon abnormalities found during histological examination
result in many physiological changes, probably the most dra- of the brain are neurofibrillary tangles and amyloid plaque
matic being loss of bone mineral density (osteopenia) that can deposition. Neurofibrillary tangles are bundles of filaments
become severe enough to increase the risk of bone fractures composed primarily of phosphorylated tau protein found
(osteoporosis). The low estrogen levels can lead to physiologi-
756 SECTION X Integrative Physiology

in the neuronal cytoplasm around the nucleus. Amyloid STUDY QUESTIONS


plaque deposits are extracellular, and contain several abnor- 1. Which of the following is true?
mal proteins. All of these abnormal proteins interfere with A) Both arterial and pulmonary compliance increase
normal neurological function. with aging.
Alzheimer disease usually progresses slowly for 10–20 B) Arterial compliance decreases and pulmonary
years. As in our patient, forgetfulness and difficulty with compliance increases with aging.
sentence formation (aphasia) are common signs. As the C) Arterial compliance increases and pulmonary
disease progresses, the loss of motor function ensues. compliance decreases with aging.
Patients develop difficulty with balance and walking, and D) Both arterial and pulmonary compliance decrease
are prone to fall. The cause of death is usually another pri- with aging.
mary disease, such as pneumonia, the susceptibility to 2. All of the following decrease with aging except
which increases dramatically due to the loss of motor func- A) number of glomeruli in the kidney
tions such as the ability to chew and swallow properly. B) creatinine clearance
C) renal plasma flow
D) risk of dehydration
3. The decrease in bone mineral density in elderly women is due
CHAPTER SUMMARY at least in part to
A) decrease in vitamin D absorption
■ It is normal for some memory and sensory functions to
B) increase in calcium absorption
decrease with age.
C) increase in estrogen secretion
■ There is typically a loss of muscle mass with aging. D) decrease in cortisol
■ An increase in arterial systolic and pulse pressure is typical 4. Which plasma hormone increases with aging?
with aging.
A) renin
■ Decreased lung elasticity increases the functional residual B) pituitary gonadotropins in women
capacity and decreased Pao2 increases the alveolar–arterial C) growth hormone
gradient for oxygen with aging. D) free testosterone in men
■ A decrease in renal glomerular number and creatinine
clearance occurs with aging.
■ In women, ovarian function invariably ceases, causing
menopause characterized by a decrease in estrogen in
the blood and loss of bone mass. A decrease in vitamin D
and calcium absorption further contributes to the loss of
bone mass.
Answers to Study Questions

Chapter 1 Chapter 12
1. B 2. D 3. C 4. B 1. B 2. E 3. B 4. D 5. E

Chapter 2 Chapter 13
1. C 2. E 1. C 2. D 3. A 4. D 5. D

Chapter 3 Chapter 14
1. C 2. E 3. D 4. E 5. C 6. D 1. C 2. E 3. C 4. E

Chapter 4 Chapter 15
1. A 2. D 3. C 4. B 1. D 2. D 3. B 4. E 5. D 6. B 7. D

Chapter 5 Chapter 16
1. A 2. D 3. A 1. E 2. B 3. C 4. E 5. D 6. D 7. E

Chapter 6 Chapter 17
1. C 2. B 3. E 4. C 5. A 6. D 1. D 2. C 3. D 4. D 5. C 6. D

Chapter 7 Chapter 18
1. A 2. D 3. B 4. B 5. C 6. C 7. D 1. C 2. E 3. E 4. D 5. C

Chapter 8 Chapter 19
1. D 2. B 3. A 4. C 1. D 2. B 3. D 4. D

Chapter 9 Chapter 20
1. B 2. C 3. B 1. C 2. D 3. C 4. D 5. A 6. C

Chapter 10 Chapter 21
1. D 2. D 3. A 4. A 1. A 2. C 3. D 4. B 5. D 6. D

Chapter 11 Chapter 22
1. A 2. C 3. B 4. C 1. E 2. D 3. E 4. B 5. B

757

Answers_757-760.indd 757 11/30/10 2:15:02 PM


758 Answers to Study Questions

Chapter 23 Chapter 37
1. B 2. E 3. C 4. B 5. A 1. C 2. D 3. B 4. A

Chapter 24 Chapter 38
1. B 2. A 3. C 4. A 5. A 1. B 2. D 3. D 4. B

Chapter 25 Chapter 39
1. B 2. D 3. A 4. C 5. C 1. B. 2. C 3. C 4. C 5. D 6. B

Chapter 26 Chapter 40
1. E 2. E 3. D 4. C 5. A 1. D 2. B 3. B 4. C 5. C

Chapter 27 Chapter 41
1. B 2. C 3. A 4. C 5. C 1. D 2. A 3. C 4. B 5. C

Chapter 28 Chapter 42
1. E 2. B 3. A 4. D 5. E 1. C 2. D 3. B 4. D 5. A

Chapter 29 Chapter 43
1. A 2. B 3. E 4. A 5. A 1. C 2. D 3. A 4. C 5. A 6. B

Chapter 30 Chapter 44
1. E 2. B 3. C 4. E 5. E 1. B 2. B 3. D 4. A 5. B 6. C

Chapter 31 Chapter 45
1. F 2. E 1. D 2. A 3. A 4. C 5. A

Chapter 32 Chapter 46
1. C 2. D 3. A 4. C 5. B 6. D 1. D 2. C 3. B 4. B 5. D

Chapter 33 Chapter 47
1. F 2. D 3. C 4. C 5. A 1. B 2. A 3. D 4. C 5. C 6. B

Chapter 34 Chapter 48
1. C 2. E 3. B 4. B 5. E 1. C 2. B 3. A 4. A 5. D 6. D

Chapter 35 Chapter 49
1. E 2. A 3. B 4. A 1. A 2. D 3. A 4. B 5. E

Chapter 36 Chapter 50
1. E 2. D 3. E 4. A 1. A 2. E 3. E 4. D 5. B 6. A
Answers to Study Questions 759

Chapter 51 Chapter 63
1. A 2. E 3. C 4. C 5. D 1. C 2. C 3. A 4. C

Chapter 52 Chapter 64
1. A 2. A 3. D 4. E 5. A 1. B 2. D 3. A 4. C 5. C

Chapter 53 Chapter 65
1. B 2. C 3. C 4. E 5. D 1. A 2. B 3. C 4. D 5. B 6. C

Chapter 54 Chapter 66
1. A 2. E 3. A 4. B 5. C 6. E 1. C 2. C 3. B 4. C 5. A

Chapter 55 Chapter 67
1. E 2. C 3. D 4. C 5. B 1. C 2. C 3. B 4. C

Chapter 56 Chapter 68
1. A 2. E 3. A 4. E 5. D 1. D 2. A 3. C 4. A 5. B

Chapter 57 Chapter 69
1. C 2. E 3. B 4. B 5. A 1. B 2. B 3. C 4. B

Chapter 58 Chapter 70
1. C 2. B 3. E 4. E 5. B 1. D 2. A 3. B 4. C

Chapter 59 Chapter 71
1. E 2. D 3. D 4. D 5. E 1. D 2. A 3. C 4. B 5. B

Chapter 60 Chapter 72
1. B 2. E 3. D 4. C 1. A 2. C 3. C

Chapter 61 Chapter 73
1. A 2. D 3. B 4. C 5. D 1. B 2. D 3. A 4. B

Chapter 62
1. B 2. A 3. C 4. B
This page intentionally left blank
761 INDEX

Index
Page numbers followed by f or t indicate figures or tables, respectively.

A metabolic acidosis, renal response to, 482 adrenal medulla, hormones


A–a oxygen gradient, 754 metabolic alkalosis, factors, 482 adrenal catecholamines, overproduction
ABC transporters, 24 respiratory acidosis, 378, 481 diseases, 667
ABC5 transporter, 568 respiratory alkalosis, 378–379, 394, 481, 736 adrenergic receptors, regulation, 667
ABC8 transporter, 568 respiratory acidosis and alkalosis, renal catecholamine physiologic effects, 667,
Abducens nerves, 143 response to, 481–482 667t
Absence seizures, 185 Acidemia, 375, 380, 464, 469, 470 catecholamine release, transport, and
Absolute refractory state, 212 Acidosis, 375, 377, 647 metabolism, 665
Absorption process, 491 Acid-sensing ion channels (ASICs), 45 chemistry and biosynthesis, 665
Accessory muscles of inspiration, 315, 316 Acini, 308, 492 target organ cellular effects, 665–667
Acclimatization process, 730 Acromegaly, 630, 631 functional anatomy and zonation, 655
Accommodation, 136 ACTH-independent Cushing’s syndrome, 664 adrenal androgens diseases, 664–665
ACE inhibitors, 453, 461 Actin, 80, 83 adrenal cortex hormones, 656–664
Acetaminophen, 732 Actin filament, 80, 83 glucocorticoids diseases, 664
Acetazolamide, 737 Action potential, 10, 47 mineralocorticoids diseases, 664
Acetylcholine (ACh), 56, 60–61, 85, 86, 178, 188, cardiac muscle, 54–55 steroid hormone, target organ cellular
206, 216, 268, 346, 510, 519, 529, 544, 671 SA and AV node, 55–56 effects, 660–661
receptors, 57, 60, 62f compound, 54 Adrenal insufficiency, 664
synapse, 62f depolarization, 48 Adrenal medulla, 56, 63, 72f, 178f, 179, 182, 268,
Acetylcholinesterase (AChE), 61, 85f, 86, 103 externally recorded, 53f 405, 464, 715
Acetyl-CoA carboxylase, 675 positive feedback cycle, 48f Adrenergic receptors, 63
ACh. See Acetylcholine (ACh) propagated, 10 α-Agonists, 667
ACh-activated K channel (KACh), 57 refractory periods, 47, 51 β-Agonists, 667
ACh receptor channels (AChRs), 20 absolute and relative, 52f β-Adrenergic blockers, 222, 273
muscarinic AChRs, 20 regenerated, 39 β1-Adrenergic receptor blockers, 231
nicotinic AChRs, 20 threshold, 51 β1-Adrenergic receptor–protein kinase
ACh receptors (AChRs), 60 Activation gate, 213 A-dependent process, 451–452
Muscarinic AChRs, 60 Activation of microglia, 112 alpha-adrenergic receptors, 64, 267, 708
nicotinic AChRs, 60 Active expiration, 317 α1−Adrenergic receptors, 267, 665
Acid–base balance, 306, 375–382, 471–472 Active hyperemia, 265 α2−Adrenergic receptors, 665
acid–base chemistry, 375–377 Active membrane transport pathways, isoproterenol, 665
acid–base paths in vivo, 378f characteristics, 528t beta-adrenergic receptors, 64, 638, 692
aldosterone, influence of, 482–483 Active spread of action potentials, 39 β1−Adrenergic receptors, 665
chloride depletion, influence of, 482 Active transport, 4 β2−Adrenergic receptors, 268, 665
extracellular volume contraction, Activin, 686 β3−Adrenergic receptors, 665
influence of, 482 Acute cholecystitis, 572 and signaling pathways, 667t
fundamentals, 472–473 Acute mountain sickness, 736, 737, 742 Adrenergic sympathetic nerves, 323
and osmolar regulation of potassium prevention and treatment, 737 Adrenocorticotropic hormone
distribution, 725 Acute respiratory distress syndrome, 321 (ACTH), 623
regulation, 398, 471 Adaptation, 118 Adult respiratory distress syndrome, 321
renal excretion, 476–478 Addison’s disease, 664 Afferent arterioles, 409
hydrogen ion excretion on ammonium, Adenohypophysis, 623 Afferent fibers, 127
478–480 Adenoids, 308 Afterload, 87, 88, 95
phosphate and organic acids as urinary Adenoma, 470 aortic pressure, 95
buffers, 477–478 Adenosine, 102, 232, 264, 457 cardiac, 232, 301
quantification, 480 Adenosine triphosphate (ATP), 11, 45, and isotonic contraction, 219–220
titratable acidity, 480 79, 607 muscle, 96
urinary buffers, hydrogen ion excretion, 477 production, 745 result in less shortening, 95
renal handling, 474–476 Adenylyl cyclase (AC), 21, 25, 607 ventricular, 227
regulation, renal glutamine control, Adequate stimulus, 43, 117 effect of changes, 228–229
metabolism and, 480–481 ADH. See Antidiuretic hormone (ADH) Ageusia, 162
sources Adjuvant, 538 Aging, 712
carbohydrate and fat, anaerobic ADP/ATP ratio, 18 cardiovascular changes, 754t
metabolism, 474 Adrenal androgens, 664. See also Androgens cardiovascular systems, 753–754
dietary protein, metabolism, 473 Adrenal cortex, 268, 405, 452f, 454, 455, 457, changes in arterial pressure with age, 754f
dietary weak acids, metabolism, 473 467, 602, 626 clinical correlation, 755–756
GI secretions, 473–474 Adrenal cortex hormones endocrine system, 755
intravenous solutions, lactated Ringer’s, 474 androgens, 664 gastrointestinal system, 755
Acid–base disorders, 377, 481 glucocorticoids, 661–662 hormone production associated
acid–base disturbances, 381t deficiency, 664 with, 712f
metabolic acidosis, 377, 379, 382, 392, 481, 679, excess, 664 muscular system, 753
725, 737, 749 mineralocorticoids, 662–664 neural system, 753
metabolic alkalosis, 379, 481, 482, 483f Adrenal gland, 178, 656f pulmonary system, 754

761
762 INDEX

Aging (continued) Aminoglycoside antibiotics Anomic aphasia, 196


related changes, 754t gentamicin, 154 Anorexia nervosa, 722, 750
renal/urinary system, 754–755 streptomycin, 154 Anosmia, 161, 165
successful/healthy, 753 α-Amino-3-hydroxyl-5-methyl-4-isoxazole- ANS. See Autonomic nervous system (ANS)
Agouti-related peptide (AgRP), 721 propionate (AMPA), 194 Antagonists, 20
Air conduction, 153 channel, 74 Anterior insula, 162
Airway resistance, 322 receptors, 194 Anterior pituitary cell type, 625t
Akinesia, 171 Amiodarone, 639 Anterior pituitary gland, 613
Albumin, 208, 404, 412, 426, 431, 569, 576, 578, Ammonia (NH3), 477, 578 diseases of
637, 647, 660, 665, 705 homeostasis, 579f hormone-producing pituitary
Aldosterone, 268, 405, 454, 467, 480, 531, 626, metabolism, principles adenoma, 630
656, 724, 731. See also Mineralocorticoids extraintestinal production, 579 hypopituitarism, 630–631
action mechanism, 456f intestinal production, 578–579 function, evaluation of, 613
deficiency, 664 role and significance, 578 functional anatomy, 623
excess, 664 urea cycle, 579 hypothalamic control of, 623–624
renal physiologic effects, 663f urea disposition, 579–580 Anterior pituitary hormones, 615,
Alerting reaction, 289 production, sources, 578f 623–630, 625f
Alerting response, 290 Ammoniagenesis, 477, 479f deficiency of, 630
Alkalemia, 375, 376, 381, 464, 469, 470, 484 and excretion, 479f glycoproteins, 625–626
Alkaline phosphatase, 647 Ammonium ion (NH4+), 478 growth hormone, 626–629
Alkaline tide, 513 AMP-activated protein kinase, 680 measurements, 631
Alkalosis, 375, 376, 377, 379, 647. See also Amphipathic, 565 prolactin family, 629–630
Metabolic alkalosis; Respiratory alkalosis character of bile acids, 567, 596 proopiomelanocortin (POMC), 626
Allodynia, 116 lipids, 16 target organs, and physiologic effects, 624f
Alpha-adrenergic receptors, 64, 102, 323 term defined, 565 Anterior wall myocardial infarctions, 222
Altitude, 735 Ampulla, 150 Anterograde conduction, 110
acclimatization to, 737–738 Amygdala, 159, 161f, 173f, 192f, 194–196 Anterograde amnesia, 196
acute effects of, 736 Amylin, 678 Anticholinergics, 384
acute mountain sickness, 736 Amyloid plaques, 753, 755 Anticholinesterases, 61, 70
adaptive response, 737 Amyloid precursor protein (APP), 194 Anticoagulants, 209, 361
physiologic responses to, 738t Amylolytic enzymes, 517 Antidiuresis, renal water handling, 446f
symptoms, 736 Amylopectin, 584 Antidiuretic hormone (ADH), 27, 64, 268,
Alveolar air equation, 338, 355, 735 structure, 585f 404–406, 443, 444, 446, 448, 458–461, 615,
Alveolar–arterial PO2 difference, 355, 383 Amylose, 584 739, 755
Alveolar–capillary diffusion, 382 structure, 585f cellular mechanism of, 618f
Alveolar–capillary unit, 309 Amyotrophic lateral sclerosis (ALS), 63, 86, magnocellular neurons, 614, 617
Alveolar dead space, 347, 354, 361 130, 176 osmoreceptor control, 459
Alveolar ducts, 308 Anabolic metabolism, 715 physiologic effects of, 618–619
Alveolar edema, 382 Anaerobic metabolism, 745 production, disorders of, 620
Alveolar gas, 337 Anal canal, anatomy, 553f release, control of, 619–620
Alveolar hypercapnia, 348 Anandamide, 75 secretion, inappropriate
Alveolar hypoxia, 348 Anaphylactic shock, 301 syndrome of, 620
Alveolar macrophage, 310 Anaphylaxis, 307, 541 signals, integration of, 619f
Alveolar PO2, 735 Anatomic dead space, 308, 334, 335 synthesis and processing of, 616f
Alveolar pressure, 314, 735 Anatomic shunts, 355 Antigen–antibody complexes, 416
Alveolar sacs, 308 Androgen-binding protein (ABP), 685 Antileukotriene, 384
Alveolar septa, 308 Androgens, 664, 687 Antrum, 508, 548
Alveolar ventilation, 331, 334, 353, 481 anabolic and metabolic effects of, 692 Aortic arterial chemoreceptors, 386
anatomic dead space and, 334–335 physiologic effects, 687 Aortic baroreceptors, 287
and carbon dioxide, 337 sex determination, 687 Aortic insufficiency, 247
measurement of, 335 sexual development, 687 Aortic regurgitation, 247
and oxygen, 338 sexual differentiation, 688–689 Aortic stenosis, 245–247
physiologic dead space, 335–336 sexual maturation, 690 Aortic valve obstruction, 229
Bohr equation, 335 Andropause, 755 Aortic valve stenosis, 247, 282
regional distribution, 338–339 Anemia, 406, 693 Aphasias, 196, 756
closing volume, 339 Anemic hypoxia, 383 Apical epithelial cell membrane. See Luminal
Alveolar vessels, 343 Anergy, 538 epithelial cell membrane
Alzheimer’s disease, 63, 193–195, 753, 755 Angina pectoris, 231, 248, 272 Apical membrane, 29, 424
familial form, 755 Angioplasty, 750 Apical sodium-dependent bile salt transporter
Ambiguous genitalia, 667 Angiotensin (asbt), 569
Amenorrhea, 631, 750 angiotensin I, 451, 658 Apnea, 388, 739
Amiloride, 163, 470 angiotensin II, 268, 302, 451, 454–456, 658 Apneustic center, 387
Amiloride-sensitive epithelial Na+ channel Angiotensin converting enzyme (ACE), Apoproteins, 597
(ENaC), 163, 662 451, 658 Apoptosis, 493
Amino acids, 61 inhibitor, 282, 293, 309, 461 Aptotagmin, 66
essential, 583, 587–588 Angiotensin II receptor blockers (ARBs), Aquaporins (AQPs), 21, 27, 426, 442, 458
naturally occurring, 588f 293, 453, 470 key features of, 619t
nonessential, 560 Angiotensinogen, 451, 658 phosphorylation of, 618
transporters, 430, 531, 590 Angle-closure glaucoma, 133 Arachidonic acid metabolites, 455
γ-Aminobutyrate (GABA), 106, 159 Anion exchanger (AE), 24 Arcuate arteries, 409
synapse, 63f Anion gap, 381–382, 679 Area postrema, chemoreceptor trigger zone, 551
INDEX 763

A receptors, 21 Aspirin, 117, 282, 732, 750 Axoplasmic transport, 60, 66


Areflexia, 168 Associative learning, 76 orthograde/retrograde., 66
Arginase, 579 Astereognosis, 195 Azotemia, 461
Arginine, 579 Asthma, 309, 326
Arginine succinate, 579 Astigmatism, 136 B
Arginine vasopressin (AVP), 444, 458, 615, 617, Astrocytes, 105, 106, 110, 176 Babinski sign, 169
755. See also Antidiuretic hormone (ADH) Astrocytic proliferation, 112 Bacteremias, 379
Aromatase, 686 Ataxia, 175, 183 Bacterial metabolism, 577
Arterial–alveolar CO2 difference, 336, 355 Atelectasis, 319, 348, 360, 388 Bacteroides, 538
Arterial baroreceptors, 287, 390 Atherosclerosis, 248, 261, 272, 462 β2-Adrenergic agonist, 384
pathway, 286f Athetosis, 171 β-Adrenergic blockers, 222, 273, 293
reflex, 286, 286f, 746 ATPase activity, 91 β1-Adrenergic receptor blockers, 231
Arterial blood gases, 380 ATP-binding cassette (ABC), 24 β1-Adrenergic receptor–protein kinase
clinical interpretation, 380–381 ATP-sensitive channels, 21 A-dependent process, 451–452
anion gap, 381–382, 679 ATP-sensitive K+ channels (KATP), 673 β1-Adrenergic receptors, 217
base excess, 381 Atrial fibrillation, 242, 420 β2-Adrenergic receptors, 268
Arterial blood pressure, 203, 210, 252, 290, Atrial flutter, 241 Balance concept. See Electrolyte balance concept;
300, 372, 444, 458, 460. See also Arterial Atrioventricular (AV) nodes, 54, 212, 236 Water, balance concept
pressure Atrophic gastritis, 538 Ballism, 171
regulation, 267, 288, 291, 398 Atropine, 60, 61, 72, 179, 432 β-Amyloid peptides (Αβ), 194
Arterial chemoreceptors, 289, 323, 351, 372, 390, Auditory agnosia, 121 Barbiturates, 63, 74
392, 394, 473, 736, 747 Auditory cortex, 153 Baroreceptors, 222, 297, 372, 450, 450f
carotid bodies, 749 Auditory meatus, external, 147 intrarenal, 451
Arterial–end-tidal CO2 difference, 355 Auditory ossicles, 147 Barotrauma, 741
Arterial gas embolism, 741 incus, 147 Basal electrical rhythm (BER), 549, 549f
Arterial hypocapnia, 737 malleus, 147 Basal ganglia, 167, 171, 172f, 173f, 183
Arterial oxygen, 754 stapes, 147 caudate nucleus, 171
Arterial pressure Auditory receptors, 149, 150, 152 globus pallidus, 171
auscultation technique, 260 electrical responses, 150 hyperkinetic diseases, 171
changes with age, 754f genesis of action potentials in, 150, 152 tremor, chorea, athetosis, ballism, 171
determinants, 260 sound transmission, 152 hypokinetic diseases, 171
arterial compliance, 261 Auditory (Eustachian) tube, 147, 153f akinesia and bradykinesia, 171
arterial pulse pressure, 260–261 Auscultation, 226, 235, 260f putamen, 171
mean arterial pressure, 260 Autoimmune disease, 86, 112, 634 substantia nigra, 171
Korotkoff sounds, 260 Autoimmunity, 538 pars compacta and pars reticulata,
long-term regulation, 290 Autonomic activity 171
fluid balance & arterial pressure, 291 reflex and central control, 183 subthalamic nucleus, 171
on urinary output rate, 291–292 Autonomic nerve activity, 181 Basal metabolic rate (BMR), 716
measurement, 259–260 responses of some effector organs to, 181–182t Basal stem cells, 159
regulation of, 285–286 Autonomic nervous system (ANS), 71, 80, 94, Basal tone, 264
short-term regulation, 286 177, 286, 523 Base deficit, 381
arterial baroreceptor reflex, 286–288 craniosacral division, 178 Base excess, 381
central command, 290 efferent fibers and motoneurons, 72f Basement membrane, 494
chemoreceptor reflexes, 289 intermediolateral column (IML), 177 circular muscle, 494
reflexes from receptors in heart & parasympathetic, 177 lamina propria, 494
lungs, 289 oculomotor, facial, glossopharyngeal mucosa, 494
reflex responses to pain, 290 nerves, 178 muscularis mucosae, 494
responses associated with emotion, 289 vagus nerves, 178 submucous (submucosal) plexus, 494
Arterial pulse pressure, 260 peripheral organization and transmitters Basilar membrane, 149
Arteriolar tone, 264 released by, 178 Basket cells, 109, 174
control of, 264 preganglionic and postganglionic neurons, 177 Basolateral membrane, 29, 424
basal tone, 264 sympathetic, 177 B cells, 536
flow responses caused by, 265–267 Autoregulation, 266, 415 Bell’s palsy, 162
hormonal influences, 268 Autotransfusion, 302 Benign prostatic hyperplasia, 755
angiotensin II, 268 AVP. See Arginine vasopressin (AVP) Benzocaine, 53
circulating catecholamines, 268 Axis deviations, ventricular, 240f Benzodiazepines
vasopressin, 268 Axon, 9, 10f, 106 diazepam (Valium) 63, 74
local influences on, 264, 266t conduction velocity, 107 Beta-adrenergic receptor blockers, 282
local metabolic influences, 264–265 degeneration, 111–112 Beta-adrenergic receptors, 94
local nonmetabolic influences, 265 of first-order neurons, 119f Beta2 (β2) receptors, 323
transmural pressure, 265 initial segment, 106 Bezoars, 551
neural influences, 267 reaction, 66 Bicarbonate
sympathetic vasoconstrictor nerves, 267 regeneration, 111–112 reabsorption, predominant proximal tubule
organ blood flow responses caused by, 267f of second-order neurons, 162 mechanism, 475f
Arteriosclerosis, 261, 753 sprouting, 111 secretion
Asbestosis, 329, 360 of sympathetic preganglionic neurons, 177 generic model, 474f
Ascending reticular system, 111f unmyelinated, 106, 615 renal contribution, 480t
Ascites, 284, 563, 664 Axonal conduction velocity, 107 Biguanides, 680
Ascorbic acid, 590 Axonal sprouting, 111 Bile
Aspartate, 430, 579 Axon collaterals, 130 composition
Aspiration, 329 Axon hillock, 72, 106 canalicular bile, 567–568
764 INDEX

Bile (continued) acidify/alkalinize process, 481t reticulospinal, 167


changes in, 571f cells, 207–208, 208t rubrospinal, 167
ductular bile, 568 flow, basic physics of, 202–203 tectospinal, 167
hepatic bile, 569 hemostasis, 208–209 vestibulospinal tracts, 167
concentration mechanism, 572f macrophages, 559 Breast milk lipase. See Cholesterol esterase
ducts, 562 plasma, 4, 208 Breathing work, 748
excretion and secretion, principles of adult, normal constituents, 209t elastic work, 737
role and significance, 565 pressure, 398–399, 411f resistance work, 737
relationship to cholesterol, 570f Blood–brain barrier, 106, 272, 392, 394, 575, Broca’s area, 195
solute entry pathways, 568f 675, 731 Brodmann’s area , 118, 141, 153, 169
Bile acids, 530, 561, 565 Blood gas analysis, 483 Bronchial blood flow, 341
circulation, quantitative aspects, 566 Blood glucose regulation, 715 Bronchial circulation, 341
conjugated bile acids, 566, 567f postprandial state, 715 Bronchioles, 307, 308
enterohepatic circulation, schematic, 561f Blood oxygen content, 363 Bronchitis, 326
conjugation, 566–567 Blood plasma, 4 chronic, 481
enterohepatic circulation Blood–testis barrier, 684, 685, 690 Bronchoconstriction, 737
hepatocyte transport mechanisms, 569 Blood urea nitrogen (BUN), 434 Brown-Séquard syndrome, 131
intestinal uptake mechanisms, 569 Body fluid compartments, 200, 437–438 Bruits, 258
synthesis and transport, regulation, 569–570 Body plethysmography, 325, 333, 334 Brush border, 29
metabolism from cholesterol, 565–566 Body temperature control, 729–734 hydrolysis, 585
physical forms, 567f clinical correlation, 732–733 membrane, 584, 586f, 596, 598
physicochemical properties, 567 effector mechanisms, 730 Buffer, 5, 375
primary, 566 feedback mechanisms, 730 Buffer systems, 4–5, 472
structures, 566f fever, 731–732 calcium-buffering system, 487
secondary, 566 infection pathway, 732f and pH, 4–5
structures, 566f heat loss/gain mechanisms, 730 phosphate and albumin, 472
Bile ducts, 562 hot/cold environment, adaptation, Bulbourethral glands, 683
Bile salt export pump (BSEP), 569 730–731 Bulimia nervosa, 750
Biliary colic, 572 Bohr effect, 366, 371, 376, 749 Bundle branch blocks, 242
Biliary ductules, 562 Bohr equation, 335, 355 α-Bungarotoxin, 70
Biliary system, 559, 561 Bone, 487 Burst lung, 741
functional anatomy, 562f fractures (See Osteoporosis)
motor functions marrow, 575 C
contraction, 571 mineral density (See Osteopenia Ca-activated K channels, 19, 75
sphincter of Oddi function, 571–572 and Osteoporosis) Ca-ATPase, 486
Bilirubin, 539 Bone conduction, 153 Cable properties, 39
conversion of heme, 576f Bone density, 652 Cadherins, 26, 74
cycling, 577f T score, 652 Caffeine, 21, 156
handling by hepatocytes, 577f Z score, 652 Ca2+ influx, 194
homeostasis Bone metabolism Ca2+ ion channels, 194
bacterial metabolism, 577 factors involved in regulation of, 650t Calbindins, 486
hyperbilirubinemia, 578 hormonal regulation, 651 Calcarine fissure, 141
urinary elimination, 577–578 childhood–adult, 651 Calcinosis, 650
metabolism, principles menopause, 652–653 Calcitonin, 487, 633, 652
hepatic transport mechanisms, 576 pregnancy and lactation, 651 Calcitonin gene–related peptide (CGRP), 64, 117,
hepatocyte conjugation, 576–577 Bone remodeling, 643, 646, 648f, 651 512, 555
role and significance, 575 Bone resorption, 644 Calcium, 79, 80, 643
pigment stones, 572 Bony labyrinth, 149 channels, 11, 55, 80, 86, 94, 102, 214, 607
Bilirubin diglucuronide, 576 Borborygmi, 540 blockers, 233, 273
Biliverdin reductase, 575 Botulinum toxin (Botox), 61 L-type, 94
Binding proteins. See Carrier proteins Botulism, 70 factors involved in regulation of, 650t
fatty acid binding protein (hepatic) FABP, 597 Bowman glands, 161 homeostasis, 647
Bioactive substances, excretion, 398 Bowman’s capsule, 400, 402, 440 clinical evaluation of abnormalities
Biogenic amines, 63 hydraulic pressure in, 412 in, 652t
Biological cell membrane oncotic pressure of fluid, 412 hormonal regulation of, 649
associated proteins in, 2f Bowman’s space, 423, 464 interaction of bone, kidney, and intestine
Biphasic action, 53 Boyle’s law, 313, 334, 739 in maintaining, 648–649
Birth, 300 Bradycardia, 221, 232, 241, 242, 289, 299, 739 role of calcitonin in, 650
Birth control pills, 261 Bradykinesia, 171–173 role of vitamin D in, 649–650
2,3-Bisphosphoglycerate (2,3-BPG), 364 Bradykinin, 116, 265, 307 malabsorption, 651
Bisphosphonates, 652 Brain, 105 reabsorption, 644
Bitemporal hemianopia, 144 endorphins in, 120 Calcium ATPase activity, 94
BK potassium channels, 466 Brain areas, concerned with somatic Calcium balance, 485–486
activity, 467f sensation, 119f effector sites
Black (or brown) widow spider venom Brain injury, 80 bone, 487
(BWSV), 61 Brain natriuretic peptide (BNP), 457 GI tract, 486
Bladder, 399 Brain rhythms hormonal control, 487–489
Blebs, 330 in wakefulness and sleep, 186 kidneys, 486
Blind spot, 134 distribution of sleep stages, 187 reabsorption, mechanism, 487f
Blobs, 141 sleep stages, 186–187 regulation, 485
Blood Brain stem pathways transcellular transport, generic method, 486f
INDEX 765

Calcium-binding proteins, 80, 486 ejection fraction (EF), 245 arterial blood, 201
Calcium channel blockers, 222, 273, 293 end-systolic pressure–volume relationship, 245 blood flow, basic physics, 202–203
Calcium channels, 86 Cardiac cycle, 224 cardiac output, 201
Calcium reabsorption, 644 diastolic pressure, 225 distribution of, 201f
Calcium-sensing receptor, 643 end-diastolic volume, 225 effects of ascent to high altitude on, 736
Calmodulin, 79, 80, 99, 102, 193f, 607 heart sounds, 226 during exercise, 745–747
Calsequestrin, 86 atrial gallop rhythm, 226 hemorrhage, 6f, 301–302
Calyces, 399 isovolumetric contraction phase, 225 homeostatic role, 200–202
cAMP response element-binding protein left heart pump, 224 myocardium, 201
(CREB), 607 mitral valve, 225 pulmonary circulation, 201, 341–352
Canalicular bile, 567 peak systolic pressure, 225 systemic circulation, 201, 199–304
composition, 568 phases, 224 Cardiovascular transport
Canaliculi, 509, 512, 561 pressure–volume & length–tension endothelial cells, 254
Canal of Schlemm, 133 relationships, 226–227 Fick principle, 252
Canals of Hering, 568 ventricular afterload, 227 lymphatic system, 255
Cannabinoid receptors (CB1), 75 ventricular preload, 226 transcapillary fluid movement, 254–255
Capacitance, 34 pulse pressure, 225 factors influencing, 255f
Capacitance vessels, 207 QRS complex, 225 transcapillary solute diffusion, 253–254
Capillary filtration coefficient, 349 right heart pump, 225–226 Cardioversion, 244
Capillary hydrostatic pressure, 349 c wave, 226 Carotid bodies, 749
Capsaicin receptor, 45 jugular venous pulse, 226 Carotid chemoreceptors, 386
Ca pumps, 23 v wave, 226 Carotid massage, 232
Carbamoyl phosphate, 579 a wave, 226 Carotid sinus baroreceptors, 287
Carbohydrates ventricular diastole, 224 Carpal tunnel syndrome, 122
assimilation, basic principles ventricular systole, 225 Carrier proteins, 603
barriers to water-soluble macromolecules, Cardiac dipoles β-Casein, 711
584 and electrocardiographic records, 237–238 Cataplexy, 187
luminal digestion, 584–585 Cardiac excitation, abnormal, 240 Catecholamines, 63–64, 231, 346, 665, 724, 731
monosaccharide uptake pathways, 587 Cardiac function physiologic effects, 667t
oligosaccharides/disaccharides, brush sympathetic neural influences on, 231 synthetic pathway, 666f
border digestion, 585–586 techniques for assessment, 235 Catechol-O-methyltransferase (COMT), 63, 665
regulation, 587 Cardiac function curve, 230, 231, 282 C cells, 633
role and significance, 583 Cardiac glycoside, 22, 218 CCK. See Cholecystokinin (CCK)
sources in diet, 584 Cardiac index, 244 CCK-A receptors, 549
digestion and absorption, 583 Cardiac muscle, 79–81, 93–97 CCK-releasing peptide (CCK-RP), 519
Carbon dioxide production, 336, 747 contraction—force–velocity, 95–96 CD4 and CD8 molecules, 25, 536
Carbon dioxide inspired and alveolar partial contraction—length–tension, 94–95 Celiac disease, 538
pressures, 736 excitation–contraction coupling, 93–94 Cell adhesion molecules, 26
Carbon dioxide transport, by blood, increases in strength of contraction in, 96–97 Cell body
368, 371f involuntary, 80 soma, 106
bicarbonate, 370 isometric contractions, 94–95 Cell–cell interactions, 6
carbamino compounds, 369–370 isotonic contractions, 95–96 Cell membrane, 1, 15
dissociation curve, 370–371 Cardiac muscle cells, 212 receptors, 605–607
chloride shift, 372 contractility, 220–221 G protein–coupled receptors, 605f, 607
Haldane effect, 370 lusitropic effect, 221 intracellular receptors, 606f
isohydric shift, 370 excitation–contraction coupling, 217 receptor kinase, 606f
physically dissolved, 368–369 mechanics, 218 receptor-linked kinase receptors, 606f
Carbonic acid, 472 relating to ventricular function, 221 α-Cells, 671
Carbonic anhydrase, 472 relaxation, 217–218 β-Cells, 671, 678
carbonic anhydrase II, 512 Cardiac output (CO), 227, 253, 341, 745 δ-Cells, 671
inhibitor (See Acetazolamide) determinants of, 227–228, 230 Cellular processes, 9
Carbon monoxide, 364 cardiac function curves, 230–231 of hypothetical three-celled organism, 10f
diffusing capacity, 360 sinoatrial (SA) node, 228 messenger to control, 19
for metabolic acidosis, 379t measurement of, 244 phosphorylation of molecules, regulatory
partial pressures of, 358f, 359 cardiac index, 244 roles, 231
poisoning, 383 Fick principle, 244 Cellular respiration
Carboxyhemoglobin (COHb), 368 Cardiac pacemaker, 93, 205, 212, 216, 222 enzyme activity, 753
Carboxypeptidases, 588 Cardiac valve function, abnormal, 245–246 waste product of, 4
carboxypeptidase A, 588 Cardiogenic shock, 301 Central chemoreceptors, 289, 392, 394, 737
carboxypeptidase B, 588 Cardiomyopathy, 222, 282 Central command, 290
Carcinoma, 489, 653, 664 Cardiopulmonary baroreceptors, 289, 297 Central delay, 127
Cardia, 495, 548 Cardiopulmonary resuscitation (CPR), 244 Central nervous system (CNS), 9, 105, 115,
Cardiac action potentials, 54 Cardiovascular adaptations, normal, 298 125, 494
cell-to-cell conduction, 215f cardiovascular changes, with normal aging, 300 modulatory neurotransmitters, 74–75
conduction, 214–216 fetal circulation & changes at birth, 299–300 synapses, 72, 74f
velocity, 215 gender-dependent differences, 300–301 axon hillock, 72
Cardiac afterload, 232 pediatric cardiovascular characteristics, 300 cell body (soma), 72
Cardiac arrhythmias, 52, 240–244, 650, 723 during pregnancy, 299 convergence and divergence, 72, 73f
Cardiac cell membrane potentials, 212–214 respiratory activity, responses to, 298–299 Central sleep apnea, 187, 391
Cardiac contractility, estimations, 245 Cardiovascular adjustments, to hemorrhage, 281f Central swallowing center, 544
echocardiography, 245 Cardiovascular system Central tendon, 315
766 INDEX

Central thermoreceptors, 730 Cholecystectomy, 572 quantification of, 418–419


Central venous compartment, 275 Cholecystitis units, 417–418
Central venous pool, 258 acute/chronic, 572 Cl–HCO3— antiporters, 475
Central venous pressure, 258, 277, 289 Cholecystokinin (CCK), 64, 499, 501, 519, 571, Cl/HCO3— exchanger, 24
abnormal, clinical implications of, 281–282 594, 719 Climbing fibers, 175
and cardiac output and venous return, 279–281 CCK-A receptors, 549 Clonus, 130
influence on venous return, 277–278 CCK releasing peptide (CCK-RP), 519 Closing capacity, 339
Central venous volume, 258 cholecystokinin (CCK)-B receptors, 510 Closing volume, 325, 339
Centrosomes, 3 effects of, 519f Clostridium botulinum, 70
Cephalic, 511, 519 role, factors causing CCK release, 519 Clostridium difficile, 534, 539
Cerebellar ataxia, 183 Cholera, 25, 529, 533 Clotting function, 272
Cerebellar cortex Cholesterol, 16, 566, 570 CNS postsynaptic receptors
basket cells, 174 intestinal handling, 597f gammaaminobutyric acid (GABAAR), 20
Golgi cells, 174 Cholesterol 7a-hydroxylase, 566 for glycine (glyR), 20
granule cells, 174 Cholesterol esterase, 595 serotonin (5HT3R), 20
Purkinje cells, 174 Cholesterol stones, 572 CNS synapse, 74
stellate cells, 174 Cholic acid, 566, 569 α-γ Coactivation, 129
Cerebellar disease, 175 Choline acetyl transferase (CAT), 60 Cobalamin, 507. See Vitamin B12
Cerebellum, 167, 173–174 Cholinergic neurons, 178 CO2-bicarbonate system, 474
cellular organization, 174–175 Cholinergic parasympathetic nerve fibers, 206 buffer system, 472
functional divisions of, 174f Cholinergic parasympathetic postganglionic Cocaine, 64
inferior peduncles, 174 nerves, 323 Cocaine-and amphetamine-regulated transcript
neural connections in, 175f Cholinesterase inhibitors, 71, 91 (CART), 721
neuronal types in cerebellar cortex, 174f Chondrogenesis, 692 Cochlea, 147
superior, middle, 174 Chorda tympani branch of the facial nerve, 162 Cockcroft–Gault formula, 420, 421
vestibulocerebellum, 174f Chorea, 171 Cogwheel rigidity, 173
Cerebral circulation, 232, 736 Chorionic somatomammotropin, 626 Colipase, 517, 595
Cerebral cortex, 107–110, 110f, 169f, 385 Choroid plexus, 272 role of, 595f
Cerebral edema, 30, 736, 738 Chromaffin cells, 665 Colitis, 538
Cerebral hyperperfusion, 736 Chromatin, 3 Collecting duct system, 438, 443–444, 464
Cerebral ischemic response, 289 Chromatolysis, 66, 111 Colloid, 633
Cerebral palsy, 340 Chronic bronchitis, 309, 333, 360 Colloid osmotic (oncotic) pressure, 29, 254,
Cerebral vascular accidents, 299 Chronic cholecystitis, 572 412, 349
Cerebrocerebellum, 174f, 175 Chronic congestive heart failure (CHF), 282 Colon, 496
Cerebrospinal fluid (CSF), 4, 106, 183, 272, 289, Chronic cor pulmonale, 742 chloride secretion, 532f
393f, 736 Chronic hypertension, 292 colonocytes, 496
Cerumen, 154 Chronic obstructive pulmonary disease (COPD), defecation, 496
CFTR. See Cystic fibrosis transmembrane 360, 392 electrogenic sodium absorption, 531
regulator (CFTR) Chronic renal failure, 653 electroneutral NaCl absorption, 532f
Chandelier cells, 109 Chronic thyroiditis, 640 epithelial layers, morphology of, 493f
Channels, 11, 16–18 Chronotropic, 220, 638 functions, 553
cell–cell, 21–22 Chronotropic effect on heart rate, positive and haustrations, 496
chemosensitive, 20–21 negative, 217, 220 ion transport mechanisms, 528t
involved in synaptic release, 65 Chvostek’s sign, 647 Color agnosia, 121
mechanosensitive, 18–19 Chylomicrons, 496, 597 Color blindness, 143
voltage-sensitive, 19 lipid-bearing, 598 Colors
water, 21 secretion of, 597f hue, intensity, and saturation, 142
Charges. See also Anion gap triglyceride-rich, 629 Color vision, 134, 142–143
separation of, 34–35 Chymotrypsin, 588 Commensal, 538
Chemical synapses, 60, 68, 73 Cilia, 134, 159 Commensal bacteria, 492
Chemoreceptors, 115, 159, 289, 311, 380, 390, Ciliary muscle, 136 Common bile duct, 562
392, 473, 737, 746 Circadian rhythms, 185, 187–188, 610, Communication, 12
peripheral, 392, 394 615, 623 Comparator, 12
reflexes, 289 Circular muscle, 494 Compensation, 372, 380, 481
Chemoreceptor trigger zone, 551 Circulatory shock, 301, 301t, 372 acid–base disorders and, 481–482
Chemosensitive channels, 11, 17, 20, 24, 44, 73 Circumventricular organs, 731 short-term cardiovascular, 297
Chenodeoxycholic acid, 566 Cirrhosis, 580, 658 Complementary color, 142
Chest wall, 305 Cisternae, 86, 94 Complex cells, 142
Chest wall compliance, 754 Citrulline, 579 Compliance, 5, 259, 318
Chief cells, 494, 508–513, 588, 643 Clara cells, 307, 308 Compound action potential, 54
Chloride Clasp-knife effect, 130 Conditioned reflex, 748
reabsorption, 439, 731 Class 1 cytokine receptor superfamily, 628 Conducting airways, 305
transport pathways for, 440f, 442f, 443f Classic sensory pathways Conducting zone, 308
secretion, regulation, 533f specific/nonspecific, 110 Conduction, 51, 52, 54, 107, 153, 204, 214–216,
shift, 372 Clathrin-coated pit, 66 236, 241, 242, 730
Chloride–iodide transporting protein, 635. See Clearance, renal Conduit vessels, 207
also Iodide channel derivation of, 418f Congenital adrenal hyperplasia, 664, 668, 692
Chloride reabsorption, 731 measuring GFR, practical method Congestive heart failure, 281–283, 482, 620
Chloride shift, 372 creatinine clearance, 419 Connecting tubule, 406
Cholangiocytes, 562, 568 plasma creatinine and urea concentrations, Connexins, 21, 214
Cholecalciferol, 650 419–420 topology of, 21f
INDEX 767

Connexons, 21 Cutaneous pain, 290 type I, 394, 587, 678–679, 726


Conn’s syndrome, 664 Cu/Zn superoxide dismutase (SOD-1), 176 type II, 292, 406, 679, 718, 753
Constant field equation, 39 Cyanide, 383 Diabetic coma, 678
Continuous positive airway pressure Cyanosis, 368, 383, 742 Diabetic ketoacidosis, 376, 379, 394, 678, 724
(CPAP), 190 Cyberneticists, 10 Diabetic neuropathy, 116
Contraception, 712 Cyclic 3′, 5′-adenosine monophosphate (cAMP), Diacylglycerol (DAG), 25, 101, 605f, 607,
principal methods for, 712t 11, 607, 618 676, 698f
Contractile element (CE), 87 Cyclic guanosine monophosphate (cGMP), Diads, 93
Contractility, 93 11, 692 Diaminopyridine, 70
Convection, 730 Cyclic nucleotide-gated channels, 11, 19, 44, 44f Diaphragm, 305, 315, 386
Convective transport, 252 Cyclic nucleotide–gated (CNG) nonspecific Diarrhea, 379, 533
Convergence, 72 cation channel, 44 Diastole, 55, 56, 94, 95, 204, 225, 227, 247,
Convergence movements, 144 Cystic duct, 562, 562f, 570, 572 259, 276, 300
Coordinated movement, 167 Cystic fibrosis, 24, 524, 584 ventricular, 204, 224
Core body temperature, 729 Cystic fibrosis transmembrane regulator (CFTR), Diastolic failure, 228
Coronary angioplasty, 273 24, 25, 521, 522, 524, 525, 532, 533, 568, 572 Diastolic murmur, 247
Coronary arteries, 269, 270, 272, 273 Cytochrome P450 sidechain cleavage (SCC) Diastolic pressure, 225
disease, 248, 272, 750 enzyme, 656 Diazepam (Valium), 63, 74
stent, 292 Cytokine receptor superfamily, class 1, 628 Diencephalon, 107, 189, 193, 613
Coronary artery bypass surgery, 750 Cytokines, 311, 384, 499t, 500, 536, 538, 731 Dietary fiber, 532, 539, 584
Coronary artery disease, 282, 750 Cytoplasm, 2 Dietary lipids, 565
Coronary artery stent, 292 Cytoplasmic/nuclear inclusions, 183 Dietary polymers, 584
Corpora cavernosa, 685 Cytoplasmic organelles, 3 Dietary potassium, 468, 723, 731
Cor pulmonale, 738 Cytosolic proteolysis, 590 Dietary salt restriction, 293
Corpus, 548 Diffusing capacity, 350, 360, 748, 750, 754
Corpus hemorrhagicum, 703 D Diffusion, 4
Corpus luteum, 696, 703 Dalton’s law, 336, 739 facilitated, 4, 26, 27, 531, 596
Correction, 6 D cells, 509, 510 simple, 4, 26, 27, 424
Cortical collecting tubule, 406 Dead space, 739 Digestion process, 491
Cortical radial arteries, 409 Deafness, 154 gastrointestinal motility, 491
Corticobulbar tract, 168 conductive, 154 Digitalis, 22, 37, 218
Corticohypothalamic pathway, 289 partial, 736 Digoxin, 420
Corticosteroids, 384 sensorineural, 154 Dihydropyridines (DHP), 55
Corticostriate pathway, 171 Decerebrate deficits in humans, 172f receptors, 80, 86, 94, 102f
Corticotropin-releasing hormone (CRH) or Decerebrate rigidity, 171 Dihydrotestosterone, 659, 664, 687, 688f,
factor (CRF), 626, 657, 725 Decerebration, 171, 172 689t, 697
Cortisol, 405, 456, 602, 604, 609, 626, 631, 657, Declarative memory, 75, 191, 196 Dihydroxyphenylalanine (DOPA), 63,
659, 662, 668, 718, 725, 726f Decomposition of movement, 175 64, 666
conversion to cortisone, 658 Decompression illness, 738, 741 1,25-Dihydroxyvitamin D, 398, 531
negative feedback, 664 Decompression sickness, 741 Diiodinated tyrosine (DIT), 636
physiologic effects, 663t Decorticate deficits in humans, 172f Diluting segment, 443
precursors, 665 Decorticate rigidity, 171 Dipalmitoyl phosphatidylcholine, 321
Cortisol-binding globulin (CBG), 658 Decortication, 171, 172f Disordered eating, 750
Cough, 390 Deep cerebellar nuclei, 175 Distal nephron
Coulomb (C), 34 Deep pain, 290 delivery of sodium, 467
Countercurrent exchange, 445 Deep tendon reflex, 126 flow rate, 468
Cranial nerves, 107, 125, 143, 155f, 164f, Deep vein thrombosis (DVT), 262, 360 Distal stump, 111
177, 619f Defecation, 299, 496, 553, 554, 556 Distal tubule, 406
functions of, 109t Defensins, 538 Distensibility, 5
Creatine phosphate (CP), 80 Defibrillation, 244 Diuresis, 430
Creatinine clearance, 419, 420, 754 Deglutition, 544, 545 Diuretics, 57, 293, 442, 443, 468, 469f, 737
Cretinism, 638 Dehydroepiandrosterone (DHEA), 686, 687 effects of, 468–469
Cribriform plate, 159 Deiodinases, 637 heavy use of, 482
Cricopharyngeal muscle, 545 Dementia, 194, 195, 580, 753, 755. See also Aging promoting calcium reabsorption, 487f
Crista ampullaris, 150 Alzheimer disease, 753 Diuretic therapy, 293
Critical micellar concentration (CMC), 567, Demyelinating diseases, 53 Divergence, 72
571, 596 Dendrites, 9, 106 Diverticula, 556, 755
Critical opening pressure, 345 Dendritic cells, 536 Diving
Crohn’s disease, 534, 538, 598 Dendritic spines, 106 breath-hold diving, 739–740
Cross-bridge cycling, 83 Denervation supersensitivity, 111 clinical problems, 741–742
Crossed extensor response, 130 Deoxycholic acid, 566 immersion up effects to neck, 739
Crura, 315 Dephosphorylation, 99 physical principles, 739
Crypt, 493 Depolarize membrane, 38 reflex, 739–740
Cryptorchidism, 689, 692 Depression, 70 electrocardiographic response, 740f
CSF. See Cerebrospinal fluid (CSF) Derecruitment of pulmonary capillaries, 345 underwater breathing apparatus, use, 740–741
Cuneate nuclei, 118 Desensitization, 69–70, 118, 608 DNA-binding transcription factors, 638
Cupula, 150 of acetylcholine receptors, 70f Dominant hemisphere, 195
Curare, 60 Desmopressin, 620 l-DOPA (levodopa), 173, 666
Cushing reflex, 289 Desmosomes, 214 DOPA decarboxylase, 63
Cushing’s disease, 664 Diabetes insipidus, 448, 620 Dopamine, 63, 629
Cushing’s syndrome, 630, 631, 664 Diabetes mellitus, 120, 442 Dopamine agonists, 631
768 INDEX

Dopamine beta-hydroxylase (DBH), 63 Elastase, 588 proliferative phase, 703


Dorsal cochlear nuclei, 153 Elasticity, 318 secretory phase, 703
Dorsal column–medial lemniscus pathway, 115 Elastic recoil, 314, 332, 748 Endopeptidases, 64, 65, 70, 588
Dorsal column system, 118 at functional residual capacity, 754 Endoperoxides, 307
Dorsal respiratory groups (DRG), 386 closing capacities, 754 Endoplasmic reticulum, 2, 15, 60, 66, 597, 603f,
Dorsal root ganglia, 125 Elastic work of breathing, 329, 737 607, 616, 672, 673f
Dorsal vagal complex, 509, 545 Elastin, 207 Endorphin, 64
Driving force, 35 Electrical dipoles, 237 β-Endorphin, 626
on ions crossing through membrane, 36f Electrical synapses, 73f Endothelial cells, 103, 206, 253, 254, 265, 401, 416,
Dry atmospheric air, 337 Electrocardiogram, 216, 224, 235–236, 750 451, 562, 597, 626, 692, 703, 706
D-Tubocurare, 70, 179 basic conventions, 236–237 Endothelin, 346
Dual-energy x-ray absorptiometry (DEXA), 652 Einthoven’s conventions, 237f endothelin-1, 102
Dubin–Johnson syndrome, 576 P, QRS, and T waves, 236 receptors, 102
Duchenne muscular dystrophy, 85 PR segment, 236 Endotherms, 729
Duct of Santorini, 518 QT interval, 236 Endotoxins, 731
Ductus arteriosus, 300 Electrocardiography, 216, 235 Endplate, 10f, 11, 68, 128f
Duodenal cluster unit, 495, 519 Electrochemical equilibrium potential, 36 End plate potential, 86
Duodenal I cells Electrochemical forces, 486 Endplate potential (EPP), 11, 68
cholecystokinin (CCK) release, 520f Electrochemical gradients, 16, 35 recording, 68–69
Duodenum, 495 Electroencephalogram (EEG), 185 End-stage renal disease (ESRD), 406
bicarbonate secretion, 533f Electrolytes, 2, 208, 731 End-systolic pressure–volume relationship, 245
Dutasteride, 687 balance concept, regulation, 397–398 End-tidal CO2, 336
DVT. See Deep vein thrombosis (DVT) exercise, effect on, 749 Energy intake, regulation of, 719
Dwarfism, 631 regulation mediators implicated in regulation, 721t
Dynamic cell processes, 9 of potassium balance, 722–725 Enkephalin, 64
Dynamic compliance, 319 of sodium balance, 722 En passant synapses, 72
Dynamic compression, 324 transport pathways, 527 ENS. See Enteric nervous system (ENS)
Dynamic equilibrium, 604 Electromotive force, 34 Enteric bacteria
Dynamic response, 126 Embolectomy, 361 metabolic effects, 539t
Dyneins, 66 Emboli, 262, 272 populations, 538t
Dynorphin, 64 Emesis, 552 Enteric flora, 538. See Microbiota
Dysdiadochokinesia, 175 Emmetropic eye, 137 Enteric nervous system (ENS), 494, 497, 510, 529,
Dysgeusia, 163 Emphysema, 319, 325, 326, 329, 332, 333, 339, 360 549, 554
Dyslexia, 195 End-capillary oxygen content, 355 enteric nerves, classification, 503t
Dyslipidemia, 718, 719 End-diastolic volume (EDV), 245 enteric neurotransmitters, 503
Dysmetria, 175 Endocardial (inner) surface, 55 little brain model, 502–503
Dyspareunia, 711 Endocrine physiology, 601–728 neurokinin A, 554
Dysphagia, 176, 556 endocrine function, assessment of pelvic nerves, 554
Dyspnea, 247, 283, 316, 329, 339, 350, 383, hormone measurements, interpretation, plexuses of, 497f
390, 470 610–611 pudendal nerves, 554
Dystonia, 61 hormone cellular effects, 605 schematic diagram, 502f
Dystrophin, 85 hormone chemistry, and mechanisms of action Enterochromaffin-like (ECL) cells, 503, 509, 529
amino acid–derived hormones, 603 Enteroendocrine cells, 464, 494, 502, 503, 509, 530
E hormone effects, 603 Enteroglucagon, 502
Ear hormone transport, 603–604, 604f Enterohepatic circulation, 561
anatomy, 147 protein/peptide hormones, 602 Enterokinase, 588, 589f
cochlea, 149–150, 150f steroid hormones, 602 Eosinophil chemotactic factors, 307
external ear, 147 hormone receptors, and signal transduction Epicardial (outer) surface, 55
middle ear, 147, 148f cell membrane receptors, 605–607 Epididymis, 684, 688, 689
inner ear, 149–150 hormone–receptor regulation, 608 Epilepsy, 52, 63, 185
cochlea, 147 intracellular receptors, 607–608 Epimerization, 566
saccule, 147 hormone release control Epinephrine (EPI), 56, 63, 268, 346, 405, 464, 665,
semicircular canals, 147 hormonal control, 608–609 666f, 676, 717f, 725, 731f
utricle, 147 neural control, 608 Epithelial cells, 619
vestibular system, 154 nutrient/ion regulation, 609–610 polarization, 425
Ear dust, 150 principles, 601 Epithelial salt and water, reabsorption, 425
Ebner gland, 162 Endocrine system, 601–602, 602f Epithelial sodium channel (ENaC), 21, 44,
ECF. See Extracellular fluid (ECF) disorders, 610 443, 531
Echocardiogram, 282 endocrine glands, 601 EPSPs. See Excitatory postsynaptic potentials
Echocardiography, 245 hormones, 601–602 (EPSPs)
Ectopeptidases, 588 physiologic functions and components, Equal pressure point, 325
Edema, 29, 207, 255, 261, 297, 736, 738. See also 601–602 Erectile dysfunction, 692
Pulmonary edema target organ, 602 Ergocalciferol, 650
Edinger–Westphal nucleus, 143 Endocytosis, 30 Error, 6
Edrophonium chloride (Tensilon), 71, 77 Endogenous opioid peptide, 626 Eructation, 741
Effective renal plasma flow, 419 Endogenous pyrogens, 731 Erythropoiesis, 737
Effector, 12 Endolymph, 149 Erythropoietin, 398, 406, 450, 602, 737
Efferent arterioles, 400, 402, 409, 411f, 722, 753 Endometrial cycle. See also Ovarian hormones Esophageal varices, 563
Efferent control, 45, 108 fertilization, 703–704 Esophageal motility, features of
Einthoven’s triangle, 236, 238, 240 implantation, 704–705 LES relaxation, 547
Elastance, 5 menstrual phase, 703 peristalsis, 545–547
INDEX 769

swallowing, 545 External anal sphincter, 494, 554 infection pathway, 732f
Esophageal motility, principles External auditory meatus, 147 Fiber, 85, 584
role and significance, 543–544 External intercostal muscles, 315 dietary, 539
Esophagus, 493, 543 Extra-alveolar vessels, 343 types, 90–91
functional anatomy and innervation, 544f Extracellular fluid (ECF), 4, 437 Fibrillation, 272
lower esophageal sphincter (LES), 544 total body water, distribution, 438f Fibrosis, 319, 322, 326, 580
upper esophageal sphincter, 544 volume, 450, 453–454 Fibrous astrocytes, 105
Estradiol, 611, 686, 687f, 693, 697–699, 705, 707f, Extrafusal fibers, 126 Fick principle, 244, 252
711, 712f, 726f Eye Fick’s first law, 26
17β−Estradiol, 659 accommodation, 137–138, 138f of diffusion, 253, 357
Estrogen, 261, 755 anatomy, 133–134, 134f Fight–or-flight response, 667
deficiency, 652 aqueous humor, 133 Filtered load, 414–415, 430
estrogen receptor–mediated (genomic) effects, canal of Schlemm, 133 Filtration, 253
705–706 choroid, 133 Filtration fraction, 414
metabolic fate of, 705f ciliary body, 133 Filtration of inspired air, 308
nongenomic effects, 706 cornea, 133 Final common pathway, 131, 167
physiologic actions, 706–707 crystalline lens, 133 Finasteride, 687
replacement therapy, 652 eyeball Fixed/nonvolatile acids, 376
synthesis, 755 sclera, 133 Fixed obstructions, 326
transport, and metabolism, 705 image-forming mechanism, 135–136 Flaccid, 129
theca and granulosa cells coordination common defects, 136–137 Flaccid paralysis, 168
forproduction of, 698f iris, 133 Flippase, 16
Estrogen receptor beta (β), 706 lens suspensary ligament (zonule), 133 Flow, 5
Ethmoid bone, 165 movements, 143–144 Flow-induced distribution
Eubacteria, 538 convergence, 144 of blood volume and pressure, 277
Eukaryotic cell, 1, 2f extraocular muscles, 143f Flow–volume curves, 326–327, 329.
Eupnea, 315 saccades, 143 See also Respiratory system
Eustachian tube, 147 smooth pursuit, 144 effort-dependent, 326
Euthyroid, 638 vestibular, 144 effort-independent, 326
Evaporation, 439, 730, 731 pupil, 133 inspiratory and expiratory, 328f
Excitability, 6, 9, 33, 52, 68, 131, 212, 243, retina, 133 maximal expiratory, 328f
463, 485, 647 vitreous humor, 133 of varying intensities, 327f
Excitation–contraction coupling, 86, 93–94 Fluid balance model, 290
Excitatory amino acid transporter (EAAT), 62 F Fluoxetine hydrochloride (Prozac), 24, 64
Excitatory postsynaptic potentials (EPSPs), Facial nerves, 162, 163f, 178, 648, 653 Follicle-stimulating hormone (FSH), 64, 614f,
11, 20, 67, 86, 125 Facilitated diffusion, 4, 26, 27 615, 623, 625f, 683, 685, 691, 697, 698f, 700f,
Excitatory spinal pathways, 287 Facilitation, 70 711, 713
Exercise, 265, 745–752 Familial dysautonomia, 162 Follicular cell, 633, 733
capacity, 753 Familial Hyper KPP, 57 Foot plate, 147
cardiovascular system, control, 746f Fanconi syndrome, 427 Foot processes, 411
changes in Farad (F), 34 Foramen ovale, 299
cardiac output, 746f Faraday’s constant, 34, 36, 39 Force, 86
heart rate, 746f Fasciculations, 168, 176 Forced expiratory volume, 325
stroke volume, 746f Fat-soluble vitamins, 565, 593, 594, 596, 597 Forced vital capacity (FVC), 325
ventilation, 747f absorption of, 598 maneuver using rolling seal spirometer,
effect on minute ventilation, 749f Fatty acid, 597 326f
muscle metabolism, 745 oxidation, 93, 745 Fovea centralis, 134
neuroendocrine response to, 717f Feature detectors, 142 Fractionate, 130
physical activity, acute effects Feedback control mechanism, 604. See also Frank–Starling mechanism, 746
cardiovascular system, 745–747 Hormones, regulation Frontal operculum, 162
fluid and electrolyte balance, 749 negative feedback, 6, 608, 610 Fructose, 586
respiratory system, 747–749 positive feedback, 12, 610 Fruit juice paradox, 473
stress tests, 750 Feedback control systems, 6f, 730 FSH. See Follicle-stimulating
training effects, 749–750 feedback device, 128 hormone (FSH)
ventilatory response to, 748 feed-forward inhibition, 175 F-type H pump, 23
Exercise capacity, 753 Female athlete triad, 750 Functional magnetic resonance imaging
Exercise stress tests, 750 Female genital tract, 697 (fMRI), 191
Exocrine pancreas, 517 Female reproductive organs, 696 Functional residual capacity (FRC), 321–322,
amylolytic enzymes, 517 age-related changes, 711 332, 739
colipase/trypsin inhibitors, 517 menopause, 711–712 Fundus, 508, 548
lipases, 517 puberty, 711
nucleases, 517 functional anatomy, 696f G
proteases, 517 Feminization, 580 GABAA receptors, 63
Exocytosis, 30 Fenestrae, 411, 562 Gain, 6
Exogenous pyrogens, 731 Fermentation, 539 Galactorrhea, 630, 631
Exophthalmos, 639, 733 Fertility, 683 Gallbladder, 562, 570
Expiratory neurons, 386 Fertilization, 695 function, principles
Expiratory reserve volume (ERV), 332, 333f, and embryo migration, 704f role and significance, 570
334f, 739 Fetal development, 299 functional anatomy
Expired air, 337 Fetal hemoglobin (HbF), 364 epithelium, 570
Explicit memory, 75 Fever, 731–732 musculature, 570–571
770 INDEX

Gallbladder (continued) Gastrin, 64, 499, 501, 508, 547 epithelium, 493
motor functions biologically active forms, 501 esophagus, 493
contraction, 571 Gastrin-releasing peptide (GRP), 504, 510, 519 liquids and absorption, 4
sphincter of Oddi function, 571–572 Gastrocolic reflex, 549 lumen, 492
neurohumoral control, 570f Gastroesophageal reflux disease (GERD), 309, neurohumoral regulation, features, 498
storage of bile 495, 556, 755 paracrine and immune mediators, 503t
bile concentration, mechanism, 571 Gastrointestinal hormones, 499 stratified squamous epithelium, 493
effects on composition, 571 candidate, 502 Gate-control hypothesis, 120
Gallstone disease, 571, 572 enteroglucagon, 502 Gating current, 49
Gamma-aminobutyric acid (GABA), 63, 74 glucagon like peptide-1, 502 G cells, 509
GABAA receptors, 20, 63, 74 ileal brake, 502 Gender-dependent differences in the
GABAB receptors, 63, 74 pancreatic polypeptide, 502 cardiovascular system, 300
Gamma-glutamyl transpeptidase (GGT), 568 peptide YY (tyrosine-tyrosine), 502 General anesthetics, 74
Gamma loop, 171 factors influencing release, 501t chloroform, 74
Ganglionic synapse, 71 sites of production, 500f ether, 74
Gap junctions, 6, 21, 93, 101, 134, 214 Gastrointestinal immune system, 492 halothane, 74
Gases, diffusion of, 357 Gastrointestinal system, 492f, 491–600 Generalized seizures, 185
diffusing capacity, measurement of, 360 anatomy of, 494f Generator potential, 125
diffusion limitation, 358–359 communication, specific modes Geniculocalcarine tract, 141
diffusion of carbon dioxide, 359–360 endocrine communication, 498–499 Genomic effects, 637–638
diffusion of oxygen, 359 immune communication, 500 GFR. See Glomerular filtration rate (GFR)
Fick’s law, 357–358 neurocrine regulation, 499–500 GH. See Growth hormone (GH)
perfusion limitation, 359 paracrine communication, 500 GH–IGF-I axis, 725
Gαs protein–coupled receptor, 676 endocrine regulation, principles GH insensitivity syndrome, 631
Gαs signaling pathway, 25 candidate GI hormones, 502 GH receptor antagonists, 631
Gastric acid gastrin/CCK family, 501 Ghrelin, 501, 627, 722
secretion, regulation, 511f GI hormones, 500 Gibbs–Donnan equilibrium, 29
cephalic phase, 511 motilin, 501 Gigantism, 630
gastric phase, 512 engineering considerations GIP (glucose-dependent insulinotropic peptide),
intestinal phase, 512 cellular specialization, 492–494 499–502, 512
Gastric glands, 495, 508 hollow organs, design, 492 Glands, 508, 523
chief cells, 588 intestine division into functional Glial cells, 105
pepsinogens, 588 segments, 494 microglia/macroglia, 105
pepsins, 588 functions principal types, in nervous system, 106f
structure, 508f digestion and absorption, 491–492 Glia-like supporting (sustentacular) cells, 159
Gastric inhibitory peptide (GIP), 499 excretion, 492 Glitazones, 680
Gastric motility, features of host defense, 492 Globulins, 208, 404, 603
basal electrical rhythm, 549 physiologic neurohumoral regulators, 499t Glomerular capillary hydraulic pressure, 412
during fasting, 551 regulation, 498 Glomerular capillary plasma
gastric emptying, 550–551 gastrointestinal tract pressure in, 413
mixing and grinding, 549–550 chemical signals, characteristics, 498 Glomerular filtration
pylorus, role, 550–551 crypt and villus structures, 493 fenestrae, 411
receptive relaxation, 549 epithelium, 493 filtered load, 414–415
vomiting, 551–552 esophagus, 493 forces involved, 413f
Gastric motility, principles lumen, 492 formation, 411–412
patterns, 550 neurohumoral regulation, features, 498 GFR, direct determinants, 412–414
role and significance, 547–548 paracrine and immune mediators, 503t summary of, 414t
Gastric musculature, functional anatomy stratified squamous epithelium, 493 Glomerular filtration rate (GFR), 404, 412, 414,
innervation, 548–549 immune system, 492 415, 418–420, 430, 449, 453, 456, 457, 462,
muscle layers, 548 neural control, 498f 486, 754
Gastric pacemaker, 548f, 549f, 550 neurocrine regulation, principles autoregulation, 415f
Gastric peristalsis, 594 enteric nervous system, little brain model, filtration coefficient, 412
Gastric phase, 512 502–503 oncotic/colloid osmotic pressures, 412
Gastric pits, 508 enteric neurotransmitters, 503 resistance, changes effect, 415f
Gastric secretion organs and systems Starling forces, 412
anatomical considerations colon, 496 Glomeruli, 159, 161, 400, 410, 412, 413, 416,
gastric cell types, 508–509 duodenal cluster unit, 495–496 456, 754
innervation, 509 neuromuscular system, 497 Glomerulonephritis, 416
stomach, functional regions, 508 oral cavity and esophagus, 495 Glomerulus, 159, 161, 400, 401f, 403, 405, 409,
basic principles small intestine, 496 426, 453f, 579
gastric secretory products, 507–508 splanchnic circulation and lymphatics, Glossopharyngeal nerves, 162, 163f, 178, 386,
role and significance, 507 496–497 390, 545
cellular basis of stomach, 495 Glucagon, 676, 715
acid secretion, 512–513 paracrine and immune regulation effects on hepatic glucose metabolism,
other products, 513–514 important mediators, 503–504 676f, 677
neural regulation, 510f mechanisms of activation, 504 physiologic effects, 676
products, 508t regulatory systems, integration, 504 receptor-mediated cellular effects, 677f
regulation, 509 Gastrointestinal (GI) tract, 4, 5, 491–600 regulation of release, 675–676
in interdigestive phase, 510–511 absorption, 5 synthesis, 675
postprandial secretion, 511–512 chemical signals, characteristics, 498 Glucagon-like peptide 1 (GLP-1), 502, 673,
regulatory strata, 510 crypt and villus structures, 493 675, 676, 680
INDEX 771

Glucagonomas, 678 G protein-coupled receptors (GPCRs), 18, 44, 62f, pumping action, 203–204
Glucoamylase, 585f 64, 71, 74, 75, 163, 521, 605f, 607, 618, 677f, Purkinje fibers, 216
Glucocorticoids, 608, 626, 651, 732 685, 697 rate, 232
diseases, 664 adrenergic receptors, 665 control of, 216–217
metabolism, 658 G proteins, 25, 44, 45, 71, 161, 164f, 533, 605, 607, sounds, 226
synthesis and release, 657–658 618, 625, 645, 665, 685 ventricular gallop rhythm, 226
Glucocorticoid therapy, 732 Gq/11 protein–coupled oxytocin receptors, 617 Heart failure, 228, 620, 664
Gluconeogenesis, 398, 405, 559, 676f, 716, 717f, Gracilis nucleus, 118 Heart rate, 232
718, 726 Gradient-limited systems, 426, 427 Heat stroke, 730
Glucose, 430 Granular cells, 403, 451–454 Helicobacter pylori, 514
glucose-6-phosphate, 745 Granule cells, 159, 174 Helicotrema, 149
liver synthesis of, 5 Graves’ disease, 634, 638–640, 733 Helium-dilution technique, 333
oligomers, 585–586 Gray rami communicans, 178 Hematocrit, 207, 372, 737
polymers, types, 584 Growth hormone (GH), 615, 623, 624, Hematuria, 416
tolerance test, 631 709, 755 Heme-containing proteins, 575
Glucose buffer function, 559 deficiency, 755 Heme oxygenase, 575
Glucose-dependent insulinotropic effects at target organs, 629 Hemianopia, 142
peptide, 499 insensitivity syndrome, 631 Hemiblocks, 242
Glucose tolerance test, 631 physiologic effects of, 628 Hemichannels, 21
Glucose transporter (GLUT), 23, 430, 675t receptor, 628–629 Hemochromatosis, 531
GLUT1, 27 receptor antagonists, 631 Hemoglobin, 363, 377, 472
GLUT2, 531, 587 regulation of, 627 chemical reaction of oxygen and, 364
GLUT3, 27 release and effects, 628f hemoglobin M, 368
GLUT4, 27, 662 stimulation of, 628t hemoglobin S, 364
GLUT5, 586, 587 Growth hormone-releasing hormone structure, 363–364
Na-GLUT, 27 (GHRH), 627 Hemophilia, 209
Glutamate, 61–63, 74, 106, 159 Gs protein–cAMP–protein kinase A, 220 Hemorrhage, 6, 171, 230, 280, 336, 348, 454
synapse, 62f Guanosine diphosphate (GDP), 25, 605, 607, Hemorrhagic shock, 618
Glutamate decarboxylase (GAD), 63 677f, 685f Hemorrhagic hypovolemic shock, 301
Glutamate receptors (gluRs), 20 Guanosine triphosphate (GTP), 25, 139, 605f, Hemostasis, 208–209, 563
metabotropic gluRs (mgluRs), 61 607, 677f, 685f activity of vitamin K, 209
NMDA gluRs, 61 Guanylate cyclase, 692 anticoagulants, 209
NMDA-type, 61, 74 Guanylin, 529, 530, 533, 534 blood clotting, 209
non-NMDA-type, 61, 74 Gustation, 161 calcium chelators, 209
umami taste, 44 Gustducin, 163 citrate, 209
Glutamine, 62, 405, 434, 478, 479f, 480 EDTA, 209
Glutarate, 579 H endogenous tissue plasminogen activator
Glycerophospholipids, 15, 16f Hair cells, 147, 150. See also Auditory receptors (tPA), 209
Glycine, 74 inner hair cells, 149 formation of thrombin, 209
Glycine receptors, 20, 63 outer hair cells, 149 heparin, 209
Glycogen phosphorylase, 675 role of tip link, 151f intrinsic pathway, 209
Glycogen synthase, 675 structure, 151f local vasoconstriction, 209
Glycolipids, 1 supporting (sustentacular) cells, 150 oxalate, 209
Glycoproteins, 1, 624 tectorial membrane, 149 platelet aggregation, 208–209
gonadotropins, 626 Haldane effect, 370, 749 thrombolytic agents, 209
thyroid-stimulating hormone, Halothane, 350 Henderson–Hasselbalch equation, 377, 380, 472
625–626 Haptocorrin, 291 Henle’s loop, 400, 402, 405, 406, 434, 435, 438,
Glycosphingolipids, 16 Hashimoto’s thyroiditis, 638 440, 442–444, 458, 464, 465, 468, 469, 475,
Goblet cells, 307–309, 494 H-ATPase, 475, 476 476, 486
Goiter, 639–640 Haustra, 496f, 553, 555 aquaporins, 442
Goldman–Hodgkin–Katz (GHK) Haustrations, 496 diluting segment, 443
equation, 39 hCG. See Human chorionic gonadotropin (hCG) loop diuretics, 442
Golgi apparatus, 2, 16, 17, 597, 603f Hearing bumetanide, 442
Golgi cells, 175 action potentials in, 153 furosemide (Lasix), 442
Golgi tendon organs, 117, 129 bone and air conduction, 152–153 Na–K–2Cl symporter, 442
Gonadal dysgenesis, 692 central pathway, 153 Henry’s law, 357, 739
Gonadal function, 686–687 loss (see Deafness) Heparin, 262, 307
Gonadotropin-releasing hormone (GnRH), 626, sound transmission, 152 Hepatic artery, 497, 560, 561f, 562
685, 697, 700, 750 sound waves, 152 Hepatic bile, 569
Gonadotropins, 623, 626, 685, 755 traveling waves, 153 Hepatic ducts, 562, 562f, 569
receptor-mediated effects of, 685f Heart, 203 Hepatic encephalopathy, 130, 580
regulation, of ovarian function, 697 autonomic neural influences, 205–206 Hepatic endothelial cells, 598
release, ovarian regulation of, 699 block, 242 fenestrae, 598
follicular phase, 699 conduction velocity, 215 Hepatic stellate cells, 562, 563, 580
luteal phase, 699 control of cardiac output, 205–206 Hepatic triad, 560
synthesis and release diastolic filling, 205 Hepatitis, 580
control of, 685–686 effective operation, requirements for, 205 Hepatitis C virus, 563
negative feedback regulation, 686f electrical activity of, 215f Hepatocytes, 496, 561, 565
Gout, 432 electrocardiograms, 216 transporters, 568t
G protein–activated inward rectifier GIRK excitation, 204–205 Hering–Breuer deflation reflex, 388
channel, 56 failure, 664 Hering–Breuer inflation reflex, 388
772 INDEX

Herpes simplex, 66 salt, 163 Hypertrophy, 292


Heteronymous hemianopia, 142, 144 sour, 163 Hyperventilation, 361, 378, 735, 740
Heterotrimeric guanine-binding (G) proteins, sweet, 163 syndrome, 378
161, 607 umami, 163 Hyperventilation syndrome, 378
Hexamethonium, 71, 179 Humoral hypercalcemia of malignancy, 489 Hypesthesia, 161
H/glutamate antiporter, 24, 27 Huntington’s disease, 63 Hypoaldosteronism, 664
High altitude pulmonary edema, 736 Hydraulic pressure in Bowman’s Hypocalcemia, 52, 57, 68, 484, 487, 644,
High-amplitude propagating contractions, 555 capsule, 412 647, 653
High-pressure nervous syndrome (HPNS), Hydrogen ions Hypocalcemic tetany, 653
741, 742 excretion of, 477f, 478 Hypocapnia, 372, 384, 736, 737
Hippocampus, 76, 108f, 191, 192f, 193–196 secretion generic model, 474f Hypogeusia, 162, 165
Histamine, 63, 64, 116, 188, 255, 307, 323, 346, tubular segments, contributions, 475t Hypoglycemia, 626, 627, 676, 717, 718, 726
510, 529 Hydrophobic groups, 16 Hypogonadism, 580, 630, 692, 693
H2 receptors, 510 Hydrostatic forces, 5 Hypogonadotropic hypogonadism, 692, 693, 713
antagonists, 510 Hydrostatic pressure, 5, 28, 254, 258, 348, 349, Hypokalemia, 463, 620, 664, 724, 725, 731
Histotoxic hypoxia, 383 412, 413 Hypomagnesemia, 644
H–K antiporters, 465 of interstitial fluid, 254 Hyponatremia, 30, 620, 665, 749
H+,K+-ATPase, 475, 512. See also Proton pump of intracapillary fluid, 254 Hyponatremic encephalopathy, 30
H/K pump, 23 Hydroxyapatite, 377, 487, 646 Hypoparathyroidism, 652t, 653
Holter monitor, 210 deposition of, 488 Hypoperfusion hypoxia, 383
Homeostasis, 1, 11 resorption of, 488 Hypophosphatemia, 651
energy, brain’s signals to control 11β-Hydroxylase, 657 Hypophysiotropic hormones, 613, 615
regulation, 720f enzymatic deficiency, 659f key aspects of, 615t
and feedback control, 12 21-Hydroxylase, 657 Hypophysis, 623
Homeotherms, 729 enzymatic deficiency, 659f Hypopituitarism, 630, 639
Homovanillic acid, 665 11β-Hydroxysteroid dehydrogenase, 658 Hyporeflexia, 168, 176
Homunculus, 118 type I, 658 Hyporeninemic hypoaldosteronism, 664
Hooke’s law, 5 type II, 658 Hyposmia, 161
Hormone-producing tumors, 678 5-Hydroxytryptamine (5-HT), (also see Hypothalamic integration, 719
Hormone–receptor agonists/antagonists, 605 serotonin) 64, 503, 529, 550 Hypothalamic neurons, 613
Hormone–receptor complex, 604, 605, 607, 608, Hyperaldosteronism Hypothalamic neuropeptides, 615
628, 661 primary, 470, 664 Hypothalamic nuclei, 613–615
Hormones, 498. See also Endocrine physiology; Hyperalgesia, 116 Hypothalamic peptides, 625
Endocrine system; Gastrointestinal Hyperbaria, 738 Hypothalamic–pituitary–adrenal axis, 660f
hormones; Ovarian hormones; Steroid Hyperbaric chambers, 738 Hypothalamic–pituitary–ovarian axis, 695, 700f
hormones Hyperbaric oxygen therapy (HBOT), 738 Hypothalamic–pituitary–thyroid axis, 634, 634f
affinity, 605 Hyperbilirubinemia, 577, 578 Hypothalamic tumors, 692
of anterior pituitary, 624–630 Hypercalcemia, 653 Hypothalamohypophysial tract, 613, 615
autocrine, 603 Hypercalcemia of malignancy, 645 Hypothalamo-pituitary hormone-mediated
biologic effect, 603 Hypercalciuria, 653 effects
endocrine, 603 Hypercapnia, 346 cellular signaling pathways, 625f
events, during ovarian and endometrial cycles, Hypercoagulable, 262 Hypothalamus, 161f, 183, 188, 189f, 287, 385,
701f Hyperemia, 265 450f, 501, 509, 602, 613, 614f, 626, 630
free/unbound hormone, 603 Hyperglycemia, 406. See also Diabetes mellitus anatomic and functional relationship, 614f
half-life, 603 Hypergonadotropic hypergonadism, 713 endocrine functions of, 615
interpretation of measurements, Hyperkalemia,38, 222, 463, 658, 665, 725 supraoptic/paraventricular nuclei, 459
610–611, 611t Hyperkalemic periodic paralysis (HyperKPP), 52 Hypothermia, 716, 731
intracrine, 603 Hypernatremia, 620 Hypothyroidism, 638
neural control, 610f Hyperopia, 136 primary hypothyroidism, 638–639
paracrine, 603 Hyperosmolarity, 722 secondary hypothyroidism, 639
precursor, 604 Hyperparathyroidism Hypotonia, 168, 175, 176
product, 625 primary, 488, 489 (see also Thyroid gland) Hypotonic encephalopathy, 30
receptor function, 611 secondary, 489, 651 Hypotonic muscle, 129
release Hyperphosphatemia, 489, 653 Hypotonic solutions, 28
patterns, 609f Hyperpnea, 388, 747 Hypoventilation, 366, 377, 382, 480
regulation of, 615–616 Hyperpolarization, 38 Hypovolemia, 484
sensitivity, 611f Hyperpolarization-activated current, 55 Hypovolemic shock, 301
specificity, 605 Hyperprolactinemia, 640, 693 Hypoxemia, 321, 379, 384
Hormone-sensitive lipase (HSL), 638, 675 Hyperreninemic hypoaldosteronism, 664 Hypoxia, 321, 346, 382, 391, 394, 730, 735
hPL. See Human placental lactogen (hPL) Hyperresonant, 329 altitude and acclimatization
H2 receptor antagonists, 510 Hypersensitivity, 111 altitude, acute effects, 736
5HT1 receptors, 551 Hypersomnolence, 187 cardiovascular system, 736
5HT3 receptors, 64, 551 Hypertension, 229, 282, 290, 292, 470, 640, 657, respiratory system, 736–738
Human body, 3f 664, 718 anemic hypoxia, 383
Human chorionic gonadotropin (hCG), 602, 625, Hyperthyroidism, 634, 638, 731, 732 classification of causes, 382t
688, 699, 703, 709 Graves’ disease, 639 effects of, 383
Human language, 195 primary, 732 histotoxic hypoxia, 383
Human placental lactogen (hPL), 626, 707f, TSH-secreting adenomas, 639 hypoperfusion hypoxia, 383
709, 711 Hypertonia, 169 hypoxic hypoxia, 382–383, 735
Human taste modalities Hypertonic (spastic) muscle, 129 Hypoxic hypoxia, 382, 735
bitter, 163 Hypertonic solutions, 28 diffusion impairment, 382–383
INDEX 773

low alveolar PO2, 382 type B, 476, 476f Intrapleural pressure, 309, 314, 739
shunts, 383 Intercalated disks, 214 Intrapulmonary shunts, 355, 366
Hypoxic pulmonary vasoconstriction, 384, 736 Intercalated ducts, 523 Intrarenal chemical messengers, 405
chronic, 742 Intercellular adhesion molecules (ICAMs), 26 Intrinsic factor, 507, 513
Hysteresis, 319, 320 Intercostal muscles, 305 Intrinsic factor-cobalamin receptor (IFCR), 591
I Interdependence of alveolar units, 315 Intrinsic proteins, 16
IgA system, 537. See also Mucosal immune system Interferon therapy, 563 Intubated patient, 309
physiological functions, 537 Interleukins, 629, 646, 699, 700f, 719, 726f, Inulin, 418
secretion of IgA across, 535, 537f, 568, 569 731, 732f renal handling, 419f
structural aspects of IgA, 536–537 Intermediolateral column (IML), 177 Invasion of immune cell, 112
IGF receptor, 629 Internal anal sphincter, 554 Inverse stretch reflex, 129
Ileal brake, 502 Interstitial/alveolar edema, 382 Inward-going pacemaker current, 216
Ileal-fatty acid binding protein (I-FABP), 597 Interstitial cells of Cajal, 549 Inward rectifier, 17, 18
Ileocecal valve, 494, 554 Interstitial fibrosis, 382 Inward rectifier K channel (Kir), 17, 18f, 55
Ileum, 496 Interstitial fluid, 4, 28 Iodide channel, 635
Immune communication, 500 Interstitial hydrostatic pressure, 349 Iodine, uptake and organification, 634
Immune responses, alterations in, 725 Interstitium, 399, 477f, 479f, 487, 499, 603f, Ion movements
Impaired glucose tolerance, 631 663f, 664 factors controlling, 35–36
Implicit memory, 76 Intestinal calcium absorption, 644 receptors channels allowing, 102
Implicit/nondeclarative memory, 191 Intestinal flatus, composition, 540f Ionotropic ligand receptors, 20
Impotence, 631 Intestinal fluid transport Ions of importance, across muscle cell
Inactivation gate, 213 anatomical considerations membrane, 35t
Incontinence, 183, 754t, 755 innervation and regulatory cells, Ion transport pathways, 522f, 525f
Incretin, 501, 675 528–529 IP3 receptors, 101, 102f, 677f, 698
Infant respiratory distress syndrome, 321 intestinal surface area, amplification of, 528 IPSPs. See Inhibitory postsynaptic potentials
Inferior colliculi, 153 cellular basis (IPSPs)
Inferior olivary nuclei, 175 absorptive mechanisms, 531–532 Ischemia, 22, 63, 117, 248, 289, 302, 360, 372,
Inflammation, 112 secretory mechanisms, 532–533 630, 703
Inflammatory bowel diseases, 534, 538 endogenous regulators, 529t Ischemic heart disease, 711
Crohn’s disease, 534, 538 integration of influences, 530f Ischemic stroke, 63
ulcerative colitis, 534, 538 principles Islets of Langerhans, 518
Inhibin B, 685 electrolytes involved, 527–528 Isohydric principle, 376
Inhibitory G proteins, 216 role and significance, 527 Isohydric shift, 370
Inhibitory postsynaptic potentials (IPSPs), water and electrolyte transport, regulation Isomaltase, 585, 586
11, 67, 125 acute regulation, 530 Isometric contraction, 85, 86
Inhibitory spinal pathways, 287 chronic adaptation, 531 Iso-osmotic process, 423
Innocent murmurs, 300 regulatory strata, 529–530 Isopotential, 40
Inositol triphosphate (IP3) receptor (IP3R), 21 Intestinal lipolysis, mediators, 595t Isoproterenol, 346, 350
Inositol trisphosphate (IP3), 607 Intestinal microecology Isosmotic solution, 28
Inotropic, 220, 638 gas generation in intestine, 540 Isotonic contraction, 85, 86
Input–process–output structural framework, 10 intestinal microbiota, development, 538–539 Isotonic solution, 28
Inspiratory capacity (IC), 332 microbiota, physiological functions, 539–540 Ito cells, 562
Inspiratory neurons, 386 Intestinal motility
Inspiratory reserve volume (IRV), 332 basic principles J
Insulin, 394, 464, 587, 671, 715, 724 role and significance in colon, 553 Jaundice, 575
effects at target organs, 674 role and significance in small intestine, differential diagnosis, 578f
early effects, 674 552–553 J chain, 536
intermediate effects, 675 esophageal musculature, functional anatomy Jejunum, 496, 499, 500, 552, 554
long-term effects, 675 innervation, 544 J receptors, 390
effects on carbohydrate, fat, and protein muscle layers, 544 Juxtaglomerular (JG) apparatus, 403, 451
metabolism, 674 features of baroreceptor, 658
effects on hepatic glucose metabolism, 676f colonic motility, 555 components, 403f
glucose-induced stimulation of, 673 defecation, 556 extraglomerular mesangial cells, 403
physiologic effects of, 673 fed versus fasted patterns, 554–555 glomerular filtration rate (GFR), 403
receptor, 25, 674 mixing and segmentation, 555 granular cells, 403
substrates, 674 peristalsis, 555 juxtamedullary nephron, 403
regulation of release, 672, 673f functional anatomy macula densa cells, 403
resistance, 662, 678–680 enteric nervous system, 554 renin, 403
sensitivity, 755 muscle layers, 553 Juxtaglomerular cells, 401f, 403, 451
synthesis, 672 sphincters, 554
Insulin/glucagon ratios, 716 Intestinal pathogens, pathophysiological K
Insulin-like growth factor-1 (IGF-1), 627–629, 755 mechanisms, 540t KAch channels, 216
Insulin-like growth factor binding proteins Intestinal phase, 519 Kainate channels, 74
(IGFBPs), 629 Intestine organization, 493f Kallmann syndrome, 692
Insulinoma, 678 Intracellular fluid (ICF), 437 K channels, 18, 37, 55, 74, 102, 214, 673f
Insulin resistance, 662 Intracellular receptors, 606f Kernicterus, 575
Integrins, 26 Intraepithelial, 536 Ketoacidosis, 376, 379, 382, 394
Intensity, 118 Intraepithelial lymphocytes, 536 Ketogenesis, 674–676, 680
Intention tremor, 175 Intrafusal fibers, 126 enzymes involved in, 680t
Intercalated cells, 403, 466 Intralaminar nuclei, 108 in insulin deficiency, 679f
type A, 475, 476f Intralobular ducts, 523 Ketone bodies, 93, 394, 474, 676, 678, 679, 717
774 INDEX

Kidney, 397–490 synaptic plasticity and, 192–193 hyperbilirubinemia, 578


anatomy, 399 habituation, 192 urinary elimination, 577–578
calyces, 399 sensitization, 192 bilirubin metabolism
cortex, 399 Left heart failure, 283 cellular heme metabolism, 575–576
functions, 397 Left ventricular failure, 226, 349, 390 hepatic transport mechanisms, 576
glucose handling by, 430f Left ventricular hypertrophy, 247, 282 hepatocyte conjugation, 576–577
Henle’s loop, 486 Length constant (λ), 40 role and significance, 575
hilum, 399 Length–tension relationship, 81, 88, 95, 95f blood vessels, bile ducts, and hepatocytes
influence of, 450f Lenticular nucleus, 171 arrangement, 561f
medulla, 399 Leptin, 501, 719–721, 750 cirrhosis, 620
pyramids, 399 Leptin receptor, 721 engineering considerations
stone, 435 Leukotrienes, 307, 323 biliary tract and gallbladder, 562
structural components, 399f Levator ani muscles, 556 blood supply, 560–561
urea handling, 434f Leydig cells, 683 hepatic parenchyma and sinusoids, 561–562
Kinesins, 66 LH. See Luteinizing hormone (LH) functions
Kinocilium, 150 Lidocaine, 53 lipid-soluble waste products, excretion, 560
Klinefelter syndrome, 692 Ligand, 16, 605f, 607, 647f, 660, 662f metabolism and detoxification, 559–560
Knee-jerk reflex, 12, 126 gated channels, 20, 607 protein metabolism and synthesis, 560
Kölliker-Fuse nucleus, 387 ionotropic ligand receptors, 20 gluconeogenesis, 559
Korotkoff sounds, 260 ligand-binding domain of, 706 glucose buffer function, 559
Krebs–Henseleit cycle, 578 metabotropic ligand receptors, 20 splanchnic circulation, 560f
Kupffer cells, 559, 561, 562, 575 osteoprotegerin, 646 Loading spindle, 127
Kyphoscoliosis, 319, 340, 382 Ligand-gated channels, 20 Local circuit interneurons, 121f
Kyphosis, 340 Limbic system, 108 Location of stimulus, 117
α-Limit dextrins, 584–586 Long-QT (LQT) syndrome, 52, 243
L Lipase, 507, 521, 594, 595, 629, 692 Long-term bed rest
Labyrinth. See Inner ear gastric and pancreatic, positional specificity, cardiovascular mechanisms involved, 298f
Labyrinth righting reflexes, 156 594f responses to cardiovascular system, 297–298
Lacrimal duct, 134 Lipases, 517 Long-term depression (LTD), 76, 193
Lacrimal gland, 134 Lipid assimilation Long-term memory, 76, 192f, 194
Lactase, 585, 586 epithelial events in Long-term potentiation (LTP), 76, 192–193
Lactated Ringer’s solution, 474 brush border events, 596–597 production of, 193f
Lactation, 617, 630, 651, 696, 703, 707f, 708, 709, 711 intracellular processing, 597 Loop diuretic, 442, 489
Lactic acid, 91, 474 lymphatic uptake of absorbed lipid, Loop of Henle. See Henle’s loop
production, 745 597–598 Loperamide, 530
Lactic acidosis, 382, 474 fat-soluble vitamins, absorption, 598 Loudness, 152
Lactic acid production, 745 intraluminal digestion Lou Gehrig’s disease, 86, 176
Lactoferrin, 523 bile acids/micelles, role of, 596 Low-density lipoprotein (LDL), 707
Lactose, 584, 586 gastric digestion, 594 Lower esophageal sphincter (LES), 495,
bush border digestion ad assimilation, 586f intestinal digestion, 594–596 508, 544
in dairy products, 591 intestinal handling of cholesterol, 597f Lower motor neurons, 168
Lactose intolerance, 591, 592 intestinal lipolysis, mediators of, 595t Low-pressure receptors, 289
Lactotrophs, 629 role of colipase, 595f Low-resistance electrical, 214
Lactulose, 580 principles Low vascular volume, responses to, 454f
Lambert–Eaton syndrome, 70 dietary and endogenous sources, 593–594 L-type (voltage-gated) calcium channels,
Lamina propria, 528 hydrophobic molecules, assimilation 94, 101
Laminar flow, 257, 322 barriers, 593 Luminal epithelial cell membrane, 619
Language role and significance, 593 Luminal/mucosal membrane, 29
disorders, 196 Lipids, 593 Luminal proteolysis
hemisphere concerned with, 195f bilayer, 1, 15, 596 gastric, 588
physiology of, 195 droplets, 2 intestinal, 588
hemisphere concerned with, 195f flip-flop, 16 Lung compliance, 318–321, 754
path taken by impulses, 196f lamellar phase, 596 Lung volumes, 331
and speech, 195 long-chain triglycerides, 593 measurement of, 332–333
Laplace’s law, 320 phosphatidylcholine, 594 body plethysmography, 334
Large intestine phospholipids, 593 helium-dilution technique, 334
anatomy, 496f rafts, 16 nitrogen-washout technique, 333
L-arginine, 265 Lipolysis, 692. See also Lipase pulmonary function tests, 333
Laron syndrome, 631 products of, 593 spirometry, 333
Latch state, 100, 101 Lipopolysaccharides, 731 and pulmonary vascular resistance,
Latent pacemaker, 214 Lithium treatment, 620 343–344
Lateral corticospinal tracts, 167 Lithocholic acid, 566 standard lung volumes, 331
Lateral geniculate bodies, 108, 139 Liver expiratory reserve volume (ERV), 332
Lateral inhibition, 117, 139, 161 ammonia metabolism, principles functional residual capacity (FRC), 332
Lateral olfactory stria, 159 extraintestinal production, 579 inspiratory capacity (IC), 332
Lateral sacs, 86 intestinal production, 578–579 inspiratory reserve volume (IRV), 332
Law of Laplace, 221, 232 role and significance, 578 residual volume (RV), 332
L-DOPA (levodopa), 173, 666 urea cycle, 579 tidal volume(VT), 331
Lead pipe rigidity, 173 urea disposition, 579–580 total lung capacity (TLC), 332
Lean body mass, 755 bilirubin homeostasis vital capacity (VC), 332
Learning, 191 bacterial metabolism, 577 Lusitropic effect, positive, 221
INDEX 775

Luteinizing hormone (LH), 64, 602, 615, 623, 626, α-Melanocyte-stimulating hormone Mitral valve prolapse, 247
683, 686, 697–699, 702, 711, 726, 750 (α-MSH), 721 Mixed micelles, 565
Lymph, 4, 255, 350, 377, 496, 563 Melatonin, 188, 188f, 189, 615, 624 Mixed venous PO2, 737
Lymphatic system, 4, 255, 311, 350, 552 Membrane-bound hydrolases, 584 Mixed venous blood, 305
Lymphedema, 255, 262 Membrane capacitance, 34, 39 MLC kinase, 100
Lymphocytes, 536 Membrane conductance, 35 Modality, 117
Lymphoid tissues/MALT, 535 Membrane potentials, 10, 34 Modiolus, 149
Lysergic acid diethylamide (LSD), 64 changes in, 39, 44f Monge’s disease, 742
Lysosomal enzymes, 307 measurement, 34 Monitor peptide, 517, 519
Lysosomes, 2, 23, 311, 431, 608, 628, 646f, 703 Membrane proteins, 1, 17, 26, 66, 485, 590 Monoamine oxidase (MAO), 63, 64, 665
Membrane pumps, localization, 23t Monoamine transporters, 665
M Membrane receptors, 24 Monoiodinated tyrosine (MIT), 636
Macroglia, 105 enzyme-linked, 25 Monomer of glutamate receptor channels
Macrophages, 105, 307, 311, 350, 536, 703, G protein–coupled, 25 (gluR), 20
731, 732f Membrane resistance, 39–41 Monophasic action, 54
Macula densa cells, 406, 452 Membrane transport, 4, 521, 532, 568, 576 Monosodium glutamate (MSG), 163
Macula lutea, 134 active pathways, characteristics of, 528t Monosynaptic reflex, 125–126
Macular sparing, 142 protein, MRP2, 576 Morphine, 64, 120
Magnetic resonance image (MRI) scan, 112, 121, 631 Membranous labyrinth, 149 Mossy fibers, 175
Magnocellular cells, 141 Ménière disease, 156 Motilin, 499, 501, 551
Magnocellular neurons, 614f, 615–617, 619f Meningiomas, 161 Motility, 72, 491, 495, 502, 529
Magnocellular pathway, 141 Menopause, 301, 652, 711, 755 esophageal, 545–547
Main pancreatic duct, 518 Menstrual cycle, 99, 611f, 696, 697, 701f, 703, 706, gastric, 547–548
Major histocompatibility complex (MHC), 536 708, 713 intestinal, 552–556
Malabsorption, 496, 512, 521 Merkel cells, 115 Motoneuron, repetitive firing, 75f
Male reproductive system, 684f Messenger RNA (mRNA) Motor cortex, 169
Male sexual differentiation, 689f synthesis of proteins, 2 premotor cortex, 169
Malignant hyperthermia, 57 Metabolic acidosis, 377, 379, 382, 392, 481, 679, primary motor cortex, 169
Maltose, 584, 585f 725, 737, 749 supplementary motor cortex, 169
Maltotriose, 584, 585f, 586 causes, 379t Motor endplate, 68, 74, 85f, 86, 101,
Mamillary bodies, 193 renal response to, 482 128f, 544
Mammalian muscle spindle, 127f Metabolic alkalosis, 379, 481–483, 483f, 725 Motor homunculus, 169
Mammalian nerve fibers, 107, 109t causes, 379t Motor neurons, 9, 85
Mammalian target of rapamycin (mTOR), 675 Metabolic clearance rate, 417 areflexia, 168
Manubrium, 147 Metabolic functions of the liver, 559 axon, 10
Mass balance system, 729 Metabolic syndrome, 435, 719 dendrites of, 9
concept of, 5f Metabolic waste, excretion, 398 fasciculations, 168
Mast cells, 64 Metabotropic, 59 flaccid paralysis, 168
Maximum voluntary ventilation (MVV) test, 387 Metabotropic glutamate receptor hyporeflexia, 168
M cells, 141, 535 (mGluR4), 163 hypotonia, 168
Mean arterial blood pressure (MABP), 258, 297, Methemoglobin, 364, 368 in facial nuclei, 168
619f, 747, 749 Methemoglobinemia, 383 in hypoglossal nuclei, 168
Mean circulatory filling pressure, 276–277 Methimazole, 639 in trigeminal nuclei, 168
Mean electrical axis, 239f Metrorrhagia, 713 muscular atrophy, 168
Mean pulmonary artery pressure, 345f Micelles, 565, 567, 568, 594, 596, 598 spasticity, 168
Mean quantal content, 69, 70 Michaelis–Menten equation, 27 synapse, 10
Mechanical nociceptors, 115 Microbiota, 538 with myelinated axon, 107
Mechanoreceptors, 115, 289, 722 controlling factors, 538–539 α-Motor neurons, 171
afferents, 746 Microelectrode, 34 γ-Motor neurons (nerves), 45, 170
Mechanosensitive channels, 11, 18 Microglia, 105, 112 Motor unit, 68, 85, 89, 91, 385
Mechanosensory transduction, 43 Microtubules, 66 Mountain sickness, acute, 736
Medial and lateral descending brain stem Microvillous membrane, 584, 596 Mountain sickness, chronic, 742
pathways, 170f Midcollicular decerebration, 171 MRP2. See Multiple organic anion transporter
Medial geniculate bodies, 108, 153 Midline nuclei, 108 (MOAT)
Medial lemniscal system, 118 Mifepristone, 708 Mucins, 523
Medial lemniscus, 118 Migrating motor complex (MMC), 548, 551, 551f, Mucociliary escalator, 308
Median eminence, 613, 623 553, 554, 554f Mucosa-associated lymphoid tissues
Median nerve, 122 Milliosmoles, calculation of, 28 (MALT), 535
Median preoptic nucleus, 619 Mineralocorticoids, 626, 662 Mucosal immune system, 535
Medium-chain fatty acids, 596 diseases, 664 CD4 cell, 536
Medulla oblongata, 287 synthesis and release, 658–659 CD8 cell, 536
Medullary cardiovascular centers, 287 Miniature endplate potentials (MEPPs), 69 enteric antigens, immune response to, 537
Medullary collecting tubule, 406 Minute ventilation, 335 autoimmunity, 538
Medullary interstitial fluid, composition, 444 Minute volume, 335 immune responsiveness, 538
Medullary osmotic gradient, components of, 444 Miosis, 182 oral tolerance, 537–538
Medullary pyramids, 168 Mitochondria, 2, 15, 23, 64, 66, 68, 93, 509, 579, features, 535
Medullary raphe neurons, 183 580, 679f, 688f, 691 functional anatomy
Medullary respiratory center, 311, 385 Mitogen-activated protein kinase (MAPK), 674 adaptive immunity, cellular mediators, 536
Meissner’s corpuscles, 115 Mitral cells, 159 innate immunity, cellular mediators,
Melanocortin receptors (MCRs), 626 Mitral regurgitation, 247 535–536
Melanocyte-stimulating hormone, 626 Mitral stenosis, 246, 247, 349 lymphoid tissues, organization, 536
776 INDEX

Mucosal immune system (continued) Na–HCO3— symporters, 475 Nerve fiber, 107
IgE, 536 Na+/K+-adenosine triphosphatase (ATPase), 662 mammalian, classification of, 109t
IgG, 536 Na,K-ATPase, 531 relative susceptibility, 110t
secretory IgA system Na,K-ATPase pumps, 425, 438, 455, 456, 465, 531 Nerve gases, 86
IgA, secretion, 537f plasma membrane pumps, 464 Nerve growth factor (NGF), 111
IgA, structural aspects, 536–537 Na–K–2Cl multiporter, 465, 479 Nervous system, 4, 45, 66, 623
physiological functions, 537 Na–K–2Cl symporter (NKCC2), 442 types of glial cells in, 106f
protective effects, mechanisms, 537 Na/K pump, 37 Net filtration pressure (NFP), 412
Mucus, 161, 307 cycle, 22f Net influx, 27
Multidrug resistance protein 3 (MDR3), 568 Named potentials, 10 Neuroblastomas, 161
Multidrug resistance (MDR) transporter, 24 Narcolepsy, 187 Neuroendocrine regulation, 715
Multiple organic anion transporter (MOAT), Nasal hairs, 308 counterregulation to acute stress, 718
568t, 569 Nasal turbinates, 307 energy metabolism during fasted state, 716
Multiple sclerosis (MS), 53, 107, 112, 162 Na/serotonin cotransporter, 24 fat, 716–717
Multiple system atrophy (MSA), 183 Nasopharynx, 307 glucose, 716
Murmurs, 258 Natriuretic peptides and hormones, 405, 457, 739 protein, 717–718
Muscarine, 60 atrial natriuretic peptide (ANP), 457 energy metabolism during fed state, 715
Muscarinic receptors, 102, 179, 206, 510, 524 brain natriuretic peptide (BNP), 457 fat, 716
AChRs, 20, 60 Nausea, 551. See also Vomiting glucose, 715–716
Muscimol, 63 NaV channels, 75 protein, 716
Muscle cell membrane, depolarization of, 6 N-CAMs, 26 long-term energy balance and fat storage,
Muscles contraction NE. See Norepinephrine (NE) maintenance of, 718–722
changes in strength of, 80 Near point of vision, 137 of stress response, 725
period, 80 Nearsightedness. See Myopia chronic/severe stress, 726f
Muscles of respiration, 305 Necrotizing enterocolitis, 598 Neuroendocrine system, 609, 692
Muscle spindles, 45, 117, 125, 390 Negative base excess, 381 Neurofibrillary tangles, 194, 753, 755
discharge, various conditions, 128f Negative-feedback system, 12, 390 Neurogenic shock, 80, 301
function, 127–128 Negative-pressure breathing, 313 Neurogenic tone, 267
structure, 126–127 Neocortex, 108, 161, 192f, 194, 195 Neurohormones, 615
Muscle tone, 129–130 Neomycin, 581 hypophysiotropic, 623
Muscle weakness, 723 Neospinothalamic tract, 120 hypothalamic, 623
Muscular atrophy, 168, 175 Neostigmine, 70 magnocellular neurons, 614f
Muscular dystrophy, 85, 340 Neosynephrine, 64 Neurohumoral regulation, 498
Muscularis mucosa, 493f, 494, 497f, 502, 553 Nephrogenic, 620 Neurohypophysis, 613
Myasthenia gravis, 68, 70, 77, 86, 91, 382 Nephrogenic diabetes insipidus, 620 Neurokinin A, 503, 554
Mydriasis, 182 Nephron Neurological disorders
Myelin, 68, 105, 106, 106f, 112 basic structure, 402f Bell’s palsy, 162
Myelination, 52 collecting ducts, 399 familial dysautonomia, 162
Myenteric plexus, 494 components, 400f multiple sclerosis, 162
Myocardial contractility, 232 juxtaglomerular apparatus, 403 primary amoeboid meningoencephalopathy, 162
Myocardial infarction, 210, 282, 350, 360 extraglomerular mesangial cells, 403 vestibular schwannoma, 162
Myocardial infarcts, 270 glomerular filtration rate (GFR), 403 Neurological examination, 120–121
Myocardial ischemia, 272, 360 granular cells, 403 Neuromuscular irritability, 647
Myocardial oxygen consumption juxtamedullary nephron, 403 Neuromuscular junction, 67f, 85–86, 85f
determinants, 231–232 macula densa cells, 403 Neuromuscular synapse, 10
Myocytes, 93 renin, 403 Neuromuscular system, 497
Myoepithelial cells, 617 renal corpuscle, 399, 400, 401f enteric nervous system, 497
Myofibroblasts, 503, 523, 529 afferent arteriole, 400 Neuronal depolarization, 613
Myogenic response mechanism, 81, 265, Bowman’s capsule, 400 Neurons, 9, 105–107
415, 547 efferent arteriole, 400 types, in mammalian nervous system, 108
Myoglobin (Mb), 90, 91, 93, 368, 369, 412, 575 glomerulus (glomeruli), 400 Neuropeptides, 64, 717f
Myometrium, 696f, 708 mesangial cell, 400 anorexigenic, 721
Myopia, 136, 138f tubule, 400–403 hypothalamic, 615, 719
Myosin, 83 ascending thin limb, 400 produced by magnocellular neurons, 616
Myosin ATPase activity, 99 connecting tubule, 402 as potential prolactin-releasing factors, 629
Myosin filament, 80, 83 descending thin limb, 400 from sensory terminals, 121f
Myosin light chain kinase (MLCK), 99, distal convoluted tubule, 402 that function as hormones, 613
100, 100f inner medullary collecting duct cells, 403 Neuropeptide Y, 721
Myosin light chain phosphatase, 99, 100, 100f intercalated cells, 403 Neurophysins, 616
Myotonia, 57 loop of Henle, 400 Neurotensin (NT), 630
macula densa, 402 Neurotransmitters, 6, 60f, 63, 64, 75, 106, 179,
N peritubular capillaries, 402, 405, 409, 426 499, 500, 530, 610f, 673, 720
Na–Ca antiporter, 486 principal cells, 403 enteric, 503
Na/Ca exchanger (NCX), 24 proximal convoluted tubule, 400 intestinal epithelial transport regulated by, 529
Na/choline cotransporter, 61 proximal straight tubule, 400 VIP and nitric oxide as, 548
Na–Cl symporter, 443 proximal tubule, 400 Neurotrophins, 110
Na–glucose cotransporter (SGLT), 24 thick ascending limb, 400 Neutrophil, 307
Na/glutamate cotransporter, 24 Nephrotic syndrome, 620 NHE3 sodium-hydrogen exchanger, 531
Na-GLUT antiporter, 27 Nernst equilibrium potential, 36 Nicotine, 60
Na-H antiporters (NHE3), 441, 455, 475 Nernst potential, 38, 39, 67, 74 Nicotinic acetylcholine receptor channels
sodium–hydrogen exchanger, 531 generation of, 36–37 (nAChR), 20, 60
INDEX 777

Nicotinic cholinergic receptor, 86 Olfactory cortex, 159–161 Osmotic pressure, 28, 254, 412
Nicotinic receptors, 86, 89, 503, 544 amygdala, 159 Ossicular conduction, 152
Niemann–Pick C1-like 1 (NPC1L1) gene anterior olfactory nucleus, 159 OST, bile acid transporter, 569
product, 596 entorhinal cortex, 159 Osteoblasts, 648f
Night blindness, 598 frontal cortex, 160 Osteoclast-differentiating factor (ODF), 646
Night terrors, 187 olfactory tubercle, 159 Osteoclasts, 648f
Night vision, 134 orbitofrontal cortex, 160 Osteocytes, 487, 648f
Nigrostriatal dopaminergic system, 171 piriform cortex, 159 Osteomalacia, 598, 650
Nitric oxide (NO), 75, 100, 265, 292, 346, 368, thalamus, 160 Osteonecrosis, 741
545, 692, 738 Olfactory discrimination, 161 Osteopenia, 755
Nitrogen narcosis, 741 Olfactory epithelium, 159, 160f Osteoporosis, 406, 487, 489, 638, 711, 750,
Nitrogen-washout technique, 333 Olfactory glomeruli, 159 753, 755
Nitroglycerin, 272 Olfactory nerve, 165 postmenopausal, 652, 713
Nitrous oxide, 350 Olfactory pathway, 161f prevention of, 652–653
N-methyl-d-aspartate (NMDA) receptors, 193 Olfactory receptors, 25 Osteosclerosis, 154
channel, 74, 76, 193 Olfactory sensory neurons, 159, 159f, 160f Otitis externa, 154
N1 nicotinic cholinergic receptors, 179 Oligoclonal bands, 112 Otitis media, 154
N2 nicotinic cholinergic receptors, 179 Oligodendrocytes, 105, 106f, 183 Otoconia, 150
Nociception, 116 Oligomenorrhea, 750 Otolithic organ (macula), 150
Nociceptive stimuli, 130 Oligospermia, 693 Otoliths, 150
Nociceptors, 45, 115, 118f, 119, 121f Olivocochlear bundle, 153 Ouabain, 22, 37
Nocturnal enuresis, 187 Omeprazole (Prilosec), 23 Oval window, 147
Nodes of Ranvier, 52, 106 On-center cell, 139 Ovarian cycle, 699
Nod2 molecule, 536 Oncotic pressure, 254, 412 Ovarian follicle, 695
Nonchloride anions, 468 in glomerular capillary plasma, 412–413 Ovarian hormones, 713
Nonessential amino acids, 560 of fluid in Bowman’s capsule, 412 overproduction and undersecretion, 713
Non-NMDA channels, 74 of interstitial fluid, 254 physiologic effects of, 705
Nonrespiratory acidosis, 379 of intracapillary fluid, 254 estrogen, 705–707
Nonsteroidal anti-inflammatory drugs (NSAIDs), Oogenesis, 699–701 placenta, 708–709
30, 116, 117, 435, 514 follicle growth and development, 702f progesterone, 707–708
Noradrenergic neurons, 179 and formation of dominant follicle, 699–701 synthesis, 697
Norepinephrine (NE), 56, 63, 178, 188, 189, 217, Open-angle glaucoma, 133 activin, 698
229, 268, 300, 346, 405, 665 Opiate drugs, 530 androgens, 697
receptors, 71 Opioid peptides, 120 estrogen, 697
Normal saline, 372 Optic chiasm, 141 follistatin, 698
NO synthase, 265 Optic disk, 134 inhibin, 698
NREM sleep, 186–190, 391 Optic neuritis, 112 progesterone, 697–698
Nuclear bag fiber Optic tract, 139 Ovulation, 701
dynamic and static, 126 Oral cavity, 495 corpus luteum, formation of, 703
Nuclear chain fiber, 126 Oral rehydration solutions, 529 follicle growth and development, 702f
Nuclear envelope, 3 Oral tolerance, 537–538 luteolysis, 703
Nucleases, 517 Organelles, 2, 3, 15, 23, 79, 95, 217 β-Oxidation of fatty acids, 678
Nucleation, 572 Organic anions, 431t Oxidative phosphorylation, 79, 80, 90, 91, 93,
Nucleolus, 3 Organic anion transporting polypeptide (OATP), 100, 383, 745
Nucleotide–gated cation channel, 163 569, 576 of glucose, 745
Nucleus, 3 Organic cations, 432t Oxidative stress, 176, 580
Nucleus ambiguus, 287, 386, 544 Organic cation transporter (OCT), 432 Oxygen
Nucleus basalis of Meynert, 193–194 Organic substances, renal handling of, 429 carrying capacity of hemoglobin, 364, 737
Nucleus of the tractus solitarius (NTS), 162, 386 organic anions, proximal secretion, 431–432 consumption, 269, 336, 746, 747
Nucleus para-ambigualis, 386 urate, 432 debt, 745
Nucleus parabrachialis medialis, 387 organic cations, proximal secretion, 432–433 delivery, 737
Nucleus retroambigualis, 386 passive reabsorption/secretion, pH inspired and alveolar partial pressures, 736
Nucleus retrofacialis, 544 dependence, 433 toxicity, 742
Nucleus tractus solitarius, 287, 509, 545 proximal reabsorption, 429 transport, factors affecting, 367–368
Nystagmus, 155 glucose, 430 Oxygen-carrying capacity, 364, 737
proteins and peptides, 430–431 Oxygen consumption, 269, 336, 746, 747
O urea, 433–435 Oxygen content of the mixed venous blood, 355
Obesity, 190, 292, 630, 718–719, 721 Organification, 634 Oxygen debt, 745
Obligatory water loss, 440 Organ of Corti, 149 Oxygen delivery, 737
Obstructive diseases, 326, 333 Organomegaly, 631 Oxygen extraction, 269
Obstructive sleep apnea (OSA), 187, 189, 329, 391 Organum vasculosum lamina terminalis, 619 Oxygen loading in lung, 365–366
Occipital lobe, 141 Orientation columns, 142 Oxygen toxicity, 742
Occlusion, 130 Ornithine, 430, 579f Oxyhemoglobin dissociation curve, 364–367,
Octreotide, 631 Oropharyngeal dysphagia, 556 737, 749
Ocular dominance columns, 142 Oropharynx, 307 Oxyntic glands, 508
Oculomotor nerves, 143, 178 Orthostatic/postural hypotension, 183, 298, 448, 620 Oxytocin, 64, 615
Odorant receptors, 159 Osmolarity, 28, 29, 208, 302, 441, 499, 620, 722, 725 physiologic effects of, 617
Off-center cell, 139 Osmoreceptor neurons, 619 release
Olfaction, 307 Osmoreceptors, 459 control of, 617
Olfactory bulbs, 159 Osmosis, 4, 27 physiologic effects and regulation of, 617f
basic neural circuits in, 160f Osmotic diuresis, 441, 468 synthesis and processing of, 616f
778 INDEX

P Parasternal intercostal muscle, 315 luminal digestion, 589f


Pacemaker, 93. See also Cardiac pacemaker; Parasympathetic ganglia, 60, 71 transporters, 589–590
Gastric pacemaker Parasympathetic nerves, 286 peptide transporter 1 (PEPT1), 531, 589
potential, 55 Parasympathetic nervous system (PNS), 80, 101, Peptide YY, 502
Pacinian corpuscles, 44, 115 177, 180f, 523, 692 Percentage of inspired oxygen (FIO2), 355
Packed red blood cells, 372 Parasympathetic tone, 216 Percussion, 329
Pain Parasympathetic vasodilator nerves, 268 Periaqueductal gray matter (PAG), 120
inflammatory pain, 116 Parathyroid adenoma, 653 Pericardium, 203
neuropathic pain, 116 Parathyroid Ca2+-sensing receptor, 644f Periglomerular cells, 159
pathological/chronic pain, 116 Parathyroid gland hyperplasia, 653 Perilymph, 149
physiological/acute pain, 116 Parathyroid glands, 487, 489, 602f, 611, 643, Perineurium, 112
Pain transmission, modulation of, 120 644, 649, 653 Peripheral chemoreceptors, 392
Paleospinothalamic tract, 120 Parathyroid hormone (PTH), 487–489, 643 Peripheral edema, 742
Palpitations, 640, 667, 668, 678, 732, 736 cellular effects of, 645–646 Peripheral motor control system, 130f
Pancreas, 492 clinical evaluation of abnormalities in, 652t Peripheral nervous system, 66, 105–107, 601
acini, 518 hyperplasia of, 653 Peripheral neuropathy, 126
duct of Santorini, 518 hypocalcemia, 487 Peripheral peaking of pulse pressure, 258
islets of Langerhans, 518 mediated osteoclast differentiation, 647f Peripheral thermoreceptors, 730
main pancreatic duct (See Wirsung’s duct) mobilization of bone calcium, 646–647 Peripheral vascular system, 252
phases of secretion production, diseases of arterioles, 207
cephalic and gastric phases, 519 primary hyperparathyroidism, 653 blood vessels, control of, 207
intestinal phase, 519 pseudohypoparathyroidism, 653 capacitance vessels, 207
sphincter of Oddi, 518 secondary hyperparathyroidism, 653 capillaries, 207
structure, 518f release, regulation of, 643–644, 644f, 645t conduit vessels, 207
Pancreatic acinar cells, 518, 595 and renal calcium rabsorption, 645f structural characteristics, 206
receptors, 521f and renal inorganic phosphate (Pi) transmural distending pressure, 207
secretory products, 518t reabsorption, 646f Peripheral venous compartment, 275
zymogen granules, 518 target organs and physiologic effects, 644–645f Peripheral venous pool, 258
Pancreatic amylase, 587 Paraventricular nucleus, 183, 459, 614f, 615t, Peripheral venous pressure, 259, 262, 278,
Pancreatic hormones, 671, 672 617f, 626 288f, 299
diseases associated with, 678–680 Paresthesia, 122 influence on venous return, 279
Pancreatic insufficiency, 525, 584 Parietal cells, 494, 508 Peristalsis, 544–547, 555
Pancreatic juice, ionic composition, 520f ion transport proteins, 513f control of, 546f, 547f
Pancreatic polypeptide, 502, 671, 677, 678 receptors, schematic representation, 511f primary, 546f
Pancreatic proteases, activation avoiding ultrastructural appearance, 509f secondary, 546, 547f
mechanism, 589f Parietal glands, 508 Peritoneal cavity, 399, 563
Pancreatic secretion Parietal lobe, 169 Permeability, 4, 21, 26, 253
anatomic considerations Parkinsonism, 183 glomerular, 431
acinar cells, 518 Parkinson’s disease, 64, 172, 183 hydraulic, 412
ductular cells, 518 Parotid glands, 522, 523 ionic, 35
cellular basis Paroxysmal hypertension, 667 to K+, 37
acinar cells, 520–521 Paroxysmal nocturnal dyspnea, 283 passive, 524
ductular cells, 521 Partial pressure of alveolar oxygen, 754 to sodium ions, 48
pathophysiology, 521–522 Partial seizures, 185, 196 to water, 438, 443, 444, 459, 618
principles Parturition, 617 Permeable membrane, 26
pancreatic secretory products, 517–518 Parvocellular cells, 141 permeant/permeate substance, 26
role and significance, 517 Parvocellular neurons, 615 Pernicious anemia, 120
regulation Parvo (P) cells, 141 Peroxisome proliferator-activated receptor-γ
phases of, 519 Passive diffusion, 253 (PPAR-γ), 719
role of CCK, 519 Passive electrical properties, 39 Perturbation, 1, 6, 459
role of secretin, 519–520 long cylindrical cell, 39–41 of acid–base balance, 471
Pancreatitis, 525 small round cell, 39 in renal handling of potassium, 468–470
Paneth cells, 494 Passive transport, 26 Pertussis, 25
Pannexons, 22 Patent foramen ovale, 741 Peyer’s patches, 535, 536
Papillae, 162, 399 Pathologic anatomic shunts, 355 structure, 536f
circumvallate papillae, 162 Pathophysiology, 1 PGF2α, 346
foliate papillae, 162 Peak expiratory flow (PEF), 326, 384 Phalen’s sign, 122
fungiform papillae, 162 Pedicels, 411 Pharyngeal dilator muscles, 322
Para-aminohippurate (PAH), 418, 432 Pelvic nerves, 554 Pharyngeal dilator reflex, 390
Paracrine, 499 Penile meatus, 692 Pharynx, 544, 545
communication, 500 Pepsin, 507 movement of food, 546f
signaling, 6 secretion, regulation, 511f Phasic contractions, 548
Paradoxical potassium retention, 470 cephalic phase, 511 Phenobarbital, 63
Paradoxical reflex, 388 gastric phase, 512 Pheochromocytomas, 665, 667, 668
Parafollicular cells, 633 intestinal phase, 512 Phonation, 306
Parageusia, 163 Pepsinogen, 507 Phosphate, 643
Parallel fibers, 174 PEPT1, 531, 590 balance, 485–486
Paralytic ileus, 723 Peptic ulcer disease, 514 regulation, 485, 651
Paraplegia, 131 Peptides, 64, 504 renal phosphate handling, 489
Parasomnias, 187 disposition in intestinal epithelial cells, homeostasis, 651
Parasternal intercartilaginous muscles, 311 590f Phosphatidylcholine (PC), 15, 568, 594
INDEX 779

Phosphatidylethanolamine (PE), 15 PNS. See Parasympathetic nervous system (PNS) cardiac, 228, 230
Phosphatidylinositol (PIP2), 15, 16, 101 Podocytes, 411 muscle, 88, 95
Phosphatidylinositol bisphosphate, 607 Poiseuille equation, 203, 258, 322 shifts, 96
Phosphatidylserine (PS), 15, 16 Poiseuille’s law, 203, 258, 322 venous return, 747
Phosphodiesterase (PDE), 607, 638, 675, 692 Polio or Poliomyelitis, 66, 340 ventricular, 226
Phospholamban, 57, 94, 221 Polycythemia, 742 effect of changes, 228
Phospholipase (PLCβ), 25 Polydipsia, 448, 678 larger, 228
Phospholipase A2, 595 Polymeric immunoglobulin receptor (pIgR), 536 Premature ventricular contractions
Phospholipase C (PLC), 21, 101, 607, 658 Polymodal nociceptors, 116 (PVCs), 243
Phospholipids, 593 Polyphagia, 678 Preoptic neurons, 188
bilayer, organization of, 2 Polysomnogram (PSG), 189 Presbycusis, 154
translocators, 15, 16 Polysynaptic reflexes, 125, 130 Presbyopia, 137
Phosphosphingolipid, 15 Polyuria, 620, 678 Pressure, 5
Photoreceptors, 43, 115 Pons, 387 transmural, 5
mechanism, 138–139 Pontine respiratory groups, 387 Pressure natriuresis, 455
current flow in visual receptors, effect of Pores of Kohn, 310 Pressure–volume curve, 318
light on, 138 Portal circulation, 492 Presynaptic cell, 9
photosensitive compounds, 139 Portal hypertension, 563, 580 Presynaptic inhibition, 75
phototransduction in rods and cones, 139f Portal triads, 561 Presynaptic neuron, 59
potentials, ionic basis of, 138 Portal vein, 560 Presynaptic processes, 60, 72
Photosensitive compounds Position agnosia, 121 Presynaptic terminals, 106
opsin and retinal, 139 Positive end-expiratory pressure (PEEP), 321, 348 Prevertebral/collateral ganglia, 178
rhodopsin, 139 Positive feedback system, 7, 12 Primary active transport, 27
scotopsin, 139 Positive-pressure ventilation or breathing, Primary adrenal insufficiency, 664. See also
Phrenic nerves, 315 313, 620 Adrenal gland
Physiological stresses, 738, 739 Positive-pressure ventilation with positive Primary amoeboid meningoencephalopathy, 162
cardiovascular responses to, 286 end-expiratory pressure, 336 Primary colors, 142
Physiologic dead space, 335 Positive-pressure ventilators, 321 Primary/essential hypertension, 292
Physiologic reserves, 750 Positron emission tomography (PET), 191 Primary hyperaldosteronism, 664
Physiologic shunt, 355 Posterior pituitary gland, 268, 448, 459, 613 Primary hyperparathyroidism, 489
Physostigmine, 70, 77 anatomic and functional relationship, 614 Primary hyperthyroidism, 732
Picrotoxin, 63 hormones of, 616–617, 618t Primary metabolic acidosis, 394. See also
Pigment epithelium, 134 synthesis and processing of, 616f Metabolic alkalosis
Pigment stones, 572 Postganglionic fibers, nerves, or neurons, 71, Primary somatosensory area, 169
Pillar cells, 149 177, 286 Primary spontaneous pneumothorax, 330
Pilocarpine, 524 Postmenopausal osteoporosis, 652 Primary uncompensated disorder, 481
Pineal gland, 188f, 189, 615 Postobstructive polyuria, 644 Primary visual cortex, 141
Pinealocytes, 189 Postsynaptic cell, 10, 59 Principal cells, 403, 443, 466
Pineal sand, 189 Postsynaptic inhibition, 128 potassium secretion, 466f
Pituitary, 623 Postsynaptic potential (PSP), 11, 59 Procedural memory, 76
adenoma, 144, 630 Postsynaptic processes, 66–67 Processivity, 66
and hypothalamus, anatomic and functional Post-tetanic potentiation, 71, 192 Procolipase, 595
relationship, 614f Posttraumatic diabetes insipidus, 620 Progesterone, 378, 697, 709
insufficiency, 630 Posture, 167 antiestrogen actions, 708
tumor, 144 Potassium balance, regulation metabolic fate of, 705f
Pituitary adenoma, 144, 630 intracellular and extracellular compartments, physiologic actions, 630f, 707–708
Pituitary adenylate cyclase activating peptide 463–464 receptor–mediated effects, 707–708
(PACAP), 503 hormonal regulation of, 724 receptors, 707
Pituitary insufficiency, 630 renal potassium handling, 464–466 Programmed cell death, 493. See Apoptosis
Pituitary tumors, 142 distal nephron secretion and regulation, Prolactin, 615, 623, 711
Placenta, 299, 625 466–468 family, 624
structure and physiologic function, 708 perturbations in, 468–470 physiologic effects of, 630, 630f
Planum temporale, 195 Potassium channel (KACh), 56 prolactin release, regulation of, 629–630
Plaques, 272 Potassium equilibrium potential, 212 Prolactinomas, 630, 631, 692
Plasma calcium concentration, physiological Potassium excretion, 731 Prolonged QT intervals, 243
responses, 488f Potassium retention, paradoxical, 470 Proopiomelanocortin (POMC), 624, 626
Plasma creatinine, 419 Potassium transport, 466f processing, 627f
steady-state relation, 420f Potential, membrane, 34 Proopiomelanocortin-derived hormones
Plasma norepinephrine, 183 Potentiation, 7 adrenocorticotropic hormone, 626
Plasma osmolality, 755 P1 receptors, 21 β−endorphin, 626
Plasma potassium, 467 Precocious puberty, 692, 713 melanocyte-stimulating hormone, 626
Plasma transcobalamin II (TC II), 591 Prednisone, 53, 77, 662, 732 Prophospholipase A2, 518
Plasma volume, 749 Preganglionic fibers or nerves, 71, 286 Propranolol, 273
Plateau phase, 55 Pregnancy, 299, 696 Proprioception, 117, 126
Platelet-activating factor, 307 and lacatation, 709 Proprioceptors, 9, 11, 156, 387, 748
Platelet aggregation, 208, 272 fetoplacental unit hormone synthesis, 710f Proptosis, 639, 733
Pleural effusion, 282 hormonal control of milk secretion, 711 Propylthiouracil, 639
P loop, 17 mammary gland development, 710–711 Prosopagnosia, 196
Plug formation, 208 parturition, hormonal control of, 709–710 Prostacyclin, 346
Pneumonia, 176, 360, 556, 756 tests, 709 Prostaglandin 15-dehydrogenase, 708
Pneumothorax, 321, 329, 330, 349, 360 Preload, 87 Prostaglandin E2, 117, 307
780 INDEX

Prostaglandins, 117, 307, 529, 533, 708, 732 lung volume and, 343–344 5HT3 receptors, 551
synthesis of, 617 passive influences on, 346t in muscle, tendons, skin, and viscera, 390
Prostaglandins G2 and H2, 307 recruitment and distention, 344–345 potential, 43
Prostate, 683 Pulmonary vascular smooth muscle pulmonary vascular, 390
Prostatic hyperplasia, 687 control of, 345–346 regulating activity of intracellular proteins,
Proteases, 517 Pulmonary wedge pressure, 282 607
Protein kinase A, 25, 57, 94, 97, 100, 231, 607, Pulse oximetry, 750 Receptor tyrosine kinases (RTK), 25
618, 675 Pumps, 16, 22–23 Reciprocal innervation, 128
Protein kinase C, 101–102, 607 Pupillary light reflex, 143 Recompression, 741
Protein kinase G, 100, 103 Purinergic channels, 102 Recruitment, spatial and temporal, 89
Protein/peptide hormones, 602 Purinergic (P2) receptors, 21, 179, 456 Rectoanal inhibitory reflex, 554
synthesis, 603f Purines, 64, 435 Rectum
Protein assimilation Purkinje cells, 174 553f
basic principles, 587–590 Purkinje fibers, 54, 214, 216 Rectus abdominis, 311, 556
barriers to water-soluble macromolecules, Purkinje system, 236 Red blood cell production, regulation, 398
584 PVR. See Pulmonary vascular resistance (PVR) 5α-Reductase inhibitors, 687
brush border hydrolysis, 588–589 P wave, 216, 224 Referred pain, 120
luminal proteolysis, 588 P2X receptors (P2XRs), 21 Reflection coefficient, 28, 349
oligopeptides/amino acids, uptake P2X ATP receptors, 64 Reflex arc, 125, 126f, 131, 499
mechanisms, 589–590 P2X4 receptors, 45 Reflexes, 1. See also Respiratory reflexes
regulation, 590 P2X3 receptor channels, 45 cardiovascular, 288
role and significance, 583 Pyelogram, 447 chemoreceptor, 289
vs. carbohydrate assimilation, 587–588 Pylorus, 494, 508, 548 hyperactive stretch, 169
digestion and absorption, 583 Pyramidal cells, 108, 159 intrinsic, 549, 550
Proteinuria, 427 Pyramids, 399 inverse stretch, 128, 129
Protein zero (P0), 106 Pyrogens, 731, 732f local, 545
Prothrombin, 209 from receptors in heart & lungs, 289
Proton pump, 512 Q respiratory, 388–390, 389t
inhibitor, 514 QRS complex, 216, 238–239 vagovagal, 510, 519, 546, 549, 550
Protoplasmic astrocytes, 105 Quadriplegia, 131 Reflux, 495
Proximal convoluted tubule, 405 Quanta disease, 547
Proximal stump, 112 of ACh, 69 Refraction, 136, 137f
Proximal tubule, 405, 423, 429, 737 Quisqualate channel, 74 Refractory fiber, 89
Pseudohypoparathyroidism, 653 Refractory period
Psilocin, 64 R absolute/relative, 51
Psilocybin, 64 Rabies, 66 Regulatory hypothalamic factor, 625
Psychogenic polydipsia, 448 Radiation, 730 Regulatory systems, 529
PTH. See Parathyroid hormone (PTH) Radioactive iodine uptake test, 733 Reissner’s membrane, 149
PTH-related protein (PTHrP), 489, 645 Raloxifene, 652 REM sleep, 186–188
Ptosis, 91 Raphe magnus nucleus, 120 Renal acid–base processing, 471–484
P-type E1–E2 pumps, 23 Raphe nucleus, 287 Renal angiogram, 461
P-type pumps, 22 Rapid eye movement (REM) sleep, 185 Renal artery, 399, 409, 414
Puberty, 189, 629, 690, 707, 711 Rapidly adapting pulmonary stretch angioplasty, 462
Puborectalis muscle, 556 receptors, 390 blockage, 462
Pudendal nerves, 554 Rapidly adapting (phasic) receptors, 118 obstruction in, 419
Pulmonary blood flow, 341, 355 Rapture of deep, 741 pressure, 415, 455
interaction of gravity and extravascular Rarefaction, 292, 754 stenosis, 453, 462
pressure, 347–348 RAS. See Renin-angiotensin systems (RAS) Renal artery stenosis, 453, 462
positive endexpiratory pressure (PEEP), 348 Rathke’s pouch, 623 Renal blood flow (RBF), 409–410, 449
regional distribution, 346–347 RBF. See Renal blood flow (RBF) afferent arterioles, 409
zones of lung, 347f R-binding protein, 591 arcuate arteries, 409
Pulmonary capillary blood volume, 360 Reaction time, 127, 130 autoregulation, 415–416, 415f
Pulmonary capillary hydrostatic pressure, 349 Reactive/post-occlusion hyperemia, 265 cortical radial arteries, 409
Pulmonary circulation, 342 Rebound phenomenon, 175 efferent arterioles, 409
nonrespiratory functions, 350 Receptive field, 43, 117 kidneys
Pulmonary edema, 30, 247, 283, 348–349, 360 Receptive relaxation process, 495, 549 flow, resistance, and blood pressure,
conditions leading to, 349–350 Receptor, 24 410–411
Pulmonary emboli, 336 Receptor activator of nuclear factor-κβ ligand peritubular capillaries, 409
Pulmonary embolism, 262, 307, 360, 361 (RANKL), 646 vasa recta, 410
Pulmonary embolus, 354, 360 Receptor desensitization, 667 Renal clearance, 417, 418
Pulmonary function tests, 333 Receptor downregulation, 608 Renal compensatory mechanisms, 380
Pulmonary resistance, 322 Receptor kinase, 606f Renal failure, 482, 580
Pulmonary sarcoidosis, 322 Receptor-linked kinase receptors, 606f, 607 chronic, 406, 489
Pulmonary stretch receptors, 388, 390 Receptor-mediated endocytosis, 30 Renal function curves, 292
Pulmonary surfactant, 307, 321 Receptor potential, 161 Renal insufficiency, 630, 664, 722
Pulmonary tissue resistance, 322 Receptor protein tyrosine kinases, 607 Renal-MAP set point, 290
Pulmonary vascular congestion, 351 Receptors, 1, 59, 390, 524, 551. See also Cell Renal microcirculation, 410f
Pulmonary vascular resistance (PVR), 300, membrane receptors Renal plasma flow (RPF), 414, 754
342–343, 748 in airways and lungs, 390 Renal processes. See also Nephron
active influences on, 346t cardiovascular, 390 catabolism, 403
distribution of, 343 5HT1 receptors, 551 chloride reabsorption, 439
INDEX 781

excretion, 403 assessment of, 325–329 (see also Flow- responses to light, 140f
filtration, 403 volume curves) horizontal cells, 134
average values, 405t distribution of, 322 optic nerve, 134
individual tubular segments increased, clinical consequences of, 329 processing of visual information in, 139
collecting duct system, 443–444 lung volume and, 323 projection, on primary visual cortex, 141f
distal convoluted tubule, 443 alveolar pressure, 314 rod and cone density, 136f
Henle’s loop, 442–443 dynamic compression of airways, 324–325 rods, 134
proximal tubule, 440–442 functions visual receptors in, 134–135
metabolism by tubules, 405 acid–base balance, 306 Retraction, 316
for sodium, chloride, and water, 437 filtration and removal of inspired particles, Retrograde amnesia, 192
fundamental elements, 404f by airways, 309 Retrograde giant contraction, 552
glomerular filtration, 404 gas exchange, 305, 306f Retrograde transport, 110
reabsorption, 403 handling of bioactive materials, 307 Retropulsion, 550, 551
average values, 405t phonation, 306 Reversal potential, 67
tubular reabsorption, 404 pulmonary defense mechanisms, 306–307 Reverse T3 (rT3), 637
regional function, overview of, 405–406 pulmonary metabolism, 307 Rhodopsin, 44f
renal function, regulation, 405 removal of material, from alveolar Rho kinase pathway, 100
secretion, 403 surface, 311 Rhythmicity, 240–241
tubular secretion, 404–405 hypoxia of altitude, 736–737 Ribosomes, 2, 603f
sodium reabsorption, 438–439 intrapleural pressure, 314 Rickets, 598, 650
synthesis, of secreted substances, 403 mechanical interaction Right atrial pressure (RAP), 747
urinary concentration between lung and chest wall, 314, 314f, Right axis deviation, 742
urea, 445–447 321–322, 338 Right heart failure, 284
vasa recta, 445 medullary respiratory center, 311 Right ventricular failure, 742
water reabsorption, 439–440 muscles of, 311, 315 Right ventricular hypertrophy, 738
Renal tubular acidosis, 427 bronchial smooth muscle, control of, Rods of Corti, 149
Renin, 658 322–323 ROMK potassium channels, 466
secretion, control, 453f expiratory muscles, 317 activity, 467f
Renin–angiotensin–aldosterone system, inspiratory muscles, 315–316 Rostral ventrolateral medulla, 183, 287
658, 722 normal tidal breath, events involved in, 317t Rough endoplasmic reticulum (RER), 2, 60,
aldosterone release, regulation, 661f volume, pressure, and airflow changes 66, 597
Renin-angiotensin system, 406, 451–453, 482 during, 318f Round window, 149
ACE inhibitors, 453 pressure, flow, and resistance, relationship Rubrospinal tracts, 167
angiotensin II receptor blockers (ARB), 453 among, 322 Ruffini corpuscles, 115
components of, 452f pressure-volume relationships in, 318 Rugae, 508
Renovascular hypertension, 462 alveolar interdependence, 321 Ryanodine receptors (RyR), 21, 80, 86, 86f,
Renshaw cell, 130 clinical evaluation of compliance, 319 94, 101
Reserpine, 64 compliance of lung and, 318–319
Reserve, 95 elastic recoil, 319–321 S
Residual volume (RV), 325, 332 pulmonary surfactant, 321 Saccades, 143
Resistance, 5 structure, 307 Saccules, 134, 147, 150, 151f, 154
Resistance vessels, 207 airways, 307–309 Safety factor, 53, 350, 366
Resistance work of breathing, 737 alveolar–capillary unit, 309–310 Saliva
Resonator, 152 alveolar septum, 310f constituents, 523
Respiratory acidosis, 377, 378, 481 human lung parenchyma, 309f ionic composition, 524f
causes, 378t pulmonary capillary, 310f Salivary amylase, 523, 584
Respiratory alkalosis, 378–379, 481, 736 transpulmonary pressure, 314 Salivary glands, 492, 495, 523
causes, 379t ventilatory response to exercise, 747–749, 748t salivatory center, 523
Respiratory compensatory mechanisms, 380 work of breathing, 329 salivatory nuclei, 523
Respiratory control system, 385 Respiratory zone, 308 salivary gland anatomy
organization of, 386f Resting membrane potential, 34, 80, 102, acinar cells, 523
response to 212, 214 ductular cells, 523
carbon dioxide, 390–392 of hair cells, 150 salivary secretion
hydrogen ions, 392–393 hyperpolarization, 216 acinar cells, 524
hypoxia, 394 Resting potential, 10, 34, 37–38 ductular cells, 524
spontaneous rhythmicity, 386–388 generation, 35 neural regulation, 523–524
Respiratory gases Kir channels supporting, 38 role and significance, 522
partial pressures of, 336–337 Restrictive disease, 326, 333, 340 salivary secretory products, 522–523
Respiratory pump, 299, 302 Reticular activating system, 110, 188 Salivary secretory products, 522–523
Respiratory rate, 329, 330, 339, 340, 350, 360, 372, Reticular formation, 110, 386 Salivatory center, 523
385, 388, 394, 427 Reticular lamina, 149 Salivatory nuclei, 523
Respiratory reflexes, 388, 389t Reticuloendothelial system, 575 Salmonella, 534
cardiovascular receptors, 390 Reticulospinal tracts, 167, 170 Salt appetite, 461
j receptors, 390 Retina, 134 Saltatory conduction, 52, 106
pulmonary stretch receptors, 388, 390 amacrine cells, 134 Salt restriction, 293
receptors bipolar cells, 134 SA node, 289
in airways, 390 cones, 134 Sarcoidosis, 319, 322, 329, 360, 382
in muscle, tendons, skin, and viscera, 390 extrafoveal portion, neural components of, Sarcolemma (SL), 79, 85, 100
Respiratory rhythm generator, 387 135f sarcoplasmic reticulum (SR) interaction, 93
Respiratory system, 305–396 ganglion cells, 134 Sarcomeres, 83
airway resistance, 322 projections from right hemiretina of, 141f pattern of striation, 84f
782 INDEX

Sarcopenia, 753 Sensory receptors, 115, 125 central sleep apnea, 187
Sarcoplasmic reticulum (SR), 79, 86, 101, 217, 231 cutaneous mechanoreceptors, 115 hypersomnolence, 187
Scalae, 149 nociceptors and thermoreceptors, 115–117 narcolepsy, 187
Scala media, 149 sensory receptors in skeletal muscles & joints, night terrors, 187
Scala tympani, 149 115–117 nocturnal enuresis, 187
Scala vestibuli, 149 Sensory systems, 4 parasomnias, 187
Scalene muscles, 311, 315 code elementary attributes of, 116f, 117 somnambulism, 187
Scanning speech, 175 improving discrimination, 139 stages, 186–187
Scar formation, 112 Sensory transduction, 43 distribution of, 187
S cells, 519 Sensory unit, 117 K complexes, 186
Schaffer collateral LTP, 193 Sepsis, 618 PET scans during REM sleep, 187
Schizophrenia, 64, 195 Septal ischemia, 222 pontogeniculooccipital (PGO)
Schwann cells, 105, 106, 107f, 112 SERCA pump, 23 spikes, 187
Scleroderma, 360 Serosal/peritubular membrane, 29 slow-wave sleep, 186
Scoliosis, 340 Serotonin (5-HT), 63, 116, 188, 307, 503 theta rhythm, 186
Scuba, 740 receptors, 20 Sleep–wake cycle, 187–188
Secondary active transport, 27 Sertoli cells, 683, 684 circadian rhythms and, 187–189
Secondary hyperparathyroidism, 651 Set point, 6, 731 and melatonin, 189
Secondary hypoaldosteronism, 664 Set point for temperature regulation, 289 Slit diaphragms, 411
Secondary hypothyroidism, 639 Severe sweating, coordinated response to, 461f Slowly adapting (tonic) receptors, 118
Secondary peristalsis, 546 Sex hormone–binding globulin (SHBG), 686 Slow-wave sleep, 186, 624
Secondary respiratory alkalosis, 394 Sex hormone synthesis, 626 SL voltage-gated calcium channels, 80
Secondary spontaneous pneumothorax, 330 SGLT-1, 531 Small intestine, 496
Secondary transporter, 23 Shallow water blackout, 740 chloride secretion, 532f
Secondary tympanic membrane, 149 Shear stress, 257 electroneutral NaCl absorption, 532f
Second-degree heart block, 242 Sheehan syndrome, 630 epithelial layers, morphology of, 493f
Secretin, 499, 501, 519, 568 Shivering thermogenesis, 730 ion transport mechanisms, 528ft
function, 520f Shock, 80, 379, 618, 665 jejunum and ileum, 496
Secretory component, 537 Shock lung syndrome, 321 role, 552
Secretory diarrheal disease, 530f Short bowel syndrome, 598 Small skeletal muscles
Secretory granules, 2 Shortening stapedius, 147
Secretory immunoglobulin (IgA) maximum velocity of, 88 tensor tympani, 147
molecules, 535 Short stature, 631 Smooth muscle, 79–81, 99–103
Segmental propulsion, 555 Short-term memory, 76, 192, 196 contraction, 99–100
Segmentation, 555 Shunt equation, 355 influences on, 102f
Selectins, 26 Shunt flow, 355 energy, for contraction and relaxation, 100
Selective estrogen receptor modulators Shunt fraction, 355 multiunit versus unitary, 100–101
(SERMs), 652 Shuntlike states, 355 smooth muscle cell types, comparison of, 100
Selective serotonin reuptake inhibitors, 64 Shunts, 383 stimulation, methods of, 101–103
Self-contained underwater breathing apparatus Shy–Drager syndrome, 183 vascular versus visceral, 100–101
(Scuba), 740 Sickle cell disease, 350, 364 Smooth pursuit movements, 144
Semen, 684 Sildenafil, 692 SNAP-25, 65
Semicircular canals, 147 Silicosis, 329 SNARE proteins, 65
Seminal fluid, 684 Simple cells, 142 Sneeze, 390
Seminiferous tubules, 683 Simple diffusion, 26 S-nitrosohemoglobin (SNO-Hb), 368
Senescence, 690 Sinoatrial (SA) node, 54, 93, 212, 228, 236 SNS. See Sympathetic nervous system (SNS)
Senile dementia, 194–195 Sinusoidal endothelium, 561, 562 Sodium
Sense organ, 115 Sinusoids, 560–563, 569 excretion, regulation
Sensitization, 541 Sitosterolemia, 596 and angiotensin II, 454–455
Sensors, 9, 12 ABCG5 transporters, 596 and autoregulation, 456–457
central thermoreceptors, 730 ABCG8 transporters, 596 and ECF volume, 453–454
peripheral thermoreceptors, 730 Skeletal muscle, 79–81, 83–91 glomerular filtration rate, 453
Sensory adaptation, 45 cell, 9 important variables, 458f
fast and slow, 45f excitation–contraction coupling, 86 long-term control, 455–456
Sensory coding, 117–118 fiber (cell) types, comparison of, 90 natriuretic peptides, 457–458
Sensory endings glycogenolysis, 715 pressure natriuresis and diuresis, 455
primary (group I) ending, 126 neuromuscular junction, 85–86 renin–angiotensin systems, 451–453
secondary (group II) endings, 126 pattern of sarcomeres, 83–84 summary, 458
Sensory fibers, 110 pump, 269, 297, 747 vascular resistance, renal
numerical classification for, 110t regulation of control of, 451
Sensory generator potential, 10, 43 contraction in skeletal muscle, 89–90 intake and loss, normal routes, 438t
all-or-none, 10 contraction-length-tension, 88–89 reabsorption, 731
encoded, 43 sarcomere within, 84 collecting duct system, 438
graded, 10 types of contractions, 86–88 comparison with water reabsorption, 439t
Sensory hair cells, 44 voluntary, 80, 85 distal convoluted tubule, 438
Sensory homunculus, 120 Skeletal muscle pump, 269, 297, 747 Henle’s loop, 438
Sensory information Sleep Na,K-ATPase pumps, 438
from peripheral receptors to cerebral and arousal, neurochemical mechanisms pathways for, 440f
cortex, 119f promoting, 188 proximal tubule, 438
Sensory neuron, 9, 12f, 108, 111, 117, 119f, 149, disorders, 187 summary of mechanisms, 441t
159, 160, 162, 502f cataplexy, 187 transport pathways, 442f, 443f
INDEX 783

Sodium-bicarbonate cotransporter (NBC), 521 rectoanal inhibitory reflex, 554 zona glomerulosa, 656
Sodium channels upper esophageal sphincter, 544 zona reticularis, 656
inactivated, 48 Sphingolipids, 15 Steroidogenesis. See Sex hormone synthesis
repolarized, 48 Sphingomyelin, 15 Steroidogenic acute regulatory (STAR)
Sodium-coupled cotransporters Sphygmomanometry, 297 protein, 656
SVCT1/SVCT2, 590 Spina bifida, 340 Steroid receptor superfamily, 607
Sodium-coupled nutrient absorption, 531f Spinal cord, 105, 385 Stethoscope, 226
Sodium-dependent glucose symporter lateral spinothalamic tract, 119 Stimulatory G proteins, 217
(SGLUT), 430 Spinal cord injury (SCI), 131 Stimulus-secretion reflex, 629
Sodium/glucose cotransporter Spinal integration, 131 Stomach, 495, 547
SGLT-1 transporter, 531, 586, 587 Spinal motor neurons, 167 antrum, 548
Sodium–hydrogen exchanger (NHE-1), 513 Spinal nerves, 107 cardia, 495, 548
Sodium–iodide (Na+/I-) symporter, 635 Spinal shock, 131 corpus, 548
Sodium/potassium/2 chloride cotransporter Spinal ventral roots, 125 functional regions, 495f, 548f
(NKCC1), 532 Spindle sensitivity, 129 antrum, 508
Sodium–proton antiporter (NHE3), 426 Spinocerebellum, 174 cardia, 508
Sodium reabsorption, 731 Spinoreticular pathway, 119 fundus, 508
Sodium taurocholate cotransporting polypeptide Spinothalamic tract, 115 gastric glands, 508
(NTCP), 569 Spiny stellate cells, 109 gastric pits, 508
Soluble N-ethylmaleimide-sensitive factor Spiral ganglion, 149 gastrin, 508
attachment protein (SNAP)-receptor Splanchnic circulation lower esophageal sphincter, 508
proteins, 65 schematic anatomy, 497f oxyntic glands, 508
Soma, 72 Splanchnic nerve, 548 rugae, 508
Somatic sensory areas Spleen, 560f, 575, 740 fundus, 548
somatic sensory area I (SI), 118 Stagnant hypoxia, 383 gastric glands, 495
somatic sensory area II (SII), 118 Staircase phenomenon, 94 intrinsic and vagovagal reflexes, 550f
Somatomedins, 629 Standard 12-lead electrocardiogram, 240 phasic and tonic contractions, 548
Somatomotor, 183, 290 Standard lung capacities, 331 pylorus, 548
peripheral organization and transmitters Standard lung volumes, 331 receptive relaxation, 495
released by, 178f Starch, 584 Stones. See also Gallstone disease; Kidney, stone
Somatosensory pathways, 118 Starling equation, 348 formation, 485
dorsal column pathway, 118–119 Starling forces, 426 Storage-operated channels (SOC), 102
dorsal horn, 118 Starling hypothesis, 254 Strabismus, 137
Somatostatin (SST), 503, 509, 627, 676, 677 Starling’s law of heart, 205, 277 Stratified squamous epithelium, 493
Somatotopic organization, 169 Stasis, 360 Strenuous exercise
Somnambulism, 187 Static compliance, 319 blood flow in skeletal muscle, 271
Sound Static response, 126 cardiac output, 227, 270, 359
amplitude, 152 Steady state, 37 diffusion limitation of oxygen transfer
bels, 152 Steatorrhea, 512 during, 359
decibel scale, 152 Stellate cells, 175, 561 lactic acidosis associated with, 474
frequency, 152 Stenosis, 272, 462 left ventricular output, distribution of, 747f
pitch, 152 aortic, 246, 248 oxygen consumption rate, 270
Sound localization, 153–154 mitral, 246f, 247, 349 Stretch receptors, 386, 388
Space constant, 40 renal artery, 453, 462 Stretch reflex, 12, 125–126
Space of Disse, 562, 576 Stents, 273, 750 pathways responsible for, 128f
Spasticity, 110, 129, 131, 168, 171, 173 Stercobilinogens, 577 Striatum, 171
Spatial and temporal facilitation, 130 Stercobilins, 577 Stricture, 556
Spatial recruitment, 89, 90 Stereocilia, 150 Stroke, 130
Spatial summation, 73 Stereognosis, 121 Stroke volume (SV), 95, 228, 258
Specificity, 24, 576, 605 Sternocleidomastoid, 311 influences on, 228
of CCK and gastrin, 501 Steroid hormone receptors, 24 cardiac muscle contractility, 229–230
of cholesterol esterase, 595 and mineralocorticoid specificity, 662f cardiac output, 230f
and high affinity, for hormones, 605 Steroid hormones, 602 Starling’s law, 228
of pepsins, 588 alterations in synthesis, 659f ventricular afterload, 228–229
positional, of lipases, 594f androgens, 655 Strong acid, 375
Specific sensory relay nuclei, 108 cholesterol esterase, 656 Structural interdependence of alveoli, 321
Sperm, 683 corticosterone, 656 Strychnine, 63
and accessory organs for production, 684t cortisol, 656 ST segment depression, 750
Spermatogenesis, 690–691 cytochrome P450 sidechain cleavage (SCC) Subfornical organ, 619
key events in, 691f enzyme, 656 Sublingual gland, 523
and functional importance, 691t DHEA and DHEA sulfate (DHEAS), 656 Submandibular gland, 523
regulation of, 691 glucocorticoids, 655 Submucosal gland cells, 307
Spermatogonia, 683 mineralocorticoids, 655 Substance P, 116, 265, 503, 545
Spermatozoa, 683 receptors and mineralocorticoid Substantia gelatinosa, 118
Spermiation, 690 specificity, 662f Sucrase, 585
Sphincter of Oddi, 494, 518, 562 steroidogenic acute regulatory (StAR) Sucrase-isomaltase, 586
Sphincters, 494, 554 protein, 656 Sucrose, 584, 586
external anal sphincter, 554 synthesis and metabolism, key bush border digestion ad assimilation, 586f
ileocecal valve, 554 enzymes, 658t Sulfonyl urea receptor, 673
internal anal sphincter, 554 target organ cellular effects, 660–661 Sulfur-containing amino acids, 473
lower esophageal sphincter (LES), 495, 508, 544 zona fasciculata, 656 Summation, 89
784 INDEX

Suppression scotoma, 137 Taste cells iodine metabolism in thyroid follicular cell,
Suprachiasmatic nucleus, 188, 615, 623 type I, II, and III, 162 regulation, 635–636
Supraventricular abnormalities, 240–241 Taste modalities, 162 key features of, 636t
Supraventricular arrhythmias, 242f Taste pathways, 161–162, 164f parafollicular cells, 633
Supraventricular tachycardia, 232, 241, 242 Tau protein, 194, 755 regulation and function, 636t
Surface mucous cells, 509 T-cells, 535 thyroglobulin, 633
Surface tension, 319 receptor, 536 thyroid hormone, 731, 732
Sustaining collateral, 111 Tectospinal tracts, 167, 170f Thyroid hormone, 731, 732
SVR. See Systemic vascular resistance (SVR) Temperature-regulating mechanisms, 731f biologic effects, 637–638
Swallowing center, 544 Temporal bone, 149 diseases of, 638
Sweat glands, 730 Temporal lobe, 191, 195, 196 hyperthyroidism, 639
Sylvian fissure, 118, 119, 195 Temporal recruitment, 89 hypothyroidism, 638–639
Sympathetically mediated vasoconstriction, 738 Temporal summation, 39, 73 inotropic and chronotropic effects of, 638
Sympathetic celiac plexus, 405 Tension, 86–88 iodine metabolism, 639–640
Sympathetic chain, 178 developed tension, 88 metabolism, 637, 637f
Sympathetic ganglia, 71 passive tension, 88 organ-specific effects, 638
Sympathetic nerves, 286 total tension, 88 release, regulation, 636
Sympathetic nervous system (SNS), 80, 94, 101, Tension pneumothorax, 330 synthesis, 634–635, 636f
177, 180f, 268, 524, 745 Terminal buttons/boutons, 106 transport and tissue delivery, 637
Sympathetic paravertebral ganglion, 178 Testosterone, 683 Thyroid hyperplasia, 634
Sympathetic preganglionic and postganglionic biosynthesis and metabolism, 688f Thyroiditis, 634, 639
fibers, 179f diseases of, 692–693 chronic, 640
Sympathetic tone, 216 receptor-mediated effects of, 687f Thyroid peroxidase, 636
Sympathetic vasoconstrictor nerves, 267 specific actions of, 689t Thyroid-stimulating hormone (TSH), 615, 623,
Sympathomimetic effects, 638 Tetanic contractions, 89 633, 634, 640, 732
Synapse, 6, 10, 40, 59, 62, 67, 71, 73, 74, 119, 126, Tetanus, 66, 89 Thyroid-stimulating immunoglobulins (TSI), 639
178, 188, 497, 571 toxin, 63 Thyroid storm, 731, 733. See Hyperthyroidism
Synaptic cleft, 11, 59, 68 Tetany, 52, 653 Thyrotoxicosis, 732
Synaptic currents Tetraiodothyronine (T4), 602, 634 thyroid storm, 733
integration, 73–74 Tetrodotoxin (TTX), 53 Thyrotropin releasing hormone (TRH),
Synaptic knobs, 106 Thalamic fasciculus, 171 625, 633
Synaptic plasticity, 192 Thalamic reticular nucleus, 108 Thyrotropin-secreting tumors, 630
Synaptic release, 64–66 Thalamostriatal pathway, 171 Thyroxine, 732
Synaptic transmission Thalamus, 107, 108, 111, 119, 154, 160, 161f, 164f, Thyroxine-binding globulin, 707
depression, 70, 71f 167, 171, 173 Tidal volume (VT), 329, 331
facilitation, 70–71, 71f Thermal nociceptors, 115 Tight junctions, 6, 29
posttetanic potentiation (PTP) of, 71f Thermoreceptors, 115 Time constant, 39
Synaptic vesicle docking, 60f cold and warm receptors, 115 Tinel’s sign, 122
Synaptotagmin, 66 Thermoregulation, 715 Tinnitus, 156
Syncope, 210, 289, 290 Thermostatic set point, 6 Tip links, 150
Syncytium, 93, 205 Thermostat signals, 6 Tissue plasminogen activator (tPA), 209
Syndrome of inappropriate ADH secretion, 620 Theta rhythm, 186 Tissue pressure hypothesis, 266
Synovial fluid, 4 Thiazide diuretics, 443 Titin, 85
Systemic anaphylaxis, 541 Thiazolidinedione drugs, 719 Titratable acidity, 480
Systemic and pulmonary circulations Third degree (total) AV nodal heart block, 221 Toll-like receptors, 536
differences in pressure, 343 Third-degree heart block, 242 Tonic–clonic seizure, 185
Systemic cardiovascular circuit, 276f Thoracic duct, 597–598 Tonic contractions, 548
Systemic hypertension, 292 Thoracoabdominal pump, 299, 747 Tonicity, 28
Systemic vascular resistance (SVR), 258, 343, 638, Threshold, 47 Tonsils, 308
739, 747 Thrombi, 272 Total body water, 200, 755
Systole, 56, 204, 221, 224, 225, 229, 247, Thrombin, 209 Total cholesterol, 707
269, 270 Thromboembolus, 361 Total CO2, 377
Systolic compression, 269 Thrombolytic drugs, 361 Total lung capacity (TLC), 322, 332, 750
Systolic heart failure, 245, 282, 283f Thrombophlebitis, 262 Total peripheral resistance (TPR), 232, 258, 264,
Thrombosis, 360 285, 292, 299, 302, 343, 450, 451, 453, 460
T Thromboxane, 208, 346 Toxic nodules, 635
Tachycardia, 232, 241, 242f, 243f, 301, 350, 361, Thymectomy, 77 Tracheobronchial tree, 307
640, 667 Thyroglobulin, 633, 634 Transcapillary fluid movement, 254–255
Tachykinins, 503 Thyroid autoimmune disease, 634 Transcapillary solute diffusion, 253–254
neurokinin A, 503 Thyroid-binding globulin (TBG), 637 pathways, 253f
substance P, 503 Thyroidectomy, 733 Transcellular processes, 425
Tachyphylaxis, 667 Thyroid gland Transcortin, 658, 707
Tachypnea, 350, 361, 372, 427 calcitonin, 633 Transcytosis, 30
Tactile acuity, 117 colloid, 633 Transducers, 43, 115
Tactile agnosia, 121 euthyroid, 638 Transducin, 44f, 45, 139, 139f
Taenia coli, 553 follicular (epithelial) cells, 633 Transepithelial transport mechanisms, 527
Taste buds, 161–163 functional anatomy Transient ischemic attack (TIA), 210
basal cells, 161 thyroid follicle, 633 Transient receptor potential (TRP), 45
dark cells, 162 hypothalamic–pituitary–thyroid axis, Transient shrinking, 29
intermediate cells, 162 evaluation, 640 Transitional zone, 308
light cells, 162 iodide concentration, mechanism, 635f Transmembrane protein, 2f, 194, 607
INDEX 785

Transmembrane solute transport, mechanisms, Type II alveolar epithelial cells, 307 Vascular smooth muscle, 263–264
424f Tyrosine hydroxylase (TH), 63, 665 Vascular tone, 264
Transmitter–receptor interaction, 69–70 Tyrosine kinases, 629 Vasculature, 206–207, 257, 267, 336, 404, 445,
Transmitters, 10–11, 59 Tyrosine-tyrosine, 502 451, 563, 619
Transmural distending pressure, 207 blood vessels, control of, 207
Transmural pressure gradient, 265, 314, 343 U Vas deferens, 683
Transport across cell membranes, 26 UDP glucuronyl transferase (UGT), 576 Vasoactive intestinal polypeptide (VIP), 64, 265,
active, 27 Ulcerative, 538 501, 503, 521, 529, 547, 630
facilitated, 27 Ulcerative colitis, 534 Vasopressin, 64, 268, 302
passive, 26–27 Unacclimatized person, 736 Vasovagal syncope, 210, 289
Transport across epithelial cells, 29–30 Uncal herniation, 171 Venoconstriction, 268
Transporters, 16, 23, 424 Uncompensated respiratory alkalosis, 361, 384 Venous admixture, 355
types of, 24f Unidirectional efflux, 27 Venous capacitance, 747
Transport mechanisms Unidirectional influx, 27 Venous function curve, 278
classification, 426 Uniporters (UT family) transports, 434 Venous return, 277, 747f
Transpulmonary pressure, 314, 318, 338–339 Unloading oxygen, at tissues, 366 Venous tone
Transthyretin, 637 Upper endoscopy, 556 control of, 268–269
Transverse tubules, 11 Upper esophageal sphincter, 544 Ventilation–perfusion mismatch, 355
Traumatic pneumothorax, 330 Upper motor neuron lesion, 130 Ventilation-perfusion ratio, 748
Traveler’s diarrhea, 534 Upper motor neurons, 168 matching, 737
Trefoil factors, 507 Urate, 432–433 mismatching, 754
Tremor, 171, 173 Urea, 433–435, 445–447 Ventilation–perfusion ratio line, 355
Tremor at rest, 173 Urea cycle, 578, 579, 580f. See also Krebs- Ventilation-perfusion ratio matching, 737
Treppe, 94 Henseleit cycle Ventilation-perfusion relationships and ratios,
T1R3 family, 163 Urea disposition, 579–580 353–356
T2R family, 163 Ureases, 579 alveolar–arterial oxygen difference, 355–356
Triiodothyronine (T3), 602f, 634, 733 Uremia, 434, 653 increased, causes of, 356t
Trochlear nerves, 143 Ureter, 399f, 400, 403, 414, 435, 444, 447 mismatched, testing for, 355
Tropic hormones, 623 Urethra, 399f, 684 physiologic dead space, 355
Tropomyosin, 83 Uric acid, 208, 398, 405, 432, 435 physiologic shunts, 355
Troponin, 80, 83, 94 Urinary system shunt equation, 355
TnC, 83 anatomy, 399 regional differences in lung, 356–357
TnI, 83 in female, 399f distribution, 357f
TnT, 83 Urobilinogens, 539, 577 ventilation–perfusion ratios, 353–355
Trousseau’s sign, 648 Urobilins, 577 high and low, consequences of, 353–355
Trypsin, 517, 588 Urolithiasis, 653 Ventral anterior/lateral nuclei, 108
Trypsin inhibitors, 517 Ursodeoxycholic acid, 566 Ventral cochlear nuclei, 153
Trypsinogen, 517, 588 Uterine contractility, 708 Ventral posterior lateral (VPL), 108
Tryptophan hydroxylase, 64 Utricle, 147, 149f, 150, 154, 155f nucleus, 118
T score, 652 Ventral posteromedial, 108
TSH receptor, 733 V nucleus, 162
t-SNARE syntaxin, 65 Vagal communication, 503 Ventral respiratory groups (VRG), 386
T-tubule, 86, 94 Vagovagal reflexes, 510, 529 Ventricular abnormalities, 242–244
T-type calcium channels, 55 Vagus nerves, 60, 162, 178, 216, 323, 390, 503, 509, Ventricular arrhythmias, 243f
D-Tubocurare, 70 545, 547, 552f Ventricular depolarization, 238, 239
Tubular maximum (Tm), 418, 426 Valsalva maneuver, 299 and generation of QRS complex, 239f
Tubular osmolality, 447f Valve abnormalities, common, 246 Ventricular fibrillation, 243
Tubular potassium transport, 465t Vanilloid receptor, 45 Ventricular gallop rhythm, 226
Tubular transport mechanisms VR1 for, 45 Ventricular repolarization, 239
limits on rate Vanillylmandelic acid (VMA), 665 and T wave, 239f
Tm and gradient-limited systems, 426–427 Variable obstructions, 326 Ventricular systole, 225
paracellular route, 424 Varicocele, 692 Ventricular tachycardia, 243
proximal tubule reabsorption, 423–426 Vasa recta, 409, 445 Ventrolateral spinothalamic tract, 119–120
transcellular and paracellular reabsorption, 424f Vascular bed, 753 Verapamil, 273
transcellular route, 424 stiffening (see Arteriosclerosis) Vermis, 174, 175
Tubuloglomerular feedback, 456 Vascular control Vertigo, 156, 552
Tubulovesicles, 509, 512 mechanisms, 269 Vesicles, 10, 60
Tufted cells, 159 in specific organs, 269 Vestibular movements, 144
Tumor necrosis factor, 731 cerebral blood flow, 271–272 Vestibular schwannoma, 162
Turbulent flow, 257, 322 coronary blood flow, 269–270 Vestibular system, 154
T wave, 215f, 216, 226, 236, 237, 239, 241, 242, skeletal muscle blood flow, 270–271 Deiters’ nucleus, 154
248, 360 Vascular function, basic, 255 vestibular apparatus, 154
Twitch, 88 arteries and veins, elastic properties, 259 vestibular nuclei, 154
Two-point discrimination test, 117 blood flow velocities, peripheral, 256–258 Vestibulo-ocular reflex, 155
Tympanic membrane, 147 blood volumes, peripheral, 258 Vestibulospinal tracts, 167
Tympanic reflex, 152 and flow in networks of vessels, 255–256 Vibratory sensibility, 120
Type A intercalated cells, 475 resistance, 255–256 Vibrio cholerae, 533
Type B intercalated cells, 476 vascular resistances, peripheral, 258 Vibrissae, 308
Type 1 diabetes mellitus, 394, 587 Vascular pressure Villus structures, 493
Type 2 diabetes mellitus, 292, 406 effect of gravity on, 296f VIP. See Vasoactive intestinal polypeptide (VIP)
Type I alveolar cells, 310 responses to changes in body position, 296–297 Virilization, 657
786 INDEX

Visceral senses, 159 Voltage clamping, 49–51 plasma volume pathway, 457f
Visual acuity, 134 circuit for squid giant axon, 49 thirst and salt appetite, 460–461
Visual agnosia, 121 Voltage-dependent Ca2+ channels, 673 gain and loss, normal routes, 439t
Visual evoked potential test, 112 Voltage-dependent K channels (KV), 49 insensible loss, 439
Visual field exam, 144 topology of monomer, 19f obligatory water loss, 440
Visual pathways, 139–141, 140f Voltage-dependent Na (NaV) channels, 19, 48 reabsorption, 439–440
optic pathways, effect of lesions in, 142 Voltage-gated channels, 213, 485 pathways for, 440f
macular sparing, 142 Voltage-sensitive Ca2+ channels (CaV), 19 Water-soluble vitamins, 429
primary visual cortex, 141–142 Voltage-sensitive channels, 11 assimilation, 590–591
Vital capacity (VC), 325, 332, 334 voltage-sensitive Ca2+ channels (CaV), 11, 19 vitamin B12 (cobalamin), 591
Vitamin A, 139 voltage-sensitive Na channels (NaV), 11, 19 vitamin C, 590
Vitamin B12, 507 role of, 47–49 digestion and absorption, 583
Vitamin D toxicity, 650 Voltage-sensitive Na channels (NaV), 11, 19 Weak acid, 375
Vitamins, 590 role of, 47–49 Weber and Schwabach tests, 154
homeostasis, 590 Voluntary movement, 167, 168f, 169 Wernicke’s area, 195
vitamin A, retinoic acid, 594 Voluntary muscle, 80, 85 White rami communicans, 178
vitamin B12, 507, 591 Vomiting, 156, 279, 379, 394, 474, 489, Willebrand factor, 208
deficiencies, 120 551–552, 563 Wirsung’s duct, 518
gastrointestinal absorption, 591f neural pathways, 552f Withdrawal reflex, 116
vitamin C, 590 (See also Ascorbic acid) v-SNARE synaptobrevin, 65 Wolff-Chaikoff effect, 636
vitamin D, 486, 643 V-type H+ pump, 23, 60 Working memory, 192–194
abnormal levels, 650 Work of breathing, 329, 748
active form, 398 W
analogs, 653 Wallerian degeneration, 66, 111 X
calciferol, 594 Warfarin, 262 Xenobiotics, 560
cellular effects of, 650 Wasting syndrome, 718
1,25-(OH)2D; calcitriol, 487 Water Z
insufficiency, 755 balance concept, regulation, 397–398 Zones of the lung, 346–348
metabolism and physiologic effects at, 649f diuresis, 444 Zone-specific, adrenal steroid hormone synthetic
production, regulation, 398 excretion, regulation, 458 pathway, 657f
synthesis and activation, 649–650 ADH secretion, baroreceptor control, Z score, 652
vitamin E, tocopherol, 594 459–460 Zymogen granules, 518
vitamin K, 594 ADH secretion, osmoreceptor control, 459
Vmax, 88, 96, 96f goals, 449
Voltage-activated L-type CaV channels, 54 mechanism, 459f, 460f

You might also like