Vous êtes sur la page 1sur 257

LWBK942-FM.

qxd 6/25/11 8:45 AM Page x


CARDIOVASCULAR PHYSIOLOGY CONCEPTS
SECOND EDITION

Klabunde_FM.indd i 6/11/2011 11:32:18 AM


Klabunde_FM.indd ii 6/11/2011 11:32:18 AM
CARDIOVASCULAR
PHYSIOLOGY CONCEPTS
SECOND EDITION

Richard E. Klabunde, Ph.D.


Associate Professor of Physiology

Department of Biomedical Sciences

Ohio University College of Osteopathic Medicine

Athens, Ohio

Klabunde_FM.indd iii 6/11/2011 11:32:18 AM


Acquisitions Editor: Crystal Taylor
Product Manager: Catherine Noonan
Marketing Manager: Joy Fisher-Williams
Vendor Manager: Bridgett Dougherty
Manufacturing Manager: Margie Orzech
Designer: Teresa Mallon
Compositor: SPi Global
Second Edition
Copyright © 2012 Lippincott Williams & Wilkins, a Wolters Kluwer business
351 West Camden Street Two Commerce Square
Baltimore, MD 21201 2001 Market Street
Philadelphia, PA 19103
First Edition, 2005
All rights reserved. This book is protected by copyright. No part of this book may be reproduced or transmitted in
any form or by any means, including as photocopies or scanned-in or other electronic copies, or utilized by any
information storage and retrieval system without written permission from the copyright owner, except for brief
quotations embodied in critical articles and reviews. To request permission, please contact Lippincott Williams &
Wilkins at Two Commerce Square, 2001 Market Street, Philadelphia, PA 19103, via email at permissions@lww.com
or via website at lww.com (products and services).
The publisher is not responsible (as a matter of product liability, negligence, or otherwise) for any injury
resulting from any material contained herein. This publication contains information relating to general principles
of medical care that should not be construed as specific instructions for individual patients. Manufacturers’ prod-
uct information and package inserts should be reviewed for current information, including contraindications,
dosages, and precautions.
Printed in China

Library of Congress Cataloging-in-Publication Data


Klabunde, Richard E.
Cardiovascular physiology concepts / Richard E. Klabunde. — 2nd ed.
p. ; cm.
Includes bibliographical references and index.
ISBN 978-1-4511-1384-6
1. Cardiovascular system—Physiology. I. Title.
[DNLM: 1. Cardiovascular Physiological Phenomena. 2. Heart Diseases—physiopathology. WG 102]
QP101.K553 2012
612.1—dc23
2011014577

DISCLAIMER
Care has been taken to confirm the accuracy of the information present and to describe generally accepted prac-
tices. However, the authors, editors, and publisher are not responsible for errors or omissions or for any con-
sequences from application of the information in this book and make no warranty, expressed or implied, with
respect to the currency, completeness, or accuracy of the contents of the publication. Application of this informa-
tion in a particular situation remains the professional responsibility of the practitioner; the clinical treatments
described and recommended may not be considered absolute and universal recommendations.
The authors, editors, and publisher have exerted every effort to ensure that drug selection and dosage set forth
in this text are in accordance with the current recommendations and practice at the time of publication. However,
in view of ongoing research, changes in government regulations, and the constant flow of information relating to
drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any change in
indications and dosage and for added warnings and precautions. This is particularly important when the recom-
mended agent is a new or infrequently employed drug.
Some drugs and medical devices presented in this publication have Food and Drug Administration (FDA)
clearance for limited use in restricted research settings. It is the responsibility of the health care provider to ascer-
tain the FDA status of each drug or device planned for use in their clinical practice.
To purchase additional copies of this book, call our customer service department at (800) 638-3030 or fax orders
to (301) 223-2320. International customers should call (301) 223-2300.
Visit Lippincott Williams & Wilkins on the Internet: http://www.lww.com. Lippincott Williams & Wilkins cus-
tomer service representatives are available from 8:30 am to 6:00 PM, EST.
9 8 7 6 5 4 3 2 1

Klabunde_FM.indd iv 6/11/2011 11:32:18 AM


~REEACE

Cardiovascular physiology textbooks have The first eight chapters discuss cardio-
traditionally emphasized biophysical princi- vascular physiology following a traditional
ples such as the behavior of flowing blood, organization of topics. The last chapter inte-
mechanics of muscle contraction, and feed- grates the material in the preceding chapters
back control systems. In the past two dec- by describing how the cardiovascular system
ades, we have gained considerable knowledge responds and adapts to increased demands by
about endothelial function, membrane recep- the body (e.g., exercise and pregnancy) or to
tors, ion channels, and signal transduction pathophysiologic conditions (e.g., hypoten-
mechanisms that regulate cardiac and vas- sion, hypertension, heart failure, and cardiac
cular function. This new insight into cellu- valve disease).
lar mechanisms has revolutionized not only Although the basic format of the second
our understanding of cardiovascular function edition is similar to the first edition, many
but also how physicians diagnose and treat chapter sections have been rewritten to
patients with cardiovascular disease. Cardio- enhance clarity and to update our knowledge
vascular Physiology Concepts was written to on specific topics. More than half the figures
provide medical, graduate, and allied health have been revised or are new in the second
science students with a firm foundation in edition. Much of the material that was for-
traditional biophysical principles and newer merly found in an accompanying CD-ROM
cellular physiology principles. has now been revised and incorporated into
This textbook incorporates several features the printed second edition.
to aid the reader in learning: (1) each chap- Cardiovascular physiology, like all areas
ter begins with a list of learning objectives to of biomedical science, can be overwhelming
direct the reader to key concepts, (2) the text is in the amount of knowledge presented to the
supplemented with problems and clinical cases reader. For this reason, I have endeavored to
used to reinforce fundamental physiological present fundamental concepts at a level suit-
concepts, (3) important concepts are summa- able for medical students in their preclini-
rized at the end of each chapter, (4) relevant cal years of training. These concepts will be
reading resources are listed in the chapters, and more than sufficient to provide a necessary
(5) review questions with explanations are pro- framework for understanding cardiovascu-
vided as a self-assessment tool for the reader. lar pharmacology and therapeutics, and car-
Many topics presented in this textbook diovascular pathophysiology. It is my hope
are placed in a medical context by describing that the reader will not only learn how the
how underlying physiologic concepts relate to cardiovascular system functions but will
disease states, such as arrhythmia, abnormal also become awed at the magnificence of the
blood pressure, and heart failure, and to clini- human body.
cal diagnosis and therapeutic intervention.
Several of the chapters contain clinical cases Richard E. Klabunde, PhD
to illustrate clinical applications of important Athens, Ohio
physiologic concepts.

Klabunde_FM.indd v 6/11/2011 11:32:18 AM


ACKNOWLEDGMENTS

I want to acknowledge the inspiration I received from my graduate advisor, Paul C. johnson,
who taught me by his example to strive for excellence in both teaching and research. I am also
grateful to the other physiology faculty at the University of Arizona in the early 1970s for their
contagious love and enthusiasm for physiology. Feedback from medical students I have taught
for more than 30 years has been invaluable in stimulating me to explore new ways to more effec-
tively teach cardiovascular physiology. I appreciate the helpful suggestions from those who criti-
cally reviewed the first edition. These individuals offered many valuable comments that served
to enrich the content and format of this second edition. I also want to thank all the talented
people at Lippincott Williams &: Wilkins who have worked with me on this textbook. Special
gratitude is reserved for my loving and patient wife Karen, our four sons, and my parents who
always encouraged me to pursue my dreams. Finally, I want to thank God for enabling me to
fulfill my dreams.

Richard E. Klabunde, PhD


Athens, Ohio

vi

Klabunde_FM.indd vi 6/11/2011 11:32:19 AM


CONTENTS

Preface ......................................................................................................................................... v
Acknowledgments .................................................................................................................. vi

1 Introduction to the Cardiovascular System ................................................ 1


THE NEED FOR A CIRCULATORY SYSTEM ................................................................... 1
THE ARRANGEMENT OF THE CARDIOVASCULAR SYSTEM ................................ 2
THE FUNCTIONS OF THE HEART AND BLOOD VESSELS .................................. ..4
Heart .......................................................................................................................................4
Vascular System ................................................................................................................. 5
Interdependence of Circulatory and Organ Function ......................................... 5
THE REGULATION OF CARDIAC AND VASCULAR FUNCTION ........................... 5
THE CONTENT OF THE FOLLOWING CHAPTERS .................................................... 6
SUMMARY OF IMPORTANT CONCEPTS ........................................................................ 7
REVIEW QUESTIONS ............................................................................................................. 7
ANSWERS TO REVIEW QUESTIONS .............................................................................. 8

2 Electrical Activity of the Heart ............................................................................. 9


INTRODUCTION ..................................................................................................................... 10
CELL MEMBRANE POTENTIALS ..................................................................................... 10
Resting Membrane Potentials ..................................................................................... 10
Maintenance of Ionic Gradients ..................................................................................12
lon Channels ...................................................................................................................... 14
Action Potentials .............................................................................................................. 16
Arrhythmias Caused by Abnormal Action Potential Generation ................... 21
CONDUCTION OF ACTION POTENTIALS WITHIN THE HEART .........................21
Electrical Conduction within the Heart.. ..................................................................21
Regulation of Conduction Velocity .......................................................................... 23
Abnormal Conduction ................................................................................................... 24
Tachycardia Caused by Reentry ............................................................................... 24
THE ELECTROCARDIOGRAM ......................................................................................... 26
ECG Tracing ...................................................................................................................... 26
Interpretation of Normal and Abnormal Cardiac Rhythms
from the ECG ................................................................................................................ 28
Volume Conductor Principles and ECG Rules of Interpretation .................. 30
ECG Leads: Placement of Recording Electrodes ............................................... 32
ELECTROPHYSIOLOGICAL CHANGES DURING CARDIAC ISCHEMIA ........... 35
SUMMARY OF IMPORTANT CONCEPTS ..................................................................... 36
REVIEW QUESTIONS ...........................................................................................................37
ANSWERS TO REVIEW QUESTIONS ........................................................................... 38
ANSWERS TO PROBLEMS AND CASES ..................................................................... 39
SUGGESTED RESOURCES ................................................................................................40

vii

Klabunde_FM.indd vii 6/11/2011 11:32:19 AM


viii CONTENTS

3 Cellular Structure and Function.........................................................................41


INTRODUCTION ....................................................................................................................41
CARDIAC CELL STRUCTURE AND FUNCTION........................................................41
Myocytes and Sarcomeres ..........................................................................................41
Excitation–Contraction Coupling ..............................................................................43
Regulation of Contraction (Inotropy) .....................................................................45
Regulation of Relaxation (Lusitropy)......................................................................48
Cardiac Myocyte Metabolism .....................................................................................49
VASCULAR STRUCTURE AND FUNCTION ................................................................49
Vascular Smooth Muscle Cells ...................................................................................50
Vascular Endothelial Cells ...........................................................................................54
SUMMARY OF IMPORTANT CONCEPTS .....................................................................55
REVIEW QUESTIONS ..........................................................................................................56
ANSWERS TO REVIEW QUESTIONS ...........................................................................57
ANSWERS TO PROBLEMS AND CASES .....................................................................58
SUGGESTED RESOURCES ................................................................................................59

4 Cardiac Function............................................................................................................60
INTRODUCTION ....................................................................................................................60
CARDIAC ANATOMY...........................................................................................................60
Functional Anatomy of the Heart ............................................................................60
Autonomic Innervation .................................................................................................61
THE CARDIAC CYCLE.........................................................................................................62
Cardiac Cycle Diagram .................................................................................................62
Phase 1. Atrial Systole ...................................................................................................62
Phase 2. Isovolumetric Contraction .........................................................................64
Phase 3. Rapid Ejection................................................................................................65
Phase 4. Reduced Ejection .........................................................................................65
Phase 5. Isovolumetric Relaxation ...........................................................................65
Phase 6. Rapid Filling ....................................................................................................66
Phase 7. Reduced Filling ..............................................................................................66
Summary of Intracardiac Pressures .........................................................................66
Ventricular Pressure–Volume Relationship ...........................................................67
CARDIAC OUTPUT...............................................................................................................67
Measurement of Cardiac Output ..............................................................................69
Influence of Heart Rate and Stroke Volume on Cardiac Output .................69
EFFECTS OF PRELOAD ON STROKE VOLUME .......................................................69
Effects of Ventricular Compliance on Preload ....................................................69
Effects of Preload on Tension Development
(Length–Tension Relationship) ..............................................................................71
Effects of Venous Return on Stroke Volume
(Frank-Starling Mechanism) ...................................................................................74
Factors Determining Ventricular Preload ..............................................................75
EFFECTS OF AFTERLOAD ON STROKE VOLUME .................................................77
Effects of Afterload on the Velocity of Fiber Shortening
(Force–Velocity Relationship) ................................................................................77
Effects of Afterload on Frank-Starling Curves ....................................................79
Effects of Afterload on Pressure–Volume Loops ...............................................79

Klabunde_FM.indd viii 6/11/2011 11:32:20 AM


CONTENTS ix

EFFECTS OF INOTROPY ON STROKE VOLUME .....................................................80


Effects of Inotropy on Length–Tension Relationship........................................80
Effects of Inotropy on Force–Velocity Relationship .........................................81
Effects of Inotropy on Frank-Starling Curves......................................................81
Effects of Inotropy on Pressure–Volume Loops .................................................81
Factors Influencing Inotropic State .........................................................................82
Cellular Mechanisms of Inotropy ..............................................................................82
INTERDEPENDENCE OF PRELOAD, AFTERLOAD, AND INOTROPY..............83
MYOCARDIAL OXYGEN CONSUMPTION ...................................................................84
How Myocardial Oxygen Consumption is Determined ....................................85
Factors Influencing Myocardial Oxygen Consumption ....................................86
SUMMARY OF IMPORTANT CONCEPTS .....................................................................87
REVIEW QUESTIONS ..........................................................................................................88
ANSWERS TO REVIEW QUESTIONS ...........................................................................89
ANSWERS TO PROBLEMS AND CASES .....................................................................91
SUGGESTED RESOURCES ................................................................................................92

5 Vascular Function..........................................................................................................93
INTRODUCTION ....................................................................................................................93
ANATOMY AND FUNCTION .............................................................................................93
Vascular Network ............................................................................................................93
Distribution of Pressures and Volumes ..................................................................95
ARTERIAL BLOOD PRESSURE .......................................................................................97
Mean Arterial Pressure ..................................................................................................97
Aortic Pulse Pressure ....................................................................................................98
HEMODYNAMICS (PRESSURE, FLOW, AND RESISTANCE) ................................100
Effects of Vessel Length, Radius, and Blood Viscosity on
Resistance to Blood Flow ........................................................................................100
Laminar versus Turbulent Flow .................................................................................102
Series and Parallel Arrangement of the Vasculature ........................................103
REGULATION OF SYSTEMIC VASCULAR RESISTANCE .......................................106
Calculation of Systemic Vascular Resistance ......................................................106
Vascular Tone ...................................................................................................................107
VENOUS BLOOD PRESSURE...........................................................................................107
Venous Blood Volume and Compliance ................................................................107
Mechanical Factors Affecting Central Venous Pressure and
Venous Return..............................................................................................................109
Summary of Factors Affecting Central Venous Pressure ...............................112
VENOUS RETURN AND CARDIAC OUTPUT .............................................................113
The Balance between Venous Return and Cardiac Output ...........................113
Systemic Vascular Function Curves ........................................................................113
Cardiac Function Curves..............................................................................................116
Interactions between Cardiac and Systemic Vascular
Function Curves ..........................................................................................................116
SUMMARY OF IMPORTANT CONCEPTS .....................................................................118
REVIEW QUESTIONS ..........................................................................................................119
ANSWERS TO REVIEW QUESTIONS ...........................................................................121
ANSWERS TO PROBLEMS AND CASES .....................................................................122
SUGGESTED RESOURCES ................................................................................................123

Klabunde_FM.indd ix 6/11/2011 11:32:20 AM


x CONTENTS

6 Neurohumoral Control of the Heart and Circulation .........................124


INTRODUCTION ....................................................................................................................124
AUTONOMIC NEURAL CONTROL .................................................................................124
Autonomic Innervation of the Heart and Vasculature .....................................124
Baroreceptor Feedback Regulation of Arterial Pressure ................................130
Chemoreceptors ..............................................................................................................134
Other Autonomic Reflexes Affecting the Heart and Circulation .................135
HUMORAL CONTROL .........................................................................................................135
Circulating Catecholamines ........................................................................................136
Renin-Angiotensin-Aldosterone System ................................................................137
Atrial Natriuretic Peptide .............................................................................................139
Vasopressin (Antidiuretic Hormone).......................................................................140
INTEGRATION OF NEUROHUMORAL MECHANISMS............................................141
SUMMARY OF IMPORTANT CONCEPTS .....................................................................142
REVIEW QUESTIONS ..........................................................................................................143
ANSWERS TO REVIEW QUESTIONS ...........................................................................145
ANSWERS TO PROBLEMS AND CASES .....................................................................146
SUGGESTED RESOURCES ................................................................................................147

7 Organ Blood Flow.........................................................................................................148


INTRODUCTION ....................................................................................................................148
DISTRIBUTION OF CARDIAC OUTPUT .......................................................................148
LOCAL REGULATION OF BLOOD FLOW ...................................................................149
Tissue Factors ..................................................................................................................149
Endothelial Factors ........................................................................................................152
Smooth Muscle (Myogenic) Mechanisms ..............................................................153
Extravascular Compression.........................................................................................153
Autoregulation of Blood Flow ...................................................................................154
Reactive and Active Hyperemia................................................................................156
SPECIAL CIRCULATIONS ..................................................................................................157
Coronary Circulation......................................................................................................157
Cerebral Circulation .......................................................................................................161
Skeletal Muscle Circulation .........................................................................................164
Cutaneous Circulation ...................................................................................................168
Splanchnic Circulation ..................................................................................................169
Renal Circulation .............................................................................................................170
Pulmonary Circulation...................................................................................................173
Summary of Special Circulations..............................................................................175
SUMMARY OF IMPORTANT CONCEPTS .....................................................................176
REVIEW QUESTIONS ..........................................................................................................176
ANSWERS TO REVIEW QUESTIONS ...........................................................................178
ANSWERS TO PROBLEMS AND CASES .....................................................................179
SUGGESTED RESOURCES ................................................................................................179

8 Exchange Function of the Microcirculation..............................................180


INTRODUCTION ....................................................................................................................180
MECHANISMS OF EXCHANGE........................................................................................180
Diffusion .............................................................................................................................181
Bulk Flow ...........................................................................................................................182
Vesicular and Active Transport .................................................................................182

Klabunde_FM.indd x 6/11/2011 11:32:20 AM


CONTENTS xi

EXCHANGE OF OXYGEN AND CARBON DIOXIDE ................................................182


Oxygen Diffusion ............................................................................................................182
Oxygen Delivery and Extraction ...............................................................................183
Carbon Dioxide Diffusion.............................................................................................186
TRANSCAPILLARY FLUID EXCHANGE .......................................................................186
Physical Mechanisms Governing Fluid Exchange ..............................................187
Capillary Exchange Model ...........................................................................................192
EDEMA FORMATION...........................................................................................................193
SUMMARY OF IMPORTANT CONCEPTS .....................................................................194
REVIEW QUESTIONS ..........................................................................................................194
ANSWERS TO REVIEW QUESTIONS ...........................................................................195
ANSWERS TO PROBLEMS AND CASES .....................................................................196
SUGGESTED RESOURCES ................................................................................................197

9 Cardiovascular Integration, Adaptation,


and Pathophysiology ..................................................................................................198
INTRODUCTION ....................................................................................................................198
CARDIOVASCULAR RESPONSES TO EXERCISE ...................................................198
Mechanisms Involved in Cardiovascular Response to Exercise....................199
Steady-State Changes in Cardiovascular Function during Exercise ..........201
Factors Influencing Cardiovascular Response to Exercise .............................203
MATERNAL CHANGES IN CARDIOVASCULAR FUNCTION DURING
PREGNANCY ......................................................................................................................205
HYPOTENSION ......................................................................................................................206
Causes of Hypotension.................................................................................................206
Compensatory Mechanisms during Hypotension ..............................................207
Decompensatory Mechanisms Following Severe and Prolonged
Hypotension ..................................................................................................................210
Physiologic Basis for Therapeutic Intervention ..................................................212
HYPERTENSION ....................................................................................................................212
Essential (Primary) Hypertension.............................................................................213
Secondary Hypertension .............................................................................................214
Physiologic Basis for Therapeutic Intervention ..................................................215
HEART FAILURE ...................................................................................................................216
Causes of Heart Failure ................................................................................................216
Systolic versus Diastolic Dysfunction .....................................................................217
Systemic Compensatory Mechanisms in Heart Failure ....................................219
Exercise Limitations Imposed by Heart Failure ..................................................221
Physiologic Basis for Therapeutic Intervention ..................................................222
VALVE DISEASE ....................................................................................................................223
Valve Stenosis ..................................................................................................................223
Valve Regurgitation .......................................................................................................226
SUMMARY OF IMPORTANT CONCEPTS .....................................................................229
REVIEW QUESTIONS ..........................................................................................................229
ANSWERS TO REVIEW QUESTIONS ...........................................................................231
ANSWERS TO CASES .........................................................................................................233
SUGGESTED RESOURCES ................................................................................................234

Index ..........................................................................................................................................235

Klabunde_FM.indd xi 6/11/2011 11:32:20 AM


Klabunde_FM.indd xii 6/11/2011 11:32:20 AM
INTRODUCTION TO 0

1
:I:
>
THE CARDIOVASCULAR 'tl
-1
m
;c
SYSTEM
Understanding the concepts presented in this chapter will enable the student to:
1. Explain why large organisms require a circulatory system, while single-cell and
small multicellular organisms do not.
2. Explain the significance of the series and parallel arrangement of the
cardiac chambers, pulmonary circulation, and major organs of the systemic
circulation .

3. Describe the pathways for the flow of blood through the heart chambers and
large vessels associated with the heart.
4. Explain the importance of negative feedback systems for the control of arterial
blood pressure.

THE NEED FOR A CIRCULATORY capillaries around cells ensures that exchange
SYSTEM can occur between blood and surrounding
cells.
All living cells require metabolic substrates Exchange between blood and the outside
(e.g., oxygen, amino acids, glucose) and a environment occurs in several different organs:
mechanism by which they can remove by- lungs, gastrointestinal tract, kidneys, and skin.
products of metabolism (e.g., carbon dioxide, As blood passes through the lungs, oxygen
lactic acid). Single-cell organisms exchange and carbon dioxide are exchanged between
these substances directly with their environ- the blood in the pulmonary capillaries and the
ment through diffusion and cellular transport gases found within the lung alveoli. Oxygen-
systems. In contrast, most cells of large organ- enriched blood is then transported to the organs
isms have limited or no exchange capacity where the oxygen diffuses from the blood into
with their environment because their cells are the surrounding cells. At the same time, carbon
not in contact with the outside environment. dioxide, a metabolic waste product, diffuses
Nevertheless, exchange with the outside from the tissue cells into the blood and is trans-
environment must occur for the cells to func- ported to the lungs, where exchange occurs
tion. To accomplish this necessary exchange, between blood and alveolar gases.
large organisms have a sophisticated system Blood passing through the intestine picks
of blood vessels that facilitates the exchange up glucose, amino acids, fatty acids, and
of substances between cells and blood and other ingested substances that have been
between blood and environment. The small- transported from the intestinal lumen into
est of these blood vessels, capillaries, are in the blood in the intestinal wall by the cells
close proximity to all cells in the body, thereby lining the intestine. The blood then delivers
permitting exchange to occur. For example, these substances to organs such as the liver
each cell in skeletal muscle is surrounded by for additional metabolic processing and to
two or more capillaries. This arrangement of cells throughout the body as an energy source.

Klabunde_Chap01.indd 1 6/10/2011 8:42:15 PM


2 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Some of the waste products of these cells are THE ARRANGEMENT OF THE
taken up by the blood and transported to CARDIOVASCULAR SYSTEM
other organs for metabolic processing and
final elimination into the outside environ- The cardiovascular system has two primary
ment through either the gastrointestinal tract components: the heart and blood vessels. A
or the kidneys. third component, the lymphatic system, does
Cells require a proper balance of water and not contain blood, but nonetheless serves an
electrolytes (e.g., sodium, potassium, and important exchange function in conjunction
calcium) to function. The circulation trans- with blood vessels.
ports ingested water and electrolytes from The heart can be viewed functionally as
the intestine to cells throughout the body, two pumps with the pulmonary and systemic
including those of the kidneys, where exces- circulations situated between the two pumps
sive amounts of water and electrolytes can be (Fig. 1.1). The pulmonary circulation is the
eliminated in the urine. blood flow within the lungs that is involved
The skin also serves as a site for exchange in the exchange of gases between the blood
of water and electrolytes (through sweating), and alveoli. The systemic circulation is com-
and for exchange of heat, which is a major by- prised of all the blood vessels within and out-
product of cellular metabolism that must be side of organs excluding the lungs. The right
removed from the body. Blood flow through side of the heart comprises the right atrium
the skin regulates heat loss from the body. and the right ventricle. The right atrium
In summary, the ultimate purpose of the receives venous blood from the systemic cir-
cardiovascular system is to facilitate exchange culation, and the right ventricle pumps it into
of gases, fluid, electrolytes, large molecules, the pulmonary circulation where oxygen and
and heat between cells and the outside envi- carbon dioxide are exchanged between the
ronment. The heart and vasculature ensure blood and alveolar gases. The left side of the
that adequate blood flow is delivered to organs heart comprises the left atrium and the left
so that this exchange can take place. ventricle. The blood leaving the lungs enters

PA Ao
RA
LA

RV LV
Pulmonary
Circulation

Systemic Circulation
■ FIGURE 1.1 Overview of the cardiovascular system. The right side of the heart, pulmonary circulation,
left side of the heart, and systemic circulation are arranged in series. RA, right atrium; RV, right ventricle;
PA, pulmonary artery; Ao, aorta; LA, left atrium; LV, left ventricle.

Klabunde_Chap01.indd 2 6/10/2011 8:42:18 PM


CHAPTER 1 • INTRODUCTION TO THE CARDIOVASCULAR SYSTEM 3

the left atrium by way of the pulmonary


veins. Blood then flows from the left atrium
into the left ventricle. The left ventricle ejects
Head
the blood into the aorta, which then distrib-
utes the blood to all the organs via the arterial
system. Within the organs, the vasculature
branches into smaller and smaller vessels, Lungs
eventually forming capillaries, which are the
primary site of exchange. Blood flow from the SVC
capillaries enters veins, which return blood Aorta
flow to the right atrium via large systemic
veins (the superior and inferior vena cava). IVC
As blood flows through organs, some of
the fluid, along with electrolytes and small
amounts of protein, leaves the circulation and Arms
enters the tissue interstitium (a process termed
fluid filtration). The lymphatic vessels, which
are closely associated with small blood vessels
within the tissue, collect the excess fluid from Liver GI
within the tissue interstitium and transport
it back into the venous circulation by way of
lymphatic ducts that empty into large veins
(subclavian veins) above the right atrium. Kidneys
It is important to note the overall arrange-
ment of the cardiovascular system. First, the
right and left sides of the heart, which are sepa-
rated by the pulmonary and systemic circula- Legs
tions, are in series with each other (see Fig. 1.1).
Therefore, all of the blood that is pumped from ■ FIGURE 1.2 Parallel arrangement of organs
the right ventricle enters into the pulmonary within the body. One major exception is the
circulation and then into the left side of the hepatic (liver) circulation, which receives blood
flow from the hepatic portal veins of the gastroin-
heart from where it is pumped into the systemic testinal (GI) circulation (series) and from the aorta
circulation before returning to the heart. This via the hepatic artery (parallel). SVC, superior vena
in-series relationship of the two sides of the cava; IVC, inferior vena cava.
heart and the pulmonary and systemic circula-
tions requires that the output (volume of blood hepatic portal system to supply the liver. The
ejected per unit time) of each side of the heart liver also receives blood from the aorta via the
closely matches the output of the other so that hepatic artery. Therefore, most of the liver circu-
there are no major blood volume shifts between lation is in series with the intestinal circulation,
the pulmonary and systemic circulations. Sec- while some of the liver circulation is in parallel
ond, most of the major organ systems of the with the intestinal circulation (see Chapter 7).
body receive their blood from the aorta, and The parallel arrangement has significant
the blood leaving these organs enters into the hemodynamic implications as described in
venous system (superior and inferior vena cava) Chapter 5. Briefly, the parallel arrangement of
that returns the blood to the heart. Therefore, major vascular beds prevents blood flow changes
the circulations of most major organ systems are in one organ from significantly affecting blood
in parallel as shown in Figure 1.2. One major flow in other organs. In contrast, when vascular
exception is the liver, which receives a large frac- beds are in series, blood flow changes in one
tion of its blood supply from the venous circula- vascular bed significantly alter blood flow to
tion of the intestinal tract that drains into the the other vascular bed.

Klabunde_Chap01.indd 3 6/10/2011 8:42:18 PM


4 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

THE FUNCTIONS OF THE HEART Ao Lungs


AND BLOOD VESSELS SVC
Heart
The heart sometimes is thought of as an organ LA
PA
that pumps blood through the organs of the RA
body. While this is true, it is more accurate to
view the heart as a pump that receives blood
from venous blood vessels at a low pressure, LV
imparts energy to the blood (raises it to a
IVC
higher pressure) by contracting around the
RV
blood within the cardiac chambers, and then
ejects the blood into the arterial blood vessels.
It is important to understand that organ
blood flow is not driven by the output of the
heart per se, but rather by the pressure gen-
erated within the arterial system as the heart
pumps blood into the vasculature, which ■ FIGURE 1.3 Blood flow within the heart. Venous
blood returns to the right atrium (RA) via the supe-
serves as a resistance network. Organ blood flow rior (SVC) and inferior vena cava (IVC). Blood passes
is determined by the arterial pressure minus the from the RA into the right ventricle (RV), which
venous pressure, divided by the vascular resistance ejects the blood into the pulmonary artery (PA).
of the organ (see Chapters 5 and 7). Pressures After passing through the lungs, the blood flows
into the left atrium (LA) and then fills the left ventri-
in the cardiovascular system are expressed in cle (LV), which ejects the blood into the aorta (Ao)
millimeters of mercury (mm Hg) above atmos- for distribution to the different organs of the body.
pheric pressure. One millimeter of mercury is
the pressure exerted by a 1-mm vertical col- ventricle. As the left ventricle contracts and
umn of mercury (1 mm Hg is the equivalent ejects blood into the systemic arterial system,
of 1.36 cm H2O hydrostatic pressure). Vascular a relatively high pressure is generated (100
resistance is determined by the size of blood to 140 mm Hg maximal or systolic pressure).
vessels, the anatomical arrangement of the vas- Therefore, the left ventricle is a high-pressure
cular network, and the viscosity of the blood pump, in contrast to the right ventricle, which is
flowing within the vasculature. a low-pressure pump. Details of the pumping
The right atrium receives systemic venous action of the heart are found in Chapter 4.
blood (venous return) at very low pressures The pumping activity of the heart is
(near 0 mm Hg) (Fig. 1.3). This venous return usually expressed in terms of its cardiac out-
then passes through the right atrium and fills put, which is the amount of blood ejected
the right ventricle; atrial contraction also with each contraction (i.e., stroke volume)
contributes to the ventricular filling. Right multiplied by the heart rate. Any factor that
ventricular contraction ejects blood from alters heart rate or stroke volume will alter the
the right ventricle into the pulmonary artery. cardiac output. The heart rate is determined
This generates a maximal pressure (systolic by specialized cells within the heart that act
pressure) that ranges from 20 to 30 mm Hg as electrical pacemakers, and their activity is
within the pulmonary artery. As the blood increased or decreased by autonomic nerves
passes through the pulmonary circulation, the and hormones (see Chapter 2). The action
blood pressure falls to about 10 mm Hg. The potentials generated by these pacemaker
left atrium receives the pulmonary venous cells are conducted throughout the heart and
blood, which then flows passively into the trigger contraction of cardiac myocytes (see
left ventricle; atrial contraction provides a Chapter 3). This results in ventricular con-
small amount of additional filling of the left traction and ejection of blood. The force of

Klabunde_Chap01.indd 4 6/10/2011 8:42:18 PM


CHAPTER 1 • INTRODUCTION TO THE CARDIOVASCULAR SYSTEM 5

ventricular contraction, and therefore stroke to the kidneys can have detrimental effects on
volume, is regulated by mechanisms intrinsic kidney function and therefore on fluid and
to the heart, by autonomic nerves and hor- electrolyte balance in the body. Furthermore,
mones (see Chapters 3, 4, and 6). renal dysfunction can lead to large increases
The heart has other important functions in blood volume, which can precipitate cardi-
besides pumping blood. The heart synthesizes ovascular changes that can lead to hyperten-
several hormones. One of these hormones, sion or exacerbate heart failure. In summary,
atrial natriuretic peptide, plays an impor- organ function is dependent on the circula-
tant role in the regulation of blood volume tion of blood, and cardiovascular function is
and blood pressure (see Chapter 6). Sensory dependent on the function of organs.
nerve receptors associated with the heart play
a role in regulating the release of antidiuretic THE REGULATION OF CARDIAC
hormone from the posterior pituitary, which AND VASCULAR FUNCTION
regulates water loss by the kidneys.
The cardiovascular system must be able to
Vascular System adapt to changing conditions and demands of
the body. For example, when a person exer-
Blood vessels constrict and dilate to regulate
cises, increased metabolic activity of contract-
arterial blood pressure, alter blood flow within
ing skeletal muscle requires large increases
organs, regulate capillary blood pressure,
in nutrient supply (particularly oxygen) and
and distribute blood volume within the body.
enhanced removal of metabolic by-products
Changes in vascular diameters are brought
(e.g., carbon dioxide, lactic acid). To meet this
about by activation of vascular smooth muscle
demand, blood vessels within the exercising
within the vascular wall by autonomic nerves,
muscle dilate to increase blood flow; however,
metabolic and biochemical signals from out-
blood flow can only be increased if the arterial
side of the blood vessel, and vasoactive sub-
pressure is maintained. Arterial pressure is
stances released by endothelial cells that line
maintained during exercise by increasing car-
the blood vessels (see Chapters 3, 5, and 6).
diac output and by constricting blood vessels
Blood vessels have other functions besides
in other organs of the body (see Chapter 9).
distribution of blood flow and exchange. The
If these changes were not to occur, arterial
endothelium lining blood vessels produces
blood pressure would fall precipitously dur-
substances that modulate hemostasis (blood
ing exercise, thereby limiting organ perfusion
clotting) and inflammatory responses (see
and exercise capacity. Therefore, a coordi-
Chapter 3).
nated cardiovascular response is required to
permit increased muscle blood flow while a
Interdependence of Circulatory
person exercises. Another example of adapta-
and Organ Function
tion occurs when a person stands up. Gravi-
Cardiovascular function is closely linked to tational forces cause blood to pool in the legs
the function of other organs. For example, the when a person assumes an upright body pos-
brain not only receives blood flow to support ture (see Chapter 5). In the absence of regu-
its metabolism but also acts as a control center latory mechanisms, this pooling will lead to
for regulating cardiovascular function. A sec- a fall in cardiac output and arterial pressure,
ond example of the interdependence between which can cause a person to faint because
organ function and the circulation is the kid- of reduced blood flow to the brain. To pre-
ney. The kidneys excrete varying amounts of vent this from happening, coordinated reflex
sodium, water, and other molecules to main- responses increase heart rate and constrict
tain fluid and electrolyte homeostasis. Blood blood vessels to maintain a normal arterial
passing through the kidneys is filtered, and blood pressure when a person stands.
the kidneys then modify the composition of It is important to control arterial blood
the filtrate to form urine. Reduced blood flow pressure because it provides the driving

Klabunde_Chap01.indd 5 6/10/2011 8:42:18 PM


6 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

AP their actions on renal function. In contrast to


(+) the rapidly acting autonomic mechanisms,
ANS Kidneys hormonal mechanisms acting on the kidneys
require hours or days to achieve their full
Cardiac
effect on blood volume. Hormonal mecha-
Stimulation
Fast
nisms include secretion of catecholamines
Vascular (chiefly epinephrine) by the adrenal glands;
Constriction release of renin by the kidneys, which triggers
Slow the formation of angiotensin II and aldos-
Blood Volume terone; and release of antidiuretic hormone
■ FIGURE 1.4 Feedback control of arterial pres-
(vasopressin) by the posterior pituitary. Hor-
sure (AP) by the autonomic nervous system mones such as angiotensin II, aldosterone,
(ANS) and kidneys. A sudden fall in AP elicits a and vasopressin are particularly important
rapid baroreceptor reflex that activates the ANS because they act on the kidneys to increase
to stimulate the heart (increasing cardiac output)
and constrict blood vessels to restore AP. The
blood volume, which increases cardiac output
kidneys respond to decreased AP by retaining Na+ and arterial pressure.
and water to increase blood volume, which helps In summary, arterial pressure is monitored
to restore AP. The (+) indicates the restoration of by the body and ordinarily is maintained
arterial pressure following the initial fall in pressure
(i.e., a negative feedback response).
within narrow limits by negative feedback
mechanisms that adjust cardiac function,
systemic vascular resistance, and blood vol-
force for organ perfusion. As described in ume. This control is accomplished by changes
Chapter 6, neural and hormonal (neurohu- in autonomic nerve activity to the heart and
moral) mechanisms regulating cardiovascular vasculature, as well as by changes in circulat-
function are under the control of pressure sen- ing hormones that influence cardiac, vascular,
sors located in arteries and veins (i.e., barore- and renal function.
ceptors). These baroreceptors, through their
afferent neural connections to the brain, pro- THE CONTENT OF THE
vide the central nervous system with informa- FOLLOWING CHAPTERS
tion regarding the status of blood pressure in
the body. A decrease in arterial pressure from This textbook emphasizes our current knowl-
its normal operating point elicits a rapid baro- edge of cellular physiology as well as the
receptor reflex that stimulates the heart to classical biophysical concepts that have been
increase cardiac output and constricts blood used for decades to describe cardiac and vas-
vessels to restore arterial pressure (Fig. 1.4). cular function. Chapter 2 describes the elec-
These cardiovascular adjustments occur trical activity within the heart, both at the
through rapid changes in autonomic nerve cellular and whole organ level. Chapter 3
activity (particularly through sympathetic builds a foundation of cellular physiology by
nerves) to the heart and vasculature. Negative emphasizing intracellular mechanisms that
feedback control mechanisms, as this exam- regulate cardiac and vascular smooth mus-
ple illustrates, can be defined as a process in cle contraction. These cellular concepts are
which a deviation from some condition (e.g., reinforced repeatedly in subsequent chap-
normal arterial pressure) leads to responses ters. Chapter 4 examines cardiac mechanical
(e.g., cardiac stimulation and vasoconstric- function. Chapter 5 summarizes concepts of
tion) that diminish the deviation. vascular function and the biophysics of blood
In addition to altering autonomic nerve flow in the context of regulation of arterial
activity, a fall in arterial pressure stimulates and venous blood pressures. Neurohumoral
the release of hormones that help to restore mechanisms regulating cardiac and vascular
arterial pressure by acting on the heart and function are described in Chapter 6. Chapter
blood vessels; they also increase arterial 7 describes the flow of blood within different
pressure by increasing blood volume through organs, with an emphasis on local regulatory

Klabunde_Chap01.indd 6 6/10/2011 8:42:18 PM


CHAPTER 1 • INTRODUCTION TO THE CARDIOVASCULAR SYSTEM 7

mechanisms. Chapter 8 addresses the ultimate Chapter 9 integrates concepts described in


purpose of the cardiovascular system, that earlier chapters by examining how the cardio-
is, the exchange of nutrients, gases, and vascular system responds to altered demands
fluid between the blood and tissues. Finally; and disease states.

SUMMARY OF IMPORTANT CONCEPTS

• Large organisms require a circulatory within the organs serve as the primary
system so that metabolic substrates site of nutrient exchange.
and by-products of cellular metabolism • Blood flow within organs is determined
can be efficiently exchanged between primarily by the arterial pressure and
cells and the outside environment, as by changes in the diameters of blood
well as transported to distant sites vessels within the organs brought
within the body. about by contraction or relaxation of
• Venous blood returns to the right smooth muscle within the walls of the
side of the heart, which pumps the blood vessels.
blood into the pulmonary circulation • Most major organ systems are in parallel
where oxygen and carbon dioxide are with each other so that blood flow in
exchanged with the gases found within one organ has relatively little influence
the lung alveoli. Oxygenated blood on blood flow in another organ.
from the lungs enters the left side
of the heart, which pumps the blood • Negative feedback mechanisms such as
at high pressure into the aorta for the baroreceptor reflex, acting through
distribution to various organs via large autonomic nerves and circulating
distributing arteries. Small capillaries hormones, help to maintain normal
arterial pressure.

REVIEW QUESTIONS

For each question, choose the one best answer: c. The right ventricle generates higher
pressures than the left ventricle
1. The cardiovascular system during contraction.
a. Aids in the transfer of heat energy d. The right ventricle receives blood
from organs deep within the body to from the pulmonary veins.
the outside environment.
b. Comprises pulmonary and systemic 3. A patient complains of becoming "light
circulations that are in parallel with headed" when he is standing upright.
each other. Blood pressure measurements reveal a
c. Transports carbon dioxide from the significant fall in arterial pressure upon
lungs to tissues within organs. standing. Which of the following is a
d. Transports oxygen from individual likely explanation of this patient's
cells to the lungs. condition?
a. Excessive activation of baroreceptor
2. Which of the following statements con- negative feedback mechanisms
cerning the heart is true? b. Excessive fluid retention by the
a. Cardiac output is the product of ven- kidneys
tricular stroke volume and heart rate. c. Increased heart rate
b. The right and left ventricles are in d. Reduced cardiac output
parallel.

Klabunde_Chap01.indd 7 6/10/2011 8:42:18 PM


8 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

1. The correct answer is "a" because blood generates much higher pressures than
flow carries heat from the deep organs the right ventricle during contraction.
within the body to the skin where the Choice "d" is incorrect because the pul-
heat energy can be given off to the monary veins empty into the left atrium.
environment. Choice "b" is incorrect 3. The correct answer is "d" because when
because the pulmonary and systemic a person stands up, blood pools in the
circulations are in series. Choice "c" legs, which reduces the filling of the
is incorrect because carbon dioxide heart and leads to a fall in cardiac out-
is transported from the tissues to the put and arterial pressure, and a decrease
lungs. Choice "d" is incorrect because in brain blood flow. Choice "a" is incor-
blood transports oxygen from the lungs rect because activation of baroreceptor
to the tissues. negative feedback mechanisms ordinar-
2. The correct answer is "a" because when ily helps to maintain arterial pressure
the volume per beat (stroke volume) is when standing. Choice "b" is incorrect
multiplied by the number of beats per because increased fluid retention by
minute (heart rate), the units become the kidneys elevates cardiac output and
volume per minute, which is the flow arterial pressure. Choice "c" is incor-
out of the heart (cardiac output). Choice rect because increased heart rate when
"b" is incorrect because the right and standing, which is brought about by the
left ventricles are in series. Choice "c" baroreceptor reflex, helps to maintain
is incorrect because the left ventricle cardiac output and arterial pressure.

Klabunde_Chap01.indd 8 6/10/2011 8:42:21 PM


n

2
:I:
>
ELECTRICAL ACTIVITY "tJ
-1
m
::0
OE II--IE I--IEARI
Understanding the concepts presented in this chapter will enable the student to:
1. Describe how changing the concentrations of sodium, potassium, and calcium ions
inside and outside the cell affect the resting membrane potential in cardiac cells.
2. Explain why the resting potential is near the equilibrium potential for potassium
and the peak of an action potential approaches the equilibrium potential for
sodium.
3. Describe the mechanisms that maintain ion concentration gradients across the
cardiac cell membrane.
4. Describe the role of voltage-gated Na+, K+, and ca++ channels in the generation
of action potentials in pacemaker and nonpacemaker cells of the heart.
5. Describe how autonomic nerves, circulating catecholamines, extracellular
potassium concentrations, thyroid hormone, and hypoxia alter pacemaker activity.
6. Describe the role of afterdepolarizations and reentry in the generation of
tachycardias.
7. Describe the normal pathways for action potential conduction within the heart
and how autonomic nerves, circulating catecholamines, and cellular hypoxia
alter conduction velocity within the heart.
8. Describe what each of the waves, intervals, and segments of a normal
electrocardiogram (ECG) tracing represents.
9. Recognize the following from an ECG rhythm strip:
a. Normal sinus rhythm
b. Sinus bradycardia and tachycardia
c. Atrial flutter and fibrillation
d. Atrioventricular (AV) blocks: first, second, and third degree
e. Premature ventricular complex
f. Ventricular tachycardia and fibrillation
10. Describe the location for placement of electrodes for each of the following
leads: I, II, Ill, aVR, aVL, and aVF, and precordial v, to V 6 •
11. Draw the axial reference system and show the position (in degrees) for the
positive electrode for each of the six limb leads.
12. Describe, in terms of vectors, how the QRS complex is generated and why the
QRS appears differently when recorded by different electrode leads.
13. Estimate the mean electrical axis for ventricular depolarization from the six
limb leads.

Klabunde_Chap02.indd 9 6/11/2011 10:28:32 AM


10 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

INTRODUCTION Myocyte
The primary function of cardiac myocytes
is to contract. Electrical changes within the
myocytes initiate this contraction. This chap-
ter examines (1) the electrical activity of indi-
K+
vidual myocytes, including resting membrane (150 mM) K+
potentials and action potentials; (2) the way (4 mM)
action potentials are conducted throughout -
the heart to initiate coordinated contraction
of the entire heart; and (3) the way electri-
Pr
Na+
cal activity of the heart is measured using the Na+ (145 mM)
electrocardiogram (ECG). (20 mM)

CELL MEMBRANE POTENTIALS Ca++


Ca++ (2.5 mM)
Resting Membrane Potentials (0.0001 mM)
Cardiac cells, like all living cells in the body,
have an electrical potential across the cell mem-
brane. This potential can be measured by insert-
-90 mV
ing a microelectrode into the cell and measuring
the electrical potential in millivolts (mV) inside ■ FIGURE 2.1 Concentrations of K+, Na+, and Ca++
the cell relative to the outside of the cell. By inside and outside a cardiac myocyte at a resting
membrane potential of −90 mV. Pr−, negatively
convention, the outside of the cell is considered charged proteins.
0 mV. If measurements are taken with a rest-
ing ventricular myocyte, a membrane potential
of about −90 mV will be recorded. This rest- (concentration difference) exists for K+ to dif-
ing membrane potential (Em) is determined by fuse out of the cell. The opposite situation is
the concentrations of positively and negatively found for Na+ and Ca++; their chemical gradi-
charged ions across the cell membrane, the rela- ents favor an inward diffusion. The concen-
tive permeability of the cell membrane to these tration differences across the cell membrane
ions, and the ionic pumps that transport ions for these and other ions are determined by
across the cell membrane. the activity of energy-dependent ionic pumps
and the presence of impermeable, negatively
EQUILIBRIUM POTENTIALS
charged proteins within the cell that affect the
Of the many different ions present inside and passive distribution of cations and anions.
outside of cells, the concentrations of Na+, To understand how concentration gra-
K+, and Ca++ are most important in determin- dients of ions across a cell membrane affect
ing the membrane potential across the cell membrane potential, consider a cell in which
membrane. Although chloride ions are found K+ is the only ion across the membrane other
inside and outside the cell, they contrib- than the large, impermeable, negatively
ute relatively little to the resting membrane charged proteins on the inside of the cell. In
potential. Figure 2.1 shows approximate this cell, K+ diffuses down its chemical gra-
concentrations of Na+, K+, and Ca++ inside dient and out of the cell because its concen-
and outside the cell. Of the three ions, K+ is tration is much higher inside than outside
the most important in determining the rest- the cell (see Fig. 2.1). As K+ diffuses out of
ing membrane potential. In a cardiac cell, the the cell, it leaves behind negatively charged
concentration of K+ is high inside and low out- proteins, thereby creating a separation of
side the cell. Therefore, a chemical gradient charge and a potential difference across the

Klabunde_Chap02.indd 10 6/11/2011 10:28:34 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 11

membrane (negative inside the cell relative to called the equilibrium potential for Na+ (ENa)
outside). The membrane potential that is nec- and is calculated using the Nernst equation,
essary to oppose the outward movement of as follows:
K+ down its concentration gradient is termed [Na+ ]i
the equilibrium potential for K+ (EK; Nernst Eq. 2-2 ENa = -61 log = + 52 mV
[Na+ ]o
potential). The Nernst potential for K+ at
37°C is as follows: in which the sodium concentration inside
+
[K ]i [Na+]i = 20 mM and the sodium concentra-
Eq. 2-1 EK = -61 log
[K+ ]o
= -96 mV tion outside [Na+]o = 145 mM. The calculated
equilibrium potential for sodium indicates
in which the potassium concentration inside that to balance the inward diffusion of Na+ at
[K+]i = 150 mM and the potassium concentra- these intracellular and extracellular concen-
tion outside [K+]o = 4 mM. The −61 is derived trations, the cell interior has to be +52 mV to
from RT/zF, in which R is the gas constant, z is prevent Na+ from diffusing into the cell.
the number of ion charges (z = 1 for K+; z = 2 for The net driving or electrochemical force act-
divalent ions such as Ca++), F is Faraday con- ing on sodium (and each ionic species) has two
stant, and T is temperature (°K). The equilibrium components. First, the sodium concentration
potential is the potential difference across the mem- gradient is driving sodium into the cell; accord-
brane required to maintain the concentration gra- ing to the Nernst calculation, the electrical force
dient across the membrane. In other words, the necessary to counterbalance this chemical gra-
equilibrium potential for K+ represents the elec- dient is +52 mV. Second, because the interior
trical potential necessary to keep K+ from diffus- of the resting cell is very negative (−90 mV), a
ing down its chemical gradient and out of the large electrical force is trying to “pull” sodium
cell. If the outside K+ concentration increased into the cell. We can derive the net electro-
from 4 to 10 mM, the chemical gradient for diffu- chemical force acting on sodium from these
sion out of the cell would be reduced; therefore, two component forces by subtracting the Em
the membrane potential required to maintain minus ENa: −90 mV − +52 mV equals −142 mV.
electrochemical equilibrium would be less nega- This large electrochemical force drives sodium
tive according to the Nernst relationship. into the cell; however, at rest, the permeability
The Em for a ventricular myocyte is about of the membrane to Na+ is so low that only a
−90 mV, which is near the equilibrium poten- small amount of Na+ leaks into the cell.
tial for K+. Because the equilibrium potential The same reasoning can be applied to Ca++
for K+ is −96 mV and the measured resting as just described for Na+. Its calculated ECa is
membrane potential is −90 mV, a net driv- +134 mV and net electrochemical force act-
ing force (net electrochemical force) acts on ing on Ca++ is −224 mV. Therefore, like Na+,
the K+, causing it to diffuse out of the cell. In there is a very large net electrochemical force
the case of K+, this net electrochemical driv- working to drive Ca++ into the resting cell;
ing force is the Em (−90 mV) minus the EK however, in the resting cell, little Ca++ leaks
(−96 mV), resulting in +6 mV. Because the into the cell because of low membrane perme-
resting cell has a finite permeability to K+ and ability to Ca++ at rest.
a small net outward driving force is acting on
K+, K+ slowly leaks outward from the cell.
IONIC CONDUCTANCES AND
Sodium ions also play a major role in deter-
MEMBRANE POTENTIAL
mining the membrane potential. Because the
Na+ concentration is higher outside the cell, As explained, the Em in a resting, nonpace-
this ion would diffuse down its chemical gra- maker cell is very near EK, and quite distant
dient into the cell. To prevent this inward flux from ENa and ECa. This occurs because the
of Na+, a large positive charge is needed inside membrane is much more permeable to K+ in
the cell (relative to the outside) to balance out the resting state than to Na+ or Ca++. Therefore,
the chemical diffusion forces. This potential is Na+ and Ca++ have little contribution to the

Klabunde_Chap02.indd 11 6/11/2011 10:28:34 AM


12 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

resting Em because Em reflects not only the g' ea++ are low. Therefore, the low relative con-
concentration gradients of individual ions ductances of Na• and Ca++ multiplied by their
(i.e., the equilibrium potentials) but also equilibrium potential values causes those ions
the relative permeability of the membrane to to contribute little to the resting membrane
those ions. If the membrane has a relatively potential. When g'Na• increases and g'K•
higher permeability to one ion over the oth- decreases (as occurs during an action poten-
ers, that ion will have a greater influence in tial), the membrane potential becomes more
determining the membrane potential. positive (depolarized) because the sodium
Membrane permeability for an ion deter- equilibrium potential has more influence on
mines the movement of an ion being driven by the overall membrane potential. Similarly; a
a net electrochemical force. Because this ion large increase in g' Ca•, particularly when g'K•
movement represents an electrical current, it is low, will also result in depolarization.
is common to speak in terms of ion conduct- In Equation 2-3, ion concentrations (which
ance (g), which is defined as the ion current determine the equilibrium potential) and ion
divided by the net voltage (net electrochemical conductances are separate variables. In reality,
force) acting on the ion. Membrane permeabil- the conductance of some ion channels is influ-
ity and ion conductance are related in that an enced by the concentration of the ion (e.g.,
increase in membrane permeability for an ion K•-sensitive K• channels) or by changes in
results in an increase in electrical conductance membrane potential (e.g., voltage-dependent
for that ion. Putting these concepts together, it Na+, K+, and Ca++ ion channels). For exam-
is possible to derive an expression that relates ple, a decrease in external K• concentration
membrane potential (Em) to the relative con- (e.g., from 4 to 3 mM) can decrease gK• in
ductances of all ions and their equilibrium some cardiac cells and lead to a small depo-
potentials as shown in the following equation: larization (less negative potential) instead of
the hyperpolarization (more negative poten-
Em= g'K.(EK) + g'Na•(ENa) tial) predicted by the Nernst relationship or
Eq. 2-3
+ g•ca••cEca> Equation 2-3. In some cells, small increases in
external K• concentration (e.g., from a normal
In Equation 2-3, the Em is the sum of the indi- concentration of 4 to 6 mM) can cause a small
vidual equilibrium potentials for K•, Na•, and hyperpolarization owing to activation of K+
Ca++, with each multiplied by the membrane channels and an increase in gK+.
conductance for that particular ion relative to
the sum of all ion conductances. For exam- PROBLEM 2-1
ple, the relative conductance for K• (g'K•) = High concentrations of potassium are
gK•f(gK• + gNa• + gca++). If the equilibrium added to cardioplegic solutions used
potentials forK+, Na+, and Ca++ are calculated to arrest the heart during surgery.
using the concentrations shown in Figure 2.1, Using the Nernst equation, calculate an
then Equation 2-3 can be depicted as follows: estimate for the new resting membrane
Em= g'K•(- 96 mV) potential (Em) when external potassium
concentration is increased from a
Eq. 2-4 + g'Na+ (+52 mV)
normal value of 4 to 40 mM. Assume
+ g'Ca++(+134 mV) that the internal concentration remains
In a cardiac cell, the individual ion concentra- at 150 mM and that K• and other ion
tion gradients change very little, even when Na• conductances are not altered.
enters and K• leaves the cell during depolariza-
tion. Therefore, changes in Em primarily result
Maintenance of Ionic Gradients
from changes in ionic conductances. The rest-
ing membrane potential (-90 mV) is near the Membrane potential depends on the main-
equilibrium potential for K• (-96 mV) because tenance of ionic concentration gradients
g'K• is high in the resting cell, while g'Na• and across the membrane. The maintenance of

Klabunde_Chap02.indd 12 6/11/2011 10:28:35 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 13

these concentration gradients requires the results in a less negative (more depolarized)
expenditure of energy (adenosine triphos- resting membrane potential primarily because
phate [ATP] hydrolysis) coupled with ionic EK becomes less negative (see Equation 2-1).
pumps. Consider the concentration gradi- Besides maintaining the Na+ and K+ concentra-
ents for Na+ and K+. Na+ constantly leaks into tion gradients, it is important to note that the
the resting cell, and K+ leaks out. Moreover, Na+/K+-ATPase pump is electrogenic because
whenever an action potential is generated, it extrudes three Na+ for every two K+ entering
additional Na+ enters the cell and additional the cell. By pumping more positive charges
K+ leaves. Although the number of ions mov- out of the cell than into it, the pump cre-
ing across the sarcolemmal membrane in a ates a negative potential within the cell. This
single action potential is small relative to the electrogenic potential may be up to −10 mV,
total number of ions, many action potentials depending on the activity of the pump. Inhi-
can lead to a significant change in the extra- bition of this pump, therefore, causes depolar-
cellular and intracellular concentration of ization resulting from changes in Na+ and K+
these ions. To prevent this change from hap- concentration gradients and from the loss of
pening (i.e., to maintain the concentration an electrogenic component of the membrane
gradients for Na+ and K+), an energy (ATP)- potential. In addition, increases in intracellu-
dependent pump system (Na+/K+-adenosine lar Na+ or extracellular K+ stimulate the activ-
triphosphatase [ATPase]), located on the ity of the electrogenic Na+/K+-ATPase pump
sarcolemma, pumps Na+ out and K+ into the and produce hyperpolarizing currents.
cell (Fig. 2.2). Normal operation of this pump Because Ca++ enters the cell, especially dur-
is essential to maintain Na+ and K+ concen- ing action potentials, it is necessary to have a
trations across the membrane. If this pump mechanism to maintain its concentration gra-
stops working (such as when ATP is lost dient. Two primary mechanisms remove cal-
under hypoxic conditions), or if the activity cium from cells (Fig. 2.2). The first involves
of the pump is inhibited by cardiac glycosides an ATP-dependent Ca++ pump that actively
such as digoxin, Na+ accumulates within the pumps calcium out of the cell and gener-
cell and intracellular K+ falls. This change ates a small negative electrogenic potential.

Ca++ Na+ K+

1 2 3

Ca++ Ca++ Na+ K+

Ca++ Na+ K+
++
1 = ATP-dependent Ca pump
+ ++
2 = Na /Ca exchanger (3:1)
+ +
3 = Na /K -ATPase pump (3:2)
■ FIGURE 2.2 Sarcolemmal ion pumps and exchangers. These pumps maintain transmembrane ionic
gradients for Na+, K+, and Ca++. Na+ and Ca++ enter the cell down their electrochemical gradient, especially
during action potentials, while K+ is leaving the cell. Ca++ is removed by an ATP-dependent, electrogenic
Ca++ pump (1) and by the electrogenic Na+/Ca++ exchanger that exchanges three Na+ for every one Ca++ (2).
Na+ is actively removed from the cell by the electrogenic Na+/K+-ATPase pump, which brings two K+ into
the cell for every three Na+ that are pumped out.

Klabunde_Chap02.indd 13 6/11/2011 10:28:37 AM


14 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

The second mechanism is the sodium–cal- efflux, thereby increasing intracellular Ca++.
cium exchanger, through which Na+ and As described in Chapter 3, this can lead to an
Ca++ are transported in opposite directions. increase in the force of myocyte contraction.
The exchanger can operate in either direc-
tion across the sarcolemma depending on the
Ion Channels
Em. In resting cells, the negative Em causes
Na+ to enter the cell in exchange for Ca++, Ions move across the sarcolemma through
which leaves the cell. Three sodium ions are specialized ion channels in the phospholipid
exchanged for each calcium ion; therefore, bilayer of the cell membrane. These channels
the exchanger generates a small (few milli- are made up of large polypeptide chains that
volts) electrogenic potential that follows the span the membrane and create an opening in
direction of Na+. The opposite occurs in depo- the membrane. Conformational changes in the
larized cells. This exchanger is also strongly ion channel proteins alter the shape of the chan-
influenced by changes in intracellular Na+ nel, thereby permitting ions to transverse the
concentration. For example, when the activ- membrane channel or blocking ion movement.
ity of the Na+/K+-ATPase pump is decreased by Ion channels are selective for different cati-
drugs such as digoxin, the increase in intra- ons and anions. For example, there are ion
cellular Na+ concentration reduces the gradi- channels selective for sodium, potassium,
ent for Na+ movement into the cell through and calcium ions (Table 2-1). Furthermore,
this exchanger, which results in less Ca++ a given ion may have several different types

TABLE 2-1 CARDIAC ION CHANNELS AND CURRENTS


CHANNELS GATING CHARACTERISTICS

Sodium
Fast Na+ (INa) Voltage Phase 0 of myocytes
Slow Na+ (If) Voltage and Contributes to phase 4 pacemaker
receptor current in SA and AV nodal cells
Calcium
L-type (ICa) Voltage Slow inward, long-lasting current; phase
2 of myocytes and phases 4 and 0 of SA
and AV nodal cells
T-type (ICa) Voltage Transient current; contributes to phase 4
pacemaker current in SA and AV nodal
cells
Potassium
Inward rectifier (IK1) Voltage Maintains negative potential in phase 4;
closes with depolarization
Transient outward (Ito) Voltage Contributes to phase 1 in myocytes
Delayed rectifier (IKr) Voltage Phase 3 repolarization
ATP-sensitive (IK, ATP) Receptor Inhibited by ATP; opens when ATP
decreases during cellular hypoxia
Acetylcholine activated (IK, ACh) Receptor Activated by acetylcholine and adenos-
ine; Gi-protein coupled; slows SA nodal
firing
Calcium activated (IK.Ca) Receptor Activated by high cytosolic calcium;
accelerates repolarization
IX, name of specific current.

Klabunde_Chap02.indd 14 6/11/2011 10:28:37 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 15

of ion channels responsible for its movement squid giant axon. In this model, two gates
across a cell membrane. For example, several regulate the movement of sodium through the
different types of potassium channels exist channel (Fig. 2.3). At a normal resting mem-
through which potassium ions can move brane potential (about −90 mV in cardiac
across the cell membrane. myocytes), the sodium channel is in a resting,
Two general types of ion channels exist: closed state. In this configuration, the m-gate
voltage-gated (voltage-operated) and receptor- (activation gate) is closed and the h-gate
gated (receptor-operated) channels. Voltage- (inactivation gate) is open. These gates are
gated channels open and close in response to polypeptides that are part of the transmem-
changes in membrane potential. Examples of brane protein channel, and they undergo con-
voltage-gated channels include several sodium, formational changes in response to changes
potassium, and calcium channels that are in voltage. The m-gates rapidly become acti-
involved in cardiac action potentials. Recep- vated and open when the membrane is rap-
tor-gated channels open and close in response idly depolarized. This permits sodium, driven
to chemical signals operating through mem- by its electrochemical gradient, to enter the
brane receptors. For example, acetylcholine, cell. As the m-gates open, the h-gates begin
which is the neurotransmitter released by the to close; however, the m-gates open more rap-
vagus nerves innervating the heart, binds to a idly than the h-gates close. The difference in
sarcolemmal receptor that subsequently leads the opening and closing rates of the two gates
to the opening of special types of potassium permits sodium to briefly enter the cell. After
channels (IK, ACh). a few milliseconds, however, the h-gates close
Ion channels have both open and closed and sodium ceases to enter the cell. The clos-
states. Ions pass through the channel only ing of the h-gates therefore limits the length
while it is in the open state. The open and of time that sodium can enter the cell. This
closed states of voltage-gated channels inactivated, closed state persists throughout
are regulated by the membrane potential. the repolarization phase as the membrane
Fast sodium channels have been the most potential recovers to its resting level. Near
extensively studied, and a conceptual model the end of repolarization, the negative mem-
has been developed based upon studies by brane potential causes the m-gates to close
Hodgkin and Huxley in the 1950s using the and the h-gates to open. These changes cause

Na+ Na+ Na+ Na+


outside
m

h
inside
Resting Activated Inactivated Resting
(closed) (open) (closed) (closed)

Depolarization Repolarization
■ FIGURE 2.3 Open and closed states of fast sodium channels in cardiac myocytes. In the resting (closed)
state, the m-gates (activation gates) are closed, although the h-gates (inactivation gates) are open. Rapid
depolarization to threshold opens the m-gates (voltage activated), thereby opening the channel and ena-
bling sodium to enter the cell. Shortly thereafter, as the cell begins to repolarize, the h-gates close and the
channel becomes inactivated. Toward the end of repolarization, the m-gates again close and the h-gates
open. This brings the channel back to its resting state.

Klabunde_Chap02.indd 15 6/11/2011 10:28:37 AM


16 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

the channel to revert back to its initial resting, The open and closed states described for
closed state. Full recovery of the h-gates can sodium channels are also found in other ion
take 100 milliseconds or longer after the rest- channels. For example, slow calcium chan-
ing membrane potential has been restored. nels have activation and inactivation gates
The response of the activation and inac- (although they have different letter designa-
tivation gates described above occurs when tions than fast sodium channels). Although
the resting membrane potential is normal this conceptual model is useful to help under-
(about −90 mV) and a rapid depolarization stand how ions transverse the membrane,
of the membrane occurs, as happens when a many of the details of how this actually occurs
normal depolarization current spreads from at the molecular level are still unknown. Nev-
one cardiac cell to another during electri- ertheless, recent research is helping to show
cal activation of the heart. The response of which regions of ion channel proteins act as
the fast sodium channel, however, is differ- voltage sensors and which regions undergo
ent when the resting membrane potential conformational changes analogous to the
is partially depolarized or the cell is slowly gates described in the conceptual model.
depolarized. For example, when myocytes
become hypoxic, the cells depolarize to a less
Action Potentials
negative resting membrane potential. This
partially depolarized state inactivates sodium Action potentials occur when the membrane
channels by closing the h-gates. The more a potential suddenly depolarizes and then
cell is depolarized, the greater the number of repolarizes back to its resting state. The two
inactivated sodium channels. At a membrane general types of cardiac action potentials
potential of about −55 mV, virtually all fast include nonpacemaker and pacemaker action
sodium channels are inactivated. If a myocyte potentials. Nonpacemaker action potentials
has a normal resting potential but then under- are triggered by depolarizing currents from
goes slow depolarization, more time is avail- adjacent cells, whereas pacemaker cells are
able for the h-gates to close as the m-gates are capable of spontaneous action potential gen-
opening. This causes the sodium channel to eration. Both types of action potentials in
transition directly from the resting (closed) the heart differ considerably from the action
state to the inactivated (closed) state. The potentials found in nerve and skeletal muscle
result is that there is no activated, open state cells (Fig. 2.4). One major difference is the
for sodium to pass through the channel, effec- duration of the action potentials. In a typical
tively abolishing fast sodium currents through nerve, the action potential duration is about 1
these channels. As long as the partial depolar-
ized state persists, the channel will not resume
Membrane Potential (mV)

its resting, closed state. As described later in +50 Nerve Cell


this chapter, these changes significantly alter
myocyte action potentials by abolishing fast Cardiac Myocyte
sodium currents during action potentials. 0
A single cardiac cell has many sodium
channels, and each channel has a slightly dif-
ferent voltage activation threshold and dura- –50
tion of its open, activated state. The amount
of sodium (the sodium current) that passes –100
through sodium channels when a cardiac cell 0 500
undergoes depolarization depends upon the Time (ms)
number of sodium channels, the duration
■ FIGURE 2.4 Comparison of action potentials
of time the channels are in the open state,
from a nerve cell and a nonpacemaker cardiac
and the electrochemical gradient driving the myocyte. Cardiac action potentials are much
sodium into the cell. longer in duration than nerve cell action potentials.

Klabunde_Chap02.indd 16 6/11/2011 10:28:37 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 17

to 2 milliseconds. In skeletal muscle cells, the the equilibrium potential for K+ because gK+,
action potential duration is approximately 2 through inward rectifying potassium channels
to 5 milliseconds. In contrast, the duration of (see Table 2-1), is high relative to gNa+ and
ventricular action potentials ranges from 200 gCa++ in resting cells (see Equation 2-4). When
to 400 milliseconds. These differences among these cells are rapidly depolarized from −90 mV
nerve, skeletal muscle, and cardiac myocyte to a threshold voltage of about −70 mV (owing
action potentials relate to differences in the to, for example, an action potential conducted
ionic conductances responsible for generating by an adjacent cell), a rapid depolarization
the changes in membrane potential. (phase 0) is initiated by a transient increase
in conductance of voltage-gated, fast Na+-
NONPACEMAKER ACTION POTENTIALS channels. At the same time, gK+ falls. These
Figure 2.5 shows the ionic mechanisms respon- two conductance changes very rapidly move
sible for the generation of “fast response” non- the membrane potential away from the potas-
pacemaker action potentials such as those sium equilibrium potential and closer to the
found in atrial and ventricular myocytes, and sodium equilibrium potential (see Equation
Purkinje fibers. By convention, the action 2-4). Phase 1 represents an initial repolariza-
potential is divided into five numbered phases. tion caused by the opening of a special type of
Nonpacemaker cells have a true resting mem- K+ channels (transient outward) and the inac-
brane potential (phase 4) that remains near tivation of the Na+ channels. However, because
of the large increase in slow inward gCa++,
the repolarization is delayed and the action
potential reaches a plateau phase (phase 2).
ERP This inward calcium movement is through
1 long-lasting (L-type) calcium channels that
2 open when the membrane potential depolar-
0
izes to about −40 mV. L-type calcium channels
mV

0 3 are the major calcium channels in cardiac and


–50 vascular smooth muscle. They are opened by
membrane depolarization (they are voltage-
4 4 operated) and remain open for a relatively
–100 long duration. These channels are blocked by
200 ms gK+
Conductances

classical L-type calcium channel blockers (e.g.,


verapamil and diltiazem). Repolarization
gCa++ (phase 3) occurs when gK+ increases through
Ion

delayed rectifier potassium channels and gCa++


gNa+
decreases. Therefore, changes in Na+, Ca++,
and K+ conductances primarily determine the
action potential in nonpacemaker cells.
Ventricular Cell During phases 0, 1, 2, and part of phase 3,
the cell is refractory (i.e., unexcitable) to the
■ FIGURE 2.5 Changes in ion conductances associ-
ated with a ventricular myocyte action potential. initiation of new action potentials. This is
Phase 0 (depolarization) primarily is due to the the effective (or absolute) refractory period
rapid increase in sodium conductance (gNa+) (ERP, or ARP) (see Fig. 2.5). During the ERP,
accompanied by a fall in potassium conductance
stimulation of the cell does not produce new,
(gK+); the initial repolarization of phase 1 is due to
opening of special potassium channels (Ito); phase propagated action potentials because the
2 (plateau) primarily is due to an increase in slow h-gates are still closed. The ERP acts as a pro-
inward calcium conductance (gCa++) through L-type tective mechanism in the heart by limiting the
Ca++ channels; phase 3 (repolarization) results from
frequency of action potentials (and therefore
an increase in gK+ and a decrease in gCa++. Phase
4 is a true resting potential that primarily reflects a contractions) that the heart can generate. This
high gK+. ERP, effective refractory period. enables the heart to have adequate time to fill

Klabunde_Chap02.indd 17 6/11/2011 10:28:37 AM


18 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

and eject blood. The long ERP also prevents site to take over as the pacemaker for the heart.
the heart from developing sustained, tetanic When this occurs, the new pacemaker outside
contractions like those that occur in skeletal of the SA node is called an ectopic focus.
muscle. At the end of the ERP, the cell is in SA nodal action potentials are divided into
its relative refractory period. Early in this three phases: phase 0, upstroke of the action
period, suprathreshold depolarization stim- potential; phase 3, the period of repolariza-
uli are required to elicit actions potentials. tion; and phase 4, the period of spontaneous
Because not all the sodium channels have depolarization that leads to subsequent gen-
recovered to their resting state by this time, eration of a new action potential (Fig. 2.6).
action potentials generated during the relative Phase 0 depolarization primarily is due
refractory period have a decreased phase 0 to increased gCa++ through L-type calcium
slope and lower amplitude. When the sodium channels. These voltage-operated channels
channels are fully recovered, the cell becomes open when the membrane is depolarized to a
fully excitable and normal depolarization threshold voltage of about −40 mV. Because the
stimuli can elicit new, rapid action potentials. movement of Ca++ through calcium channels
is not rapid compared to fast sodium channels
(hence, the term “slow calcium channels”),
PACEMAKER ACTION POTENTIALS
the rate of depolarization (the slope of phase 0)
Pacemaker cells have no true resting poten- is much slower than that found in other
tial, but instead generate regular, spontaneous cardiac cells (e.g., in Purkinje cells). As the
action potentials. Unlike most other cells that calcium channels open and the membrane
exhibit action potentials (e.g., nerve cells, and potential moves toward the calcium
muscle cells), the depolarizing current of the equilibrium potential, a transient decrease in
action potential is carried primarily by relatively
slow, inward Ca++ currents (through L-type cal-
cium channels) instead of by fast Na+ currents. 0
The rate of depolarization of pacemaker cells 0 3
mV

is slow compared to “fast response” nonpace-


maker cells, and therefore they are sometimes 4
called “slow response” action potentials. –50 4
Cells within the sinoatrial (SA) node, located
Conductances

within the posterior wall of the right atrium gK+


(RA), constitute the primary pacemaker site
gCa++
within the heart. Other pacemaker cells exist
Ion

within the AV node and ventricular conduction


system, but their firing rates are driven by the
higher rate of the SA node because the intrinsic
pacemaker activity of the secondary pacemak- If
ers is suppressed by a mechanism termed over-
drive suppression. This mechanism causes the SA Node
secondary pacemaker to become hyperpolar- ■ FIGURE 2.6 Changes in ion conductances
ized when driven at a rate above its intrinsic associated with a sinoatrial (SA) nodal pacemaker
rate. Hyperpolarization occurs because the action potential. Phase 0 (depolarization) primar-
ily is due to an increase in calcium conductance
increased action potential frequency stimulates
(gCa++) through L-type Ca++ channels accompa-
the activity of the electrogenic Na+/K+-ATPase nied by a fall in potassium conductance (gK+);
pump as a result of enhanced entry of sodium phase 3 (repolarization) results from an increase in
per unit time into these cells. If the SA node gK+ and a decrease in gCa++. Phase 4 undergoes a
spontaneous depolarization owing to a pacemaker
becomes depressed, or its action potentials
current (If) carried in part by Na+; decreased gK+
fail to reach secondary pacemakers, overdrive and increased gCa++ also contribute to the sponta-
suppression ceases, which permits a secondary neous depolarization.

Klabunde_Chap02.indd 18 6/11/2011 10:28:37 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 19

gK+ occurs, which contributes to the depolari- per minute. Heart rate, however, can vary
zation as shown in the following equation: between low resting values of 50 to 60 beats/
min and over 200 beats/min. These changes
Eq. 2-5 in rate primarily are controlled by autonomic
Em = g'K( - 96 mV) + g'Ca( + 134 mV)
nerves acting on the SA node. At low resting
Depolarization causes voltage-operated, delayed heart rates, vagal influences are dominant
rectifier potassium channels to open, and the over sympathetic influences. This is termed
increased gK+ repolarizes the cell toward the vagal tone. Autonomic nerves increase SA
equilibrium potential for K+ (phase 3). At the nodal firing rate by both decreasing vagal tone
same time, the slow inward Ca++ channels that and increasing sympathetic activity on the SA
opened during phase 0 become inactivated, node in a reciprocal manner. An increase in
thereby decreasing gCa++ and contributing heart rate is a positive chronotropic response
to the repolarization. Phase 3 ends when the (or positive chronotropy), whereas a reduc-
membrane potential reaches about −65 mV. The tion in heart rate is a negative chronotropic
phase of repolarization is self-limited because response (or negative chronotropy).
the potassium channels begin to close again as Autonomic influences alter the rate of pace-
the cell becomes repolarized. maker firing through the following mechanisms:
The ionic mechanisms responsible for the (1) changing the slope of phase 4; (2) altering the
spontaneous depolarization of the pacemaker threshold voltage for triggering phase 0; and (3)
potential (phase 4) are not entirely clear, but altering the degree of hyperpolarization at the
probably involve multiple ionic currents. First, end of phase 3. Any of these three mechanisms
early in phase 4, gK+ is still declining. This fall will either increase or decrease the time to reach
in gK+ contributes to depolarization. Second, in threshold. Sympathetic activation of the SA node
the repolarized state, a pacemaker current (If), increases the slope of phase 4 (Fig. 2.7) and low-
or “funny” current, has been identified (see ers the threshold, thereby increasing pacemaker
Fig. 2.6). This depolarizing current involves, frequency (positive chronotropy). In this mech-
in part, a slow inward movement of Na+. Third, anism, norepinephrine released by sympathetic
in the second half of phase 4, there is a small adrenergic nerves binds to β1-adrenoceptors
increase in gCa++ through T-type calcium chan- coupled to a stimulatory G-protein (Gs-protein),
nels. T-type (“transient”) calcium channels dif- which activates adenylyl cyclase and increases
fer from L-type calcium channels in that they cyclic adenosine monophosphate (cAMP; see
open briefly only at very negative voltages Chapter 3). This effect leads to an increase in If
(−50 mV) and are not blocked by the classi- and an earlier opening of L-type calcium chan-
cal L-type calcium channel blockers. Fourth, nels, both of which increase the rate of depolari-
as the depolarization begins to reach threshold, zation. Repolarization is also accelerated, which
the L-type calcium channels begin to open, shortens overall cycle length and may increase
causing a further increase in gCa++ until thresh- maximal hyperpolarization.
old is reached and phase 0 is initiated. Vagal stimulation releases acetylcholine at
To summarize, “slow response” action the SA node, which decreases the slope of phase
potentials found in SA nodal cells primarily 4 (by inhibiting “funny” currents), hyperpolar-
depend on changes in gCa++ and gK+ conduct- izes the cell, and increases the threshold voltage
ances, with “funny” currents (If) and changes required to trigger phase 0. All of these effects
in gCa++ and gK+ conductances playing a role cause the pacemaker potential to take longer
in the spontaneous depolarization. to reach threshold, thereby slowing the rate
(negative chronotropy). The rate of repolariza-
tion is reduced, which contributes to increas-
REGULATION OF SA NODAL PACEMAKER
ing overall cycle length. Acetylcholine acts by
ACTIVITY
binding to muscarinic receptors (M2). This
The SA node displays intrinsic automaticity decreases cAMP via the inhibitory G-protein
at a rate of 100 to 110 depolarizations (Gi-protein), the opposite effect of sympathetic

Klabunde_Chap02.indd 19 6/11/2011 10:28:38 AM


20 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

SA Nodal Cell
Sympathetic Normal Vagal
0

Normal
mV Threshold

-50
Normal Maximal
Hyperpolarization
■ FIGURE 2.7 Effects of sympathetic and parasympathetic (vagal) stimulation on sinoatrial (SA) nodal pace-
maker activity. Sympathetic stimulation increases the firing rate by increasing the slope of phase 4 and lower-
ing the threshold for the action potential. Vagal stimulation has the opposite effects, and it hyperpolarizes the
cell. Horizontal dashed lines represent threshold and maximal hyperpolarization potentials for normal cell.

activation (see Chapter 3). Acetylcholine also released by sympathetic nerves. Hyperthyroid-
activates a special type of potassium channel ism induces tachycardia, and hypothyroidism
(KACh channel) that hyperpolarizes the cell by induces bradycardia (abnormally low heart
increasing potassium conductance. rate). Changes in the serum concentration of
Nonneural mechanisms also alter pacemaker ions, particularly potassium, can cause changes
activity (Table 2-2). For example, circulating in SA node firing rate. Hyperkalemia induces
catecholamines (epinephrine and norepineph- bradycardia or can even stop SA nodal firing,
rine) cause tachycardia (abnormally high heart whereas hypokalemia increases the rate of
rate) by a mechanism similar to norepinephrine phase 4 depolarization and causes tachycar-
dia, apparently by decreasing potassium con-
ductance during phase 4. Cellular hypoxia
TABLE 2-2 FACTORS INCREASING OR depolarizes the membrane potential, causing
DECREASING THE SA NODE
bradycardia and abolition of pacemaker activ-
FIRING RATE
ity. Increased body temperature (e.g., fever)
INCREASING DECREASING
leads to increased rate of SA nodal firing.
Sympathetic Parasympathetic Various drugs used to treat abnormal heart
stimulation stimulation rhythm (i.e., antiarrhythmic drugs) also affect
Muscarinic receptor Muscarinic receptor SA nodal rhythm. Calcium channel blockers,
antagonist agonists for example, cause bradycardia by inhibiting
b-Adrenoceptor b-Blockers L-type calcium channels, which reduces slow
agonists inward Ca++ currents during phase 4 and
Circulating cat- Ischemia/hypoxia phase 0. Drugs affecting autonomic control
echolamines or autonomic receptors (e.g., β-blockers and
Hypokalemia Hyperkalemia M2 receptor antagonists; β-adrenoceptor ago-
nists) alter pacemaker activity. Digoxin causes
Hyperthyroidism Sodium and calcium
channel blockers
bradycardia by increasing parasympathetic
activity and inhibiting the sarcolemmal Na+/
Hyperthermia Hypothermia
K+-ATPase, which leads to depolarization.

Klabunde_Chap02.indd 20 6/11/2011 10:28:38 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 21

Arrhythmias Caused by Abnormal Early Afterdepolarizations


Action Potential Generation
0
ABNORMAL AUTOMATICITY

mV
“Fast response” nonpacemaker action poten- -50
tials do not ordinarily display automatic-
ity because they are characterized as having
a true resting membrane potential that does -100
not undergo spontaneous depolarization. If Delayed Afterdepolarizations
the fast sodium channels that are responsible
0
for the rapid depolarization during phase 0
are blocked pharmacologically, or inactivated

mV
by depolarization caused by cellular hypoxia, -50
the slope and amplitude of phase 0 are sig-
nificantly depressed, and the action potential
appears much like a “slow response” action -100
Time
potential. The depolarization phase of the
action potential under these conditions is ■ FIGURE 2.8 Early (top panel) and delayed
(bottom panel) afterdepolarizations. If the magni-
brought about by slow inward calcium cur- tude of spontaneous depolarization is sufficient, it
rents carried through L-type calcium channels. can trigger self-sustaining action potentials.
Furthermore, like SA nodal pacemakers, these
cells may display spontaneous depolarization during ischemia, digoxin toxicity, and excessive
during phase 4. This abnormal automaticity catecholamine stimulation.
in these transformed “fast response” cells can
result in spontaneous action potential genera- CONDUCTION OF ACTION
tion, thereby producing arrhythmias. POTENTIALS WITHIN THE HEART

TRIGGERED ACTIVITY Electrical Conduction within


the Heart
A second mechanism that can lead to abnormal
generation of action potentials is called triggered The action potentials generated by the SA node
activity. Nonpacemaker cells may undergo spread throughout the atria primarily by cell-
spontaneous depolarizations either during phase to-cell conduction (Fig. 2.9). When a single
3 or early in phase 4, triggering abnormal action
potentials. These spontaneous depolarizations
– – –
(termed afterdepolarizations), if of sufficient – – – –
magnitude, can trigger self-sustaining action + + + + + + – –
+
potentials resulting in tachycardia (Fig. 2.8). + – – – – – –
+
Early afterdepolarizations occur during phase ++ + + + – – –
3 and are more likely to occur when action ++ + – –

potential durations are prolonged. Because these – – –
afterdepolarizations occur at a time when fast – –
– –
Na+ channels are still inactivated, slow inward
Ca++ carries the depolarizing current. Another
■ FIGURE 2.9 Cell-to-cell conduction. Cardiac
type of afterdepolarization, delayed afterdepo- cells are connected together by low-resistance
larization, occurs at the end of phase 3 or early gap junctions between the cells, forming a
in phase 4. It, too, can lead to self-sustaining functional syncytium. When one cell depolarizes,
depolarizing currents can pass through the gap
action potentials and tachycardia. This form of junctions (red arrows) and depolarize adjacent
triggered activity appears to be associated with cells, resulting in a cell-to-cell propagation of
elevations in intracellular calcium, as occurs action potentials.

Klabunde_Chap02.indd 21 6/11/2011 10:28:38 AM


22 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

myocyte depolarizes, positive charges accu- excitation–contraction coupling is initiated


mulate just inside the sarcolemma. Because (see Chapter 3).
individual myocytes are joined together by low- Nonconducting connective tissue separates
resistance gap junctions located at the inter- the atria from the ventricles. Action potentials
calated disks (see Chapter 3), ionic currents normally have only one pathway available to
can flow between two adjoining cells. When enter the ventricles, a specialized region of
these ionic currents are sufficient to rapidly cells called the AV node. The AV node, located
depolarize the adjoining cell to its threshold in the inferior–posterior region of the intera-
potential, an action potential is elicited in trial septum separating the left from the right
the second cell. This is repeated in every cell, atrium, is a highly specialized conducting tis-
thereby causing action potentials to be propa- sue (cardiac, not neural in origin) that slows
gated throughout the atria. Action potentials in the impulse conduction velocity to about
the atrial muscle have a conduction velocity of 0.05 m/s. This is one-tenth the velocity found
about 0.5 m/s (Fig. 2.10). Although the con- in atrial or ventricular myocytes (see Fig. 2.10).
duction of action potentials within the atria is The delay in conduction between the atria
primarily between myocytes, some functional and ventricles at the AV node is physiologically
evidence (although controversial) points to the important. First, it allows sufficient time for
existence of specialized myocytes that serve as complete atrial depolarization, contraction, and
conducting pathways within the atria, termed emptying of atrial blood into the ventricles prior
internodal tracts (e.g., Bachmann bundle). As to ventricular depolarization and contraction
action potentials originating from the SA node (see Chapter 4). Second, the low conduction
spread across and depolarize the atrial muscle, velocity helps to limit the frequency of impulses

Atrial Muscle
(~0.5 m/sec)
SA Node
AV Node
LA (~0.05 m/sec)

Bundle of His
(~2 m/sec)
RA

Left & Right


RV LV Bundle Branches
(~2 m/sec)

Purkinje Ventricular
Fibers Muscle
(~0.5 m/sec)
(~4 m/sec)
■ FIGURE 2.10 Conduction system within the heart. Conduction velocities of different regions are noted in
parentheses. Note that Purkinje fibers have the highest conduction velocity and the atrioventricular (AV)
node has the lowest conduction velocity. SA, sinoatrial; RA, right atrium; LA, left atrium; RV, right ventricle;
LV, left ventricle.

Klabunde_Chap02.indd 22 6/11/2011 10:28:39 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 23

traveling through the AV node and activating nels increases the rate of depolarization. The
the ventricle. This is important in atrial flutter more rapidly one cell depolarizes, the more
and fibrillation, in which excessively high atrial quickly an adjoining cell depolarizes. There-
rates, if transmitted to the ventricles, can lead fore, conditions that decrease the availability
to a very high ventricular rate. This can reduce of fast sodium channels (e.g., depolarization
cardiac output because of inadequate time for caused by cellular hypoxia), decrease the rate
ventricular filling (see Chapter 4). and magnitude of phase 0, thereby decreas-
Action potentials leaving the AV node ing conduction velocity within the heart. In
enter the base of the ventricle at the bundle of AV nodal tissue in which slow inward calcium
His and then follow the left and right bundle primarily determines phase 0 of the action
branches along the interventricular septum potential, alterations in calcium conductance
that separates the two ventricles. These spe- alter the rate of depolarization and therefore
cialized bundle branch fibers conduct action the rate of conduction between AV nodal cells.
potentials at a high velocity (about 2 m/s). Extrinsic factors can influence conduction
The bundle branches divide into an exten- velocity, including autonomic nerves, circulat-
sive system of Purkinje fibers that conduct ing hormones (particularly catecholamines),
the impulses at high velocity (about 4 m/s) and various drugs (Table 2-3). Autonomic nerve
throughout the ventricles. The Purkinje fiber activity significantly influences the conduction
cells connect with ventricular myocytes, of electrical impulses throughout the heart, par-
which become the final pathway for cell-to- ticularly in the specialized conduction system.
cell conduction within the ventricles. An increase in sympathetic firing (or increased
The conduction system within the heart is circulating catecholamines) increases conduc-
important because it permits rapid, organized, tion velocity via norepinephrine binding to
near-synchronous depolarization and contrac- β1-adrenoceptors. The activation of parasym-
tion of ventricular myocytes, which is essen- pathetic (vagal) nerves decreases conduction
tial to generate pressure efficiently during velocity via the action of acetylcholine on M2
ventricular contraction. If the conduction receptors. This is most prominent at the AV
system becomes damaged or dysfunctional, as node, which has a high degree of vagal inner-
can occur during ischemic conditions or myo- vation. The signal transduction mechanisms
cardial infarction, this can lead to altered path- coupled to β1-adrenoceptors and M2 receptors
ways of conduction and decreased conduction (Gs- and Gi-proteins) are the same as described
velocity within the heart. The functional con- in Chapter 3 (see Fig. 3.6) for the regulation
sequence is that it diminishes the ability of the of cardiac contraction. A number of drugs can
ventricles to generate pressure. Furthermore,
damage to the conducting system can precipi- TABLE 2-3 EXTRINSIC FACTORS
tate arrhythmias as described later. INCREASING OR DECREASING
CONDUCTION VELOCITY
WITHIN THE HEART
Regulation of Conduction
INCREASING DECREASING
Velocity
Sympathetic Parasympathetic
The rate of cell-to-cell conduction is deter- stimulation stimulation
mined by several intrinsic and extrinsic fac- Muscarinic recep- Muscarinic receptor
tors. Intrinsic factors include the electrical tor antagonists agonists
resistance between cells and the nature of the b-Adrenoceptor b-Blockers
action potential, particularly in the initial rate agonists
of depolarization (phase 0). As discussed ear-
Circulating Ischemia/hypoxia
lier in this chapter, fast sodium channels are catecholamines
responsible for the rapid upstroke velocity of
Hyperthyroidism Sodium and calcium
nonpacemaker action potentials. Increasing
channel blockers
the number of activated fast sodium chan-

Klabunde_Chap02.indd 23 6/11/2011 10:28:39 AM


Normal Reentry
Partial
Conduction
Block

1 2 1 2

3 3

Klabunde_Chap02.indd 24 6/11/2011 10:28:39 AM


AV Node
LA
SA Node
RA

Local Reentry
Site

Bypass Tract LV
(e.g., Bundle of Kent) RV

Global AV
Reentry

Klabunde_Chap02.indd 25 6/11/2011 10:28:40 AM


26 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

THE ELECTROCARDIOGRAM ECG Tracing


As cardiac cells depolarize and repolarize,
The ECG is a crucial diagnostic tool in clini-
electrical currents spread throughout the
cal practice. It is especially useful in diagnos-
body because the tissues surrounding the
ing rhythm disturbances, changes in electrical
heart are able to conduct electrical currents
conduction, and myocardial ischemia and
generated by the heart. When these electri-
infarction. The remaining sections of this
cal currents are measured by an array of elec-
chapter describe how the ECG is generated
trodes placed at specific locations on the body
and how it can be used to examine changes in
surface, the recorded tracing is called an ECG
cardiac electrical activity.

T
P

Q S
PR ST
QT

0 0.2 0.4 0.6 0.8


Time (sec)
■ FIGURE 2.13 Components of the ECG trace. An enlargement of one of the repeating waveform units
in the rhythm strip shows the P wave, QRS complex, and T wave, which represent atrial depolarization,
ventricular depolarization, and ventricular repolarization, respectively. The PR interval represents the time
required for the depolarization wave to transverse the atria and the AV node; the QT interval represents
the period of ventricular depolarization and repolarization; and the ST segment is the isoelectric period
when the entire ventricle is depolarized. Each small square is 1 mm.

Klabunde_Chap02.indd 26 6/11/2011 10:28:41 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 27

(Fig. 2.13). The repeating waves of the ECG aberrant conduction, or it can occur when an
represent the sequence of depolarization and ectopic ventricular pacemaker drives ventric-
repolarization of the atria and ventricles. The ular depolarization. Such ectopic foci nearly
ECG does not measure absolute voltages, but always cause impulses to be conducted over
voltage changes from a baseline (isoelectric) slower pathways within the heart, thereby
voltage. ECGs are generally recorded on paper increasing the time for depolarization and the
at a speed of 25 mm/s and with a vertical cali- duration of the QRS complex.
bration of 1 mV/cm. The isoelectric period (ST segment) follow-
By convention, the first wave of the ECG is ing the QRS is the period at which the entire
the P wave (Fig. 2.13). It represents the wave ventricle is depolarized and roughly corre-
of depolarization that spreads from the SA node sponds to the plateau phase of the ventricular
throughout the atria; it is usually 0.08 to 0.1 sec- action potential. The ST segment is impor-
onds in duration (Table 2-4). No distinctly vis- tant in the diagnosis of ventricular ischemia,
ible wave represents atrial repolarization in the in which the ST segment can become either
ECG because it is masked by ventricular depo- depressed or elevated, indicating nonuniform
larization and is of relatively small amplitude. membrane potentials in ventricular cells. The
The brief isoelectric (zero voltage) period after T wave represents ventricular repolarization
the P wave represents the time in which the (phase 3 of the action potential) and lasts
atrial cells are depolarized and the impulse is longer than depolarization.
traveling within the AV node, where conduction During the QT interval, both ventricular
velocity is greatly reduced. The period of time depolarization and repolarization occur. This
from the onset of the P wave to the beginning interval roughly estimates the duration of
of the QRS complex, the PR interval, normally ventricular action potentials. The QT interval
ranges from 0.12 to 0.20 seconds. This interval can range from 0.2 to 0.4 seconds depending
represents the time between the onset of atrial on heart rate. At high heart rates, ventricular
depolarization and the onset of ventricular action potentials are shorter, decreasing the QT
depolarization. If the PR interval is >0.2 sec- interval. Because prolonged QT intervals can be
onds, a conduction defect (usually within the diagnostic for susceptibility to certain types of
AV node) is present (e.g., first-degree AV block). arrhythmias, it is important to determine if a
The QRS complex represents ventricular given QT interval is excessively long. In prac-
depolarization. The duration of the QRS com- tice, the QT interval is expressed as a corrected
plex is normally 0.06 to 0.1 seconds, indi- QT (QTc) interval by taking the QT interval and
cating that ventricular depolarization occurs dividing it by the square root of the RR interval
rapidly. If the QRS complex is prolonged (the interval between ventricular depolariza-
(>0.1 seconds), conduction is impaired tions). This calculation allows the QT interval
within the ventricles. Impairment can occur to be assessed independent of heart rate. Nor-
with defects (e.g., bundle branch blocks) or mal corrected QTc intervals are <0.44 seconds.

TABLE 2-4 SUMMARY OF ECG WAVES, INTERVALS, AND SEGMENTS


ECG COMPONENT REPRESENTS NORMAL DURATION (S)

P wave Atrial depolarization 0.08–0.10


QRS complex Ventricular depolarization 0.06–0.10
1
T wave Ventricular repolarization
PR interval Atrial depolarization plus AV nodal delay 0.12–0.20
1
ST segment Isoelectric period of depolarized ventricles
QT interval Length of depolarization plus repolarization— 0.20–0.402
corresponds to action potential duration
1
Duration not normally measured.
2
High heart rates reduce the action potential duration and therefore the QT interval.

Klabunde_Chap02.indd 27 6/11/2011 10:28:41 AM


28 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Interpretation of Normal
and Abnormal Cardiac Rhythms
from the ECG Normal
One important use of the ECG is to enable a
physician to evaluate abnormally slow, rapid,
or irregular cardiac rhythms. Atrial and ven-
tricular rates of depolarization can be deter- Atrial Flutter
mined from the frequency of P waves and
QRS complexes by recording a rhythm strip.
A rhythm strip is usually generated from a
single ECG lead (often lead II). In a normal Atrial Fibrillation
ECG (Fig. 2.14), a consistent, one-to-one
correspondence exists between P waves and
the QRS complex; that is, each P wave is fol-
First-Degree AV Block
lowed by a QRS complex. This correspond-
ence indicates that ventricular depolarization
is being triggered by atrial depolarization.
Under these normal conditions, the heart
is said to be in sinus rhythm, because the Second-Degree AV Block (2:1)
SA node is controlling the cardiac rhythm.
Normal sinus rhythm can range from 60 to
100 beats/min. Although the term “beats” is
Third-Degree AV Block
being used here, strictly speaking, the ECG
gives information only about the frequency of
electrical depolarizations. However, a depo-
larization usually results in contraction and
therefore a “beat.”
Abnormal rhythms (arrhythmias) can be Premature Ventricular Complex
caused by abnormal formation of action poten-
tials. A sinus rate <60 beats/min is termed
sinus bradycardia. The resting sinus rhythm,
as previously described, is highly dependent
on vagal tone. Some people, especially highly Ventricular Tachycardia
conditioned athletes, may have normal rest-
ing heart rates that are significantly <60 beats/
min. In other individuals, sinus bradycardia Ventricular Fibrillation
may result from depressed SA nodal func-
■ FIGURE 2.14 ECG examples of abnormal
tion. A sinus rate of 100 to 180 beats/min, rhythms. AV, atrioventricular.
sinus tachycardia, is an abnormal condition
for a person at rest; however, it is a normal
response when a person exercises or becomes therefore, the ventricular rate (as determined
excited. by the frequency of QRS complexes) may
In a normal ECG, a QRS complex follows be less than half of the atrial rate. In atrial
each P wave. Conditions exist, however, when fibrillation, the SA node does not trigger the
the frequency of P waves and QRS complexes atrial depolarizations. Instead, depolarization
may be different (Fig. 2.14). For example, currents arise from many sites throughout
atrial rate may become so high in atrial flut- the atria, leading to uncoordinated, low-
ter (250 to 350 beats/min) that not all of the voltage, high-frequency depolarizations with
impulses are conducted through the AV node; no discernable P waves. In this condition,

Klabunde_Chap02.indd 28 6/11/2011 10:28:41 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 29

the ventricular rate is irregular and usually CASE 2·2


rapid. Atrial fibrillation and flutter illustrate
A patient is being treated for
an important function of the AV node; it lim-
hypertension with a ~-blocker (a drug
its the frequency of impulses that it conducts,
that blocks ~-adrenoceptors in the
thereby limiting ventricular rate. This feature
heart) in addition to a diuretic. A routine
is important because when ventricular rates
ECG reveals that the patient's PR
become very high (e.g., >200 beats/min), car-
interval is 0.24 seconds (first-degree AV
diac output falls owing to inadequate time for
nodal block). Explain how removal of
ventricular filling between contractions.
the ~-blocker might improve A V nodal
Atrial rate is greater than ventricular
conduction.
rate in some forms of AV block (see Fig.
2.14). This is an example of an arrhythmia
caused by abnormal (depressed) impulse A condition can arise in which ventricular
conduction. With AV block, atrial rate is nor- rate is greater than atrial rate; that is, the fre-
mal, but every atrial depolarization may not quency of QRS complexes is greater than the
be followed by a ventricular depolarization. frequency of P waves (see Fig. 2.14). This con-
A second-degree AV block may have two or dition is termed ventricular tachycardia (100
three P waves preceding each QRS complex to 200 beats/min) or ventricular flutter (>200
because the AV node does not successfully beats/min). The most common causes of ven-
conduct every impulse. In a less severe form tricular tachycardias are reentry circuits caused
of AV block, the conduction through the AV by abnormal impulse conduction within the
node is delayed, but the impulse is still able ventricles or rapidly firing ectopic pacemaker
to pass through the AV node and excite the sites within the ventricles (which may be
ventricles. With this condition, termed first- caused by afterdepolarizations). With ven-
degree AV block, a consistent one-to-one cor- tricular tachycardias, there is a complete dis-
respondence remains between the P waves sociation between atrial and ventricular rates
and QRS complexes; however, the PR interval because ventricular depolarizations are not
is found to be >0.2 seconds. In an extreme being triggered by atrial sites. Both ventricular
form of AV nodal blockade, third-degree AV tachycardia and ventricular flutter are serious
block, no atrial depolarizations are conducted clinical conditions because they impair ventric-
through the AV node into the ventricles, and ular filling, reduce stroke volume, and can lead
P waves and QRS complexes are completely to ventricular fibrillation (see Fig. 2.14). This
dissociated. The ventricles still undergo depo- latter condition is seen in the ECG as rapid,
larization because of the expression of a sec- low-voltage, uncoordinated depolarizations
ondary, latent pacemaker site (e.g., within the (having no discernable QRS complexes), which
AV junction or from some ectopic foci within results in cardiac output going to zero. This
the ventricles); however, the ventricular rate lethal condition can sometimes be reverted to a
is generally slow (<40 beats/min). Ventricu- sinus rhythm by applying strong but brief elec-
lar bradycardia occurs because the intrinsic trical currents to the heart by placing electrodes
firing rate of secondary, latent pacemakers is on the chest (electrical defibrillation).
much slower than in the SA node. For exam- The ECG can reveal another type of
ple, pacemaker cells within the AV node and arrhythmia, premature depolarizations (see
bundle of His have rates of 50 to 60 beats/min, Fig. 2.14). These depolarizations can occur
whereas those in the Purkinje system have within either the atria (premature atrial com-
rates of only 30 to 40 beats/min. If the ectopic plex) or the ventricles (premature ventricular
foci are located within the ventricles, the QRS complex). They are usually caused by ectopic
complex will have an abnormal shape and pacemaker sites within these cardiac regions
be wider than normal because depolariza- and appear as extra (and early) P waves or QRS
tion does not follow the normal conduction complexes. These premature depolarizations
pathways. are often abnormally shaped, particularly in

Klabunde_Chap02.indd 29 6/11/2011 10:28:41 AM


30 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

ventricles, because the impulses generated by vectors. At any given instant, many individual
the ectopic site are not conducted through instantaneous electrical vectors exist; each
normal pathways. one represents action potential conduction in
a different direction. An instantaneous mean
Volume Conductor Principles and electrical vector can be derived by summing
the individual instantaneous vectors.
ECG Rules of Interpretation
In the heart, the mean electrical vector
The previous section defined the components changes its orientation as different regions of
of the ECG trace and what they represent in the heart undergo depolarization or repolariza-
terms of electrical events within the heart. tion. The direction of the mean electrical vec-
This section examines in more detail how the tor relative to the axis between positive and
appearance of the recorded ECG waveform negative recording electrodes determines the
depends on (1) location of recording electrodes polarity and influences the magnitude of the
on the body surface; (2) conduction pathways recorded voltage as illustrated in Figure 2.16.
and speed of conduction; and (3) changes in This illustration depicts the sequence of
muscle mass. To interpret the significance of depolarization within the ventricles by show-
changes in the appearance of the ECG, we ing four different mean vectors representing
must first understand the basic principles of different times during depolarization. In this
how the ECG is generated and recorded. model, the septum and free walls of the left
and right ventricles are shown, and each of
VECTORS AND MEAN ELECTRICAL AXIS the four vectors is depicted as originating from
The ECG records time-dependent changes in the top of the septum where the left and right
electrical activity within the heart. At a given bundle branches divide. The size of the vector
instant in time, the recording electrodes “see” arrow is related to the mass of tissue undergo-
a summation of all the regions of the heart ing depolarization; the larger the arrow (and
that are undergoing depolarization or repo- tissue mass), the greater the measured voltage.
larization. To help understand this concept, The placement of the positive recording elec-
Figure 2.15 illustrates waves of depolarization trodes represents leads II and aVL (described
originating within the SA node and then later in this chapter). Before the ventricles
spreading into the atrial muscle. When the SA undergo depolarization (Panel A), there are
node fires, many separate depolarization waves no electrical vectors so the voltage recording
emerge from the SA node and travel throughout in either lead will be zero. Early during ven-
the atria. These separate waves can be depicted tricular activation (Panel B), the first region
as arrows representing individual electrical to depolarize is the interventricular septum,
which normally depolarizes from left to right
as depicted by the mean electrical vector. The
SA Node vector is small because the tissue mass is small.
Because the vector is heading away from the
aVL positive electrode, this results in a negative
Mean Electrical
voltage in that lead (Q wave of the QRS). The
Vector same mean vector, however, when recorded
using lead II will not show a change in volt-
age (no Q wave) because the mean vector is
oriented perpendicular to the lead II axis.
Atrial Muscle About 20 milliseconds later (Panel C), the sep-
tum is completely depolarized and the apex
■ FIGURE 2.15 Electrical vectors. Individual of the heart begins to depolarize. At this time,
instantaneous vectors of depolarization (black the mean electrical vector points downward
arrows) spread across the atria after the sinoatrial
(SA) node fires. The mean electrical vector (red
toward the apex and is heading roughly per-
arrow) represents the sum of the individual vec- pendicular to the aVL lead axis, thereby gener-
tors at a given instant in time. ating only a very small positive voltage in aVL.

Klabunde_Chap02.indd 30 6/11/2011 10:28:42 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 31

aVL aVL
+ +
Septum

LV free
wall

RV free Apex
wall
A B
+ +
II II

aVL aVL
+ +

C
+
D +
II II

aVL aVL
+ +

Mean
Electrical
Axis

E + F +
II II

■ FIGURE 2.16 Generation of QRS complex from two different recording electrodes. A. Ventricles prior to
depolarization; isoelectric (zero) voltage recorded by electrodes aVL and II. B. Septal depolarization; volt-
age aVL < II. C. Apical depolarization; voltage aVL < II. D. Left ventricular depolarization (primarily); voltage
aVL > II. E. Left ventricular depolarization; voltage in aVL > II. F. Ventricles depolarized; isoelectric voltage
in aVL and II; red arrow represents mean electrical axis.

In contrast, the mean vector is heading almost electrode and is almost perpendicular to the
directly towards the lead II positive electrode, lead II axis. Therefore, this vector produces a
which results in a very tall, positive deflection large positive voltage in lead aVL and a rela-
(R wave of the QRS). After another 20 milli- tively small positive voltage in lead II. The last
seconds (Panel D), the apex and most of the regions of the left ventricle to depolarize (Panel
right ventricular free wall are completely depo- E) result in a mean vector that is heading some-
larized. At this time, the left ventricular free what toward lead aVL, and away from lead II.
wall depolarizes from the endocardial (inside) Therefore, aVL will still record a small positive
to epicardial (outside) surface. The resulting voltage, whereas lead II will record a small neg-
mean vector is mostly heading toward the aVL ative voltage (S wave of the QRS). When the

Klabunde_Chap02.indd 31 6/11/2011 10:28:42 AM


32 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

ventricles are completely depolarized (Panel each arm and leg, and six electrodes are placed
F), the voltage in all recording leads will be at defined locations on the chest. Three basic
zero. It is important to note that the placement types of ECG leads are recorded by these elec-
of the recording electrode determines the shape trodes: standard limb leads, augmented limb
of the QRS complex that is recorded. leads, and chest leads. These electrode leads are
If the four mean vectors in Figure 2.16 (Pan- connected to a device that measures potential
els B-E) are summed, the resultant vector (Panel differences between selected electrodes to pro-
F, large red arrow) is the mean electrical axis. duce the characteristic ECG tracings. The limb
The mean electrical axis represents the aver- leads are sometimes referred to as bipolar leads
age of all of the instantaneous mean electrical because each lead uses a single pair of positive
vectors occurring sequentially during ven- and negative electrodes. The augmented leads
tricular depolarization. The determination and chest leads are unipolar leads because
of mean electrical axis is of particular signifi- they have a single positive electrode with the
cance for the ventricles and is used diagnosti- other electrodes coupled together electrically
cally to identify left and right axis deviations, to serve as a common negative electrode.
which can be caused by a number of factors
including conduction blocks in a bundle ECG LIMB LEADS
branch and ventricular hypertrophy.
Based on the previous discussion, the follow- Standard limb leads are shown in Figure 2.17.
ing rules can be used in interpreting the ECG: Lead I has the positive electrode on the left arm
and the negative electrode on the right arm,
1. A wave of depolarization (instantaneous therefore measuring the potential difference
mean electrical vector) traveling toward across the chest between the two arms. In this
a positive electrode results in a posi- and the other two limb leads, an electrode on
tive deflection in the ECG trace. (Corol- the right leg is a reference electrode for record-
lary: A wave of depolarization traveling ing purposes. In the lead II configuration, the
away from a positive electrode results in a positive electrode is on the left leg and the
negative deflection.)
2. A wave of repolarization traveling toward
a positive electrode results in a negative
deflection. (Corollary: A wave of repolari- RA _ LA
zation traveling away from a positive elec- +
trode results in a positive deflection.) _ _
3. A wave of depolarization or repolariza-
tion oriented perpendicular to an elec-
trode axis produces no net deflection.
4. The instantaneous amplitude of the
measured potentials depends upon the ori-
entation of the positive electrode relative
to the mean electrical vector.
5. Voltage amplitude (positive or negative) is
directly related to the mass of tissue under-
going depolarization or repolarization. + +

ECG Leads: Placement of RL LL


Recording Electrodes ■ FIGURE 2.17 Placement of the standard ECG
limb leads (leads I, II, and III) and the location of
The ECG is recorded by placing an array of
the positive and negative recording electrodes for
electrodes at specific locations on the body sur- each of the three leads. RA, right arm; LA, left arm;
face. Conventionally, electrodes are placed on RL, right leg; LL, left leg.

Klabunde_Chap02.indd 32 6/11/2011 10:28:42 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 33

negative electrode is on the right arm. Lead III lead III when the depolarization wave travels
has the positive electrode on the left leg and the parallel to the axis between the left arm and
negative electrode on the left arm. These three left leg.
limb leads roughly form an equilateral triangle If the three limbs of Einthoven triangle are
(with the heart at the center), called Einthoven broken apart, collapsed, and superimposed
triangle in honor of Willem Einthoven who over the heart (Fig. 2.18), the positive elec-
developed the ECG in 1901. Whether the trode for lead I is defined as being at zero
limb leads are attached to the end of the limb degrees relative to the heart (along the hori-
(wrists and ankles) or at the origin of the limbs zontal axis; see Fig. 2.18). Similarly, the posi-
(shoulder and upper thigh) makes virtually no tive electrode for lead II is +60° relative to the
difference in the recording because the limb heart, and the positive electrode for lead III is
can be viewed as a wire conductor originating +120° relative to the heart, as shown in Fig-
from a point on the trunk of the body. ure 2.18. This new construction of the electri-
When using the ECG rules described in cal axis is called the axial reference system.
the previous section, a wave of depolariza- Although the designation of lead I as being 0°,
tion heading toward the left arm gives a posi- lead II as being +60°, and so forth is arbitrary,
tive deflection in lead I because the positive it is the accepted convention. With this axial
electrode is on the left arm. Maximal positive reference system, a wave of depolarization
deflection of the tracing occurs in lead I when oriented at +60° produces the greatest positive
a wave of depolarization travels parallel to the deflection in lead II. A wave of depolarization
axis between the right and left arms. If a wave oriented +90° relative to the heart produces
of depolarization heads away from the left equally positive deflections in both leads II
arm, the deflection is negative. In addition, and III. In the latter case, lead I shows no net
a wave of repolarization moving away from deflection because the wave of depolarization
the left arm is seen as a positive deflection. is heading perpendicular to the 0°, or lead I,
Similar statements can be made for leads axis (see ECG rules).
II and III, with which the positive electrode Three augmented limb leads exist in addi-
is located on the left leg. For example, a wave tion to the three bipolar limb leads described.
of depolarization traveling toward the left Each of these leads has a single positive elec-
leg gives a positive deflection in both leads trode that is referenced against a combination
II and III because the positive electrode for of the other limb electrodes. The positive elec-
both leads is on the left leg. A maximal posi- trodes for these augmented leads are located
tive deflection is obtained in lead II when the on the left arm (aVL), the right arm (aVR), and
depolarization wave travels parallel to the axis the left leg (aVF; the “F” stands for “foot”).
between the right arm and left leg. Similarly, In practice, these are the same positive elec-
a maximal positive deflection is obtained in trodes used for leads I, II, and III. (The ECG

RA I LA

I 0° Lead I
II III

III II
LL +120° +60°
Lead III Lead II
Einthoven’s Triangle Axial Reference System

■ FIGURE 2.18 Transformation of leads I, II, and III from Einthoven triangle into the axial reference system. Leads
I, II, and III correspond to 0°, +60°, and +120° in the axial reference system. RA, right arm; LA, left arm; LL, left leg.

Klabunde_Chap02.indd 33 6/11/2011 10:28:43 AM


aVR -150° -30° aVL

+120° +90° +60°


aVF

Klabunde_Chap02.indd 34 6/11/2011 10:28:43 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 35

ELECTROPHYSIOLOGICAL
CHANGES DURING CARDIAC
ISCHEMIA

The ECG is a key tool for diagnosing myocar-


V2 V6 dial ischemia and infarction. A 12-lead ECG
V1
can identify the extent, location, and progress
V3 V4 V5 of damage to the heart following ischemic
injury. For example, altered conduction can
result in exaggerated Q waves in specific leads
following some types of myocardial infarction.
Posterior Ischemia can also damage conduction path-
ways, leading to arrhythmias or changes in
the shape of the QRS complex. Furthermore,
LV V6 Left ischemia can produce injury currents flow-
RV Lateral ing from the depolarized ischemic regions to
V5 normal regions that can shift the isoelectric
V4 portions of the ECG, resulting in upward or
V1 V2 V3 downward shifts in the ST segment recorded
Anterior by overlying electrodes.
The mechanisms by which ischemia and
V1 V2 V3 infarction alter the ECG are complex and not
fully understood. We do know, however, that
tissue hypoxia caused by ischemia results
in membrane depolarization. As ATP levels
decline during hypoxia, there is a net loss of
K+ as it leaks out of cells through KATP channels
V4 V5 V6 (normally inhibited by ATP) and as a result
of decreased activity of the Na+/K+-ATPase
pump. Increased extracellular K+, coupled
■ FIGURE 2.20 Placement of the six precordial with decreased intracellular K+, causes mem-
chest leads and the normal appearance of the brane depolarization. This depolarization
ECG recording for leads V1 − V6. These electrodes inactivates fast sodium channels as previously
record electrical activity in the horizontal plane,
described, thereby decreasing action poten-
which is perpendicular to the frontal plane of the
limb leads. tial upstroke velocity. One result is decreased
conduction velocity. Changes in refractory
period and conduction velocity can lead to
and V6 overlies the left ventricular lateral wall. reentry currents and tachycardia. Membrane
The rules of interpretation are the same as for the depolarization also alters pacemaker activity
limb leads. For example, a wave of depolarization and can cause latent pacemakers to become
traveling toward a particular electrode on the active, leading to changes in rhythm and
chest surface elicits a positive deflection. Normal ectopic beats. Finally, cellular hypoxia results
electrical activation of the ventricles results in a in the accumulation of intracellular calcium,
net negative deflection in V1 and a net positive which can lead to afterdepolarizations and
deflection in V6 as shown in Figure 2.20. tachycardia.

Klabunde_Chap02.indd 35 6/11/2011 10:28:43 AM


36 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

SUMMARY OF IMPORTANT CONCEPTS

• The membrane potential is determined • At rest, SA nodal pacemaker activity


primarily by the concentration of sodium, is strongly influenced by vagal activ-
potassium, and calcium ions across the ity (vagal tone), which significantly
cell membrane, and by the relative con- reduces the intrinsic SA nodal firing
ductances of the membrane to these ions. rate. Pacemaker activity is increased
• The resting membrane potential is very by sympathetic activation and vagal
close to the potassium equilibrium inhibition.
potential (calculated from Nernst equa- • Conduction of action potentials within
tion) because the relative conductance the heart is primarily cell-to-cell,
of potassium is much higher than the although specialized conduction path-
relative conductances of sodium and ways exist within the heart that ensure
calcium in the resting cell. rapid distribution of the conducted
• Ions move across the cell membrane action potentials. Conduction velocity
through ion-selective channels, which is increased by activation of sympa-
have open (activated) and closed thetic nerves and decreased by para-
(inactivated) states that are regulated sympathetic activation.
by either membrane voltage or by • The low conduction velocity within the
receptor-coupled mechanisms. AV node ensures sufficient time for
• Concentrations of sodium, potassium, atrial contraction to contribute to ven-
and calcium across the cell membrane tricular filling.
are maintained by the Na+jK+-ATPase • Cells located within the AV node and
pump, the Na+/Ca++ exchanger, and the ventricular conducting system can also
Ca++-ATPase pump. serve as pacemakers if the SA node
• Nonpacemaker cardiac action potentials fails or conduction is blocked between
are characterized as having very nega- the atria and ventricles (AV block).
tive resting potentials (approximately • The ECG evaluates rhythm and con-
-90 mV), a rapid phase 0 depolarization duction by examining the appearance
produced primarily by a transient (amplitude, duration, and shape) of
increase in sodium conductance, and specific waveforms that represent atrial
a prolonged plateau phase (phase 2) depolarization (P wave), ventricular
generated primarily by inward calcium depolarization (QRS complex), and
currents through L-type calcium chan- ventricular repolarization (T wave).
nels; increased potassium conductance • Different ECG leads view the electri-
repolarizes the cells during phase 3. cal activity of the heart from different
• Pacemaker action potentials (e.g., angles. Each limb lead can be repre-
those found in SA nodal cells) spon- sented by an electrical axis on a fron-
taneously depolarize during phase 4, tal plane from which the direction of
owing in part to special pacemaker cur- depolarization and repolarization vec-
rents (1 1). Upon reaching the threshold tors within the heart can be determined
for action potential generation, cal- using standard rules of interpretation
cium conductance increases as L-type (e.g., a wave of depolarization traveling
calcium channels become activated, toward a positive electrode produces
which causes depolarization (phase 0). a positive voltage in the ECG). Chest
As the calcium channels close, potas- leads (V, to V 6 ) measure the electri-
sium conductance increases and the cal activity in a horizontal plane that is
cell repolarizes (phase 3). perpendicular to the frontal plane.

Klabunde_Chap02.indd 36 6/11/2011 10:28:44 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 37

REVIEW QUESTIONS

For each question, choose the one best 5. The normal sequence of conduction
answer: within the heart is
a. SA node ~ atrioventricular (AV)
l. Which one of the following depolar- node ~ bundle of His ~ bundle
izes the resting membrane potential in a branches ~ Purkinje fibers
cardiac myocyte? b. SA node ~bundle of His ~ AV node
a. Decreased calcium conductance ~bundle branches ~ Purkinje fibers
b. Decreased sodium conductance c. AV node ~ SA node ~ bundle of His
c. Increased potassium conductance ~ bundle branches ~ Purkinje fibers
d. Inhibition of the sarcolemmal Na+fK+- d. SA node ~ AV node ~ bundle of His
ATPase ~ Purkinje fibers ~bundle branches

2. In nonnodal cardiac tissue, fast sodium 6. A patient is found to have a PR interval


channels are inactivated that is >0.2 seconds. Which of the fol-
lowing interventions would most likely
a. During phase 0. reduce the PR interval?
b. When the h-gates open.
c. By slow depolarization of the cell. a. Blocking ~-adrenoceptors
d. More slowly than L-type calcium b. Blocking muscarinic (M2) receptors
channels are inactivated. c. Blocking L-type calcium channels
d. Enhancing vagal nerve activity

3. The relative potassium conductance 7. In a normal ECG,


is highest during which of the fol-
lowing phases of a ventricular action a. The PR interval is >0.2 seconds.
potential? b The ST segment represents the dura-
tion of the ventricular action potential.
a. Phase 0 c. The T wave represents ventricular
b. Phase 2 repolarization.
c. Early phase 3 d. The duration of the QRS is >0.2 seconds.
d. Late phase 4
8. A patient is found to have normal QRS
4. The rate of sinoatrial (SA) nodal action but inverted T waves in leads II, III, and
potential firing increases during exercise. aVF Which of the following is the most
Which of the following mechanisms can likely explanation for these findings?
cause this increase in rate? a. The direction of ventricular repolari-
a. ~-adrenoceptoractivation increasing zation is reversed from normal.
"funny" currents (Ir) b. The polarity of the recording elec-
b. Decreasing the slope of phase 4 by trodes is reversed.
vagal (parasympathetic) activation c. Ventricular depolarization and
c. Inactivation of fast sodium channels repolarization are occurring in
d. Increased potassium conductance opposite directions.
during phase 4 d. Ventricular depolarization is abnormal.

Klabunde_Chap02.indd 37 6/11/2011 10:28:46 AM


38 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

9. The following ECG results are obtained 10. An ECG rhythm strip shows a complete
from a patient: The QRS is equally dissociation between P waves and QRS
biphasic in lead II (no net deflection), complexes. The atrial rate is 95 beats/min
and the QRS has a net positive voltage in and regular, and the ventricular rate is
lead aVL. What is the approximate mean about 60 beats/min and regular. The QRS
electrical axis? complexes are of normal shape and dura-
a. -30° tion. This ECG represents
b. 0° a. First-degree AV nodal block.
c. +60° b. Second-degree AV nodal block.
d. +120° c. Third-degree AV nodal block.
d. Premature ventricular complexes.

1. The correct answer is "d" because and "c" are incorrect because the overall
the sarcolemmal Na+fK+-ATPase is potassium conductance is reduced dur-
an electrogenic pump that generates ing phases 0 through 2, and it begins to
hyperpolarizing currents; inhibition recover only during early phase 3.
of this pump results in depolarization. 4. The correct answer is "a" because one
Furthermore, inhibition of the pump effect of ~-adrenoceptor activation is to
leads to an increase in intracellular sodi- increase If' which enhances the rate of
urn and a decrease in intracellular potas- spontaneous depolarization. Choice "b"
sium, both of which cause depolariza- is incorrect because vagal stimulation
tion. Choices "a" and "b" are incorrect reduces pacemaker firing rate, in part,
because decreased calcium and sodium by decreasing the slope of phase 4.
conductance reduces the inward move- Choice "c" is incorrect because fast
ment of positive charges that normally sodium channels do not play a role
depolarize the membrane. Choice "c" is in SA nodal action potentials; inward
incorrect because increased potassium calcium currents are responsible for
conductance hyperpolarizes the mem- phase 0. Choice "d" is incorrect because
brane (see Equation 2-4). increasing potassium conductance dur-
2. The correct answer is "c" because ing phase 4 hyperpolarizes the cell so it
slow depolarization leads to closure of takes longer to reach threshold.
the h-gates, which inactivates the fast 5. The correct sequence of activation and
sodium channels. Choice "a" is incor- conduction within the heart is choice "a".
rect because the m-gates open at the 6. The correct answer is "b" because ace-
onset of phase 0, which activates the tylcholine released by the vagus nerve
fast sodium channels. Choice "b" is binds to M2 receptors, which decreases
incorrect because it is the closure of AV nodal conduction velocity and
the h-gates that inactivates the channel. increases the PR interval. Removal of
Choice "d" is incorrect because L-type vagal tone through the use of a musca-
(long-lasting) calcium channels have rinic receptor antagonist (e.g., atropine)
a prolonged phase of activation before leads to an increase in conduction
they become inactivated. velocity. Choice "a" is incorrect because
3. The correct answer is "d" because the blocking ~-adrenoceptors would
membrane potential during phase 4 is decrease the influence of sympathetic
primarily determined by the high potas- nerves on the AV node and lead to
sium conductance. Choices "a," "b," a decrease in conduction velocity.

Klabunde_Chap02.indd 38 6/11/2011 10:28:46 AM


CHAPTER 2 • ELECTRICAL ACTIVITY OF THE HEART 39

Choice "c" is incorrect because L-type wave are upright. Choice "d" is incorrect
calcium channel blockers reduce con- because the QRS, which represents ven-
duction velocity by decreasing the rate of tricular depolarization, is normal.
calcium entry into the cells during depo- 9. The correct answer is "a" because when
larization, which decreases the slope lead II is biphasic, the mean electrical
of phase 0 in AV nodal cells, thereby axis must be perpendicular to that lead,
decreasing conduction velocity. Choice and therefore it is either -30° or +150°.
"d" is incorrect because enhancing vagal Because aVL is positive, the mean electri-
activity decreases AV nodal conduction cal axis must be -30° because that is the
velocity and increases the PR interval. axis for aVL. All the other choices are
7. The correct answer is "c" because the therefore incorrect.
T wave represents repolarization of the 10. The correct answer is "c" because
ventricular muscle. Choice "a" is incor- a complete dissociation between P
rect because the normal PR interval is waves and QRS complexes indicates a
between 0.12 and 0.20 seconds. Choice complete (third-degree) AV nodal block.
"b" is incorrect because the duration Furthermore, the rate of ventricular
of the ventricular action potential is depolarizations and the normal shape
most closely associated with the QT and duration of the QRS complexes
interval. Choice "d" is incorrect because suggest that the pacemaker driving
the duration of the QRS complex is ventricular depolarization lies within
normally <0.1 seconds. the AV node or bundle of His so that
8. The correct answer is "a" because the T conduction follows normal ventricu-
wave is normally positive when the last lar pathways. Choice "a" is incorrect
cells that depolarize are the first to repo- because a first-degree AV nodal block
larize. When the direction of repolariza- increases only the PR interval. Choice
tion is reversed, the T wave becomes "b" is incorrect because all of the QRS
inverted. Choice "b" is incorrect because complexes would still be preceded by a
accidental reversal of the electrode polar- P wave in a second-degree block. Choice
ity would lead to an inverted QRS and "d" is incorrect because premature
inverted T wave. Choice "c" is incorrect ventricular complexes normally have
because when depolarization and repo- an irregular discharge rhythm and the
larization occur in opposite directions QRS is abnormally shaped and has a
(which is normal), both the QRS and T longer-than-normal duration.

PROBLEM 2-1 of electrogenic pumps to the membrane


Using Equation 2-1, the membrane potential potential. Nevertheless, a high concentration
(actually, the equilibrium potential for potas- of external potassium causes a large depolari-
sium) with 4 mM external potassium would zation, as predicted by the Nernst equation.
be -96 m V. Solving the equation for 40 mM
external potassium results in a membrane PROBLEM 2-2
potential of -35 m V. This is the membrane Because phase 0 of myocyte action poten-
potential predicted by the Nernst equation tials is generated by activation of fast sodium
assuming that no other ions contribute to the channels, partial inactivation of these chan-
membrane potential (see Equation 2-3). This nels would decrease the upstroke velocity of
calculation also neglects any contribution phase 0 (decrease the slope of phase 0). Partial

Klabunde_Chap02.indd 39 6/11/2011 10:28:46 AM


40 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

inactivation also would decrease the maximal Therefore, taking the patient off the β-blocker
degree of depolarization. These changes in might improve AV nodal conduction and
phase 0 would reduce the conduction velocity thereby decrease the PR interval to within the
within the ventricle. Blockade of fast sodium normal range (0.12 to 0.20 seconds).
channels is the primary mechanism of action
of Class I antiarrhythmic drugs such as quini- CASE 2-3
dine and lidocaine. The QRS complex has no net voltage in lead
I (i.e., equally positive and negative voltages),
CASE 2-1
which indicates that the mean electrical axis
Reentry requires that cells can be prematurely is perpendicular (90°) to lead I (see Rule 3);
reexcited by action potentials emerging from therefore, it is either at −90° or +90° because the
adjacent conducting pathways. By increas- axis for lead I is 0° by definition. Because the
ing the ERP of these cells, the action poten- QRS is positive in leads II and III, the mean
tial emerging from adjacent pathways may electrical axis must be oriented toward the
encounter tissue that is still refractory and positive electrode on the left leg, which is used
therefore unexcitable, thereby preventing or for leads II and III. Therefore, the mean elec-
abolishing reentry. trical axis cannot be −90°, but is instead +90°.
CASE 2-2 Both aVL and aVR leads would have net negative
Sympathetic nerve activity increases conduc- QRS voltages because the direction of the mean
tion velocity within the AV node (positive electrical axis is away from these two leads,
dromotropic effect). This effect on the AV which are oriented at −30° and −150°, respec-
node is mediated by norepinephrine binding tively (see Fig. 2.19). Furthermore, the net neg-
to β-adrenoceptors within the nodal tissue. ative deflections in these two augmented leads
A β-blocker would remove this sympathetic would be of equal magnitude because each lead
influence and slow conduction within the AV axis differs from the mean electrical axis by the
node, which might prolong the PR interval. same number of degrees.

SUGGESTED RESOURCES Lilly LS. Pathophysiology of Heart Disease. 5th Ed.


Dubin D. Rapid Interpretation of EKGs. 6th Ed. Tampa: Philadelphia: Lippincott Williams & Wilkins, 2011.
Cover Publishing, 2000. Opie LH. The Heart: Physiology from Cell to Circula
Katz AM. Physiology of the Heart. 4th Ed. Philadelphia: tion. 4th Ed. Philadelphia: Lippincott Williams &
Lippincott Williams & Wilkins, 2006. Wilkins, 2004.

Klabunde_Chap02.indd 40 6/11/2011 10:28:47 AM


0

3
:I:
>
CELLULAR STRUCTURE 'tl
-1
m
;c
AND FUNCTION
Understanding the concepts presented in this chapter will enable the student to:
1. Describe the structure and function of the following cellular components of
cardiac myocytes: sarcolemma, intercalated disks, transverse (T)-tubules,
myofilaments, sarcomeres, sarcoplasmic reticulum, and terminal cisternae.
2. List the steps of excitation-contraction coupling, and describe the cellular
mechanisms involved in its regulation .

3. List in order of preference the metabolic substrates used by the heart, and
summarize the importance of oxidative metabolism relative to anaerobic
metabolism.
4. Describe the major histological structures of a muscular artery and the function
of these structures.
5. Contrast the organization of actin and myosin in vascular smooth muscle with
the organization of these myofilaments in cardiac myocytes.
6. Describe the mechanisms and regulation of vascular smooth muscle contraction
and relaxation.
7. Compare the major G-protein signal transduction pathways of cardiac muscle
and vascular smooth muscle and how these pathways regulate contraction.
8. Describe the effects of endothelial-derived nitric oxide (NO), prostacyclin
(PGI 2 ), and endothelin-1 (ET-1) on vascular function.

INTRODUCTION Although cardiac muscle shares some struc-


tural and functional similarities with skeletal
Many different cell types are associated with muscle, it has several important differences.
the cardiovascular system. This chapter exam- Cardiac myocytes are generally single nucle-
ines the structure and function of three major ated and have a diameter of approximately
types of structural cells that serve important 25 Jlm and a length of about 100 Jlm in the
roles in cardiovascular function: cardiac myo- ventricle (atrial myocytes are smaller). In con-
cytes, vascular smooth muscle, and vascular trast, although some types of skeletal muscle
endothelium. myocytes may have a similar diameter, their
cell lengths run the entire length of the mus-
CARDIAC CELL STRUCTURE cle and therefore can be many centimeters
AND FUNCTION long. Cardiac myocytes form a branching net-
work of cells that is sometimes referred to as
Myocytes and Sarcomeres a functional syncytium, which results from
a fusion of cells. Individual myocytes con-
Cardiac myocytes represent a type of stri- nect to each other by way of specialized cell
ated muscle, so called because crossbands or membranes called intercalated disks. Gap
cross striations are observed microscopically. junctions within these intercellular regions
41

Klabunde_Chap03.indd 41 6/10/2011 8:52:44 PM


42 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

serve as low-resistance pathways between The sarcomere contains thick and thin
cells, permitting cell-to-cell conduction of filaments, which represent about 50% of
electrical (ionic) currents. Therefore, if one the cell volume (see Fig. 3.1). Thick fila-
cardiac myocyte is electrically stimulated, ments are comprised of myosin, whereas thin
cell-to-cell conduction ensures that the elec- filaments contain actin and other associated
trical impulse will travel to all of the intercon- proteins. Chemical interactions between the
nected myocytes. This arrangement allows actin and myosin filaments during the pro-
the heart to contract as a unit (i.e., as a syn- cess of excitation–contraction coupling (see
cytium). In contrast, individual skeletal mus- the next section) cause the sarcomere to
cle cells are innervated by motor neurons, shorten as the myosin and actin filaments
which utilize neuromuscular transmission to slide past each other, thereby shortening the
activate individual muscle fibers to contract. distance between the Z-lines. Within the sar-
No cell-to-cell electrical conduction occurs in comere, a large, filamentous protein called
skeletal muscle. titin exists. It connects the myosin filament to
The cardiac myocyte is composed of bun- the Z-lines, which helps to keep the thick fila-
dles of myofibrils that contain myofilaments ment centered within the sarcomere. Because
(Fig. 3.1). When myocytes are viewed micro- of its elastic properties, titin plays an impor-
scopically, distinct repeating lines and bands tant role in the passive mechanical properties
can be seen, each of which represents differ- of the heart (see Chapter 4). In addition to
ent myofilament components. The segment titin, myosin, and actin, a number of other
between two Z-lines represents the basic con- proteins form the cytoskeleton of myocytes,
tractile unit of the myocyte, the sarcomere. connecting the internal and external cell
The length of each sarcomere under physi- components.
ologic conditions ranges from about 1.6 to Myosin is a large molecular weight protein.
2.2 μm in human hearts. As described later Within each sarcomere, myosin molecules
and in Chapter 4, the length of the sarcomere are bundled together so that there are about
is an important determinant of the force of 300 molecules of myosin per thick filament.
myocyte contraction. Each myosin molecule contains two heads,

Intercalated
Sarcomere disc

Myofilaments

Myofibrils

Myocyte

Z Z
Myosin
Actin Titin
■ FIGURE 3.1 Structure of cardiac myocytes. Myocytes are joined together by intercalated disks to form a
functional syncytium (right side of the figure). Myocytes are composed of myofibrils, each of which con-
tains myofilaments that are composed largely of actin (thin filaments) and myosin (thick filaments) (left
side of the figure). Myosin is anchored to the Z-line by the protein titin. The sarcomere, or basic contractile
unit, lies between two Z-lines.

Klabunde_Chap03.indd 42 6/10/2011 8:52:47 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 43

which serve as the site of myosin adenosine subunits: troponin-T (TN-T), which attaches
triphosphatase (myosin ATPase), an enzyme to the tropomyosin; troponin-C (TN-C),
that hydrolyzes adenosine triphosphate which serves as a binding site for Ca++ dur-
(ATP). ATP is required for the cross-bridge ing excitation–contraction coupling; and
formation between the thick and thin fila- troponin-I (TN-I), which inhibits myosin
ments. The molecule’s heads interact with a binding to actin. The troponin complex holds
binding site on actin (Fig. 3.2). Regulatory tropomyosin in position to prevent binding
subunits (myosin light chains) that can alter of myosin heads to actin. When Ca++ binds to
the ATPase activity when phosphorylated are TN-C, a conformational change occurs in the
associated with each myosin head. troponin complex such that the troponin–
Each thick filament is surrounded by a tropomyosin complex moves away from the
hexagonal arrangement of six thin filaments. myosin-binding site on the actin, thereby
The thin filaments are composed of actin, making the actin accessible to the myosin
tropomyosin, and troponin (Fig. 3.2). Actin head for binding. When Ca++ is removed from
is a globular protein arranged as a chain of the TN-C, the troponin–tropomyosin com-
repeating globular units, forming two heli- plex resumes its inactivated position, thereby
cal strands. Interdigitated between the actin inhibiting myosin–actin binding. As a clini-
strands are rod-shaped proteins called tropo- cal aside, both TN-I and TN-T are used as
myosin. Each tropomyosin molecule is asso- diagnostic markers for myocardial infarction
ciated with seven actin molecules. Attached to because of their release into the circulation
the tropomyosin at regular intervals is the tro- when myocytes die.
ponin regulatory complex, made up of three
Excitation–Contraction Coupling
TRANSVERSE TUBULES AND THE
Myosin Myosin SARCOPLASMIC RETICULUM
Heads
The coupling between myocyte action poten-
tials and contraction is called excitation–
contraction coupling. To understand this
TN-C process, the internal structure of the myo-
TN-I TN-T cyte needs to be examined in more detail.
The sarcolemmal membrane of the myocyte
surrounds the bundle of myofibrils and has
deep invaginations called transverse (T)
tubules (Fig. 3.3), particularly in ventricular
Tropomyosin myocytes. The T tubules, being a part of the
Actin external sarcolemma, are open to the exter-
■ FIGURE 3.2 Composition of cardiac thick and nal environment of the cell. This permits ions
thin myofilaments. The thick filaments are com- to exchange between extracellular and intra-
posed of myosin molecules, with each molecule
cellular compartments to occur deep within
having two myosin heads, which serve as the
site of the myosin ATPase. Thin filaments are the myocyte during electrical depolarization
composed of actin, tropomyosin, and regulatory and repolarization of the myocyte. Within
proteins (troponin complex, TN) having three sub- the cell, and in close association with the
units: TN-T (binds to tropomyosin), TN-C (binds to
calcium ions), and TN-I (inhibitory troponin, which
T tubules, is an extensive, branching tubular
inhibits myosin binding to actin). Calcium binding network called the sarcoplasmic reticulum
to TN-C produces a conformation change in the that surrounds the myofilaments. The pri-
troponin–tropomyosin complex that exposes a mary function of this structure is to regulate
myosin-binding site on the actin, leading to ATP
hydrolysis. For simplicity, this figure shows only
intracellular calcium concentrations, which
one actin strand and its associated tropomyosin is involved with contraction and relaxation.
filament. Terminal cisternae are end pouches of the

Klabunde_Chap03.indd 43 6/10/2011 8:52:47 PM


44 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

++
Sarcolemma Ca

Myosin

Ca++ Ca++ L-type


TN-C calcium
channels
SR
Actin RyR
SERCA Phospho- “feet”
T-tubule
lamban

■ FIGURE 3.3 Role of calcium (Ca++) in cardiac excitation–contraction coupling. During action potentials,
Ca++ enters cell through L-type Ca++ channels. This so-called trigger Ca++ is sensed by the “feet” of the
calcium-release channel (ryanodine receptor, RyR) of the sarcoplasmic reticulum (SR), which releases
Ca++ into the cytoplasm. This Ca++ binds to troponin-C (TN-C), inducing a conformational change in the
troponin–tropomyosin complex so that movement of the troponin–tropomyosin complex exposes a
myosin-binding site on actin, leading to ATP hydrolysis and movement of actin relative to myosin. Ca++ is
resequestered into the SR by an ATP-dependent Ca++ pump, sarcoendoplasmic reticulum calcium ATPase
(SERCA) that is inhibited by phospholamban. Not shown are Ca++ pumps that remove Ca++ from the cell.

sarcoplasmic reticulum that are adjacent to receptors, or ryanodine-sensitive calcium-


the T tubules. Between the terminal cisternae release channels) associated with the termi-
and the T tubules are electron-dense regions nal cisternae. This triggers the subsequent
called feet that are believed to sense calcium release of large quantities of calcium stored in
between the T tubules and the terminal cister- the terminal cisternae through the calcium-
nae. Closely associated with the sarcoplasmic release channels, which increases intracel-
reticulum are large numbers of mitochondria, lular calcium concentrations 100-fold, from
which provide the energy necessary for myo- about 10−7 to 10−5 M. Therefore, the calcium
cyte contraction. that enters the cell during depolarization is
sometimes referred to as “trigger calcium.”
The free calcium binds to TN-C in a
CALCIUM CYCLING AND THE FUNCTION
concentration-dependent manner. This induces
OF REGULATORY PROTEINS
a conformational change in the regulatory
When an action potential causes depolariza- complex such that the troponin–tropomyosin
tion of a myocyte (see Chapter 2), it initi- complex moves away from and exposes a
ates excitation–contraction coupling. When myosin-binding site on the actin molecule.
the myocyte is depolarized, calcium ions The binding of the myosin head to the actin
enter the cell during the action potential results in ATP hydrolysis, which supplies
through long-lasting (L-type) calcium chan- energy so that a conformational change can
nels located on the external sarcolemma and occur in the actin–myosin complex. This
T tubules (see Fig. 3.3). It is important to results in a movement (“ratcheting”) between
note that a relatively small amount of calcium the myosin heads and the actin. The actin
enters the cell during depolarization. By itself, and myosin filaments slide past each other,
this calcium influx does not significantly thereby shortening the sarcomere length (this
increase intracellular calcium concentrations is referred to as the sliding filament theory
except in local regions just inside the sarco- of muscle contraction) (Fig. 3.4). Ratcheting
lemma. This calcium is sensed by the “feet” cycles will occur as long as the cytosolic
of the calcium-release channels (ryanodine calcium remains elevated. Toward the end of

Klabunde_Chap03.indd 44 6/10/2011 8:52:47 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 45

the myocyte action potential, calcium entry TABLE 3-1 SUMMARY OF EXCITATION–
into the cell diminishes and the sarcoplas- CONTRACTION COUPLING
mic reticulum sequesters calcium by an ATP- 1. Ca++ enters cell during depolarization
dependent calcium pump, sarcoendoplasmic and triggers release of Ca++ by terminal
reticulum calcium ATPase (SERCA; see cisternae.
Fig. 3-3). As intracellular calcium concentra- 2. Ca++ binds to TN-C, inducing a
tion declines, calcium dissociates from TN-C, conformational change in the troponin
which causes a conformational change in the complex.
troponin–tropomyosin complex; this again 3. Myosin heads bind to actin, leading to
leads to troponin–tropomyosin inhibition of cross-bridge movement (requires ATP
the actin-binding site. At the end of the cycle, hydrolysis) and reduction in sarcomere
a new ATP binds to the myosin head, displac- length.
ing the adenosine diphosphate, and the ini- 4. Ca++ is resequestered by sarcoplasmic
tial sarcomere length is restored. Thus, ATP reticulum by the SERCA pump.
is required both for providing the energy of 5. Ca++ is removed from TN-C, and myo-
contraction and for relaxation. In the absence sin unbinds from actin (requires ATP);
of sufficient ATP as occurs during cellu- this allows the sarcomere to resume its
lar hypoxia, cardiac muscle contraction and original, relaxed length.
relaxation will be impaired. The events associ- ATP, adenosine triphosphate; SERCA, sarcoendoplasmic
ated with excitation–contraction coupling are reticulum calcium ATPase; TN-C, troponin-C.

summarized in Table 3-1.


Regulation of Contraction ultimately affect calcium handling by the cell.
Changes in contraction resulting from altered
(Inotropy)
calcium handling and myosin ATPase activity
Several cellular mechanisms regulate contrac- are referred to as inotropic changes (inotropy).
tion (Fig. 3.5). Most of these mechanisms Inotropy is modulated by (1) calcium entry

Z Titin Z
Actin
Myosin

Actin-myosin + Calcium
binding

– Calcium

■ FIGURE 3.4 Sarcomere shortening and the sliding filament theory. Calcium binding to TN-C permits
actin–myosin binding (cross-bridge formation) and ATP hydrolysis. This results in the thin filaments slid-
ing over the myosin during cross-bridge cycling, thereby shortening the sarcomere (distance between
Z-lines). Removal of calcium from the TN-C inhibits actin–myosin binding so that cross-bridge cycling
ceases and the sarcomere resumes its relaxed length.

Klabunde_Chap03.indd 45 6/10/2011 8:52:48 PM


46 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

6
Ca++

++
Ca
Myosin
4 2
++
Ca Ca
++

TN-C
3 1
SR
Actin 5 RyR L-type
calcium
SERCA Phospho- channels
lamban

■ FIGURE 3.5 Intracellular mechanisms regulating inotropy. Inotropy can be increased by increasing
Ca++ influx through L-type Ca++ channels (site 1); increasing release of Ca++ by the sarcoplasmic reticulum
(SR) (site 2); increasing troponin-C (TN-C) affinity for Ca++ (site 3); increasing myosin–ATPase activity
through phosphorylation of myosin heads (site 4); increasing sarcoendoplasmic reticulum calcium ATPase
(SERCA) activity by phosphorylation of phospholamban (site 5); or inhibiting Ca++ efflux across the
sarcolemma (site 6).

into the cell through L-type calcium channels; different sites within the cell. One important
(2) calcium release by the sarcoplasmic reticu- site of phosphorylation is the L-type calcium
lum; (3) calcium binding to TN-C; (4) myosin channel. Phosphorylation increases the per-
phosphorylation; (5) SERCA activity; and (6) meability of the channel to calcium, thereby
calcium efflux across the sarcolemma. increasing calcium influx during action
potentials. This increase in trigger calcium
CALCIUM ENTRY INTO MYOCYTES enhances calcium release by the sarcoplasmic
The amount of calcium that enters the cell reticulum, thereby increasing inotropy. There-
during depolarization (Fig. 3.5, site 1) is fore, norepinephrine and epinephrine are pos-
regulated largely by phosphorylation of the itive inotropic agents.
L-type calcium channel. The primary mech- Another G-protein, the inhibitory G-protein
anism for this regulation involves cyclic (Gi-protein), inhibits adenylyl cyclase and
adenosine monophosphate (cAMP), the for- decreases intracellular cAMP. Therefore, acti-
mation of which is coupled to β-adrenocep- vation of this pathway decreases inotropy. This
tors (Fig. 3.6). Norepinephrine released by pathway is coupled to muscarinic receptors
sympathetic nerves, or circulating epineph- (M2) that bind acetylcholine released by para-
rine released by the adrenal glands, binds sympathetic (vagal) nerves within the heart.
primarily to β1-adrenoceptors located on the Adenosine receptors (A1) also are coupled to
sarcolemma. This receptor is coupled to a the Gi-protein. Therefore, acetylcholine and
specific guanine nucleotide-binding regula- adenosine are negative inotropic agents.
tory protein (stimulatory G-protein; Gs-pro-
CALCIUM RELEASE BY THE
tein), that activates adenylyl cyclase, which
SARCOPLASMIC RETICULUM
in turn hydrolyzes ATP to cAMP. The cAMP
acts as a second messenger to activate protein Enhanced calcium release by the sarcoplasmic
kinase A (cAMP-dependent protein kinase, reticulum also can increase inotropy (Fig. 3.5,
PK-A), which is capable of phosphorylating site 2). During β-adrenoceptor and cAMP

Klabunde_Chap03.indd 46 6/10/2011 8:52:48 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 47

+ L-type
DAG PK-C Calcium
+ ++ SR
+ Ca Channel
PIP2 IP3
+
NE
PL-C Ca++ Ca ++
AII R +
ET-1 + +
Gq Contraction
PK-A
+
cAMP
ATP

_
R Gs
+ AC R
Gi

NE ACh
Epi Ado
■ FIGURE 3.6 Signal transduction pathways regulating cardiac myocyte contraction. The two major path-
ways involve formation of either cyclic adenosine monophosphate (cAMP) or inositol 1,4,5-triphosphate
(IP3), both of which affect Ca++ release by sarcoplasmic reticulum and therefore affect contraction. R, recep-
tor; Gs, stimulatory G-protein; Gi, inhibitory G-protein; Gq, phospholipase C-coupled G-protein; AC, adenylyl
cyclase; PL-C, phospholipase C; PIP2, phosphatidylinositol 4,5-bisphosphate; DAG, diacylglycerol; PK-C,
protein kinase C; PK-A, protein kinase A; SR, sarcoplasmic reticulum; ATP, adenosine triphosphate; NE,
norepinephrine; AIl, angiotensin II; ET-1, endothelin-1; Epi, epinephrine; ACh, acetylcholine; Ado, adenosine.

activation, PK-A phosphorylates sites on the calcium to TN-C (Fig. 3.5, site 3). The bind-
sarcoplasmic reticulum, leading to an increase ing of calcium to TN-C is determined by the
in calcium release. free intracellular concentration of calcium
Besides the cAMP pathway, a second path- and the binding affinity of TN-C to calcium.
way within myocytes can affect calcium release The greater the intracellular calcium concen-
by the sarcoplasmic reticulum, although this tration, the more the calcium that is bound
pathway appears to be less important physi- to TN-C, and the more the force that is gen-
ologically than the cAMP/PK-A pathway. This erated between actin and myosin. Increasing
second pathway involves a class of G-proteins the affinity of TN-C for calcium increases
(Gq-proteins; Fig. 3.6) that are associated with binding at any given calcium concentra-
α1-adrenoceptors (bind norepinephrine), angi- tion, thereby increasing force generation.
otensin II receptors (AT1), and endothelin-1 Acidosis, which occurs during myocardial
receptors (ETA). Activation of these receptors hypoxia, has been shown to decrease TN-C
stimulates phospholipase C to form inositol affinity for calcium. This may be one mecha-
triphosphate (IP3) from phosphatidylinositol nism by which acidosis decreases the force of
4,5-bisphosphate (PIP2), which stimulates cal- contraction.
cium release by the sarcoplasmic reticulum. Changes in calcium sensitivity may explain
in part how increases in sarcomere length
CALCIUM BINDING TO TN-C
(also known as preload; see Chapter 4) leads
Another mechanism by which inotropy to an increase in force generation. It appears
can be modulated is by altered binding of that increased preload increases calcium

Klabunde_Chap03.indd 47 6/10/2011 8:52:48 PM


48 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

sensitivity of TN-C, thereby increasing cal- can increase inotropy because more calcium
cium binding. The mechanism by which is available to be taken up by the sarcoplasmic
changes in length increase calcium affinity by reticulum and subsequently released.
TN-C is unknown. Digoxin and related cardiac glycosides
inhibit the Na+/K+-ATPase, which increases
MYOSIN ATPASE ACTIVITY intracellular Na+ (see Chapter 2). This leads
The myosin heads have sites (myosin light to an increase in intracellular Ca++ through the
chains) that can be phosphorylated by the Na+/Ca++ exchange pump, leading to enhanced
enzyme myosin light chain kinase (Fig. 3.5, inotropy. Cellular hypoxia also decreases the
site 4). Increased cAMP is known to be asso- activity of the Na+/K+-ATPase pump, as well as
ciated with increased phosphorylation of the the Ca++-ATPase pump, by reducing ATP avail-
myosin heads, which may increase inotropy. ability. This leads to calcium accumulation in
The physiologic significance of this mecha- the cell; however, inotropy is not increased, in
nism, however, is uncertain. part, because the lack of ATP decreases myo-
sin ATPase activity.
CALCIUM UPTAKE BY SARCOPLASMIC
RETICULUM
Regulation of Relaxation
In addition to influencing relaxation, increas- (Lusitropy)
ing calcium transport into the sarcoplasmic
The rate of myocyte relaxation (lusitropy) is
reticulum by the SERCA pump can indirectly
determined by the ability of the cell to rap-
increase the amount of calcium released by the
idly reduce the intracellular concentration
sarcoplasmic reticulum (Fig. 3.5, site 5). PK-A
of calcium following its release by the
phosphorylation of phospholamban, which
sarcoplasmic reticulum. This reduction in
removes the inhibitory effect of phospholamban
intracellular calcium causes calcium that is
on SERCA, increases the rate of calcium trans-
bound to TN-C to be released, thereby permit-
port into the sarcoplasmic reticulum. SERCA
ting the troponin–tropomyosin complex to
activity can also be stimulated by increased
resume its resting, inactivated conformation.
intracellular calcium caused by increased cal-
Several intracellular mechanisms help to
cium entry into the cell or decreased cellular
regulate lusitropy, most of which influence
efflux. Enhanced sequestering of calcium by
intracellular calcium concentrations.
the sarcoplasmic reticulum increases subse-
quent release of calcium by the sarcoplasmic 1. The rate at which calcium enters the cell
reticulum, thereby increasing inotropy. Because at rest and during action potentials influ-
the SERCA pump requires ATP, hypoxic con- ences intracellular concentrations. Under
ditions that reduce ATP production by the cell some pathologic conditions (e.g., myo-
can diminish the pump activity, thereby reduc- cardial ischemia), the cell becomes more
ing subsequent release of calcium by the sarco- permeable to calcium, leading to “calcium
plasmic reticulum and decreasing inotropy. overload,” which impairs relaxation.
2. The rate with which calcium leaves the cell
REGULATION OF CALCIUM EFFLUX
through the sarcolemmal calcium ATPase
FROM THE MYOCYTE
pump and the Na+/Ca++ exchange pump
The final mechanisms that can modulate inot- (see Chapter 2) affects intracellular concen-
ropy are the sarcolemmal Na+/Ca++ exchange trations. Inhibiting these transport systems
pump and the ATP-dependent calcium pump can cause intracellular calcium concen-
(Fig. 3.5, site 6). As described in Chapter 2, trations to increase sufficiently to impair
these pumps transport calcium out of the cell, relaxation.
thereby preventing the cell from becoming 3. The activity of the SERCA pump, which
overloaded with calcium. If calcium extrusion pumps calcium back into the sarcoplasmic
is inhibited, the rise in intracellular calcium reticulum, has a major role in determining

Klabunde_Chap03.indd 48 6/10/2011 8:52:48 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 49

intracellular calcium concentrations. Lusi- short, cardiac muscle contracts one to three
tropy can be increased by increasing SERCA times per second throughout life. Repetitive
activity through phosphorylation of phos- cycles of contraction and relaxation require
pholamban, a regulatory protein associated an enormous amount of ATP, which the heart
with SERCA. Phosphorylation of phos- must produce aerobically. This is why car-
pholamban removes its inhibitory effect diac myocytes contain such large numbers of
on SERCA. This is a normal physiologic mitochondria. In the absence of oxygen, myo-
mechanism in response to ~-adrenoceptor cytes can contract for no more than a minute.
stimulation, which increases cAMP and Unlike some types of skeletal muscle fibers
PK-A, the latter of which phosphorylates (e.g., fast twitch, glycolytic), cardiac myo-
phospholamban. Impairment of the activ- cytes have only a limited anaerobic capacity
ity of the SERCA pump, as occurs in some for meeting ATP requirements. This limited
forms of heart failure, causes intracellular anaerobic capacity coupled with a high use
calcium concentrations to rise, leading to of ATP explains why cellular ATP concentra-
impaired relaxation. tions fall and contractions weaken so rapidly
4. The binding affinity of TN-C for calcium under hypoxic conditions.
also influences lusitropy. Calcium binding Unlike many other cells in the body, car-
to TN-C can be modulated by PK-A phos- diac myocytes can use a variety of substrates
phorylation ofTN-1. This increases calcium to regenerate ATP oxidatively. For example, in
dissociation from TN-C, thereby increas- an overnight fasted state, the heart uses pri-
ing relaxation. The increased lusitropy marily fatty acids (-60%) and carbohydrates
caused by ~-adrenoceptor stimulation (-40%). Following a high-carbohydrate meal,
may be partly related to TN-I phospho- the heart can adapt to using carbohydrates
rylation. Some drugs used to increase the (primarily glucose) almost exclusively. Lac-
force of contraction (inotropic drugs) do tate can be used in place of glucose, and it
so by increasing TN-C affinity for calcium. becomes an important substrate during exer-
Although this may increase inotropy, it also cise when circulating concentrations of lac-
may lead to reduced lusitropy because the tate increase. The heart also can use amino
calcium is more tightly bound to the TN-C. acids and ketones (e.g., acetoacetate) instead
of fatty acids.
PROBLEM 3-1 Myocyte ATP use and oxygen consumption
increase dramatically when the frequency of
Describe the mechanisms by which
contraction (i.e., heart rate) and the force of
norepinephrine, after being released by
contraction are increased. Under these condi-
sympathetic nerve activation, increases
tions, more oxygen must be delivered to the
myocardial inotropy and lusitropy. Note
heart by the coronary circulation to support
that norepinephrine primarily binds to
myocyte metabolic demands. As Chapter 8 dis-
~,-adrenoceptors, although it also can
cusses, biochemical signals from the myocytes
bind to a,-adrenoceptors.
dilate the coronary blood vessels to supply addi-
tional blood flow and oxygen to meet greater
oxygen demands. This ensures that the heart is
Cardiac Myocyte Metabolism
able to generate ATP by aerobic mechanisms.
The maintenance of ionic pumps and other
transport systems in living cells requires VASCULAR STRUCTURE
significant amounts of energy, primarily in AND FUNCTION
the form of ATP. Cardiac myocytes have an
exceptionally high metabolic rate because Large blood vessels, both arterial and venous,
their primary function is to contract repeti- are composed of three layers-intima, media,
tively. Unlike skeletal muscle, in which con- and adventitia (Fig. 3.7). The intima, or
traction is often intermittent and relatively innermost layer, is composed of a single layer

Klabunde_Chap03.indd 49 6/10/2011 8:52:48 PM


50 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Adventitia
Collagen
Media Fibroblasts
Smooth muscle Vasa vasorum
Collagen Nerves
Elastin
Lumen

Intima
Endothelium

■ FIGURE 3.7 Blood vessel components. Blood vessels, except capillaries and small postcapillary venules,
are composed of three layers: intima, media, and adventitia. Capillaries and small postcapillary venules do
not have media and adventitia. The primary components are given for each layer.

of thin endothelial cells, which are separated The adventitia contains collagen, fibroblasts,
from the media by a basal lamina. In larger blood vessels (vasa vasorum found in large
vessels, a region of connective tissue also vessels), lymphatics, and autonomic nerves
exists between the endothelial cells and the (primarily sympathetic adrenergic). The
basal lamina. The media contains smooth smallest vessels, capillaries, are composed of
muscle cells, imbedded in a matrix of col- endothelial cells and a basal lamina; they are
lagen, elastin, and various glycoproteins. devoid of smooth muscle.
Depending on the size of the vessel, there
may be several layers of smooth muscle cells, Vascular Smooth Muscle Cells
some arranged circumferentially and others
CELLULAR STRUCTURE OF VASCULAR
arranged helically along the longitudinal axis
SMOOTH MUSCLE
of the vessel. The smooth muscles cells are
organized so that their contraction reduces Vascular smooth muscle cells are typically 5 to
the vessel diameter. The ratio of smooth mus- 10 μm in diameter and vary from 50 to 300 μm
cle, collagen, and elastin, each of which has in length. Numerous small invaginations (cav-
different elastic properties, determines the eolae) found in the cell membrane significantly
overall mechanical properties of the vessel. increase the surface area of the cell (Fig. 3.8).
For example, the aorta has a large amount of The sarcoplasmic reticulum is poorly devel-
elastin, which enables it to passively expand oped compared with the sarcoplasmic reticu-
and contract as blood is pumped into it from lum found in cardiac myocytes. Contractile
the heart. This mechanism enables the aorta proteins (actin and myosin) are present; how-
to dampen the arterial pulse pressure (see ever, the actin and myosin in smooth muscle are
Chapter 5). In contrast, smaller arteries and not organized into distinct bands of repeating
arterioles have a relatively large amount of units as they are in cardiac and skeletal mus-
smooth muscle, which is required for these cle. Instead, bands of actin filaments are joined
vessels to contract and thereby regulate arte- together and anchored by dense bodies within
rial blood pressure and organ blood flow. The the cell or dense bands on the inner surface
outermost layer, or adventitia, is separated of the sarcolemma, which function like Z-lines
from the media by the external elastic lamina. in cardiac myocytes. Each myosin filament is

Klabunde_Chap03.indd 50 6/10/2011 8:52:49 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 51

Dense
Calveolae Band

Dense
Body
Enlarged cross-section
of actin and myosin
■ FIGURE 3.8 Vascular smooth muscle cell structure. Actin and myosin filaments are connected by dense
bodies and dense bands. Each myosin filament is surrounded by several actin filaments. N, nucleus.

surrounded by several actin filaments. Similar hormones (e.g., epinephrine, angiotensin II),
to cardiac myocytes, vascular smooth muscle substances released by the endothelium lining
cells are electrically connected by gap junc- the vessel, and vasoactive substances released
tions. These low-resistance intercellular con- by the tissue surrounding the blood vessel.
nections allow propagated responses along the Vascular smooth muscle contraction
length of the blood vessels. For example, elec- can be initiated by electrical, chemical, and
trical depolarization and contraction of a local mechanical stimuli. Electrical depolarization
site on an arteriole can result in depolarization of the vascular smooth muscle cell membrane
at a distant site along the same vessel, indicat- using electrical stimulation elicits contraction
ing cell-to-cell propagation of the depolarizing primarily by opening voltage-dependent
currents. calcium channels (L-type calcium channels),
which causes an increase in the intracellular
VASCULAR SMOOTH MUSCLE
concentration of calcium. Membrane depolar-
CONTRACTION
ization can also occur through changes in ion
Contractile characteristics and the mecha- concentrations (e.g., depolarization induced
nisms responsible for contraction differ by high concentrations of extracellular potas-
considerably between vascular smooth mus- sium) or by the receptor-coupled opening of
cle and cardiac myocytes. Vascular smooth ion channels, particularly calcium channels.
muscle tonic contractions are slow and sus- Many different chemical stimuli, such as
tained, whereas cardiac muscle contractions norepinephrine, epinephrine, angiotensin II,
are rapid and relatively short (a few hundred vasopressin, endothelin-1, and thromboxane A2
milliseconds). In blood vessels, the smooth can elicit contraction. Each of these substances
muscle is normally in a partially contracted binds to specific receptors on the vascular
state, which determines the resting tone or smooth muscle cell. Different signal transduc-
diameter of the vessel. This tonic contraction tion pathways converge to increase intracellu-
is determined by stimulatory and inhibitory lar calcium, thereby eliciting contraction.
influences acting on the vessel. As described Mechanical stimuli in the form of passive
in later chapters, the most important of these stretching of smooth muscle in some arter-
are sympathetic adrenergic nerves, circulating ies can cause a contraction that originates

Klabunde_Chap03.indd 51 6/10/2011 8:52:49 PM


52 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

from the smooth muscle itself and is therefore Intracellular calcium concentrations,
termed a myogenic response. This probably therefore, are very important in regulating
results from stretch-induced activation of smooth muscle contraction. The concentra-
ionic channels that leads to calcium influx. tion of intracellular calcium depends on the
Figure 3.9 illustrates the mechanism balance between the calcium that enters the
by which an increase in intracellular cal- cells, the calcium that is released by intracel-
cium stimulates vascular smooth muscle lular storage sites, and the movement of cal-
contraction. An increase in free intracellu- cium either back into intracellular storage
lar calcium can result from either increased sites or out of the cell. Calcium is reseques-
entry of calcium into the cell through L-type tered by the sarcoplasmic reticulum by an
calcium channels or release of calcium from ATP-dependent calcium pump similar to the
internal stores (e.g., sarcoplasmic reticulum). SERCA pump found in cardiac myocytes. Cal-
The free calcium binds to a special calcium- cium is removed from the cell to the external
binding protein called calmodulin. The cal- environment by either an ATP-dependent cal-
cium–calmodulin complex activates myosin cium pump or the sodium–calcium exchanger,
light chain kinase, an enzyme that phospho- as in cardiac muscle (see Chapter 2).
rylates myosin light chains in the presence of Several signal transduction mechanisms
ATP. Myosin light chains are regulatory subu- modulate intracellular calcium concentration
nits found on the myosin heads. Myosin light and therefore the state of vascular tone. This
chain phosphorylation leads to cross-bridge section describes three different pathways:
formation between the myosin heads and (1) IP3 via Gq-protein activation of phospho-
the actin filaments, thus leading to smooth lipase C; (2) cAMP via Gs-protein activation
muscle contraction. of adenylyl cyclase; and (3) cyclic guanosine

Ca++
L-type
Calcium
Channel

Ca++ + Calmodulin
SR

Ca++– Calmodulin
cAMP
+ –
MLC ATP
MLCK Pi
Phosphatase

■ FIGURE 3.9 Regulation of vascular smooth muscle contraction by myosin light chain kinase (MLCK).
Increased intracellular calcium, by either increased entry into the cell (through L-type Ca++ channels) or
release from the sarcoplasmic reticulum (SR), forms a complex with calmodulin, activating MLCK, which
phosphorylates myosin light chains (MLC), causing contraction. Cyclic adenosine monophosphate (cAMP)
inhibits MLCK, thereby causing relaxation. Dephosphorylation of myosin light chains by MLC phosphatase
also produces relaxation. ATP, adenosine triphosphate; Pi, phosphate group.

Klabunde_Chap03.indd 52 6/10/2011 8:52:49 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 53

monophosphate (cGMP) via nitric oxide (NO) as isoproterenol causes relaxation. The
activation of guanylyl cyclase (Fig. 3.10). mechanism for this process is cAMP inhibi-
The IP3 pathway in vascular smooth muscle tion of myosin light chain kinase (see Fig.
is similar to that found in the heart. Norepi- 3.9), which decreases myosin light chain
nephrine and epinephrine (via α1-adrenocep- phosphorylation, thereby inhibiting the inter-
tors), angiotensin II (via AT1 receptors), actions between actin and myosin. Adenosine
endothelin-I (via ETA receptors), vasopressin and prostacyclin (PGI2) also activate Gs-pro-
(via V1 receptors) and acetylcholine (via M3 tein through their receptors, leading to an
receptors) activate phospholipase C through increase in cAMP and smooth muscle relaxa-
the Gq-protein, causing the formation of IP3 tion. Epinephrine binding to β2-adrenoceptors
from PIP2. IP3 then directly stimulates the relaxes vascular smooth muscle through the
sarcoplasmic reticulum to release calcium. Gs-protein.
The formation of diacylglycerol from PIP2 A third important mechanism for regulat-
activates protein kinase C, which can modu- ing vascular smooth muscle contraction is the
late vascular smooth muscle contraction as NO–cGMP system. Many endothelial-depend-
well via protein phosphorylation. ent vasodilator substances (e.g., acetylcho-
Receptors coupled to the Gs-protein line, bradykinin, substance P), when bound
stimulate adenylyl cyclase, which catalyzes to their respective endothelial receptors,
the formation of cAMP. In vascular smooth stimulate the conversion of L-arginine to
muscle, unlike cardiac myocytes, an increase NO by activating NO synthase. The NO dif-
in cAMP by a β2-adrenoceptor agonist such fuses from the endothelial cell to the vascular

+ L-type
DAG PK-C Calcium
PIP2
+
IP3
+ Ca++ SR Channel
NE
A Ca ++ Ca ++
PL-C +
ET-1 R _
ACh + MLCK + Contraction
AVP Gq _
GTP _
cGMP
GDP cAMP GC
ATP
GTP GDP GTP
+
+ AC
R Gs
NO
Epi
Ado
PGI2
■ FIGURE 3.10 Receptors and signal transduction pathways that regulate vascular smooth muscle contrac-
tion. R, receptor; Gs, stimulatory G-protein; Gq, phospholipase C-coupled G-protein; AC, adenylyl cyclase;
PL-C, phospholipase C; PIP2, phosphatidylinositol 4,5-bisphosphate; IP3, inositol triphosphate; DAG, diacylg-
lycerol; PK-C, protein kinase C; SR, sarcoplasmic reticulum; MLCK, myosin light chain kinase; Ado, adeno-
sine; PGI2, prostacyclin; Epi, epinephrine; NO, nitric oxide; GC, guanylyl cyclase; AII, angiotensin II; ET-1,
endothelin-1; NE, norepinephrine; ACh, acetylcholine; AVP, arginine vasopressin; GDP, guanosine diphos-
phate; GTP, guanosine triphosphate; ATP, adenosine triphosphate; cAMP, cyclic adenosine monophosphate;
cGMP, cyclic guanosine monophosphate.

Klabunde_Chap03.indd 53 6/10/2011 8:52:49 PM


Platelets
Leukocytes
Blood
– –

ET-1 PGI 2 NO
EC

VSM + – –
Contraction

Klabunde_Chap03.indd 54 6/10/2011 8:52:49 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 55

In addition, endothelial cells synthesize hypertension, diabetes, and hypercholester-


endothelin-1 (ET-1), a powerful vasoconstric- olemia. Endothelial dysfunction results in
tor (see Fig. 3.11). Synthesis is stimulated less NO and PGI 2 production, which causes
by angiotensin II, vasopressin, thrombin, vasoconstriction, loss of vasodilatory capacity,
cytokines, and shearing forces, and it is inhib- thrombosis, and vascular inflammation. Evi-
ited by NO and PGI2 • ET-1leaves the endothe- dence exists that enhanced ET-1 production
lial cell and can bind to receptors (ETA) on contributes to hypertension and other vascu-
vascular smooth muscle, which causes cal- lar disorders. Physical damage to the endothe-
cium mobilization and smooth muscle con- lium at the capillary level increases capillary
traction. The smooth muscle actions of ET-1 permeability (see Chapter 8), which leads to
occur through activation of the IP3 signaling increased capillary fluid filtration and tissue
pathway (see Fig. 3.10). edema.
PGI 2 is a product of arachidonic acid
metabolism within endothelial cells. The two PROBLEM 3·3
primary roles of PGI 2 formed by endothelial
When acetylcholine is infused
cells are smooth muscle relaxation and inhi-
into normal coronary arteries, the
bition of platelet aggregation (see Fig. 3.11),
vessels dilate; however, if the vessel
both of which are induced by the formation of
is diseased and the endothelium
cAMP (see Fig. 3.10).
damaged, acetylcholine can cause
The importance of normal endothelial
vasoconstriction. Explain why
function is made clear from examining how
acetylcholine can have opposite effects
endothelial dysfunction contributes to dis-
on vascular function depending on the
ease states. For example, endothelial dam-
integrity of the vascular endothelium.
age and dysfunction occur in atherosclerosis,

SUMMARY OF IMPORTANT CONCEPTS

• The basic contractile unit of a cardiac ATP, which is generated primarily by


myocyte is the sarcomere, which oxidative metabolism of fatty acids and
contains thick filaments (myosin) and carbohydrates, although the heart is
thin filaments (actin, troponin, and flexible in its use of substrates and can
tropomyosin) that are involved in also metabolize amino acids, ketones,
muscle contraction. and lactate.
• Excitation-contraction coupling is • Arteries and veins are arranged as
initiated by depolarization of the three layers: adventitia, media, and
cardiac myocyte, and is controlled intima. Autonomic nerves and small
by changes in intracellular calcium, blood vessels (vasa vasorum in large
which binds to regulatory proteins on vessels) are found in the adventitia;
the thin filaments; ATP is required for vascular smooth muscle is found in the
contraction and relaxation. media; and the intima is lined by the
• Relaxation of cardiac myocytes endothelium.
(lusitropy) is primarily regulated by the • Vascular smooth muscle contains
reuptake of calcium into the sarcoplasmic actin and myosin; however, these
reticulum by the SERCA pump. components are not arranged in the
same repetitive pattern as that found in
• The contractile function of cardiac
cardiac myocytes. Unlike cardiac muscle
myocytes requires large amounts of

(Continued)

Klabunde_Chap03.indd 55 6/10/2011 8:52:50 PM


56 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

contraction, vascular smooth muscle contraction/relaxation primarily by


contraction Is slow and sustained. regulating intracellular calcium.
• Cardiac muscle contraction is regulated • The vascular endothelium synthesizes
by various substances that bind to nitric oxide and prostacyclin, both of
receptors coupled to G-proteins. which relax vascular smooth muscle.
Vascular smooth muscle contraction/ Endothelin-1, which is also synthesized
relaxation is additionally regulated by the endothelium, contracts vascular
by NO/cGMP-dependent pathways. smooth muscle.
All these pathways largely affect

REVIEW QUESTIONS

For each question, choose the one best answer: 5. Vascular smooth muscle contraction is
enhanced by
l. Which of the following is common to a. Activation of myosin light chain
both cardiac myocytes and vascular kinase.
smooth muscle cells? b. Activation of myosin light chain
a. Dense bodies phosphatase.
b. Myosin light chain kinase c. Calcium binding to troponin-C.
c. Terminal cisternae d. Dephosphorylation of myosin light
d. T tubules chains.

2. Thick filaments within cardiac myocytes 6. Angiotensin II causes contraction of


contain vascular smooth muscle by
a. Actin. a. Activating Gs-protein.
b. Myosin. b. Increasing cAMP.
c. Tropomyosin. c. Increasing IP3.
d. Troponin. d. Inhibiting release of calcium by
sarcoplasmic reticulum.
3. During excitation-contraction coupling
in cardiac myocytes, 7. A patient in circulatory shock is
a. Calcium binds to myosin causing ATP treated with norepinephrine to raise
hydrolysis. arterial pressure by stimulating the
b. Calcium binds to troponin-1. heart through ~-adrenoceptor activa-
c. Myosin heads bind to actin. tion and constricting blood vessels
d. SERCA pumps calcium out of the through a. 1-adrenoceptor activation.
sarcoplasmic reticulum. The cardiac and vascular effects can be
explained by
4. Cardiac inotropy is enhanced by a. Increased cardiac cAMP and increased
a. Agonists coupled to Gi-protein. vascular cGMP.
b. Decreased calcium binding to b. Increased cardiac cAMP and increased
troponin-C. vascular IP3 .
c. Decreased release of calcium by c. Increased cardiac and vascular cAMP.
terminal cisternae. d. Increased cardiac IP 3 and increased
d. Protein kinase A phosphorylation of vascular cAMP.
L-type calcium channels.

Klabunde_Chap03.indd 56 6/10/2011 8:52:52 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 57

8. A patient with a complaint of leg pain a. Increased endothelial production of


is found to have a blood clot in a large nitric oxide and prostacyclin.
artery in his leg; he is subsequently b. Diminished endothelial production of
diagnosed with peripheral artery disease. cGMP.
Because peripheral artery disease is c. Increased endothelial production of
associated with endothelial dysfunction, prostacyclin and decreased produc-
which of the following could have con- tion of endothelin-1.
tributed to the formation of the blood d. Decreased endothelial production of
clot? nitric oxide.

l. The correct answer is "b" because incorrect because it is the calcium that
myosin light chain kinase is involved in is released by the terminal cisternae of
myosin phosphorylation in both types of the sarcoplasmic reticulum that binds to
muscle. Choice "a" is incorrect because TN-C leading to contraction.
dense bodies are specialized regions 5. The correct answer is "a" because myo-
found only within vascular smooth mus- sin light chain kinase activation by cal-
de cells where bands of actin filaments dum-calmodulin phosphorylates myosin
are joined together. Choices "c" and "d" light chains, which induces contraction.
are incorrect because these structures Choices "b" and "d" are incorrect because
are found in cardiac muscle cells, not myosin light chain phosphatase activa-
smooth muscle cells. tion dephosphorylates the myosin light
2. The correct answer is "b" because chains, which causes relaxation. Choice
myosin is the major component of the "c" is incorrect because there is no tropo-
thick filament. Choices "a," "c," and nin C in vascular smooth muscle.
"d" are incorrect because they are all 6. The correct answer is "c" because angio-
components of the thin filament. tensin II receptors (AT) are coupled to
3. The correct answer is "c" because a the Gq-protein and activates phospholi-
myosin-binding site is exposed on the pase C, which increases IP3 . Choice "a"
actin after calcium binds to TN-C. is incorrect because angiotensin II acti-
Choices "a" and "b" are incorrect vates the Gq-protein, not the Gs-protein.
because calcium binds to TN-C, not Choice "b" is incorrect because the
myosin or TN-I. Choice "d" is incorrect Gq-protein stimulates IP3 formation, not
because SERCA pumps calcium back cAMP. Choice "d" is incorrect because
into the sarcoplasmic reticulum. the increase in IP3 stimulates calcium
4. The correct answer is "d" because release from the sarcoplasmic reticulum.
phosphorylation of the L-type calcium 7. The correct answer is "b" because car-
channels by protein kinase A increases diac ~-adrenoceptors are coupled to
the permeability of the channel to cal- the Gs-protein and cAMP formation,
cium, thereby permitting more calcium and the vascular 0.1-adrenoceptors are
to enter the cell during depolarization, coupled to the Gq-protein and IP3 for-
which triggers the release of calcium mation. Choice "a" is incorrect because
by the sarcoplasmic reticulum. Choice cGMP is increased by nitric oxide in
"a" is incorrect because Gi-protein blood vessels, not by Gq-protein activa-
activation decreases cAMP formation, tion. Choice "c" is incorrect because
thereby decreasing inotropy. Choice "b" vascular 0.1-adrenoceptors are not
is incorrect because calcium binding to coupled to the Gs-protein. Choice "d"
TN-C enhances inotropy. Choice "c" is is incorrect because vascular cAMP is

Klabunde_Chap03.indd 57 6/10/2011 8:52:53 PM


58 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

not increased by Gq-proteins linked to production can lead to clot formation.


a.1-adrenoceptors, and ~-adrenoceptors Choice "a" is incorrect because the pro-
in the heart are not coupled to IP3 ; duction of nitric oxide and prostacyclin
however, IP 3 may increase in the heart are decreased when the endothelium is
because norepinephrine also binds to damaged or dysfunctional. Choice "b" is
cardiac a.1-adrenoceptors. incorrect because decreased endothelial
8. The correct answer is "d" because endo- cGMP does not affect platelet function.
thelial-derived nitric oxide normally Choice "c" is incorrect because dysfunc-
inhibits platelet aggregation and clot for- tional endothelium results in decreased
mation; therefore, decreased nitric oxide prostacyclin production.

PROBLEM 3-1 urn from the sarcoplasmic reticulum, leading


Sympathetic nerve stimulation releases nor- to an increase in inotropy.
epinephrine, which binds to ~ 1 -adrenoceptors
PROBLEM 3-2
and a.1-adrenoceptors found on cardiac myo-
Increasing cAMP in the heart activates pro-
cytes. ~ 1 -adrenoceptor activation stimulates
tein kinase A, which phosphorylates differ-
cAMP production through the Gs-protein.
ent sites within the cells (see the answer to
cAMP production activates protein kinase A
Problem 3-1). Phosphorylation enhances
(PK-A), which phosphorylates L-type calcium
calcium influx into the cell and calcium
channels, leading to an increase in calcium
release by the sarcoplasmic reticulum, lead-
influx during the action potential. Increased
ing to an increase in inotropy. In vascular
calcium influx triggers increased calcium
smooth muscle, myosin light chain kinase,
release by the sarcoplasmic reticulum, lead-
when activated by calcium-calmodulin,
ing to increased calcium binding by TN-C.
phosphorylates myosin light chains to
Calcium binding increases myosin ATPase
stimulate smooth muscle contraction. cAMP
activity and force generation. PK-A also
inhibits myosin light chain kinase; therefore,
phosphorylates phospholamban and removes
an increase in cAMP by a phosphodiester-
its inhibition of SERCA, which leads to
ase inhibitor such as milrinone inhibits the
increased calcium reuptake by the sarcoplas-
myosin light chain kinase, thereby reducing
mic reticulum and increases the rate of relax-
smooth muscle contraction.
ation, or lusitropy. Increased calcium within
the sarcoplasmic reticulum subsequently PROBLEM 3-3
enhances the release of calcium from the Acetylcholine has two effects on blood
sarcoplasmic reticulum. In addition, PK-A vessels. When acetylcholine binds to M2
may phosphorylate sites on the sarcoplasmic receptors on the vascular endothelium, it
reticulum to enhance calcium release. PK-A stimulates the formation of nitric oxide
phosphorylation of TN-I also may contrib- (NO) by NO synthase. The NO can then
ute to enhanced lusitropy by altering TN-C diffuse from the endothelial cell into the
affinity for calcium. Although physiologically adjacent smooth muscle cells, where it
less important than the ~ 1 -adrenoceptor-Gs activates guanylyl cyclase to form cGMP.
protein pathway, norepinephrine binding to Increased cGMP relaxes vascular smooth
0.1-adrenoceptors increases the formation of muscle cells by inhibiting calcium entry
IP3 via Gq-protein and phospholipase Cacti- into the cell and by other mechanisms.
vation, which stimulates the release of calci- Acetylcholine, however, also can bind

Klabunde_Chap03.indd 58 6/10/2011 8:52:54 PM


CHAPTER 3 • CELLULAR STRUCTURE AND FUNCTION 59

to M3 receptors located on the smooth If the endothelium is intact, stimulation


muscle. This activates the IP3 pathway of the NO–cGMP pathway dominates
and stimulates calcium release by the over the actions of the IP3 pathway;
sarcoplasmic reticulum, which leads to therefore, acetylcholine normally causes
increased smooth muscle contraction. vasodilation.

SUGGESTED RESOURCES Rhodin JAG. Architecture of the vessel wall. In: Bohr
Goldstein MA, Schroeter JP. Ultrastructure of the heart. DF, Somlyo AP, Sparks HV, eds. Handbook of
In: Page E, Fozzard HA, Solaro RJ, eds. Handbook of Physiology, vol 2. Bethesda: American Physiological
Physiology, vol 1. Bethesda: American Physiological Society, 1980; 1–31.
Society, 2002; 3–74. Sanders KM. Invited review: mechanisms of calcium
Katz AM. Physiology of the Heart. 4th Ed. Philadelphia: handling in smooth muscles. J Appl Physiol
Lippincott Williams & Wilkins, 2006. 2001;91:1438–1449.
Moss RL, Buck SH. Regulation of cardiac contraction Somlyo AV: Ultrastructure of vascular smooth
by calcium. In: Page E, Fozzard HA, Solaro RJ, eds. muscle. In: Bohr DF, Somlyo AP, Sparks HV, eds.
Handbook of Physiology, vol 1. Bethesda: American Handbook of Physiology, vol 2. Bethesda: American
Physiological Society, 2002; 420–454. Physiological Society, 1980; 33–67.
Opie LH. The Heart: Physiology from Cell to
Circulation. 4th Ed. Philadelphia: Lippincott
Williams & Wilkins, 2004.

Klabunde_Chap03.indd 59 6/10/2011 8:52:54 PM


0

4
:I:
>
'tl
-1
m
;o
CARDIAC FUNCTION
Understanding the concepts presented in this chapter will enable the student to:
1. Describe the basic anatomy of the heart, including the names of vessels enter-
ing and leaving the heart, cardiac chambers, and heart valves; trace the flow of
blood through the heart.
2. Describe the changes in cardiac pressures and volumes, and associated electri-
cal events and heart sounds, that occur during one cardiac cycle .

3. Draw and label ventricular pressure-volume loops derived from ventricular
pressure and volume changes during the cardiac cycle.
4. Calculate stroke volume, cardiac output, and ejection fraction from ventricular
end-diastolic and end-systolic volumes and heart rate.
5. Describe the factors that determine or modify ventricular preload, afterload,
and inotropy.
6. Show how changes in preload, afterload, and inotropy affect ventricular end-
diastolic volume, end-systolic volume, and stroke volume by using Frank-
Starling curves and ventricular pressure-volume loops.
7. Describe how changes in preload, afterload, and inotropy alter the length-
tension and force-velocity relationships for cardiac muscle.
8. Calculate myocardial oxygen consumption given coronary blood flow, and coro-
nary arterial and venous oxygen contents.
9. Explain how changes in stroke volume, stroke work, afterload, heart rate, and
inotropy affect myocardial oxygen consumption.

INTRODUCTION the mechanisms that regulate cardiac output,


particularly those mechanisms that influence
The heart is a specialized muscular organ the amount of blood ejected into the aorta with
that rhythmically contracts and pumps blood each contraction of the left ventricle. The last
from the low-pressure venous side to the high- section of this chapter discusses the relationship
pressure arterial side of the circulation. Efficient between myocardial oxygen consumption and
pumping occurs because of the orderly contrac- the mechanical activity of the heart.
tion sequence of the different heart chambers
and the presence of valves within the heart that CARDIAC ANATOMY
ensure a unidirectional flow of blood. This chap-
ter describes the basic anatomy of the heart-its
Functional Anatomy of the Heart
chambers, valves, and vessels entering and leav-
ing the heart-and the sequence of electrical The heart consists of four chambers: right
and mechanical events that occur during a cycle atrium, right ventricle, left atrium, and left ven-
of contraction and relaxation. It then describes tricle (Fig. 4.1). The right atrium receives blood

60

Klabunde_Chap04.indd 60 6/10/2011 10:49:35 PM


CHAPTER 4 • CARDIAC FUNCTION 61

Aorta
Pulmonary Artery
SVC
Pulmonary
Pulmonic Veins
Valve LA
RA Mitral Valve
Tricuspid
Valve IVC Aortic Valve
LV
Chordae
Tendineae RV

Papillary
Muscle
Septum
■ FIGURE 4.1 Anatomy of the heart. SVC, superior vena cava; IVC, inferior vena cava; RA, right atrium; RV,
right ventricle; LA, left atrium; LV, left ventricle.

from the superior and inferior vena cavae, The tricuspid and mitral valves have fibrous
which carry blood returning from the systemic strands (chordae tendineae) on their leaflets
circulation. The right atrium is a highly disten- that attach to papillary muscles located on
sible chamber that can easily expand to accom- the respective ventricular walls. The papillary
modate the venous return at a low pressure (0 muscles contract when the ventricles con-
to 4 mm Hg). Blood flows from the right atrium, tract. This generates tension on the valve leaf-
across the tricuspid valve (right atrioventricu- lets via the chordae tendineae, preventing the
lar [AV] valve), and into the right ventricle. The valves from bulging back and leaking blood
free wall of the right ventricle wraps around into the atria (i.e., preventing regurgitation)
part of the larger and thicker left ventricle. The as the ventricles develop pressure. The semi-
outflow tract of the right ventricle is the pulmo- lunar valves (pulmonic and aortic) do not
nary artery, which is separated from the ven- have analogous attachments.
tricle by the semilunar pulmonic valve. Blood
returns to the heart from the lungs via four pul-
Autonomic Innervation
monary veins that enter the left atrium. Blood
flows from the left atrium, across the mitral Autonomic innervation of the heart plays an
valve (left AV valve), and into the left ventricle. important role in regulating cardiac function.
The left ventricle has a thick muscular wall that The heart is innervated by parasympathetic
allows it to generate high pressures during con- (vagal) and sympathetic efferent fibers (see
traction. The left ventricle ejects blood across Chapter 6 for details on the origin of these
the aortic valve and into the aorta. autonomic nerves). The right vagus nerve

Klabunde_Chap04.indd 61 6/10/2011 10:49:37 PM


62 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

preferentially innervates the sinoatrial (SA) side. Furthermore, the timing of mechanical
node, whereas the left vagus nerve inner- events in the right side of the heart is very
vates the AV node; however, significant over- similar to that of the left side. The main differ-
lap can occur in the anatomical distribution. ence is that the pressures in the right side of
Atrial muscle is also innervated by vagal the heart are much lower than those found in
efferents; the ventricular myocardium is only the left side. For example, the right ventricu-
sparsely innervated by vagal efferents. Sym- lar pressure typically changes from about 0 to
pathetic efferent nerves are present through- 4 mm Hg during filling to a maximum of 25 to
out the atria (especially in the SA node) and 30 mm Hg during contraction.
ventricles, and in the conduction system of A catheter can be placed in the ascending
the heart. aorta and left ventricle to obtain the pressure
Vagal activation of the heart decreases heart and volume information shown in the cardiac
rate (negative chronotropy), decreases con- cycle diagram and to measure simultaneous
duction velocity (negative dromotropy), and changes in aortic and intraventricular pres-
decreases contractility (negative inotropy) of sure as the heart beats. This catheter can also
the heart. Vagal-mediated inotropic influences be used to inject a radiopaque contrast agent
are moderate in the atria and relatively weak into the left ventricular chamber. This per-
in the ventricles. Activation of the sympa- mits fluoroscopic imaging (contrast ventricu-
thetic nerves to the heart increases heart rate, lography) of the ventricular chamber, from
conduction velocity, and inotropy. Sympa- which estimates of ventricular volume can be
thetic influences are pronounced in both the obtained; however, real-time echocardiogra-
atria and ventricles. phy and nuclear imaging of the heart are more
As Chapter 6 describes in more detail, the commonly used to obtain clinical assessment
heart also contains vagal and sympathetic of volume and function.
afferent nerve fibers that relay information In the following discussion, a complete
from stretch and pain receptors. The stretch cardiac cycle is defined as the cardiac events
receptors are involved in feedback regula- initiated by the P wave in the electrocardio-
tion of blood volume and arterial pressure, gram (ECG) and continuing until the next
whereas the pain receptors produce chest pain P wave. The cardiac cycle is divided into two
when activated during myocardial ischemia. general categories: systole and diastole. Sys-
tole refers to events associated with ventricu-
lar contraction and ejection. Diastole refers to
THE CARDIAC CYCLE the rest of the cardiac cycle, including ventric-
ular relaxation and filling. The cardiac cycle
is further divided into seven phases, begin-
Cardiac Cycle Diagram
ning when the P wave appears. These phases
To understand how cardiac function is regu- are atrial systole, isovolumetric contraction,
lated, one must know the sequence of mechan- rapid ejection, reduced ejection, isovolumet-
ical events during a complete cardiac cycle ric relaxation, rapid filling, and reduced fill-
and how these mechanical events relate to ing. The events associated with each of these
the electrical activity of the heart. The cardiac phases are described below.
cycle diagram in Figure 4.2 (sometimes called
the Wiggers diagram) depicts changes in the Phase 1. Atrial Systole: AV Valves
left side of the heart (left ventricular pressure Open; Aortic and Pulmonic
and volume, left atrial pressure, and aortic
Valves Closed
pressure) as a function of time. Although not
shown in this figure, pressure and volume The P wave of the ECG represents electrical
changes in the right side of the heart (right depolarization of the atria, which initiates
atrium and ventricle and pulmonary artery) contraction of the atrial musculature. As the
are qualitatively similar to those in the left atria contract, the pressures within the atrial

Klabunde_Chap04.indd 62 6/10/2011 10:49:38 PM


CHAPTER 4 • CARDIAC FUNCTION 63

Dias Sys Dias

Phase: 1 2 3 4 5 6 7
Aortic
Valve
Aortic Closes
120 Valve AP
Opens

80
Pressure Mitral LVP
(mmHg) Valve
40 Closes Mitral
LAP Valve
a c v Opens
0
x x’ y
120 LVEDV

LV
Volume 80
(ml)
R
LVESV
40
P T
ECG Q
S
Heart
Sounds S4 S1 S2 S3

0 0.4 0.8
Seconds
■ FIGURE 4.2 Cardiac cycle. The seven phases of the cardiac cycle are (1) atrial systole; (2) isovolumetric
contraction; (3) rapid ejection; (4) reduced ejection; (5), isovolumetric relaxation; (6) rapid filling; and
(7) reduced filling. Sys, systole; Dias, diastole; AP, aortic pressure; LVP, left ventricular pressure; LAP, left
atrial pressure; a, a wave; c, c wave; v, v wave; x, x descent; x’, x’ descent; y, y descent; LV, left ventricle;
ECG, electrocardiogram; LVEDV, left ventricular end-diastolic volume; LVESV, left ventricular end-systolic
volume, S1–S4, four heart sounds.

chambers increase; this drives blood from the into the venous vessels (i.e., pulmonary veins
atria, across the open AV valves, and into the and vena cava). On the right side of the heart,
ventricles. Retrograde atrial flow back into the this produces the “a wave” of the jugular
vena cava and pulmonary veins is impeded pulse. This can be observed when a person is
by the inertial effect of venous return and by recumbent and the jugular vein in the neck
the wave of contraction throughout the atria, expands with blood, which permits pulsa-
which has a “milking effect.” Atrial contrac- tions to be visualized.
tion produces a small transient increase in left Atrial contraction normally accounts for
and right atrial pressure that is called the “a only about 10% of left ventricular filling
wave.” The a wave is also reflected proximally when a person is at rest and the heart rate

Klabunde_Chap04.indd 63 6/10/2011 10:49:38 PM


64 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

is low, because most of the ventricular filling Phase 2. Isovolumetric


occurs before the atria contract. Therefore, Contraction: All Valves Closed
ventricular filling is mostly passive and
depends on the venous return. However, at This phase of the cardiac cycle, which is the
high heart rates (e.g., during exercise), the beginning of systole, is initiated by the QRS
period of diastolic filling is shortened con- complex of the ECG, which represents ven-
siderably (because overall cycle length is tricular depolarization. As the ventricles depo-
decreased), and the amount of blood that larize, myocyte contraction leads to a rapid
enters the ventricle by passive filling is increase in intraventricular pressure. The
reduced. Under these conditions, the relative abrupt rise in pressure causes the AV valves to
contribution of atrial contraction to ventricu- close as the intraventricular pressure exceeds
lar filling increases greatly and may account atrial pressure. Contraction of the papillary
for up to 40% of ventricular filling. In addi- muscles with their attached chordae tendineae
tion, atrial contribution to ventricular fill- prevents the AV valve leaflets from bulging
ing is enhanced by an increase in the force back or prolapsing into the atria and becom-
of atrial contraction caused by sympathetic ing incompetent (i.e., “leaky”). Closure of the
nerve activation. Enhanced ventricular filling AV valves results in the First Heart Sound
owing to increased atrial contraction is some- (S1). This heart sound is generated when sud-
times referred to as the “atrial kick.” During den closure of the AV valves results in oscil-
atrial fibrillation (see Chapter 2), the con- lation of the blood, which causes vibrations
tribution of atrial contraction to ventricular (i.e., sound waves) that can be heard with
filling is lost. This leads to inadequate ven- a stethoscope overlying the heart. The first
tricular filling, particularly when ventricular heart sound is normally split (∼0.04 second)
rates increase during physical activity. because mitral valve closure precedes tricus-
After atrial contraction is complete, the atrial pid closure; however, because this very short
pressure begins to fall, which causes a slight time interval normally cannot be perceived
pressure gradient reversal across the AV valves. through a stethoscope, only a single sound is
This fall in atrial pressure following the peak heard.
of the a-wave is termed the “x descent.” As the During the time between the closure of the
pressures within the atria fall, the AV valves AV valves and the opening of the aortic and
float upward (preposition) before closure. pulmonic semilunar valves, ventricular pres-
At the end of this phase, which represents sures rise rapidly without a change in ventric-
the end of diastole, the ventricles are filled to ular volumes (i.e., no ejection of blood into
their end-diastolic volume (EDV). The left the aorta or pulmonary artery occurs). Ven-
ventricular EDV (typically about 120 mL) tricular contraction, therefore, is said to be
is associated with end-diastolic pressures “isovolumic” or “isovolumetric” during this
of about 8 mm Hg. The right ventricular phase. During this phase, some individual fib-
end-diastolic pressure is typically about ers shorten when they contract, whereas oth-
4 mm Hg. ers generate force without shortening or can be
A heart sound is sometimes heard dur- mechanically stretched as they are contracting
ing atrial contraction (Fourth Heart Sound, because of nearby contracting cells. Ventricu-
S4). The sound is caused by vibration of the lar chamber geometry changes considerably
ventricular wall as blood rapidly enters the as the heart becomes more spheroid in shape,
ventricle during atrial contraction. This sound although the volume does not change. Early
generally is noted when the ventricle com- in this phase, the rate of pressure develop-
pliance is reduced (i.e., “stiff” ventricle), as ment becomes maximal. The maximal rate
occurs in ventricular hypertrophy (described of pressure development, abbreviated “dP/dt
later in this chapter). The sound is com- max,” is the maximal slope of the ventricular
monly present in older individuals because of pressure tracing plotted against time during
changes in ventricular compliance. isovolumetric contraction.

Klabunde_Chap04.indd 64 6/10/2011 10:49:38 PM


CHAPTER 4 • CARDIAC FUNCTION 65

Atrial pressures transiently increase during Phase 4. Reduced Ejection:


this phase owing to continued venous return Aortic and Pulmonic Valves
and possibly to bulging of AV valves back Open; AV Valves Remain Closed
into the atrial chambers, which results in a
“c wave” noted in the atria and their proximal Approximately 150 to 200 milliseconds after
veins (e.g., in the jugular vein). the QRS, ventricular repolarization (T wave)
occurs. This causes ventricular active tension
to decrease (i.e., muscle relaxation occurs) and
Phase 3. Rapid Ejection: Aortic the rate of ejection (ventricular emptying) to
and Pulmonic Valves Open; AV fall. Ventricular pressure falls slightly below
Valves Remain Closed outflow tract pressure; however, outward flow
When the intraventricular pressures exceed still occurs owing to kinetic (or inertial) energy
the pressures within the aorta and pulmonary of the blood that helps to propel the blood into
artery, the aortic and pulmonic valves open and the aorta and pulmonary artery. Atrial pres-
blood is ejected out of the ventricles. Ejection sures gradually rise during this phase owing to
occurs because the total energy of the blood continued venous return into the atrial cham-
within the ventricle exceeds the total energy bers. The end of this phase concludes systole.
of blood within the aorta. The total energy of
the blood is the sum of the pressure energy Phase 5. Isovolumetric
and the kinetic energy; the latter is related to
Relaxation: All Valves Closed
the square of the velocity of the blood flow. In
other words, ejection occurs because an energy As the ventricles continue to relax and intra-
gradient is present (mostly owing to pressure ventricular pressures fall, a point is reached at
energy) that propels blood into the aorta and which the total energy of blood within the ven-
pulmonary artery. During this phase, ventric- tricles is less than the energy of blood in the
ular pressure normally exceeds outflow tract outflow tracts. When this total energy gradient
pressure by only a few millimeters of mercury reversal occurs, the aortic and pulmonic valves
(mm Hg). Although blood flow across the to abruptly close. At this point, systole ends and
valves is high, the relatively large valve open- diastole begins. Valve closure causes the Sec-
ing (i.e., providing low resistance) requires ond Heart Sound (S2), which is physiologically
only a few mm Hg of a pressure gradient to and audibly split because the aortic valve closes
propel flow across the valve. Maximal outflow before the pulmonic valve. Normally, little or
velocity is reached early in the ejection phase, no blood flows backward into the ventricles as
and maximal (systolic) aortic and pulmonary these valves close. Valve closure is associated
artery pressures are achieved, which are typi- with a characteristic notch (incisura) in the
cally about 120 and 25 mm Hg in the aorta aortic and pulmonary artery pressure tracings.
and pulmonary artery, respectively. Unlike in the ventricles, where pressure rapidly
While blood is being ejected and ventric- falls, the decline in aortic and pulmonary artery
ular volumes decrease, the atria continue to pressures is not abrupt because of potential
fill with blood from their respective venous energy stored in their elastic walls and because
inflow tracts. Although atrial volumes are systemic and pulmonic vascular resistances
increasing, atrial pressures initially decrease impede the flow of blood into distributing arter-
(x' descent) as the base of the atria is pulled ies of the systemic and pulmonary circulations.
downward, expanding the atrial chambers. Ventricular volumes remain constant (iso-
No heart sounds are ordinarily heard dur- volumetric) during this phase because all valves
ing ejection. The opening of healthy valves are closed. The residual volume of blood that
is silent. The presence of a sound during remains in a ventricle after ejection is called the
ejection (i.e., ejection murmurs) indicates end-systolic volume (ESV). For the left ven-
valve disease or intracardiac shunts (see tricle, this is approximately 50 mL of blood.
Chapter 9). The difference between the EDV (120 mL)

Klabunde_Chap04.indd 65 6/10/2011 10:49:38 PM


66 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

and the ESV (50 mL) represents the stroke Phase 7. Reduced Filling:
volume (SV) of the ventricle, which is about AV Valves Open; Aortic and
70 mL. In a normal ventricle, about 60% or Pulmonic Valves Closed
more of the EDV is ejected. The SV (EDV –
ESV) divided by the EDV is called the ejection No clear demarcation exists between the phases
fraction (EF) of the ventricle, which normally of rapid and reduced ventricular filling. The
is >0.55 (or 55%). Although ventricular vol- reduced filling phase is the period during dias-
ume does not change during isovolumetric tole when passive ventricular filling is nearing
relaxation, atrial volumes and pressures con- completion. This is sometimes referred to as
tinue to increase owing to venous return. the period of ventricular diastasis. As the ven-
tricles continue to fill with blood and expand,
they become less compliant (i.e., “stiffer”).
Phase 6. Rapid Filling: AV Valves This causes the intraventricular pressures
Open; Aortic and Pulmonic to rise, as described later in this chapter.
Valves Closed Increased intraventricular pressure reduces
When the ventricular pressures fall below the pressure gradient across the AV valve (the
atrial pressures, the AV valves open and ven- pressure gradient is the difference between
tricular filling begins. Initially, the ventricles the atrial and ventricular pressure) so that the
are still relaxing, which causes intraventricular rate of filling declines, even though atrial pres-
pressures to continue to fall by several mm Hg sures continue to increase slightly as venous
despite ongoing ventricular filling. The rate of blood continues to flow into the atria. Aortic
initial filling is enhanced by the fact that atrial pressure and pulmonary arterial pressure con-
volumes are maximal just prior to AV valve tinue to fall during this period as blood flows
opening. Once the valves open, the elevated into the systemic and pulmonary circulations.
atrial pressures coupled with declining ventric- It is important to note that Figure 4.2
ular pressures (ventricular diastolic suction) depicts the cardiac cycle at a relatively low
and the low resistance of the opened AV valves heart rate (75 beats/min). At low heart rates,
results in rapid, passive filling of the ventri- the length of time allotted to diastole is rela-
cles. Once the ventricles are fully relaxed, their tively long, which lengthens the time of the
pressure begins to rise as they fill. reduced filling phase. High heart rates reduce
The opening of the AV valves causes a rapid the overall cycle length and are associated
fall in atrial pressures. The peak of the atrial pres- with reductions in the duration of both sys-
sure just before the valve opens is the “v wave.” tole and diastole, although diastole shortens
This peak is followed by the “y descent” as much more than systole. Without compen-
blood leaves the atria. The v wave and y descent satory mechanisms, this cycle length reduc-
are transmitted into the proximal venous ves- tion would lead to less ventricular filling (i.e.,
sels such as the jugular vein on the right side of reduced EDV). Compensatory mechanisms
the heart and pulmonary veins on the left side. are important for maintaining adequate ven-
Clinically, changes in atrial pressures and jugu- tricular filling during exercise (see Chapter 9).
lar pulses are useful in the diagnosis of altered
cardiac function (see Chapter 9).
Summary of Intracardiac
If the AV valves are functioning normally, no
Pressures
prominent sounds will be heard during filling.
When a Third Heart Sound (S3) is audible dur- It is important to know normal values of
ing ventricular filling, it may represent tensing intracardiac pressures, as well as the pressures
of chordae tendineae and the AV ring, which is within the veins and arteries entering and
the connective tissue support for the valve leaf- leaving the heart, because abnormal pressures
lets. This S3 heart sound is normal in children, can be used to diagnose certain types of car-
but it is considered pathologic in adults because diac disease and dysfunction. Figure 4.3 sum-
it is often associated with ventricular dilation. marizes normal, typical pressures in an adult

Klabunde_Chap04.indd 66 6/10/2011 10:49:38 PM


CHAPTER 4 • CARDIAC FUNCTION 67

120/80 25/10 ventricular ejection (c), and isovolumetric


relaxation (d). The EDV is the maximal vol-
Ao ume achieved at the end of filling, and ESV
LA is the minimal volume (i.e., residual volume)
RA PA 8 of the ventricle found at the end of ejection.
The width of the loop, therefore, represents
4 120/8 the difference between EDV and ESV, which is
LV the SV. The area within the pressure–volume
loop is the ventricular stroke work.
RV
The filling phase moves along the end-dias-
25/4 tolic pressure–volume relationship (EDPVR),
or passive filling curve for the ventricle. The
slope of the EDPVR at any point along the
■ FIGURE 4.3 Summary of normal pressures curve is the reciprocal of ventricular compli-
within the cardiac chambers and great vessels. ance, as described later in this chapter.
The higher of the two pressure values (expressed
in mm Hg) in the right ventricle (RV), left ventricle
The maximal pressure that can be devel-
(LV), pulmonary artery (PA), and aorta (Ao) rep- oped by the ventricle at any given left ventric-
resent the normal peak pressures during ejection ular volume is described by the end-systolic
(systolic pressure), whereas the lower pressure pressure–volume relationship (ESPVR). The
values represent normal end of diastole pres-
sure (ventricles) or the lowest pressure (diastolic pressure–volume loop, therefore, cannot cross
pressure) found in the PA and Ao. Pressures in the over the ESPVR, because the ESPVR defines
right atrium (RA) and left atrium (LA) represent the maximal pressure that can be generated
average values during the cardiac cycle.
at any given volume under a given inotropic
state, as described later in this chapter.
heart. Note that the pressures on the right The changes in pressures and volumes
side of the heart are considerably lower than described in the cardiac cycle diagram and
those on the left side of the heart, and that by the pressure–volume loop are for normal
the pulmonary circulation has low pressures adult hearts at resting heart rates. Pressure–
compared to the systemic arterial system. The volume loops appear very differently in the
pressures shown for the right and left atria presence of valve disease and heart failure as
indicate an average atrial pressure during the described in Chapter 9.
cardiac cycle—atrial pressures change by sev- CARDIAC OUTPUT
eral mm Hg as they fill and contract.
The primary function of the heart is to impart
Ventricular Pressure–Volume energy to blood to generate and sustain an
Relationship arterial blood pressure sufficient to adequately
Although measurements of pressures and perfuse organs. The heart achieves this by con-
volumes over time can provide important tracting its muscular walls around a closed
insights into ventricular function, pressure– chamber to generate sufficient pressure to pro-
volume loops provide another powerful tool pel blood from the left ventricle, through the
for analyzing the cardiac cycle, particularly aortic valve, and into the aorta. Each time the
ventricular function. left ventricle contracts, a volume of blood is
Pressure–volume loops (Fig. 4.4, bottom ejected into the aorta. This SV, multiplied by the
panel) are generated by plotting left ventric- number of beats per minute (heart rate, HR),
ular pressure against left ventricular volume equals the cardiac output (CO) (Equation 4-1).
at many time points during a complete car-
Eq. 4-1 CO = SV ⭈ HR
diac cycle (Fig. 4.4, top panel). In Figure 4.4,
the letters represent the periods of ventricu- Therefore, changes in either SV or heart rate
lar filling (a), isovolumetric contraction (b), alter cardiac output.

Klabunde_Chap04.indd 67 6/10/2011 10:49:38 PM


Aortic Aortic
Valve Valve
100 Opening Closing

LV Mitral
Pressure Valve
Mitral
Closing
(mmHg) Valve
Opening
0
EDV
LV 100
Volume ESV
(ml) 0
a b c d a

200
ESPVR
Aortic
Valve Aortic
Closing Valve
LV c Opening
Pressure 100
(mmHg) d Mitral
Mitral SV Valve
Valve Closing
Opening b
a
EDPVR
0
0 100 200
ESV EDV
LV Volume (ml)

Klabunde_Chap04.indd 68 6/10/2011 10:49:39 PM


CHAPTER 4 • CARDIAC FUNCTION 69

Measurement of Cardiac Output increase by <50%. These changes in heart rate


are brought about primarily by changes in
In experimental settings, cardiac output can sympathetic and parasympathetic nerve activ-
be measured by electromagnetic or Dop- ity at the SA node (see Chapter 2).
pler flowmeters placed around the pulmo- A change in heart rate does not necessar-
nary artery. Obviously, this approach cannot ily result in a proportionate change in cardiac
be used in humans; therefore, indirect tech- output. The reason is that changes in heart
niques are used. The most commonly used is rate can inversely affect SV. For example, dou-
the thermodilution technique, which uses a bling heart rate from 70 to 140 beats/min by
special multilumen, thermistor-tipped cath- pacemaker stimulation alone does not double
eter (Swan-Ganz) that is inserted into the pul- cardiac output because SV falls when heart
monary artery from a peripheral vein. A cold rate is elevated. This occurs because the ven-
saline solution of known temperature and tricular filling time decreases as the length of
volume is injected into the right atrium from a diastole shortens, thereby resulting in less ven-
proximal port on the catheter. The cold injec- tricular filling. However, when normal physi-
tate mixes into the blood and cools the blood, ological mechanisms during exercise cause the
which then passes through the right ventricle heart rate to double, cardiac output more than
and into the pulmonary artery. The thermistor doubles because SV actually increases. This
at the catheter tip measures the blood temper- increase in SV, despite the elevation in heart
ature, and a cardiac output computer is used rate, is brought about by several mechanisms
to calculate flow (cardiac output). Doppler acting on the heart and systemic circulation
echocardiography can be used to estimate (see Chapter 9). When these mechanisms fail,
real-time changes in flow within the heart, SV cannot be maintained at elevated heart rates.
pulmonary artery, or ascending aorta. Echo- Therefore, it is important to understand the
cardiography and various radionuclide tech- mechanisms that regulate SV because impaired
niques can also be used to measure changes SV regulation can lead to a state of heart failure
in ventricular dimensions during the cardiac and limited exercise capacity (see Chapter 9).
cycle in order to calculate SV, which, when
multiplied by heart rate, gives cardiac out-
put. Although used less frequently, the Fick
EFFECTS OF PRELOAD ON
method permits time-averaged cardiac output STROKE VOLUME
(CO; mL/min) calculations from measure-
ments of arterial and venous blood oxygen Preload is the initial stretching of the cardiac
content (CaO2 and CvO2, respectively; mL O2/ myocytes prior to contraction; therefore, it is
mL blood), and whole body oxygen consump- related to the sarcomere length at the end of dias-
. tole. Sarcomere length cannot be determined in
tion (VO2; mL O2/min). This method is based
on the following relationship (Fick Principle): the intact heart, so indirect indices of preload,
such as ventricular EDV or pressure, must be
 used. These measures of preload are not ideal
VO 2
CO = because they may not always reflect sarcomere
(CaO2 - CvO2 )
length because of changes in the structure and
mechanical properties of the heart. Despite these
Influence of Heart Rate and limitations, acute changes in end-diastolic pres-
Stroke Volume on Cardiac Output sure and volume are useful indices for examin-
ing the effects of acute preload changes on SV.
Although cardiac output is determined by
both heart rate and SV, changes in heart rate
Effects of Ventricular Compliance
are generally more important quantitatively
in producing changes in cardiac output. For on Preload
example, heart rate may increase by 100% As the ventricle fills with blood, the pressure
to 200% during exercise, whereas SV may generated at a given volume is determined

Klabunde_Chap04.indd 69 6/10/2011 10:49:40 PM


70 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

by the compliance of the ventricle, in which is, the ventricle becomes less compliant or
compliance is defined as the ratio of a change “stiffer” at higher volumes.
in volume divided by a change in pressure. Nor- Ventricular compliance is determined by
mally, compliance curves are plotted with vol- the physical properties of the tissues mak-
ume on the Y-axis and pressure on the X-axis, ing up the ventricular wall and the state of
so that the compliance is the slope of the ventricular relaxation. For example, in ven-
line at any given pressure (i.e., the slope of tricular hypertrophy, the increased muscle
the tangent at a particular point on the line). thickness decreases the ventricular compli-
For the ventricle, however, it is common to ance; therefore, ventricular end-diastolic
plot pressure versus volume (Fig. 4.5) and pressure is higher for any given EDV. This
to refer to this pressure–volume relationship is shown in Figure 4-5, in which the filling
as the filling curve for the ventricle. Plotted curve of the hypertrophied ventricle shifts
in this manner, the slope of the tangent at a upward and to the left. From a different per-
given point on the curve is the reciprocal of spective, for a given end-diastolic pressure,
the compliance. Therefore, the steeper the a less compliant ventricle will have a smaller
slope of the pressure–volume relationship, EDV (i.e., filling will be decreased). If ven-
the lower the compliance. This means that tricular relaxation (lusitropy) is impaired, as
the ventricle becomes “stiffer” when the slope occurs in some forms of diastolic ventricular
of the passive filling curve is greater; there- failure (see Chapter 9), the functional ven-
fore, compliance and stiffness are reciprocally tricular compliance will be reduced. This will
related. impair ventricular filling and increase end-
The relationship between pressure and diastolic pressure. If the ventricle becomes
volume is nonlinear in the ventricle (as in chronically dilated, as occurs in other forms
most biological tissues); therefore, compli- of heart failure, the filling curve shifts down-
ance decreases with increasing pressure or ward and to the right. This enables a dilated
volume. When pressure and volume are plot- heart to have a greater EDV without causing a
ted as in Figure 4.5, we find that the slope large increase in end-diastolic pressure.
of the filling curve (the EDPVR described in The length of a sarcomere prior to contrac-
Fig. 4.4) increases at higher volumes; that tion, which represents its preload, depends on

100

Decreased
LV Compliance
Pressure (e.g., hypertrophy)
(mmHg) 50 Increased
Compliance
Normal (e.g., dilation)

(EDP)
0
0 100 200 300
(EDV)
LV Volume (ml)
■ FIGURE 4.5 Left ventricular compliance (or filling) curves. The slope of the tangent of the passive pres-
sure–volume curve at a given volume represents the reciprocal of the ventricular compliance. The slope
of the normal compliance curve is increased by a decrease in ventricular compliance (e.g., ventricular
hypertrophy), whereas the slope of the compliance curve is reduced by an increase in ventricular compli-
ance (e.g., ventricular dilation). Decreased compliance increases the end-diastolic pressure (EDP) at a given
end-diastolic volume (EDV), whereas increased compliance decreases EDP at a given EDV. LV, left ventricle.

Klabunde_Chap04.indd 70 6/10/2011 10:49:40 PM


CHAPTER 4 • CARDIAC FUNCTION 71

the ventricular EDV. This, in turn, depends pressure and ventricular compliance, can
on the ventricular end-diastolic pressure and alter the preload on sarcomeres in cardiac
compliance. Although end-diastolic pressure muscle cells. This change in preload will alter
and EDV are sometimes used as indices of the ability of the myocyte to generate force
preload, care must be taken when interpret- when it contracts. The length–tension rela-
ing the significance of these values in terms tionship examines how changes in the initial
of how they relate to the preload of indi- length of a muscle (i.e., preload) affect the
vidual sarcomeres. An elevated end-diastolic ability of the muscle to develop force (ten-
pressure may be associated with sarcomere sion). To illustrate this relationship, a piece
lengths that are increased, decreased, or of cardiac muscle (e.g., papillary muscle) is
unchanged, depending on the ventricular isolated and placed within an in vitro bath
volume and compliance at that volume. For containing an oxygenated, physiologic salt
example, a stiff, hypertrophied ventricle may solution. One end of the muscle is attached
have an elevated end-diastolic pressure with to a force transducer to measure tension, and
a reduced EDV owing to the reduced compli- the other end is attached to an immovable
ance. Because the EDV is reduced, the sar- support rod (Fig. 4.6, left side). The end that
comere length will be reduced despite the is attached to the force transducer is mov-
increase in end-diastolic pressure. As another able so that the initial length (preload) of
example, a larger than normal EDV may not the muscle can be fixed at a desired length.
be associated with an increase in sarcomere The muscle is then electrically stimulated to
length if the ventricle is chronically dilated contract; however, the length is not permit-
and structurally remodeled such that new ted to change and therefore the contraction
sarcomeres have been added in series, thus is isometric.
maintaining normal individual sarcomere If the muscle is stimulated to contract
lengths. at a relatively short initial length (low
preload), a characteristic increase in tension
Effects of Preload on Tension (termed “active” tension) will occur, last-
Development (Length–Tension ing about 200 milliseconds (Fig. 4.6, right
side, curve a). By stretching the muscle to
Relationship)
a longer initial length, the passive tension
We have seen how ventricular EDV, which will be increased prior to stimulation. The
is determined by ventricular end-diastolic amount of passive tension depends on the

Increased
Resting Preload
Tension Length c For curve c
Transducer Stimulate
Tension

b Active
L L Total
Muscle a
Passive

Fixed Time
■ FIGURE 4.6 Effects of increased preload on tension development by an isolated strip of cardiac muscle.
The left side shows how muscle length and tension are measured in vitro. The bottom of the muscle strip
is fixed to an immovable rod, whereas the top of the muscle is connected to a tension transducer and
a movable bar that can be used to adjust initial muscle length (L). The right side shows how increased
preload (initial length) increases both passive and active (developed) tension. The greater the preload, the
greater the active tension generated by the muscle.

Klabunde_Chap04.indd 71 6/10/2011 10:49:41 PM


72 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

elastic modulus (“stiffness”) of the tissue. Total


The elastic modulus of a tissue is related to Tension
the ability of a tissue to resist deformation;
therefore, the higher the elastic modulus,

Tension
the “stiffer” the tissue. When the muscle is
stimulated at the increased preload, there
will be a larger increase in active tension
(curve b) than had occurred at the lower Passive
preload. If the preload is again increased, c Tension
there will be a further increase in active a b
tension (curve c). Therefore, increases in
preload lead to an increase in active tension.
Not only is the magnitude of active tension Active
increased, but also the rate of active tension Tension

Tension
development (i.e., the maximal slope with
respect to time of the tension curve during
contraction). The duration of contraction
and the time-to-peak tension, however, are c
not changed. b
If the results shown in Figure 4.6 are plot-
a
ted as tension versus initial length (preload),
a length–tension diagram is generated
(Fig. 4.7). In the top panel, the passive ten- Length
sion curve is the tension that is generated ■ FIGURE 4.7 Length–tension relationship for
as the muscle is stretched prior to contrac- cardiac muscle undergoing isometric contraction.
tion. Points a, b, and c on the passive curve The top panel shows that increasing the preload
correspond to the passive tensions and ini- length from points a to c increases the passive
tension. Furthermore, increasing the preload
tial preload lengths for curves a, b, and c in increases the total tension during contraction as
Figure 4.6 prior to contraction. The total shown by arrows a, b, and c, which correspond to
tension curve represents the maximal tension active tension changes depicted by curves a, b,
and c in Figure 4.6. The length of the arrow is the
that occurs during contraction at different
active tension, which is the difference between
initial preloads. The total tension curve is the the total and passive tensions. The bottom panel
sum of the passive tension and the additional shows that the active tension increases to a
tension generated during contraction (active maximum value as preload increases.
tension). The active tension, therefore, is
the difference between the total and passive
tension curves; it is plotted separately in the
bottom panel of Figure 4-7. The active ten- (i.e., with no change in length). Cardiac
sion diagram demonstrates that as preload muscle fibers, however, normally shorten
increases, there is an increase in active when they contract (i.e., undergo isotonic
tension up to a maximal limit. The maxi- contractions). If a strip of cardiac muscle in
mal active tension in cardiac muscle corre- vitro is set at a given preload length and stim-
sponds to a sarcomere length of about 2.2 ulated to contract, it will shorten and then
μm. Because of the passive mechanical prop- return to its resting preload length (Fig. 4.8).
erties of cardiac myocytes, their length sel- If the initial preload is increased and the mus-
dom exceeds 2.2 μm at maximal ventricular cle stimulated again, it will ordinarily shorten
EDVs. to the same minimal length, albeit at a higher
This discussion described how changes in velocity of shortening.
preload affect the force generated by cardiac The length–tension relationship, although
muscle fibers during isometric contractions usually used to describe the contraction of

Klabunde_Chap04.indd 72 6/10/2011 10:49:41 PM


CHAPTER 4 • CARDIAC FUNCTION 73

Increased
Preload
Resting Increased
Length Preload

B dL/dt

Length
Muscle DL
DL A DL

Resting
Load Length
Contracted
Length

A B Time
■ FIGURE 4.8 Effects of increased initial muscle length (increased preload) on muscle shortening
(isotonic contractions). The left panel shows a muscle lifting a load (afterload) at two different preload
lengths (A and B). The right panel shows how increasing the preload leads to increased shortening (DL)
and increased velocity of shortening (dL/dt; change in length with respect to time). The muscle shortens
to the same minimal length when preload is increased.

isolated muscles, can be applied to the whole ric ventricular pressure development occurs
heart. By substituting ventricular volume for during ventricular contraction, analogous
length and ventricular pressure for tension, to what is observed with a single papillary
the length–tension relationship becomes a muscle (see Fig. 4.7). This can be observed
pressure–volume relationship for the ven- experimentally in the ventricle by occluding
tricle. This can be done because a quantita- the aorta during ventricular contraction at
tive relationship exists between tension and different ventricular volumes and measuring
pressure and between length and volume the peak systolic pressure generated by the
that is determined by the geometry of the ventricle under this isovolumetric condition.
ventricle. Figure 4.9 shows that as ventricu- The peak systolic pressure curve is analogous
lar EDV increases, an increase in isovolumet- to the ESPVR shown in Figure 4.4 because
this is the maximal pressure that can be gen-
erated by the ventricle at a given ventricular
Peak-Systolic volume.
Pressure
What mechanisms are responsible
Ventricular
Pressure

Developed for the increase in force generation with


c Pressure increased preload in the heart? In the past,
b it was thought that changes in active ten-
a End-Diastolic sion caused by altered preload could be
Pressure explained by the overlap of actin and myo-
sin and therefore by a change in the number
Ventricular Volume
of actin and myosin cross bridges formed
■ FIGURE 4.9 Effects of increasing ventricular (see Chapter 3). However, unlike skeletal
volume (preload) on ventricular pressure develop- muscle that can operate under a very wide
ment. Increasing ventricular volume from a to c
and then stimulating the ventricle to contract
range of sarcomere lengths (1.3 to 3.5 µm),
isovolumetrically increases the developed pressure the intact heart under physiologic condi-
and the peak-systolic pressure. tions operates within a narrow range of

Klabunde_Chap04.indd 73 6/10/2011 10:49:41 PM


74 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

sarcomere lengths (1.8 to 2.2 μm). These 100


and other observations have led to the con- B
cept of length-dependent activation. Exper-
imental evidence supports three possible A Increased
explanations. First, studies have shown that Venous Return
increased sarcomere length sensitizes the SV 50 Decreased
regulatory protein troponin C to calcium (ml) C Venous Return
without necessarily increasing intracellular
release of calcium. This increases calcium
binding by troponin C, leading to an increase
in force generation as described in Chapter 0
3. A second explanation is that fiber stretch- 0 10 20
ing alters calcium homeostasis within the
LVEDP (mmHg)
cell so that increased calcium is available to
bind to troponin C. A third explanation is ■ FIGURE 4.10 Frank-Starling mechanism.
that as a myocyte (and sarcomere) length- Increasing venous return to the left ventricle
ens, the diameter must decrease because the increases left ventricular end-diastolic pres-
volume has to remain constant. It has been sure (LVEDP) by increasing ventricular volume;
this increased preload increases stroke volume
proposed that this would bring the actin (SV) from point A (normal operating point) to B.
and myosin molecules closer to each other Decreasing venous return decreases preload and
(decreased lateral spacing), which would stroke volume (point C).
facilitate their interactions.

Effects of Venous Return on venous return to the heart is increased and


the end-diastolic pressure is increased, this
Stroke Volume (Frank-Starling
will lead to an increase in SV (Point B). A
Mechanism) decrease in venous return (Point C) would
Altered preload is an important mechanism result in less ventricular filling, leading to a
by which the ventricle changes its force lower end-diastolic pressure and a reduced
of contraction and therefore its SV. When SV along this Frank-Starling curve.
venous return to the heart is increased, The Frank-Starling mechanism plays an
ventricular filling increases, and therefore important role in balancing the output of the
its preload. This stretching of the myocytes two ventricles. For example, when venous
causes an increase in force generation, which return increases to the right side of the heart
enables the heart to eject the additional during physical activity, the Frank-Starling
venous return and thereby increase SV. This mechanism enables the right ventricular SV
is called the Frank-Starling mechanism in to increase, thereby matching its output to
honor of the scientific contributions of Otto the increased venous return. The increased
Frank (late 19th century) and Ernest Starling right ventricular output increases the venous
(early 20th century). Another term for this return to the left side of the heart, and the
mechanism is “Starling’s law of the heart.” Frank-Starling mechanism operates to increase
In summary, the Frank-Starling mechanism the output of the left ventricle. This mecha-
states that increasing venous return and ven- nism ensures that the outputs of the two ven-
tricular preload leads to an increase in SV. tricles are matched over time; otherwise blood
Figure 4.10 shows the Frank-Starling rela- volume would shift between the pulmonary
tionship for the left ventricle. Assume that and systemic circulations.
the left ventricle is normally operating at This analysis using Frank-Starling curves
an end-diastolic pressure of 8 mm Hg and shows how changes in venous return and ven-
is ejecting an SV of 70 mL (Point A). If the tricular preload lead to changes in SV. These

Klabunde_Chap04.indd 74 6/10/2011 10:49:41 PM


200
ESPVR Increased
LV Pressure (mmHg)

Venous
Return

100
Control
Loop

SV

 EDV
0
0 100 200
LV Volume (ml)

Klabunde_Chap04.indd 75 6/10/2011 10:49:42 PM


Outflow
Resistance
& Afterload
Atrial Heart
Inotropy Rate

Ventricular Ventricular Ventricular


Compliance Preload Inotropy

Venous Inflow
Pressure Resistance

Venous Venous
Compliance Blood Volume
Total blood volume
Venous return

Klabunde_Chap04.indd 76 6/10/2011 10:49:42 PM


CHAPTER 4 • CARDIAC FUNCTION 77

EFFECTS OF AFTERLOAD Effects of Afterload on the


ON STROKE VOLUME Velocity of Fiber Shortening
(Force–Velocity Relationship)
Afterload is the “load” against which the heart
Afterload influences the contraction of cardiac
must contract to eject blood. A major compo-
muscle fibers. Increased afterload decreases the
nent of the afterload for the left ventricle is
velocity of fiber shortening, whereas decreased
the aortic pressure, or the pressure the ventri-
afterload increases the velocity of shortening.
cle must overcome to eject blood. The greater
This inverse relationship between afterload
the aortic pressure, the greater the afterload
and velocity of fiber shortening is basis for the
on the left ventricle. For the right ventricle,
force–velocity relationship. To illustrate this,
the pulmonary artery pressure represents the
a papillary muscle is placed in an in vitro bath,
major afterload component.
set at a fixed initial length and passive ten-
Ventricular afterload, however, involves
sion (preload), and a load is attached to one
factors other than the pressure that the ven-
end (Fig. 4.13, left panel). When the muscle
tricle must develop to eject blood. One way to
is stimulated to contract, the fiber first gener-
estimate the afterload on the individual cardiac
ates active tension isometrically, that is, active
fibers within the ventricle is to examine ven-
tension is developed with no change in length
tricular wall stress (σ), which is proportional
(right panel, a to b). Once the developed ten-
to the product of the intraventricular pressure
sion exceeds the load imposed on the muscle,
(P) and ventricular radius (r), divided by the
the muscle fiber begins to shorten, and the ten-
wall thickness (h) (Equation 4-2). This rela-
sion remains constant and equal to the load that
tionship for wall stress assumes that the ven-
is being lifted (b to c). The maximal velocity of
tricle is a sphere. The determination of actual
shortening (rate of shortening) occurs shortly
wall stress is complex and must consider not
after the muscle begins to shorten. The muscle
only ventricular geometry, but also muscle
continues to shorten until the muscle begins to
fiber orientation. Nonetheless, Equation 4-2
relax. When active tension falls below the load
helps to illustrate the factors that contribute
(point c), the muscle resumes its resting length
to wall stress and therefore afterload on the
and tension (i.e., preload) (point c). Active ten-
muscle fibers.
sion continues to fall isometrically (c to d) until
P⋅r only the passive tension remains (point d).
Eq. 4-2 σ∝ If this experiment with the papillary muscle
h
were repeated with increasing loads, a decrease
Wall stress can be thought of as the aver- would occur in both the maximal velocity of
age tension that individual muscle fibers fiber shortening (maximal slope of line) and the
within the ventricular wall must generate to degree of shortening, as shown in Figure 4.14.
shorten against the developed intraventricu- Plotting the maximal velocity of shortening
lar pressure. At a given intraventricular pres- against the load that the muscle fiber must
sure, wall stress is increased by an increase shorten against (i.e., the afterload) generates an
in radius (ventricular dilation). Therefore, inverse relationship between velocity of short-
afterload is increased whenever intraven- ening and afterload (force–velocity relation-
tricular pressures are elevated during systole ship; Fig. 4.15). In other words, the greater the
and by ventricular dilation. On the other afterload, the slower the velocity of shortening.
hand, a thickened, hypertrophied ventricle To further illustrate the force–velocity rela-
will have reduced wall stress and afterload on tionship, consider the following example. If a
individual fibers. Ventricular wall hypertro- person holds a 2-lb dumbbell at their side while
phy can be thought of as an adaptive mecha- standing, and then contracts their biceps mus-
nism by which the ventricle is able to offset cle at maximal effort, the weight will be lifted at
the increase in wall stress that accompanies a relatively high velocity as the biceps muscle
increased ventricular systolic pressures or shortens. If the weight is increased to 20 lb, and
ventricular dilation. the weight once again is lifted at maximal effort,

Klabunde_Chap04.indd 77 6/10/2011 10:49:43 PM


78 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

b c
Maximal

Tension DT
Muscle a d
DL Resting

Preload

Load Length DL
Minimal

Time
■ FIGURE 4.13 Cardiac muscle isotonic contractions. The left panel shows how muscle length and tension
are measured in vitro. The lower end of the muscle is attached to a weight (load) that is lifted up from an
immovable platform as the muscle develops tension and shortens (DL). A bar attached to the top of the
muscle can be moved to adjust initial muscle length (preload). The right panel shows changes in tension
and length during contraction. The periods from a to b and from c to d represent periods of isometric con-
traction and relaxation, respectively. Muscle shortening (DL) occurs between b and c, which occurs when
the developed tension (DT) exceeds the load.

the velocity will be slower. Higher weights Vmax represents the intrinsic capability of the
further reduce the velocity until the weight muscle fiber to generate force independent of
can no longer be lifted and the contraction load, and therefore changes when inotropy is
of the biceps muscle becomes isometric. The altered, as discussed later in this chapter.
x-intercept in the force–velocity diagram (see It is important to note that a cardiac mus-
Fig. 4.15) is the point at which the afterload is cle fiber does not operate on a single force–
so great that the muscle fiber cannot shorten. velocity curve (Fig. 4.16). As previously
The x-intercept therefore represents the maxi-
mal isometric force. The y-intercept represents
Shortening Velocity

an extrapolated value for the maximal velocity Vmax


(Vmax) that would be achieved if there was no
afterload. The value is extrapolated because it a
cannot be measured experimentally (a muscle
will not contract in the absence of any load). Maximal
b Isometric
Force
a c = increasing afterload c
Preload

c
Decreasing b Afterload (Force)
Length
■ FIGURE 4.15 Force–velocity relationship.
a Increased afterload (which requires increased force
generation) decreases velocity of shortening by
Time the muscle fiber. The x-intercept represents the
maximal isometric force that occurs when the load
■ FIGURE 4.14 Effects of afterload on myocyte exceeds the muscle’s force-generating capacity,
shortening. Increased afterload (curves a to c) thus preventing muscle shortening; the y-intercept
decreases the degree of muscle shortening and represents the maximal velocity of shortening
maximal velocity of shortening at a given preload, (Vmax) extrapolated to zero load. Points a, b, and c
which is measured as the change in length over represent the maximal shortening velocity gener-
time shortly after muscle begins to shorten. ated in Figure 4.14 for three increasing afterloads.

Klabunde_Chap04.indd 78 6/10/2011 10:49:43 PM


CHAPTER 4 • CARDIAC FUNCTION 79

100
Shortening Velocity

Increasing Preload a c

Stroke Volume (ml)


Afterload
c
Control
b
50
a Afterload

Afterload (Force)
■ FIGURE 4.16 Effects of increasing preload (shift 0
from curve a to c) on the force–velocity relation-
ship. At a given afterload (vertical dashed line), 0 10 20
increasing the preload increases the velocity of LVEDP(mmHg)
shortening. Furthermore, increasing the preload
shifts the x-intercept to the right, which represents ■ FIGURE 4.17 Effects of afterload on Frank-
an increase in isometric force generation. Note Starling curves. An increase in afterload shifts
that y-intercept, which is the maximal velocity of the Frank-Starling curve downward, whereas a
shortening (Vmax) extrapolated to zero load, does decrease in afterload shifts the Frank-Starling
not change with increasing preload. curve upward. Therefore, at a given preload (verti-
cal dashed line) increased afterload decreases
stroke volume, and decreased afterload increases
stroke volume.
discussed, changes in preload also affect the
velocity of fiber shortening (see Fig. 4.8). If
preload is increased, a cardiac muscle fiber will
have a greater velocity of shortening at a given reduced, and indeed, this is what occurs, as
afterload. This occurs because the length–ten- shown in Figure 4.17. An increase in after-
sion relationship requires that as the preload load rotates the Frank-Starling curve down
is increased, there is an increase in active ten- and to the right. Therefore, at a given preload
sion development. Once the fiber begins to (left ventricular end-diastolic pressure
shorten, an increased preload with an increase [LVEDP] in Fig. 4.17), an increase in after-
in tension-generating capability causes a load decreases SV. Conversely, decreasing
greater shortening velocity. In other words, afterload shifts the curves up and to the left,
increasing the preload enables the muscle to thereby increasing the SV at a given preload.
contract faster against a given afterload; this As discussed in Chapter 9, reducing ventric-
shifts the force–velocity relationship to the ular afterload in heart failure patients is an
right (see Fig. 4.16). Note that increasing the important therapeutic approach to enhance
preload increases the maximal isometric force SV.
(x-intercept) as well as the shortening velocity
at a given afterload (a to b to c). Changes in Effects of Afterload on
preload, however, do not alter Vmax. Therefore, Pressure–Volume Loops
an increase in preload on a cardiac myocyte can
help to offset the reduction in velocity that occurs The effects of afterload on ventricular
when afterload is increased. function can be depicted using ventricu-
lar pressure–volume loops as shown in
Effects of Afterload on Figure 4.17. Increasing afterload by increas-
ing aortic pressure at a constant preload
Frank-Starling Curves
(EDV) causes a decrease in SV (width of the
We have just seen how an increase in after- loop) and an increase in ESV. The ventricle
load at a given preload decreases the velocity will generate increased pressure to overcome
and extent of fiber shortening. This being the the elevated aortic pressure, but at the cost
case, we should expect ventricular SV to be of a reduced SV. A reduction in afterload

Klabunde_Chap04.indd 79 6/10/2011 10:49:44 PM


160
LV Pressure (mmHg)

PAo

80 Control

PAo

0
0 100 200
ESV EDV
LV Volume (ml)

Increased
Inotropy
Total
Tension
Tension

Passive
Tension

Length

Klabunde_Chap04.indd 80 6/10/2011 10:49:44 PM


CHAPTER 4 • CARDIAC FUNCTION 81

Effects of Inotropy on

Shortening Velocity
Increasing Inotropy a c
Force–Velocity Relationship
Changes in inotropy also alter the force– c
velocity relationship. If the inotropic state
of the myocyte is increased, the force– b
velocity curve exhibits an upward parallel
a
shift, resulting in an increase in both Vmax
(y-intercept) and maximal isometric force
(x-intercept) (Fig. 4.20). The increase in veloc- Afterload (Force)
ity at any given afterload (a to b to c) results ■ FIGURE 4.20 Effects of increasing inotropy
from the increased inotropy enhancing force (parallel shift from curve a to c) on the force–
generation by the actin and myosin filaments and velocity relationship. Increased inotropy increases
the velocity of shortening at any given afterload
increasing the rate of cross-bridge turnover. The
(vertical dashed line), and increases Vmax (y-inter-
increase in Vmax represents an increased intrinsic cept). Furthermore, increased inotropy increases
capability of the muscle fiber to generate force maximal isometric force (x-intercept).
independent of load. In contrast, changes in
preload do not alter Vmax (see Fig. 4-16). increase in ESV. As described later in this chap-
ter, changes in inotropy in a normal, healthy
Effects of Inotropy on Frank- heart will also lead to secondary changes in
Starling Curves preload and afterload that are not shown in
The change in velocity of muscle shortening Figure 4.22.
associated with a change in inotropy results Changes in inotropy change the ejection
in an increase in SV at any given preload and fraction, which is defined as the SV divided
afterload and therefore causes the Frank- by the EDV. In Figure 4.22, this would be
Starling curve to shift up or down (Fig. 4.21). represented by the ratio of the width of the
If, at a given preload, inotropy is enhanced, pressure–volume loop divided by the EDV.
SV will increase. Conversely, a decrease in A normal EF is >0.55 (or 55%). Increasing
inotropy at a given preload will decrease SV.
100
Effects of Inotropy on Pressure–
Stroke Volume (ml)

Volume Loops Inotropy

The increased velocity of fiber shortening Control


that occurs with increased inotropy causes an
increased rate of ventricular pressure devel- 50
Inotropy
opment (dP/dt), which increases ejection
velocity and SV, and reduces ESV, as shown
in Figure 4.22. When inotropy is increased,
the ESPVR is shifted to the left and becomes
0
steeper, because the ventricle can gener-
ate increased pressure at any given volume.
0 10 20
The ESPVR sometimes is used experimen- LVEDP (mmHg)
tally to define the inotropic state of the ven-
■ FIGURE 4.21 Effects of inotropy on Frank-
tricle. It is analogous to the upward shift that Starling curves. An increase in inotropy shifts the
occurs in the total tension curve in the length– Frank-Starling curve upward, whereas a decrease
tension relationship (Fig. 4.19) when ino- in inotropy shifts the Frank-Starling curve down-
ward. Therefore, at a given preload (vertical
tropy increases. Conversely, a decrease in ino-
dashed line), increased inotropy increases stroke
tropy (decreased ESPVR slope) decreases volume, and decreased inotropy decreases stroke
the rate of ejection and SV, which leads to an volume.

Klabunde_Chap04.indd 81 6/10/2011 10:49:45 PM


82 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

160 and norepinephrine) have positive inotropic


LV Pressure (mmHg)

Inotropy Control Inotropy effects similar to sympathetic activation. In


humans and some other mammalian hearts, an
abrupt increase in afterload can cause a modest
increase in ino-tropy (Anrep effect) by a mecha-
80
nism that is not fully understood. In addition,
an increase in heart rate can cause a positive ino-
tropic effect (also termed the Bowditch effect,
treppe, or frequency-dependent activation).
0 This latter phenomenon probably is due to an
0 100 200 inability of the Na+/K+-ATPase to keep up with
ESV EDV
LV Volume (ml) the sodium influx at the higher frequency of
action potentials at elevated heart rates, leading
■ FIGURE 4.22 Effects of increasing inotropy to an accumulation of intracellular calcium via
on steady-state left ventricular pressure–volume
loops. Increased inotropy shifts the ESPVR (see Fig.
the sodium–calcium exchanger (see Chapter 2).
4.4) up and to the left, thereby increasing stroke Increased inotropy brought about by sym-
volume and decreasing end-systolic volume (ESV). pathetic activation and increased heart rate
Decreased inotropy shifts the end-diastolic pres- is particularly important during exercise (see
sure–volume relationship down and to the right,
thereby decreasing stroke volume and increasing
Chapter 9) because it helps to maintain SV at
end-systolic volume. LV, left ventricle. Preload and high heart rates. Recall that increased heart
aortic pressure are held constant in this illustration. rate alone decreases SV because reduced dias-
tolic filling time decreases EDV. When ino-
inotropy increases EF, whereas decreasing tropic state increases at the same time, this
inotropy decreases EF. Therefore, EF often is decreases ESV to help maintain SV despite
used as a clinical index for evaluating the ino- reduced EDV.
tropic state of the heart. Systolic failure that results from cardio-
myopathy, ischemia, valve disease, arrhyth-
Factors Influencing mias, and other conditions is characterized
Inotropic State by a loss of intrinsic inotropy (see Chapter 9).
Furthermore, there are many inotropic drugs
Several factors influence ventricular inot- that are used clinically to increase inotropy
ropy (Fig. 4.23); the most important of these in acute and chronic heart failure. These
is the activity of sympathetic nerves. Sympa- drugs include digoxin (inhibits sarcolem-
thetic nerves, by releasing norepinephrine that mal Na+/K+-ATPase), β-adrenoceptor agonists
binds to β1-adrenoceptors on myocytes, serve (e.g., dopamine, dobutamine, epinephrine,
a prominent role in ventricular and atrial ino- isoproterenol), and cAMP-dependent phos-
tropic regulation (see Chapter 3). Elevated lev- phodiesterase inhibitors (e.g., milrinone).
els of circulating catecholamines (epinephrine Although the above discussion focuses on
the regulation of ventricular inotropy, it is
Sympathetic Circulating important to note that many of these same
Activation Catecholamines factors influence atrial inotropy. Unlike the
ventricles, the atria are richly innervated with
parasympathetic nerves (vagal efferents),
Ventricular and activation of this autonomic pathway
Inotropy decreases atrial inotropy.

Afterload Heart Rate Cellular Mechanisms of Inotropy


(Anrep Effect) (Bowditch Effect)
As previously stated, inotropy can be thought
■ FIGURE 4.23 Factors that increase inotropy. of as a length-independent activation of the

Klabunde_Chap04.indd 82 6/10/2011 10:49:45 PM


CHAPTER 4 • CARDIAC FUNCTION 83

contractile proteins. Any cellular mechanism to a small increase in ESV that will partially
that ultimately alters myosin ATPase activity attenuate the increase in SV brought about by
at a given sarcomere length alters force the increased preload as shown in Figure 4.24
generation and therefore can be considered (panel A). The increased preload still results
an inotropic mechanism. Most of the signal in an increase in SV, but the increase is less
transduction pathways that regulate inot- than what would have occurred had the after-
ropy involve Ca++ (see Chapter 3 for details). load not increased.
Briefly, the following calcium-related intracel- An increase in afterload, as previously dis-
lular mechanisms play an important role in cussed, leads to a decrease in SV and an increase
regulating inotropy: in ESV as shown in Figure 4.24 (panel B,
solid red loop). However, because the ESV is
1. Increasing Ca++ influx across the sarco-
increased, changes in afterload produce sec-
lemma during the action potential
ondary changes in preload (dashed red loop).
2. Increasing the release of Ca++ by the sarco-
The increased ESV inside the ventricle is added
plasmic reticulum
to the venous return, thereby increasing EDV.
3. Sensitizing troponin C to Ca++
After several beats, a steady state is achieved in
which the increase in ESV is greater than the
INTERDEPENDENCE secondary increase in EDV so that the differ-
OF PRELOAD, AFTERLOAD, ence between the two—the SV—is decreased
AND INOTROPY (i.e., the width of the pressure–volume loop is
decreased). This increase in preload secondary
Previous discussion focused on the independ- to an increase in afterload activates the Frank-
ent effects of preload, afterload, and inotropy Starling mechanism, which partially compen-
on ventricular function; however, it is impor- sates for the reduction in SV caused by the
tant to understand that these determinants of initial increase in afterload.
ventricular function are also interdependent. The direct, independent effects of an increase
For example, a change in preload leads to inotropy are an increase in SV and a decrease in
secondary changes in afterload that can alter ESV (Fig. 4.24, panel C, solid red line). How-
the initial response to the change in preload. ever, the increased SV increases cardiac output
Furthermore, a change in afterload leads to and arterial pressure, which increases afterload
changes in preload, and a change in inotropy on the ventricle (dashed red line). Increased
can alter both preload and afterload. afterload tends to increase ESV, which par-
Let us first consider how ventricular tially offsets the effects of increased inotropy
responses to a change in preload can be modi- on ESV. With a decrease in ESV from control,
fied by secondary changes in afterload. Similar less blood remains in the ventricle that can be
to Figure 4.11, panel A of Figure 4.24 (solid added to the venous return, so the EDV will
red loop) shows that the independent effect be smaller, although this will be partially off-
of an increase in preload (EDV) is an increase set by the tendency of the increased afterload
in SV (width of pressure–volume loop) with- to increase EDV. After a new steady state is
out a change in ESV. However, because SV is reached following the increase in inotropy, the
increased, cardiac output is increased, and net effect of these changes is an increase in SV,
this will likely lead to an increase in arterial which is accompanied by a reduction in ESV
pressure, which increases afterload. Further- and a smaller reduction in EDV.
more, the increase in EDV increases ven- The interactions between preload, after-
tricular wall stress (see Equation 4-2), which load, and inotropy can also be visualized
represents an increase in afterload. Therefore, using Frank-Starling curves (Fig. 4.25).
a change in preload is normally accompanied In this figure, the left ventricle under con-
by a secondary change in afterload. If after- trol conditions has a SV of 60 mL at an
load increases when there is an increase in end-diastolic pressure (index of preload) of
preload (dashed red loop), then this will lead about 8 mm Hg. Decreasing the afterload or

Klabunde_Chap04.indd 83 6/10/2011 10:49:46 PM


A B C
200
Afterload & Inotropy Inotropy &
LV Pressure (mmHg)

Preload & Preload Afterload &


Afterload Preload

Afterload
100
Preload

Control Control Control

0
0 100 200 0 100 200 0 100 200
LV Volume (ml) LV Volume (ml) LV Volume (ml)

100
Afterload
Inotropy
Control

Afterload
SV 50 Inotropy
(ml)

0
0 10 20
LVEDP(mmHg)

Klabunde_Chap04.indd 84 6/10/2011 10:49:46 PM


CHAPTER 4 • CARDIAC FUNCTION 85

that affects either the generation of force by oxygen consumption varies considerably
myocytes or their frequency of contraction depending on the state of mechanical activity.
will alter oxygen consumption. In addition, Although myocardial oxygen consump-
even in noncontracting cells, ATP utilized tion can be calculated as described above,
by ion pumps and other transport functions generally it is not feasible to measure CBF and
requires oxygen for the resynthesis of ATP. coronary venous oxygen content except in
experimental studies. CBF can be measured
How Myocardial Oxygen by placing flow probes on coronary arteries
Consumption is Determined or a thermodilution catheter within the coro-
nary sinus. Arterial oxygen content can be
Oxygen consumption is defined as the vol- taken from a peripheral artery, but the venous
ume of oxygen consumed per min (e.g., mL oxygen content has to be obtained from the
Ojmin) and is sometimes expressed per 100 coronary sinus by inserting a catheter into
g of tissue weight (mL Ojmin per 100 g). the right atrium and then into the coronary
The myocardial oxygen consumption (MV0 2) sinus.
can be calculated by knowing the coronary Indirect indices of myocardial oxygen
blood flow ( CBF) and the arterial and venous consumption have been developed to esti-
oxygen contents (Ca0 2 and Cv0 2) according mate myocardial oxygen consumption when
to the following equation that uses the Fick it is not feasible to measure it. Although no
principle: index has proven to be satisfactory over a
Eq. 4-3 wide range of physiologic conditions, one
simple index sometimes used in clinical stud-
Myocardial oxygen consumption, therefore, is ies is the pressure-rate product (also called
equal to the CBF multiplied by the amount the double product). This index can be meas-
of oxygen extracted from the blood (the arte- ured noninvasively by multiplying heart rate
rial-venous oxygen difference). The content and systolic arterial pressure (mean arterial
of oxygen in blood is usually expressed as mL pressure sometimes is used instead of systolic
Oj100 mL blood (or, vol% 0 2). The oxygen arterial pressure). The pressure-rate product
content of arterial blood is normally about assumes that the pressure generated by the
20 mL OjlOO mL blood. To calculate the ventricle is not significantly different than the
myocardial oxygen consumption in the cor- aortic pressure (i.e., there is no aortic valve
rect units, mL Oj100 mL blood is converted stenosis). Experiments have shown that a rea-
to mL OjmL blood; with this conversion, the sonable correlation exists between changes
arterial oxygen content is 0.2 mL OjmL blood. in the pressure-rate product and myocardial
For example, if CBF is 80 mUmin per 100 g, oxygen consumption. For example, if arterial
the Ca02 is 0.2 mL OjmL blood and Cv0 2 is pressure, heart rate, or both become elevated,
0.1 mL OjmL blood, then MV02 = 8 mL o; oxygen consumption will increase.
min per 100 g. This value of myocardial oxy-
gen consumption is typical for what is found
in a heart contracting at resting heart rates PROBLEM 4-3
against normal aortic pressures. During heavy
In an experimental study, administration
exercise, myocardial oxygen consumption can
of an inotropic drug is found to increase
increase to 70 mL Ojmin per 100 g, or more.
CBF from 50 to 150 ml/min and
If contractions are arrested (e.g., by depolari-
increase the arterial-venous oxygen
zation of the heart with a high concentration
difference (Ca0 2 - Cv0 2 ) from 10 to
of potassium chloride), the myocardial oxy-
14 ml 0/100 ml blood. Calculate the
gen consumption decreases to about 2 mL
percent increase in myocardial oxygen
Ojmin per 100 g. This value represents the
consumption (MV0 2 ) caused by infusion
energy costs of cellular functions not associ-
of this drug.
ated with contraction. Therefore, myocardial

Klabunde_Chap04.indd 85 6/10/2011 10:49:47 PM


86 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Factors Influencing Myocardial must generate increased contractile force (i.e.,


Oxygen Consumption wall stress) to develop a higher pressure. The
contractile force must be increased even fur-
Part of the difficulty in finding a suitable index ther to generate the same elevated pressure if
of oxygen consumption is that several factors the ventricular radius is increased. For exam-
determine myocyte oxygen consumption, ple, if the ventricle is required to generate
including frequency of contraction, inotropic 50% more pressure than normal to eject blood
state, afterload, and preload (Table 4-1). For because of elevated aortic pressure, the wall
example, doubling heart rate approximately stress that individual myocytes must generate
doubles oxygen consumption, because myo- will be increased by approximately 50%. This
cytes are generating twice the number of ten- will increase the oxygen consumption of these
sion cycles per minute. Increasing inotropy myocytes by about 50% because changes in
increases oxygen consumption because both oxygen consumption are closely related to
the rate of tension development and the mag- changes in wall stress. As a second example,
nitude of tension are increased, and they both if the radius of the ventricle is increased by
are associated with increased ATP hydroly- 50%, the wall stress needed by the myocytes
sis and oxygen consumption. An increase in to eject blood at a normal pressure will be
afterload likewise increases oxygen consump- increased by about 50%. On the other hand,
tion because it increases the tension that must if the ventricular EDV is increased by 50%
be developed by myocytes. Increasing SV and the pressure and wall thickness remain
by increasing preload (EDV) also increases unchanged, the wall stress will be increased
oxygen consumption. by only about 14%. The reason for this is
Quantitatively, increased preload has less that a large change in ventricular volume (V)
impact on oxygen consumption than does an requires only a small change in radius (r). If
increase in afterload (e.g., aortic pressure). To we assume that the shape of the ventricle is a
understand why, we need to examine the rela- sphere, then
tionship between wall stress, pressure, and
radius of the ventricle. As discussed earlier 4
V= π ⋅ r3
(see Equation 4-2), ventricular wall stress (σ) 3
is proportional to the intraventricular pressure
(P) multiplied by the ventricular internal radius By rearranging this relationship, we find that
(r) and divided by the wall thickness (h).
r∝3V
P⋅r
σ∝ Substituting this into the wall stress equation
h
results in
Wall stress is related to the tension an individ-
ual myocyte must develop during contraction Eq. 4-4 P⋅ 3
V
σ∝
to generate a given ventricular pressure. At a h
given radius and wall thickness, a myocyte
Although no single acceptable model for the
shape of the ventricle exists because its shape
TABLE 4-1 FACTORS INCREASING
MYOCARDIAL OXYGEN
changes during contraction, a sphere serves
CONSUMPTION as a convenient model for illustrating why
changes in volume have a relatively small
↑ Heart rate
affect on wall stress and oxygen consump-
↑ Inotropy tion. Using this model, Equation 4-4 shows
↑ Afterload that increasing the EDV by 50% (by a factor of
↑ Preload1 1.5) represents only a 14% (cube root of 1.5)
1
Changes in preload affect oxygen consumption much less
increase in wall stress at a given ventricular
than do changes in the other factors. pressure, whereas a 50% increase in pressure

Klabunde_Chap04.indd 86 6/10/2011 10:49:48 PM


CHAPTER 4 • CARDIAC FUNCTION 87

increases wall stress by 50%. Therefore, by disease. A less efficient heart performs less
increasing pressure by a given percentage work per unit oxygen consumed (i.e., it gen-
increases wall stress about four times more erates less pressure and SV).
than the same change in volume. The concepts described above have impli-
Relating the wall stress equation to oxygen cations for treating patients with coronary
consumption helps to explain why increases artery disease (CAD). For example, drugs that
in pressure generation have a much greater decrease afterload, heart rate, and inotropy
influence on oxygen consumption than a sim- are particularly effective in reducing myocar-
ilar percentage increase in ventricular preload. dial oxygen consumption and relieving symp-
It is important, however, not to use the wall toms of chest pain (i.e., angina), which results
stress equation to estimate oxygen demands from inadequate oxygen delivery relative to
by the whole heart. The reason for this is that the oxygen demands of the myocardium.
wall stress estimates the tension required by CAD patients are counseled to avoid activi-
individual myocytes to generate pressure as ties such as lifting heavy weights that lead to
they contract. This wall stress, in large part, large increases in arterial blood pressure. In
determines the oxygen consumption of indi- contrast, CAD patients are often encouraged
vidual myocytes, but oxygen consumption to participate in exercise programs such as
of the whole heart is the sum of the oxygen walking that utilize preload and SV changes
consumed by all of the myocytes. A hypertro- to augment cardiac output by the Frank-Star-
phied ventricle with a thicker wall, which has ling mechanism. It is important to minimize
reduced wall stress, will not have a reduction stressful situations in these patients because
in overall oxygen consumption as suggested stress causes sympathetic activation of the
by Equation 4-4. In fact, because of its greater heart and vasculature that increases heart
muscle mass, oxygen consumption may be rate, inotropy, and afterload, all of which lead
significantly increased in a hypertrophied to significant increases in oxygen demand by
heart, particularly if its efficiency is impaired the heart.

SUMMARY OF IMPORTANT CONCEPTS

• The cardiac cycle is divided into two • Ventricular SV is the difference


general phases: diastole and systole. between the end-diastolic and end-
Diastole refers to the period of time systolic volumes. Ventricular ejection
that the ventricles are undergoing fraction (EF) is calculated as the SV
relaxation and filling with blood from divided by the EDV.
the atria. Ventricular filling is primarily • Cardiac output is normally influenced
passive, although atrial contraction more by changes in heart rate than
has a variable effect on the final extent by changes in SV; however, impaired
of ventricular filling (EDV). Systole regulation of SV can have a significant
represents the time when the ventricles adverse affect on cardiac output, as
are contracting and ejecting blood occurs during heart failure.
(SV). The volume of blood remaining
in the ventricle at the end of ejection is • Ventricular preload is related to the
the ESV. extent of ventricular filling (EDV)
and the sarcomere length. Increased
• Normal heart sounds (51 and 5 2 ) preload increases the force of
originate from abrupt closure of heart contraction and SV.
valves.

(Continued)

Klabunde_Chap04.indd 87 6/10/2011 10:49:50 PM


88 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

• Ventricular afterload can be estimated velocity, and SV at a given preload and


by ventricular wall stress, which is afterload.
the product of ventricular pressure • Preload, afterload, and inotropy are
and ventricular radius divided by the interdependent, meaning that a change
ventricular wall thickness. Increased in one usually leads to secondary
afterload decreases the velocity of changes in the others.
fiber shortening during contraction,
which decreases the SV. • Myocardial oxygen consumption can
be calculated using the Fick Principle,
• lnotropy is the property of a cardiac in which oxygen consumption equals
myocyte that enables it to alter its the product of the CBF and the
tension development independent of arteriovenous oxygen difference.
changes in preload length. Increased Myocardial oxygen consumption
inotropy enhances active tension is strongly influenced by changes
development by individual muscle in arterial pressure, heart rate, and
fibers and increases ventricular inotropy; it is less influenced by changes
pressure development, ejection inSV.

REVIEW QUESTIONS

For each question, choose the one best d. Pulmonary artery diastolic pressure is
answer: less than mean right atrial
pressure.
1. During the phase of rapid ventricular
filling, 3. Right ventricular preload is increased by
a. 54 may sometimes be heard. which of the following?
b. The aortic valve is open. a. Decreased atrial contractility
c. The mitral valve is open. b. Decreased blood volume
d. Ventricular pressure is higher than c. Decreased heart rate
aortic pressure. d. Decreased ventricular compliance

2. A patient with valve disease undergoes car- 4. A 78-year-old female patient with a his-
diac catheterization to compare vascular tory of left ventricular failure complains
and intracardiac pressures against normal of difficulty breathing when lying down.
values. Which of the following is found in Which of the following occurs to the
a heart with normal valve function? cardiac muscle fibers that can lead to an
a. Aortic diastolic pressure is less than increase in right ventricular output and
pulmonary artery systolic pressure. pulmonary congestion when this patient
b. Left ventricular end-diastolic pressure lies down?
is less than mean right atrial a. Active tension development increases.
pressure. b. Degree of muscle shortening is dimin-
c. Mean left atrial pressure is normally ished.
greater than mean right atrial pressure c. Preload decreases.
by<lOmmHg. d. Velocity of shortening decreases.

Klabunde_Chap04.indd 88 6/10/2011 10:49:52 PM


CHAPTER 4 • CARDIAC FUNCTION 89

5. Left ventricular end-diastolic pressure is a. Decrease left ventricular end-systolic


increased by volume
a. Decreased afterload. b. Decrease the velocity of muscle fiber
b. Decreased venous return. shortening
c. Increased inotropy. c. Decrease ventricular preload
d. Ventricular hypertrophy. d. Increase stroke volume

6. A 67-year-old male patient complains 8. Increasing the inotropic state of the


of excessive shortness of breath during myocardium will
exertion. The report from a follow-up a. Increase end-systolic volume.
echocardiogram says that his left ven- b. Increase the width of the pressure-
tricular stroke volume is 50 mL, with an volume loop.
ejection fraction of 25%. Which of the c. Increase ventricular end-diastolic
following statements is true concerning volume.
this patient's left ventricle? d. Shift the force-velocity relationship to
a. End-diastolic volume is elevated the left.
above normal.
b. End-systolic volume is less than normal. 9. If each of the following is increased by 25%,
c. Inotropy is increased. which one would cause the smallest change
d. Preload is reduced. in myocardial oxygen consumption?
a. Heart rate
7. A patient is experiencing an acute b. Ventricular end-diastolic volume
hypertensive crisis. This large increase in c. Mean arterial pressure
arterial pressure will have which of the d. Ventricular radius
following direct effects on the heart?

1. The correct answer is "c" because the about 4 mm Hg. Choice "a" is incorrect
mitral valve is open throughout ven- because aortic diastolic pressure is about
tricular filling. Choice "a" is incorrect 80 mm Hg compared to a pulmonary
because 54 , when heard, is associated artery systolic pressure of 25 mm Hg.
with atrial contraction and frequently is Choice "b" is incorrect because left
heard in hypertrophied hearts. Choice atrial and left ventricular end-diastolic
"b" is incorrect because the aortic valve pressures are normally higher than their
is open only during ventricular ejec- corresponding pressures on the right
tion. Choice "d" is incorrect because the side of the heart. Choice "d" is incorrect
ventricular pressure is higher than aortic because pulmonary artery diastolic pres-
pressure only during the phase of rapid sure is about IO mm Hg, whereas right
ejection. atrial pressure is about 4 mm Hg.
2. The correct answer is "c" because aver- 3. The correct answer is "c" because more
age left atrial pressure is about 8 mm Hg time is available for filling at reduced
and average right atrial pressure is heart rates (diastole is lengthened);

Klabunde_Chap04.indd 89 6/10/2011 10:49:54 PM


90 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

therefore, preload is increased at Choices “b,” “c,” and “d” are incorrect
reduced heart rates. Choices “a,” “b,” because this low EF (normally >55%)
and “d” are incorrect because decreased indicates ventricular failure (loss of inot-
atrial contractility, blood volume, and ropy), which leads to a reduced SV and
ventricular compliance lead to reduced an elevated end-systolic volume. Preload
ventricular filling and therefore reduced (end-diastolic volume) is increased (as
preload. calculated above) because the elevated
4. The correct answer is “a” because end-systolic volume leads to a secondary
increased preload resulting from increase in preload, and because of other
increased venous return when lying compensatory mechanisms discussed in
down causes length-dependent acti- Chapter 9.
vation of actin and myosin, which 7. The correct answer is “b” because an
increases active tension development. increased arterial pressure increases left
This is the basis for the Frank-Starling ventricular afterload, which decreases
mechanism. Being in heart failure, the velocity of fiber shortening as
increased output of the right ventricle shown by the force–velocity relation-
may not lead to a corresponding ship. Choices “a,” “c,” and “d” are
increase in left ventricular stroke, there- incorrect because increased afterload
by causing pulmonary congestion and decreases the velocity of fiber shorten-
difficulty breathing. Choices “b,” “c,” ing, which decreases stroke volume.
and “d” are incorrect because decreased This leads to an increase in left ventricu-
muscle shortening, preload, and veloc- lar end-systolic volume and a second-
ity of shortening all lead to a decrease in ary increase in preload (end-diastolic
stroke volume. volume).
5. The correct answer is “d” because ven- 8. The correct answer is “b” because an
tricular hypertrophy reduces ventricular increase in inotropy increases stroke
compliance, which results in elevated volume, which is represented by the
end-diastolic pressures when the ventri- width of the pressure–volume loop.
cle fills. Choice “a” is incorrect because Choice “a,” “c,” and “d” are incorrect
decreased afterload leads to a reduction because increased inotropy causes a par-
in end-systolic volume, which results allel, upward shift in the force–velocity
in a secondary fall in end-diastolic vol- relationship, which leads to an increase
ume and pressure. Choice “b” is incor- in velocity of fiber shortening and there-
rect because decreased venous return fore an increase in stroke volume at any
decreases ventricular filling, which given afterload. Increased stroke volume
decreases ventricular end-diastolic vol- decreases end-systolic volume and leads
ume and pressure. Choice “c” is incor- to a secondary decrease in left ventricu-
rect because increased inotropy reduces lar end-diastolic volume (preload).
end-systolic volume, which results in a 9. The correct answer is “b” because an
secondary fall in end-diastolic volume increase in end-diastolic volume will
and pressure. increase stroke volume; however, stroke
6. The correct answer is “a” because with volume changes are about one-fourth as
an ejection fraction (EF) of 25% and effective in changing myocardial oxygen
a stroke volume (SV) of 50 mL, this consumption as are changes in heart
patient’s end-diastolic volume (EDV) rate, mean arterial pressure, or ventricu-
is 200 mL, which is much greater than lar radius because of the relationships
normal (usually <150 mL). The calcula- between oxygen consumption, wall
tion is based on: EF = (SV/EDV) × 100, stress, ventricular pressure, and ventric-
and therefore EDV = (SV/EF) × 100 ular radius. For this reason, choices “a,”
when EF is expressed as a percentage. “c,” and “d” are incorrect.

Klabunde_Chap04.indd 90 6/10/2011 10:49:55 PM


200

LV Pressure (mmHg)
Control
Loop
Decreased
100 Compliance
(hypertrophy)

0
0 100 200
LV Volume (ml)

Klabunde_Chap04.indd 91 6/10/2011 10:49:55 PM


92 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

100 CASE 4-3


Non-failing Reduced ventricular filling time caused by
Stroke Volume (ml)

Heart tachycardia leads to a decrease in EDV and


SV. This preload change alone, however, will
B
not alter ESV. The addition of a fall in arterial
50 pressure decreases afterload on the ventricle,
Failing
A which independently increases stroke volume
Heart and reduces ESV. This can lead to a further
decrease in EDV. The net effect of these two
0 conditions will be a large reduction in EDV
0 10 20 coupled with a small reduction in ESV, and
A LVEDP (mmHg) large fall in stroke volume. Because of the
hypotension (both systolic and diastolic), the
ejection phase of the pressure–volume loop
200 starts at a lower pressure, and the peak ven-
Heart tricular systolic pressure is also reduced.
LV Pressure (mmHg)

Failure
Loop
160
LV Pressure (mmHg)

100
Heart Failure Control
plus Loop
Arterial Dilator 80
Reduced
0 Filling &
Hypotension
0 100 200
B LV Volume (ml)

0
0 80 160
LV Volume (ml)

SUGGESTED RESOURCES
Braunwald E, Ross J, Sonnenblick EH. Mechanisms of Lilly LS. Pathophysiology of Heart Disease. 5th Ed.
Contraction of the Normal and Failing Heart. 2nd Philadelphia: Lippincott Williams & Wilkins, 2011.
Ed. Boston: Little, Brown & Co., 1976. Opie LH. The Heart: Physiology from Cell to
Covell JW, Ross J. Systolic and diastolic function Circulation. 4th Ed. Philadelphia: Lippincott
(mechanics) of the intact heart. In: Page E, Fozzard HA, Williams & Wilkins, 2004.
Solaro RJ, eds. Handbook of Physiology, vol. 1. Bethesda: Sagawa K, Maughan L, Suga H, Sunagawa K. Cardiac
American Physiological Society, 2002; 741–785. Contraction and the Pressure-Volume Relationship.
Fuchs F, Smith SH. Calcium, cross-bridges, and the New York: Oxford University Press, 1988.
Frank-Starling relationship. News Physiol Sci Solaro, RJ. Integration of Myocyte Response to Ca2+ with
2001;16:5–10. Cardiac Pump Regulation and Pump Dynamics. Am.
Katz AM. Physiology of the Heart. 4th Ed. Philadelphia: J. Physiol. 1999;277(Adv. Physiol. Educ. 22):
Lippincott Williams & Wilkins, 2006. S155–S163.

Klabunde_Chap04.indd 92 6/10/2011 10:49:56 PM


0

5
:I:
>
'tl
-1
m
;c
VASCULAR FUNCTION
Understanding the concepts presented in this chapter will enable the student to:
1. Name the different types of vessels constituting the vascular network of the
body and describe the general function of each.
2. Describe how changes in cardiac output, systemic vascular resistance, and
central venous pressure affect mean arterial pressure.

• 3. Describe the factors that determine arterial pulse pressure.


4. Describe in quantitative terms how changes in vessel radius, vessel length,
blood viscosity, and perfusion pressure affect blood flow.
5. Explain how turbulent flow alters the relationship between pressure and flow.
6. Calculate total resistance from series or parallel resistance networks.
7. Explain why the pressure drop across small arteries and arterioles is much
greater than the pressure drop across other vessel types.
8. Define vascular tone and list factors that alter vascular tone.
9. Explain how each of the following affects central venous pressure: blood
volume, venous compliance, gravity, respiration, and muscle contraction.
10. Using cardiac and systemic function curves, explain how changes in blood
volume, venous compliance, vascular resistance, and cardiac performance
influence the equilibrium between right atrial pressure and cardiac output.

INTRODUCTION ANATOMY AND FUNCTION

The vascular system serves two basic func- Vascular Network


tions: distribution and exchange. Distribution
The left ventricle ejects blood into the aorta,
includes transporting blood to and away from
which then distributes the blood flow through-
organs. Exchange involves the movement of
gases, nutrients, and fluid between the blood out the body using a network of arterial ves-
sels, which branch into successively smaller
and tissues. This chapter focuses on vascu-
vessels until they reach the smallest vascular
lar anatomy and the general hemodynamic
unit, capillaries, within organs and tissues.
principles involved in the regulation of blood
Capillaries then converge into successively
pressure and the distribution of blood flow
larger vessels (veins), which return the blood
in the body. Chapters 6 and 7 describe these
physiologic control mechanisms in more to the heart. These vessels are illustrated in
Figure 5.1. Table 5-1 summarizes the relative
detail. The exchange function is described in
sizes and functions of different blood vessels.
Chapter 8.

93

Klabunde_Chap05.indd 93 6/11/2011 11:10:01 AM


94 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Aorta
Vena
Cava
Small
Artery
Large
Artery Capillaries Vein

Arteriole Venule

Distribution/Resistance Exchange Capacitance


■ FIGURE 5.1 Major types of blood vessels found within the circulation.

The different types of vessels can be grouped (e.g., carotid, mesenteric, and renal arteries)
by their primary function: distribution/ distribute the blood flow to specific organs
resistance (aorta, large and small distributing or regions of the body. These large arteries,
arteries), exchange (capillaries, small ven- although capable of constricting and dilating,
ules), and capacitance (large venules, veins, serve no significant role in the regulation of
vena cavae). pressure and blood flow under normal physi-
ologic conditions. Once the distributing artery
DISTRIBUTION/RESISTANCE VESSELS
reaches the organ to which it supplies blood,
The aorta, besides being the main vessel to it branches into small arteries that distribute
distribute blood from the heart to the arte- blood flow within the organ. These smaller
rial system, dampens the pulsatile pressure arteries continue branching into smaller
that results from the intermittent ejection of and smaller vessels. Once they reach diam-
blood from the left ventricle. The dampen- eters of <200 μm, they are termed arterioles.
ing is a function of the aortic compliance, No clear demarcation between small arteries
which is discussed in more detail later in this and arterioles exists; therefore, no consen-
chapter. Large arteries branching off the aorta sus has been reached regarding the point at

TABLE 5-1 SIZE AND FUNCTION OF DIFFERENT TYPES OF BLOOD VESSELS


IN THE SYSTEMIC CIRCULATION
VESSEL TYPE DIAMETER (mm) FUNCTION

Aorta 25 Pulse dampening and distribution


Large arteries 1.0–10.0 Distribution
Small arteries 0.2–1.0 Distribution and resistance
Arterioles 0.01–0.20 Resistance (pressure/flow regulation)
Capillaries 0.006–0.010 Exchange
Venules 0.01–0.20 Exchange, collection, and capacitance
Veins 0.2–10.0 Capacitance function (blood volume)
Vena cava 35 Collection

Klabunde_Chap05.indd 94 6/11/2011 11:10:04 AM


CHAPTER 5 • VASCULAR FUNCTION 95

which a small artery becomes an arteriole. are exchanged across the capillary endothe-
Many investigators speak of different branch- lium between the plasma and the surrounding
ing orders of arterial vessels within a tissue or tissue interstitium (see Chapter 8). Capillaries,
organ. Most would agree that arterioles have therefore, are the primary exchange vessels
only a few layers of vascular smooth muscle within the body.
and are, in general, <200 μm in diameter. When capillaries join together, they form
Together, the small arteries and arteri- small, postcapillary venules, which are still
oles represent the primary resistance ves- devoid of smooth muscle. They, like capil-
sels that regulate arterial blood pressure laries, serve as exchange vessels for fluid
and blood flow within organs. Resistance and macromolecules because of their high
vessels are highly innervated by autonomic permeability.
nerves (particularly sympathetic adrener-
gic), and they constrict or dilate in response CAPACITANCE VESSELS
to changes in nerve activity. The resistance
vessels are richly endowed with receptors As small postcapillary venules converge and
that bind circulating hormones (e.g., cat- form larger venules, smooth muscle reap-
echolamines, angiotensin II), which can pears. These vessels, like the resistance ves-
alter vessel diameter (see Chapters 3 and 6). sels, are capable of dilating and constricting.
They also respond to various substances (e.g., Changes in venular diameter regulate capil-
adenosine, potassium ion, and nitric oxide) lary pressure and venous blood volume. Ven-
produced by the tissue surrounding the vessel ules converge to form larger veins. Together,
or by the vascular endothelium. venules and veins are the primary capacitance
vessels of the body, that is, the site where most
of the blood volume is found and regional
EXCHANGE VESSELS blood volume is regulated. Constriction of
As arterioles become smaller in diameter veins decreases venous blood volume and
(<10 μm), they lose their smooth muscle. increases venous pressure, which can alter
Vessels that have no smooth muscle and are cardiac output by affecting right atrial pres-
composed of only endothelial cells and a sure and ventricular preload. The final venous
basement membrane are termed capillaries. vessels are the inferior and superior vena
Although they are the smallest vessels within cavae, which carry the blood back to the right
the circulation, they have the greatest cross− atrium of the heart.
sectional area because they are so numerous.
Because the total blood flow of all capillaries Distribution of Pressures
in the body is the same as the flow within the and Volumes
aorta leaving the heart, and because the capil-
lary cross−sectional area is about 1000 times Mean blood pressure is highest in the aorta
greater than the aorta, the mean velocity of (about 95 mm Hg in a normal adult) and pro-
blood flowing within capillaries (∼0.05 cm/s) gressively decreases as the blood flows further
is about 1000−fold less than the velocity in the away from the heart (Fig. 5.2). The reason why
aorta (∼50 cm/s). The reason for this is that pressure falls as blood flows through vessels is
flow (F) is the product of mean velocity (V) because energy is lost as heat owing to friction
times cross−sectional area (A) (F = V . A). within the moving blood (related to blood vis-
When this expression is rearranged, we find cosity) and between the blood and the vessel
that the mean velocity is inversely propor- wall. Between any two points along the length
tional to cross−sectional area (V = F/A). of an artery, for example, the drop in pressure
Capillaries have the greatest surface area (ΔP) is related to the flow (F) and resistance to
for exchange. Oxygen, carbon dioxide, water, flow (R) as shown in Equation 5−1.
electrolytes, proteins, metabolic substrates
Eq. 5-1 ΔP = F ⋅ R
and by−products, and circulating hormones

Klabunde_Chap05.indd 95 6/11/2011 11:10:04 AM


96 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

100
Mean Pressure
Mean Percent Volume
Pressure 80
(mmHg)
60
or

Percent 40
Total
Volume 20

a
les

ns
ta

ries

es

ies

Cav
Aor

Vei
riol

u
llar
Arte

Ven
Arte

a
i
Cap

Ven
■ FIGURE 5.2 Distribution of pressures and volumes in the systemic circulation. The greatest pressure drop
occurs across small arteries and arterioles; most of the blood volume is found within the veins and venules.

Equation 5−1 is a hydrodynamic form of Ohm be 25 to 30 mm Hg, depending on the organ. It


law (ΔV = I . R), where the voltage difference is important that the capillary pressure is rela-
(ΔV; analogous to ΔP) is equal to the current tively low; otherwise, large amounts of fluid
(I; analogous to F) times the resistance (R). would leak through the capillaries (and post-
This equation generally applies in the body capillary venules), causing tissue edema (see
when blood is flowing under nonturbulent, Chapter 8). The pressure falls further as blood
laminar flow conditions. travels through veins back to the heart; how-
The mean blood pressure does not fall ever, the pressure drop is small compared to
much as the blood flows down the aorta and the pressure drop across the small arteries and
through large distributing arteries because arterioles because the resistance of the veins
these vessels have a low resistance relative is very low compared to the arterial resistance
to their flow, and therefore, little loss of pres- vessels. Pressure within the thoracic vena
sure energy (ΔP) occurs along their lengths. cava near the right atrium is very close to zero
In contrast, when the blood flows through millimeters of mercury (mm Hg), although it
the small arteries and arterioles (the primary fluctuates by a few mm Hg during the cardiac
resistance vessels), there is a large fall in mean cycle and because of respiratory activity.
arterial blood pressure. The reason is that The greatest volume (60% to 80%) of blood
these vessels, as a group, have a high resist- within the circulation resides within the venous
ance relative to their flow, and therefore, ΔP vasculature. This is why veins are referred to
across this group of vessels is large. In fact, as capacitance vessels. The relative volume of
approximately 50% to 70% of the pressure blood between the arterial and venous sides of
drop within the vasculature occurs within the the circulation can vary considerably depend-
resistance vessels. By the time blood reaches ing on total blood volume, intravascular pres-
the capillaries, the mean blood pressure may sures, and vascular compliance as described

Klabunde_Chap05.indd 96 6/11/2011 11:10:05 AM


CHAPTER 5 • VASCULAR FUNCTION 97

later in this chapter. Vascular compliance varies Mean Arterial Pressure


depending on the state of venous smooth mus-
cle contraction, which is primarily regulated by Because of the shape of the aortic pressure
sympathetic nerves innervating the veins. pulse, the value for the mean pressure (geo-
metric mean) is less than the arithmetic
ARTERIAL BLOOD PRESSURE average of the systolic and diastolic pres-
sures as shown in Figure 5.3. At normal
Ejection of blood into the aorta by the left resting heart rates, mean aortic (or arterial)
ventricle results in a characteristic aortic pres- pressure (MAP) can be estimated from the
sure pulse (Fig. 5.3). The peak pressure of diastolic (Pdias) and systolic (Psys) pressures
the aortic pulse is termed the systolic pres- by Equation 5−2:
sure. Shortly after the peak systolic pres- Eq. 5-2 MAP ≅ Pdias + 1 (Psys - Pdias )
3
sure, there appears a notch (dicrotic notch
or incisura) followed by the appearance of For example, if systolic pressure is 120 mm Hg
a small increase in pressure (dicrotic wave) and diastolic pressure is 80 mm Hg, the
prior to the pressure falling toward its mini- mean arterial pressure will be approximately
mal value, the diastolic pressure. The dif- 93 mm Hg. At high heart rates, however, mean
ference between the systolic and diastolic arterial pressure is more closely approxi-
pressures is the aortic pulse pressure. If, for mated by the arithmetic average of systolic
example, the systolic pressure is 130 mm Hg and diastolic pressure because the shape
and the diastolic pressure is 85 mm Hg, then of the arterial pressure pulse changes (it
the pulse pressure is 45 mm Hg. Therefore, becomes narrower) as the period of diastole
any factor that affects either systolic or dias- shortens more than does systole. Therefore,
tolic pressures affects pulse pressure. The to determine mean arterial pressure accu-
systolic and diastolic pressures are those that rately, analog electronic circuitry or digital
are measured with an arm blood pressure cuff techniques are used, usually in conjunction
(sphygmomanometer). While these values with an indwelling arterial catheter.
are very important clinically, neither value is No single value exists for normal mean
the primary pressure that drives blood flow arterial pressure. In infant children, the mean
in organs. That pressure is the mean arterial arterial pressure may be only 70 mm Hg,
pressure, which is the average pressure over whereas in older adults, mean arterial pres-
time. This pressure needs to be determined sure may be 100 mm Hg. With increasing
when hemodynamic information is required age, the systolic pressure generally rises more
to assess vascular function. than diastolic pressure; therefore, the pulse
pressure increases with age. Small differences
exist between men and women, with women
Incisura having slightly lower pressures at equivalent
Psystolic Dicrotic
ages. In adults, arterial pressure is considered
Wave

Pmean normal when the systolic pressure is <120 mm


Hg (but > 90 mm Hg) and the diastolic
pressure is <80 mm Hg (but > 60 mm Hg),
Pdiastolic which represents a normal mean pressure of
<95 mm Hg. Abnormally low and elevated
Pulse Pressure = Psystolic- Pdiastolic arterial pressures are discussed in Chapter 9.
What factors determine mean arterial
■ FIGURE 5.3 Pressure pulse within the aorta. The pressure? As blood is pumped into the resist-
pulse pressure is the difference between the maxi- ance network of the systemic circulation,
mal pressure (systolic) and the minimal pressure
pressure is generated within the arterial vas-
(diastolic). The mean pressure is approximately
equal to the diastolic pressure plus one−third the culature. The mean arterial (or aortic) pres-
pulse pressure. sure (MAP) is determined by the cardiac

Klabunde_Chap05.indd 97 6/11/2011 11:10:05 AM


98 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

output (CO), systemic vascular resistance one variable can change each of the other
(SVR), and central venous pressure (CVP) as variables). For example, increasing systemic
shown in Equation 5−3. vascular resistance increases the afterload on
the heart, which decreases cardiac output and
Eq. 5-3 MAP = (CO ⋅ SVR) + CVP
alters CVP, as described in more detail later in
This equation is based on Equation 5−1, this chapter. Furthermore, extrinsic control
where ΔP = F . R. The ΔP in Equation 5−3 rep- mechanisms acting on the heart and circulation
resents the pressure drop across the entire sys- can affect these variables. If, for example, car-
temic circulation, which is MAP − CVP; the diac output suddenly falls by 20% (as can occur
CO and SVR are the F and R, respectively, of when standing), mean arterial pressure will not
Equation 5−1. Therefore, from Equation 5−3, decrease by 20% because the body compen-
changes in cardiac output, systemic vascular sates by increasing systemic vascular resistance
resistance, or CVP affect mean arterial pres- through baroreceptor mechanisms to maintain
sure. If cardiac output and systemic vascular constant pressure (see Chapter 6).
resistance change reciprocally and propor-
tionately, MAP will not change. For example, Aortic Pulse Pressure
if cardiac output is reduced by one−half and
systemic vascular resistance is doubled, mean As blood flows down the aorta and into dis-
arterial pressure will remain unchanged. tributing arteries, characteristic changes take
Figure 5.4, which is based upon Equation 5−3, place in the shape of the pressure wave con-
shows that as cardiac output is increased, a tour. As the pressure pulse moves away from
linear increase occurs in arterial pressure the heart, the systolic pressure rises, and the
(assuming that resistance and venous pressure diastolic pressure falls. The change in the
remain constant). An increase in systemic vas- shape of the pressure pulse is related to a num-
cular resistance (increased slope of the line) ber of factors including (1) decreased compli-
results in a greater arterial pressure for any ance of distal arteries and (2) reflective waves,
given cardiac output. Conversely, a decrease particularly from arterial branch points,
in resistance results in a lower arterial pressure which summate with the pulse wave trave-
for any given cardiac output. ling down the aorta and arteries. In addition,
Cardiac output, systemic vascular resistance, mean arterial pressure declines as the pres-
and venous pressure are constantly changing, sure pulse travels down distributing arteries
and they are interdependent (i.e., changing owing to the resistance of the arteries; how-
ever, the reduction in mean pressure is small
(just a few mm Hg) because the distributing
Mean Arterial Pressure

SVR
arteries have a relatively low resistance. There-
fore, the values measured for arterial pressure
differ depending on the site of measurement.
When the arterial pressure is measured using a
SVR sphygmomanometer (i.e., blood pressure cuff)
on the upper arm, the pressure measurement
represents the pressure within the brachial
CVP
artery. The measured pressures, however, are
Cardiac Output not identical with the systolic and diastolic
pressures found in the aorta or the pressures
■ FIGURE 5.4 The relationship between cardiac
output (CO), systemic vascular resistance (SVR), measured in other distributing arteries.
mean arterial pressure (MAP), and central venous The compliance of the aorta and the
pressure (CVP). Increasing SVR increases MAP at ventricular stroke volume determine pulse
any given cardiac output (dotted line), whereas
pressure. Compliance is defined by the
decreasing SVR decreases MAP at a given cardiac
output. This figure is based on Equation 5-3, in relationship between volume and pressure, in
which MAP = (CO . SVR) + CVP. which compliance (C) equals the slope of that

Klabunde_Chap05.indd 98 6/11/2011 11:10:06 AM


CHAPTER 5 • VASCULAR FUNCTION 99

relationship, or the change in volume (ΔV) tricular ejection. However, as blood is ejected
divided by the change in pressure (ΔP) at a into the aorta, the walls of the aorta expand
given pressure: to accommodate the increase in blood vol-
ume contained within the aorta because the
ΔV
Eq. 5-4 C = or, ΔV = C ⋅ ΔP aorta is compliant. As the aorta expands, the
ΔP
increase in pressure is determined by the
Therefore, a highly compliant vessel will change in aortic volume divided by the com-
display a relatively small increase in pressure pliance of the aorta at that particular range of
for a given increase in volume. Conversely, a volumes (Fig. 5.5, panel A). The less compli-
less compliant vessel (i.e., “stiffer” vessel) will ant the aorta, the greater the pressure change
display a relatively large increase in pressure (i.e., pulse pressure) at any given change in
for a given increase in volume. aortic volume (Fig. 5.5, panel B). Age and
The compliance of a blood vessel is deter- arteriosclerotic disease decrease aortic com-
mined in large part by the relative propor- pliance, which increases aortic pulse pres-
tion of elastin fibers versus smooth muscle sure. It is not uncommon for elderly people
and collagen in the vessel wall (see Fig. 3.7). to have aortic pulse pressures of 60 mm Hg
Elastin fibers offer the least resistance to or more, whereas younger adults have aortic
stretch, whereas collagen offers the greatest pulse pressures of about 40 to 45 mm Hg at
resistance. A vessel such as the aorta that has resting heart rates.
a greater proportion of elastin fibers versus A change in compliance affects only
smooth muscle and collagen has a relatively pulse pressure and not the mean pressure,
low resistance to stretch and, therefore, has a which remains unchanged as long as cardiac
compliance that is greater than that found in a output and systemic vascular resistance do
muscular artery that has more smooth muscle not change. In contrast, a change in stroke
and less elastin. volume normally changes mean aortic pres-
The relatively high compliance of the sure in addition to pulse pressure because
aorta dampens the pulsatile output of the left the cardiac output changes. For example,
ventricle, thereby reducing the pulse pres- if stroke volume and cardiac output are
sure. If the aorta were a rigid tube, the pulse increased by an increase in inotropy, both
pressure would be very high with each ven- pulse pressure and mean arterial pressure

Decreased Increased Increased


Normal Compliance Stroke Volume Mean Aortic Pressure
Aortic Volume

V
V V V

P P P P
A B C D
40 80 120 160 40 80 120 160 40 80 120 160 40 80 120 160
Aortic Pressure Aortic Pressure Aortic Pressure Aortic Pressure
(mmHg) (mmHg) (mmHg) (mmHg)

■ FIGURE 5.5 Effects of stroke volume, aortic compliance, and mean aortic pressure on aortic pulse
pressure. Panel A. At a given stroke volume (DV), the pulse pressure (DP) is determined by the aortic
compliance (red line). Panel B. Decreasing the aortic compliance (slope of red line) increases the pulse
pressure at a given stroke volume. Panel C. Increasing the stroke volume into the aorta increases the pulse
pressure. Panel D. At higher mean aortic pressures (dotted line), a given stroke volume produces a greater
pulse pressure because the aortic compliance is less at higher pressures and volumes. Panels A–C assume
a constant mean aortic pressure.

Klabunde_Chap05.indd 99 6/11/2011 11:10:07 AM


100 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

increase. If, however, cardiac output is changes stroke volume (e.g., ventricular
not changed when stroke volume changes preload, afterload, and inotropy; heart rate)
(e.g., if a decrease in heart rate accompa- or aortic compliance (e.g., age, arteriosclero-
nies the increase in stroke volume), then sis, hypertension) alters aortic pulse pressure.
only the pulse pressure changes—the Beat−to−beat changes in pulse pressure occur
mean aortic pressure does not change owing to changes in stroke volume. In con-
(Fig. 5.5, panel C). trast, chronic, long−term increases in pulse
No single value for aortic compliance pressure are commonly due to decreased
exists because the relationship between vol- aortic compliance.
ume and pressure (compliance curve; red
line in Fig. 5.5) is not linear. At higher aortic HEMODYNAMICS (PRESSURE,
volumes and pressures, the slope of the rela- FLOW, AND RESISTANCE)
tionship decreases and compliance decreases
(see Fig. 5.5, panel D). Therefore, at elevated The term hemodynamics describes the physi-
mean arterial pressures, the reduced compli- cal factors governing blood flow within the
ance results in an increase in pulse pressure at circulatory system. Blood flow through an
a given stroke volume. organ is determined by the pressure gradient
In summary, aortic pulse pressure is deter- (ΔP) driving the flow divided by the resist-
mined by ventricular stroke volume and ance (R) to flow (Equation 5−5), which is a
aortic compliance (Fig. 5.6). Any factor that rearrangement of Equation 5−1. The pres-
sure gradient (or perfusion pressure) driving
flow through an organ is the arterial minus
the venous pressure. For an individual blood
Preload Afterload vessel, the pressure gradient is the pressure
difference between two defined points along
Inotropy Heart Rate the vessel.

Eq. 5-5 ΔP
F=
Stroke R
Volume Blood flow through organs is determined
largely by changes in resistance because
arterial and venous pressures are normally
maintained within a narrow range by vari-
Aortic Pulse ous feedback mechanisms. Therefore, it is
Pressure important to understand what determines
resistance in individual vessels and within
vascular networks.
Aortic
Effects of Vessel Length,
Compliance
Radius, and Blood Viscosity on
Resistance to Blood Flow
Age Three factors determine the resistance (R)
to blood flow within a single vessel: vessel
Arteriosclerosis length (L), blood viscosity (η), and diameter
Hypertension (or radius, r) of the vessel. These are described
■ FIGURE 5.6 Factors affecting aortic pulse pres-
by Equation 5−6 as follows:
sure. Pulse pressure is increased by those factors
η⋅L
that increase stroke volume or decrease aortic Eq. 5-6 R∝
compliance. r4

Klabunde_Chap05.indd 100 6/11/2011 11:10:07 AM


CHAPTER 5 • VASCULAR FUNCTION 101

Resistance is directly proportional to vessel radius reduces resistance. Furthermore, a


length. Therefore, a vessel that is twice as change in radius alters resistance inversely to
long as another vessel with the same radius the fourth power of the radius. For example, a
will have twice the resistance to flow. In twofold increase in radius decreases resistance
the body, individual vessel lengths do not 16−fold! Therefore, vessel resistance is exqui-
change appreciably; therefore, changes in sitely sensitive to changes in radius. Because
vessel length have only a minimal effect on changes in radius and diameter are directly
resistance. proportional, diameter can be substituted for
Resistance to flow is directly related to the radius in Equation 5−6.
viscosity of the blood. Viscosity is related to If the expression for resistance (Equation
friction generated by interactions between 5−6) is combined with the equation describ-
fluid molecules in the plasma and suspended ing the relationship between flow, pressure,
formed substances (e.g., red blood cells) as and resistance (F = ΔP/R; Equation 5−5), the
the blood is flowing. Viscosity also takes into following relationship is obtained:
account the friction generated between the
blood and the lining of the vessel. Therefore, ΔP ⋅ r 4
Eq. 5-7 F∝
viscosity can be thought of as a force that η⋅L
opposes blood flow. If viscosity increases two-
fold, the resistance to flow increases twofold, This relationship (Poiseuille’s equation)
thereby decreasing flow by one−half at con- was first described by the French physician
stant ΔP. At normal body temperatures, the Poiseuille (1846). The full equation contains π
viscosity of plasma is about 1.8 times the vis- in the numerator, and the number 8 in the
cosity of water. The viscosity of whole blood denominator (a constant of integration).
is about three to four times the viscosity of Equation 5−7 describes how flow is related to
water owing to the presence of red cells and perfusion pressure, radius, length, and viscos-
proteins. Blood viscosity normally does not ity. In the body, however, flow does not con-
change much; however, it can be significantly form precisely to this relationship because the
altered by changes in hematocrit and tem- equation assumes the following: (1) the vessels
perature and by low flow states. Hematocrit are long, straight, rigid tubes; (2) the blood
is the volume of red blood cells expressed behaves as a Newtonian fluid in which viscos-
as a percentage of a given volume of whole ity is constant and independent of flow; and
blood. If hematocrit increases from a normal (3) the blood is flowing under steady laminar
value of 40% to an elevated value of 60% (this flow (nonturbulent) conditions. Despite these
is termed polycythemia), the blood viscos- assumptions, which clearly are not always
ity approximately doubles. Decreasing blood achieved in vivo, the relationship is important
temperature increases viscosity by about because it describes the dominant influence
2% per degree centigrade. The flow rate of of vessel radius on resistance and flow, and
blood also affects viscosity. At very low flow therefore provides a conceptual framework to
states in the microcirculation—as occurs understand how physiologic and pathologic
during circulatory shock—the blood vis- changes in blood vessels and blood viscosity
cosity can increase severalfold. This occurs affect pressure and flow.
because at low flow states, cell−to−cell and The relationship between flow and radius
protein−to−cell adhesive interactions increase, (Equation 5−7) for a single vessel is shown
which can cause erythrocytes to adhere to one graphically in Figure 5.7. In this analysis,
another and increase the blood viscosity. laminar flow conditions are assumed, and
Of the three independent variables in driving pressure, viscosity, and vessel length
Equation 5−6, vessel radius is the most are held constant. As vessel radius decreases
important quantitatively for determining from a relative value of 1.0, a dramatic fall in
resistance to flow. Because radius and resist- flow occurs because flow is directly related
ance are inversely related, an increase in to radius to the fourth power. For example,

Klabunde_Chap05.indd 101 6/11/2011 11:10:09 AM


1.0

0.8 F r4
Relative Flow

@ constant P
0.6

0.4

0.2

0
0 0.2 0.4 0.6 0.8 1.0
Relative Radius

Klabunde_Chap05.indd 102 6/11/2011 11:10:09 AM


CHAPTER 5 • VASCULAR FUNCTION 103

Normal Flow

Laminar Flow
D P = 10 mmHg

2-Times Normal Flow

Turbulent Flow
■ FIGURE 5.8 Laminar versus turbulent flow. In
laminar flow, blood flows smoothly in concentric
layers parallel with the axis of the blood vessel,
with the highest velocity in the center of the ves-
sel and the lowest velocity next to the endothelial D P = 35 mmHg
lining of the vessel. When laminar flow becomes
disrupted (e.g., by a atherosclerotic plaque), it ■ FIGURE 5.9 Effects of flow on turbulence.
becomes turbulent; blood no longer flows in A twofold increase in flow across a stenotic lesion
concentric, parallel layers, but rather moves in dif- causes a disproportionate increase in the pressure
ferent paths, often forming vortices. drop (DP) across the lesion due to increased tur-
bulence. In this illustration, DP may increase three-
fold or fourfold instead of twofold as predicted by
Poiseuille relationship when flow is doubled.
Turbulence causes increased energy loss
and a greater pressure drop along a ves-
sel length than predicted by the Poiseuille across a resistance than predicted simply by
relationship (Equation 5−7). For example, the radius and length of the resistance ele-
as illustrated in Figure 5.9, if blood flow is ment because of increased energy losses asso-
increased twofold across a stenotic arterial ciated with turbulence.
segment that already has mild turbulence,
the pressure drop across the stenosis may Series and Parallel Arrangement
increase threefold or fourfold, and the turbu-
of the Vasculature
lence enhanced. The Poiseuille relationship
predicts a twofold increase in the pressure It is crucial that Poiseuille’s equation should
drop across the lesion because the pressure be applied only to single vessels. If, for
drop is proportionate to flow under laminar example, a single arteriole within the kid-
flow conditions (see Fig. 5.10). Turbulence, ney were constricted by 50%, although the
however, alters the relationship between flow resistance of that single vessel would increase
and perfusion pressure so that the relation- 16−fold, the vascular resistance for the entire
ship is no longer linear and proportionate renal circulation would not increase 16−fold.
as described by the Poiseuille relationship. The change in overall renal resistance would
Instead, a greater perfusion pressure is be so small that it would be immeasurable.
required to propel the blood at a given flow This is because the single arteriole is one of
rate when turbulence is present. Alternatively, many resistance vessels within a complex net-
a given flow causes a greater pressure drop work of vessels, and therefore, it constitutes

Klabunde_Chap05.indd 103 6/11/2011 11:10:10 AM


104 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Laminar
Flow

Flow
Turbulent
Flow
Turbulence
Begins

Perfusion Pressure
■ FIGURE 5.10 Effects of turbulence on the pressure–flow relationship. Turbulence decreases flow at any
given perfusion pressure, or requires a greater perfusion pressure to drive a given flow.

only a small fraction of the resistance for the a network of parallel resistances is less than
whole organ. To help understand this com- the resistance of the single lowest resistance;
plex arrangement of vessel architecture, it is therefore, parallel vessels greatly reduce
necessary to examine the vascular compo- resistance. For example, assume that R1 = 5,
nents in terms of series and parallel elements. R2 = 10, and R3 = 20. When the equation is
The parallel arrangement of organs and solved, RT = 2.86, a value that is less than the
their circulations (see Fig. 1.2) is important lowest individual resistance. The resistance
because parallel vessels decrease total vascular calculation for parallel networks explains
resistance. When there is a parallel arrange- why capillaries constitute a relatively small
ment of resistances, the reciprocal of the total fraction of the total vascular resistance of an
resistance is equal to the sum of the reciprocals organ or microvascular network. Although
of the individual resistances. For example, the capillaries have the highest resistance of
total resistance (RT) of three parallel resistances individual vessels because of their small
(R1, R2, R3) would be diameter, they also form a large network of
parallel vessels. This reduces their resistance
1 1 1 1 as a group of vessels. The second principle is
= + +
RT R1 R2 R3 that when many parallel vessels exist, chang-
or, solving for RT, ing the resistance of a small number of these
vessels will have little effect on total resistance.
1 Within an organ, the vascular arrangement
RT =
Eq. 5-8 1 1 1 is a combination of series and parallel
+ +
R 1 R2 R3 elements. In Figure 5.11, the artery, arteri-
oles, capillaries, venules, and vein as groups
Two important principles emerge from of vessels are in series with each other. All
Equation 5−8. First, the total resistance of of the blood that flows through the artery

Klabunde_Chap05.indd 104 6/11/2011 11:10:10 AM


Artery Arterioles Capillaries Venules Vein

Klabunde_Chap05.indd 105 6/11/2011 11:10:11 AM


106 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

CASE 5·1 This calculation is done by rearranging


Equation 5-3 as follows:
A patient was found to have
5-T segment depression in his
Eq. 5-10 SVR =(MAP- CVP)
electrocardiogram during an exercise co
stress test, suggesting the presence of
coronary artery disease. A follow-up Although systemic vascular resistance can be
coronary angiogram revealed that the calculated from mean arterial pressure and
diameter of the left main coronary artery cardiac output, its value is not determined by
(see Chapter 7, Fig. 7-6) was reduced by either of these variables (although its value
50%. If this vessel normally contributes changes depending upon the pressure-see
to 1% of the total coronary vascular below). Systemic vascular resistance is deter-
resistance under resting flow conditions, mined by vascular diameters, length, anatomi-
how much will this reduction in diameter cal arrangement of vessels, and blood viscosity.
increase total coronary vascular Because vessels are compliant, increasing
resistance? Assume no change in the intravascular pressure expands the vessels,
resistance of the vessels downstream thereby causing a small reduction in resist-
from the narrowed coronary artery. ance. Nonetheless, the decrease in systemic
Express your answer as a percentage vascular resistance that occurs when pressure
increase. increases is not owing to the pressure directly
but rather is caused by passive increases in
vessel diameter. Mathematically, systemic vas-
REGULATION OF SYSTEMIC cular resistance is the dependent (calculated)
VASCULAR RESISTANCE variable in Equation 5-10; however, physi-
ologically, systemic vascular resistance and
Systemic vascular resistance, which is cardiac output are the independent variables
sometimes called total peripheral resistance normally, and mean arterial pressure is the
(TPR), is the resistance to blood flow offered dependent variable; that is, mean arterial pres-
by all of the systemic vasculature, excluding sure changes in response to changes in cardiac
the pulmonary vasculature. Systemic vascular output and systemic vascular resistance.
resistance primarily is determined by changes When calculating systemic vascular resist-
in vascular diameters, although changes in ance, it is customary to use the units of mm
blood viscosity also affect systemic vascular Hg/mL · min- 1 (peripheral resistance units,
resistance. Mechanisms that cause generalized PRU) or the units of dynes· s/cm5 (in which
vasoconstriction will increase systemic vas- pressure is expressed in dynes/cm2 instead of
cular resistance, and mechanisms that cause mm Hg; 1 mm Hg = 1330 dynes/cm2) and flow
vasodilation will decrease systemic vascular is expressed in cm3/s. When calculating resist-
resistance. The increase in systemic vascular ance in PRU, pressure has the units of mm Hg
resistance in response to sympathetic stimula- and cardiac output is expressed in mUmin.
tion, for example, depends on the degree of
sympathetic activation, the responsiveness of PROBLEM 5·3
the vasculature, and the number of vascular Infusion of a drug is found to increase
beds involved. cardiac output by 30% and decrease
mean arterial pressure by 10%. By what
Calculation of Systemic Vascular percentage does this drug change
Resistance systemic vascular resistance? Is this
drug a vasodilator or vasoconstrictor?
Systemic vascular resistance (SVR) can be
Assume that CVP is 0 mm Hg and does
calculated if cardiac output (CO), mean
not change.
arterial pressure (MAP), and CVP are known.

Klabunde_Chap05.indd 106 6/11/2011 11:10:12 AM


CHAPTER 5 • VASCULAR FUNCTION 107

Vascular Tone others promote smooth muscle relaxation and


vascular dilation (e.g., endothelial−derived
Under normal physiologic conditions, nitric oxide and tissue metabolites such as
changes in the diameter of precapillary resist- adenosine and hydrogen ion). Therefore, at
ance vessels (small arteries and arterioles) any given time, vasoconstrictor and vasodi-
represent the most important mechanism lator influences are competing to determine
for regulating systemic vascular resistance. the vascular tone. The extrinsic and intrin-
Resistance vessels are normally in a partially sic mechanisms regulating vascular tone are
constricted state that is referred to as the vas- described in more detail in Chapters 6 and 7.
cular tone of the vessel. This tone is gener- In general, the vasoconstrictor mechanisms
ated by smooth muscle contraction within are important for maintaining systemic vascu-
the wall of the blood vessel. From this par- lar resistance and arterial pressure, whereas
tially constricted state, a vessel can constrict vasodilator mechanisms regulate blood flow
further and thereby increase resistance, or it within organs. For example, if the body needs
can dilate by smooth muscle relaxation and to maintain arterial blood pressure when a
thereby decrease resistance. Venous vessels person stands up, vasoconstrictor mecha-
likewise possess a level of vascular tone. nisms (primarily sympathetic adrenergic) are
Extrinsic and intrinsic mechanisms deter- activated to constrict resistance vessels and
mine the degree of smooth muscle activation increase systemic vascular resistance. If an
(Fig. 5.12). Extrinsic mechanisms, such as organ requires more blood flow and oxygen
sympathetic nerves and circulating hormones, delivery (e.g., exercising muscle), vasodila-
originate outside of the organ or tissue. tor mechanisms will predominate and over-
Intrinsic mechanisms originate from within ride vasoconstrictor influences. Therefore,
the blood vessel or the tissue surrounding the competition between vasoconstrictor and
the vessel. Examples of intrinsic mechanisms vasodilator influences can be thought of as
include endothelial−derived factors, smooth competition between maintenance of arterial
muscle myogenic tone, locally produced blood pressure and organ perfusion.
hormones, and tissue metabolites. Some of
these extrinsic and intrinsic factors promote
vasoconstriction (e.g., sympathetic nerves, VENOUS BLOOD PRESSURE
angiotensin II, and endothelin−1), whereas
Venous pressure is a general term that
Extrinsic Intrinsic represents the average blood pressure within
the venous compartment. A more specific term,
Arteriole Tissue central venous pressure (CVP), describes the
Metabolites blood pressure in the thoracic vena cava near
Neural
Local the right atrium. This pressure is important
Hormones because it determines the filling pressure of
Myogenic the right ventricle, and thereby determines
Humoral
ventricular stroke volume through the Frank−
Endothelial Starling mechanism as discussed in Chapter 4.
Factors

Constriction Dilation
Venous Blood Volume
and Compliance
Several factors influence CVP: cardiac output,
■ FIGURE 5.12 Vascular tone. The state of vessel respiratory activity, contraction of skeletal mus-
tone is determined by the balance between constric- cles (particularly leg and abdominal muscles),
tor and dilator influences. Extrinsic influences origi-
nate outside of the tissue, whereas intrinsic influences
sympathetic vasoconstrictor tone, and gravi-
originate from the vessel or surrounding tissue. tational forces. All of these factors ultimately

Klabunde_Chap05.indd 107 6/11/2011 11:10:13 AM


108 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

change CVP (ΔPV) by changing either venous of venous tone, and the slope of a tangent
blood volume (ΔVV) or venous compliance line at any point on the curve represents the
(CV) as described by Equation 5−11. compliance. Looking at a single curve, it is
evident that an increase in venous volume
ΔVV
Eq. 5-11 ΔPV ∝ will increase venous pressure (point A to B).
CV The amount by which the pressure increases
for a given change in volume depends on the
Equation 5−11 is a rearrangement of the
slope of the relationship between the volume
equation used to define compliance (Equa-
and pressure (i.e., the compliance). As with
tion 5−4), in which compliance (in this
arterial vessels (see Fig. 5.5), the relationship
case venous compliance) equals a change in
between venous volume and pressure is not
venous volume divided by a change in venous
linear (see Fig. 5.13). The slope of the compli-
pressure. Therefore, an increase in venous vol-
ance curve (ΔV/ΔP) is greater at low pressures
ume increases venous pressure by an amount
and volumes than at higher pressures and
determined by the compliance of the veins.
volumes. The reason for this is that at very
Furthermore, a decrease in venous compli-
low pressures, a large vein collapses. As the
ance, as occurs during sympathetic activation
pressure increases, the collapsed vein assumes
of veins, increases venous pressure.
a more cylindrical shape with a circular cross−
The relationship described by Equation 5−11
section. Until a cylindrical shape is attained,
can be depicted graphically as shown in
the walls of the vein are not stretched appreci-
Figure 5.13, in which venous blood volume
ably. Therefore, small changes in pressure can
is plotted against venous blood pressure. The
result in a large change in volume by changes
different curves represent different states
in vessel geometry rather than by stretching
the vessel wall. At higher pressures, when the
vein is cylindrical in shape, increased pressure
can increase the volume only by stretching the
Vein Shape vessel wall, which is resisted by the structure
B and composition of the wall (particularly by
collagen, smooth muscle, and elastin com-
A ponents). Therefore, at higher volumes and
Volume

C
pressures, the change in volume for a given
change in pressure (i.e., compliance) is less.
The smooth muscle within veins is ordinar-
Increased ily under some degree of tonic contraction. Like
Tone arteries and arterioles, a major factor determin-
(¯ Compliance) ing venous smooth muscle contraction is sym-
pathetic adrenergic stimulation, which occurs
under basal conditions. Changes in sympa-
Pressure thetic activity can increase or decrease the
contraction of venous smooth muscle, thereby
■ FIGURE 5.13 Compliance curves for a vein.
Venous compliance (the slope of line tangent to a altering venous tone. When this occurs, a
point on the curve) is very high at low pressures change in the volume–pressure relationship
because veins collapse. As pressure increases, the (or compliance curve) occurs, as depicted in
vein assumes a more circular cross−section and its
Figure 5.13. For example, increased sympa-
walls become stretched; this reduces compliance
(decreases slope). Point A is the control pressure thetic activation shifts the compliance curve
and volume. Point B shows how pressure increases down and to the right, decreasing its slope
along the compliance curves as volume increases. (compliance) at any given volume (from point
Point C shows how pressure increases as vol-
A to C in Fig. 5.13). This rightward diagonal
ume decreases when venous tone is increased
(decreased compliance) by sympathetic stimula- shift in the venous compliance curve results in
tion of the vein, for example. a decrease in venous volume and an increase

Klabunde_Chap05.indd 108 6/11/2011 11:10:14 AM


CHAPTER 5 • VASCULAR FUNCTION 109

in venous pressure. Drugs that reduce venous (supine position), systemic blood vessels
tone (e.g., nitrodilators) will decrease venous are positioned near the hydrostatic level of
pressure while increasing venous volume by the heart, which causes a generally uniform
shifting the compliance curve to the left. distribution of the blood volume between
the head, thorax, abdomen, and legs. When
Mechanical Factors Affecting supine, CVP averages about 2 mm Hg, and
Central Venous Pressure and venous pressure in the legs is only a few mm
Venous Return Hg above CVP. When a person changes from
supine to a standing posture, gravity acts on
Several of the factors affecting CVP can be
the vascular volume, causing blood to accu-
classified as mechanical (or physical) factors.
mulate in the lower extremities (Fig. 5.14).
These include gravitational effects, respira-
Because venous compliance is much higher
tory activity, and skeletal muscle contraction.
than arterial compliance, the shift in blood
Gravity passively alters CVP and volume,
volume to the legs increases their venous pres-
and respiratory activity and muscle contrac-
sure and volume. In fact, venous pressures in
tion actively promote or impede the return of
the feet when a person is standing still may
blood into the central venous compartment,
reach 90 mm Hg because of the increased
thereby altering CVP and volume.
hydrostatic pressure owing to the influence of
gravity. The shift in blood volume from the
GRAVITY
thorax to the dependent limbs causes thoracic
Gravity exerts significant effects on CVP and venous volume and CVP to fall. This reduces
venous return. When a person is reclining right ventricular filling pressure (preload)

CVP CVP

Thorax Thorax
Gravity

Vena Cava Aorta

Legs Legs
Pv > CVP Pv >> CVP
Pv Pv

Reclining Standing
■ FIGURE 5.14 Effects of gravity on central venous pressure (CVP) and venous pressure (PV) in the lower
leg. When horizontal (reclining), thoracic blood volume and CVP are relatively high, and PV is only a few
mm Hg above CVP. When standing upright, the force of gravity causes a large increase in venous pressure
in the legs, expanding the compliant veins and increasing their volume. This translocation of blood volume
to the veins in the legs reduces thoracic volume and pressure.

Klabunde_Chap05.indd 109 6/11/2011 11:10:14 AM


110 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

and stroke volume by the Frank−Starling


mechanism. Left ventricular stroke volume
subsequently falls because of reduced pulmo-
nary venous return to the left ventricle; the
reduced stroke volume causes cardiac out-
put and arterial blood pressure to decrease. If
systemic arterial pressure falls by more than
20 mm Hg upon standing, this is termed
orthostatic or postural hypotension. When
this occurs, cerebral perfusion may fall and a
person may become “light headed” and expe-
rience a transient loss of consciousness (syn-
cope). Normally, baroreceptor reflexes (see
Chapter 6) are activated to restore arterial Relaxed Contracted
pressure by causing peripheral vasoconstric-
■ FIGURE 5.15 Rhythmic contraction of skeletal
tion and cardiac stimulation (increased heart muscle compresses veins, particularly in the lower
rate and inotropy). Furthermore, as seen in limbs, and propels blood toward the heart through
the next section, limb movement (e.g., walk- a system of one−way valves.
ing) in the upright position facilitates venous
return, thereby lowering venous pressures in
the leg and partially restoring CVP. helps to counteract gravitational forces when a
The influence of gravity on venous volume person stands up by facilitating venous return
can be visualized by looking at the veins on and lowering venous and capillary pressures
the back of the hand when below the level in the feet and lower limbs. For example,
of the heart and then raising the hand above human experiments have shown that when a
the head. When this is done, the veins col- person is standing still, the venous pressure
lapse because they are above the level of the measured at the ankle is about 90 mm Hg.
heart and the negative hydrostatic pressure When the person begins to walk, the venous
caused by gravity acting on the column of pressure falls to 30 to 40 mm Hg after sev-
blood reduces the pressure within the veins. eral steps. When the venous valves become
If the hand is suddenly brought down below incompetent, as occurs when veins become
the heart, the veins will refill with blood as enlarged (varicose veins), muscle pumping
the positive hydrostatic pressure increases the becomes ineffective. Besides the loss of mus-
venous pressure and distends the veins. cle pumping in aiding venous return, blood
volume and pressure increase in the veins of
the dependent limbs, which increases cap-
SKELETAL MUSCLE PUMP illary pressure and may cause edema (see
Veins, particularly in extremities, contain Chapter 8).
one−way valves that permit blood flow
RESPIRATORY ACTIVITY
toward the heart and prevent retrograde flow.
(ABDOMINOTHORACIC OR
Deep veins in the lower limbs are surrounded
RESPIRATORY PUMP)
by large groups of muscle that compress the
veins when the muscles contract. This com- Venous return to the right atrium from the
pression increases the pressure within the abdominal vena cava is determined by the
veins, which closes upstream valves and pressure difference between the abdominal
opens downstream valves, thereby function- vena cava and the right atrial pressure, as well
ing as a pumping mechanism (Fig. 5.15). This as by the resistance to flow, which is primar-
pumping mechanism plays a significant role ily determined by the diameter of the thoracic
in facilitating venous return during exercise. vena cava. Therefore, increasing right atrial
Rhythmical contraction of leg muscles also pressure impedes venous return, whereas

Klabunde_Chap05.indd 110 6/11/2011 11:10:15 AM


CHAPTER 5 • VASCULAR FUNCTION 111

lowering right atrial pressure facilitates intrapleural pressure causes the transmural
venous return. These changes in venous pressure to increase across the chamber walls.
return significantly influence stroke volume The transmural pressure is the difference
through the Frank−Starling mechanism. between the pressure within the chamber and
Pressures and volumes in the right atrium the pressure outside the chamber (Ppl). When
and thoracic vena cava depend on the sur- transmural pressure increases, the chamber
rounding intrapleural pressure. This pres- volume increases, which increases sarcomere
sure is measured in the space between the length and myocyte preload. For example, if
thoracic wall and the lungs and is generally intrapleural pressure is normally −4 mm Hg
negative (subatmospheric). During inspi- at end−expiration and right atrial pressure is
ration, the chest wall expands and the dia- 0 mm Hg, the transmural pressure (the pres-
phragm descends (red arrows on chest wall sure that distends the atrial chamber) is 4 mm
and diaphragm in Fig. 5.16). This causes Hg. During inspiration, if intrapleural pres-
the intrapleural pressure (Ppl) to become sure decreases to −8 mm Hg and atrial pressure
more negative, causing expansion of the decreases to −2 mm Hg, the transmural pres-
lungs, atrial and ventricular chambers, and sure across the atrial chamber increases from
vena cava (smaller red arrows). This expan- 4 to 6 mm Hg, thereby expanding the cham-
sion decreases the pressures within the ves- ber. At the same time, because blood pressure
sels and cardiac chambers. As right atrial within the atrium is diminished, this leads
pressure falls during inspiration, the pres- to an increase in venous return to the right
sure gradient for venous return to the heart atrium from the abdominal vena cava. Similar
is increased. During expiration the opposite increases in right ventricular transmural pres-
occurs, although the net effect of respiration sure and preload occur during inspiration. The
is that the increased rate and depth of ventila- increase in sarcomere length during inspira-
tion facilitates venous return and ventricular tion augments right ventricular stroke volume
stroke volume. by the Frank−Starling mechanism. In addition,
Although it may appear paradoxical, the changes in intrapleural pressure during inspi-
fall in right atrial pressure during inspiration ration influence the left atrium and ventricle;
is associated with an increase in right atrial however, the expanding lungs and pulmonary
and ventricular preloads and right ventricular vasculature act as a capacitance reservoir
stroke volume. This occurs because the fall in (pulmonary blood volume increases) so that

Chest Wall Inspiration Expiration


Expiration
-4
_ _
_ Diaphram Ppl -8

Air Lungs 0
_ PRA
_ -2
_
_ RV _ Ppl _
SVC RA IVC Venous
Venous Venous Return
_ _
Return Return

■ FIGURE 5.16 Effects of respiration on venous return. Left panel. During inspiration, intrapleural pressure
(Ppl) decreases as the chest wall expands and the diaphragm descends (large red arrows). This increases
the transmural pressure across the superior and inferior vena cava (SVC and IVC), right atrium (RA), and
right ventricle (RV), which causes them to expand. This facilitates venous return and leads to an increase
in atrial and ventricular preloads. Right panel. During inspiration, Ppl and right atrial pressure (PRA) become
more negative, which increases venous return. During expiration, Ppl and PRA become less negative and
venous return falls. Numeric values for Ppl and PRA are expressed as mm Hg.

Klabunde_Chap05.indd 111 6/11/2011 11:10:15 AM


112 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

the left ventricular filling is not enhanced Summary of Factors Affecting


during inspiration. During expiration, how- Central Venous Pressure
ever, blood is forced from the pulmonary
vasculature into the left atrium and ventricle, As previously discussed, CVP plays a very
thereby increasing left ventricular filling and important role in cardiac filling and ventricular
stroke volume. The net effect of respiration is stroke volume (via the Frank−Starling mecha-
that increasing the rate and depth of respiration nism). Conditions that elevate CVP increase car-
increases venous return and cardiac output. diac output, whereas conditions that decrease
If a person exhales forcefully against a closed CVP decrease cardiac output. Furthermore, as
glottis (Valsalva maneuver), intrapleural pres- described in Chapter 8, elevation in CVP can
sure becomes very positive, which causes lead to peripheral edema. Therefore, it is impor-
the transmural pressure to become negative, tant to understand how the following different
thereby collapsing the thoracic vena cava. This conditions influence CVP (see Fig. 5.17):
dramatically increases resistance to venous 1. An increase in total blood volume
return and reduces venous return. Because (hypervolemia), as occurs in renal failure
of the accompanying decrease in transmural or with activation of the renin−angioten-
pressure across the ventricular chamber walls, sin−aldosterone system (see Chapter 6),
ventricular volume decreases (particularly increases thoracic blood volume and there-
the more compliant right ventricle) despite fore CVP.
the large increase in the pressure within the 2. A decrease in cardiac output, caused by
chamber. Decreased chamber volume (i.e., decreased heart rate (e.g., bradycardia) or
decreased preload) leads to a fall in ven- stroke volume (e.g., ventricular failure),
tricular stroke volume by the Frank−Starling causes blood to back up into the venous
mechanism. Similar changes can occur when circulation (increased venous volume)
a person strains while having a bowel move- as less blood is pumped into the arterial
ment or when a person lifts a heavy weight circulation. The resultant increase in
while holding his or her breath. thoracic blood volume elevates CVP.

Total Blood Muscle


Volume Pump
+ +
Cardiac Respiratory
Output + + Pump
CVP
SVR + + Valsalva
+ +
Venous Reclining or
Compliance Squatting
■ FIGURE 5.17 Summary of conditions that alter central venous pressure (CVP). (+), increase CVP; (−),
decrease CVP.

Klabunde_Chap05.indd 112 6/11/2011 11:10:15 AM


CHAPTER 5 • VASCULAR FUNCTION 113

3. A decrease in systemic vascular resist- and does not show what factors determine
ance by selective arterial dilation increases venous return from the capillaries. Venous
blood flow from the arterial into the venous return from capillaries is determined by the dif-
compartments, thereby increasing venous ference between the mean capillary and right
volume and CVP, while at the same time atrial pressures divided by the resistance of all
reducing arterial volume and pressure (dis- the postcapillary vessels. If we consider venous
cussed later in this chapter). return as being all the systemic flow return-
4. Constriction of peripheral veins (reduced ing to the heart, venous return is determined
venous compliance) elicited by sympa- by the difference between the mean aortic and
thetic activation or circulating vasocon- right atrial pressures divided by the systemic
strictor substances (e.g., catecholamines, vascular resistance. Therefore, the pressures
angiotensin II) causes blood volume to and resistances that are used as the hemody-
be translocated from peripheral veins namic variables for determining venous return
into the thoracic compartment, thereby depend on whether one is defining venous
increasing CVP. return from specific locations in the systemic
5. Postural changes such as moving from a vasculature, or if one is viewing venous return
standing to a reclining or squatting posi- as the flow of blood throughout all the systemic
tion diminishes venous pooling in the legs circulation as it travels back to the heart.
caused by gravity, which increases thoracic An important concept to note is the follow-
volume and CVP. ing: under steady-state conditions, venous return
6. A forceful expiration against a high resist- equals cardiac output when averaged over time.
ance (Valsalva maneuver) causes external The reason for this is that the cardiovascular
compression of the thoracic vena cava system is essentially a closed system. Strictly
(decrease in functional compliance), which speaking, the cardiovascular system is not a
increases CVP. closed system because fluid is lost through the
7. Increased respiratory activity (abdomino- kidneys and by evaporation through the skin,
thoracic pump) facilitates venous return and fluid enters the circulation through the
into the thorax, thereby helping to main- gastrointestinal tract. Nevertheless, a balance is
tain CVP when cardiac output is elevated maintained between fluid entering and leaving
during exercise. the circulation during steady−state conditions.
8. Rhythmic muscular contraction (muscle Therefore, it is appropriate to view the system
pump), particularly of the limbs during as closed, and therefore cardiac output and
exercise, compresses the veins and facili- venous return as being equal. There may occur
tates venous return into the thoracic transient imbalances, such as when a person
compartment, which increases CVP. suddenly starts to run and venous return is
augmented by the muscle and abdominotho-
VENOUS RETURN AND racic pumps; however, this augmentation leads
CARDIAC OUTPUT to an increase in cardiac output by the Frank−
Starling mechanism and cardiac stimulation
The Balance between Venous so that shortly after starting to run the cardiac
output once again equals the venous return,
Return and Cardiac Output
although at a higher level of cardiac output.
Venous return is the flow of blood back to
the heart. It was previously described how the
Systemic Vascular Function Curves
venous return to the right atrium from the
abdominal vena cava is determined by the pres- Blood flow through the entire systemic
sure gradient between the abdominal vena cava circulation, whether viewed as the flow
and the right atrium, divided by the resistance leaving the heart (cardiac output) or return-
of the vena cava. However, that analysis looks ing to the heart (venous return), depends on
at only a short segment of the venous system both cardiac and systemic vascular function.

Klabunde_Chap05.indd 113 6/11/2011 11:10:15 AM


114 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

As described in more detail below, cardiac sure, cardiac output, and systemic vascular
output under normal physiologic conditions resistance (see Equation 5−3). As cardiac
depends on systemic vascular function. Cardiac output is reduced to zero, right atrial pres-
output is limited to a large extent by the prevail- sure continues to rise and mean aortic
ing state of systemic vascular function. There- pressure continues to fall, until both pres-
fore, it is important to understand how changes sures are equivalent, which occurs when
in systemic vascular function affect cardiac out- systemic blood flow ceases. When all flow
put and venous return (or total systemic blood ceases, pressures throughout all the sys-
flow because cardiac output and venous return temic circulation are equal. The pressure at
are equal under steady−state conditions). zero systemic flow, which is called the mean
The best way to show how systemic circulatory filling pressure, is about 7 mm
vascular function affects systemic blood flow Hg. This value is found experimentally when
is by use of systemic vascular and cardiac baroreceptor reflexes are blocked; otherwise
function curves. Credit for the conceptual the value for mean circulatory filling pres-
understanding of the relationship between sure is higher because of vascular smooth
cardiac output and systemic vascular func- muscle contraction and decreased vascular
tion goes to Arthur Guyton and colleagues, compliance owing to sympathetic activation.
who conducted extensive experiments in the The reason right atrial pressure increases
1950s and 1960s. To develop the concept of in response to a decrease in cardiac output is
systemic vascular function curves, we must that less blood per unit time is translocated by
understand the relationship between car- the heart from the venous to the arterial vas-
diac output, mean aortic pressure, and right cular compartment. This leads to a reduction
atrial pressure. Figure 5.18 shows that at a in arterial blood volume and pressure, and to
cardiac output of 5 L/min, the right atrial an increase in venous blood volume and pres-
pressure is near zero and mean aortic pres- sure, which increases right atrial pressure.
sure is about 95 mm Hg. If cardiac output When the heart is completely stopped and
is reduced experimentally, right atrial pres- there is no flow in the systemic circulation,
sure increases and mean aortic pressure the intravascular pressure found throughout
decreases. The fall in aortic pressure reflects the entire vasculature is a function of total
the relationship between mean aortic pres- blood volume and vascular compliance.
Finally, it is important to note in Figure 5.18
100 that if one attempts to increase cardiac out-
Mean put above 5 L/min by increasing heart rate,
Pressure (mmHg)

Aortic for example, cardiac output will not increase


Pressure much above 5 L/min. The reason is that right
atrial pressure falls below zero, which collapses
50 the vena cava at the level of the diaphragm
where it enters the thorax from the abdomen.
Right This increases the resistance of the vena cava,
Atrial thereby limiting venous return into the thorax,
Pressure which limits the cardiac output.
Pmc
0 The magnitude of the relative changes in
aortic and right atrial pressures from a normal
0 5 cardiac output to zero cardiac output is deter-
Cardiac Output (L/min)
mined by the ratio of venous to arterial com-
■ FIGURE 5.18 Effects of cardiac output on mean pliances. If venous compliance (CV) equals
aortic and right atrial pressures. Decreasing car- the change in venous volume (ΔVV) divided
diac output results in a rise in right atrial pressure
by the change in venous pressure (ΔPV), and
and a fall in aortic pressure. When cardiac output
is zero, both pressures equilibrate at the mean arterial compliance (CA) equals the change in
circulatory filling pressure (Pmc). arterial volume (ΔVA) divided by the change in

Klabunde_Chap05.indd 114 6/11/2011 11:10:16 AM


CHAPTER 5 • VASCULAR FUNCTION 115

arterial pressure (ΔPA), the ratio of venous to the systemic vascular function curve. This rela-
arterial compliance (CV/CA) can be expressed tionship can be thought of as either the effect of
by the following equation: cardiac output on right atrial pressure (cardiac
output being the independent variable) or the
CV ΔVV / ΔPV effect of right atrial pressure on venous return
Eq. 5-12 =
CA ΔVA / ΔPA (right atrial pressure being the independent
variable). When viewed from the latter
When the heart is stopped, the decrease perspective, systemic vascular function curves
in arterial blood volume (ΔVA) equals the are sometimes called venous return curves.
increase in venous blood volume (ΔVV). The value of the x−intercept in Figure 5.19
Because ΔVA equals ΔVV, Equation 5−12 can is the mean circulatory filling pressure, which
be simplified to the following relationship: is the pressure throughout the vascular sys-
CV ΔPA tem when there is no blood flow. This value
Eq. 5-13 ∝ depends on the vascular compliance and
CA ΔPV
blood volume (Fig. 5.19, panel A). Increased
Equation 5−13 shows that the ratio of venous blood volume or decreased venous compli-
to arterial compliance is proportional to the ance causes a parallel shift of the vascular
ratio of the changes in arterial to venous pres- function curve to the right, which increases
sures when the heart is stopped. This ratio is mean circulatory filling pressure. Decreased
usually in the range of 10 to 20. If, for exam- blood volume or increased venous compli-
ple, the ratio of venous to arterial compliance ance causes a parallel shift to the left and a
is 15, there is a 1 mm Hg increase in right decrease in the mean circulatory filling pres-
atrial pressure for every 15 mm Hg decrease sure. Therefore, at a given cardiac output, an
in mean aortic pressure. increase in total blood volume (or decreased
If the right atrial pressure curve from Figure venous compliance) is associated with an
5.18 is plotted as cardiac output versus right increase in right atrial pressure.
atrial pressure (i.e., reversing the axis), the rela- Decreased systemic vascular resistance
tionship shown in Figure 5.19 (black curve in increases the slope without appreciably chang-
both panels) is observed. This curve is called ing mean circulatory filling pressure (Fig. 5.19,

A B
10
Cardiac Output (L/min)

Vol
SVR
Cv
5

Vol
SVR
Cv
0
0 10 0 10
PRA (mmHg) Pmc PRA (mmHg) Pmc
■ FIGURE 5.19 Systemic function curves. Panel A shows the effects of changes in cardiac output on
right atrial pressure (PRA) and mean circulatory filling pressures (Pmc). Changes in blood volume (Vol) and
venous compliance (Cv) cause parallel shifts in the curves and changing Pmc. Panel B shows how changes
in systemic vascular resistance (SVR) alter the slope of the systemic function curves without changing Pmc.

Klabunde_Chap05.indd 115 6/11/2011 11:10:16 AM


116 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

panel B). Increased systemic vascular resistance


decreases the slope while keeping the same
10 Enhanced Normal
mean circulatory filling pressure. Therefore, at
a given cardiac output, a decrease in systemic
Cardiac
vascular resistance increases right atrial pres-
Output Depressed
sure, whereas an increase in systemic vascu- 5
lar resistance decreases right atrial pressure. (L/min)
These changes can be difficult to conceptual-
ize, but the following explanation might help
to clarify. When small resistance vessels dilate 0
at a constant cardiac output, the rate of blood 0 10
flow from the arteries into the capillaries and
veins increases. This causes a transient imbal-
PRA (mmHg)
ance between the rate of flow into the arterial ■ FIGURE 5.20 Cardiac function curves. Cardiac
system (cardiac output) and the rate of flow output is plotted as a function of right atrial pres-
out of the arterial system (more blood leaves sure (PRA); normal (solid black), enhanced (red), and
depressed curves (red) are shown. Cardiac perfor-
than enters the arterial system per unit time). mance, measured as cardiac output, is enhanced
The increase in venous volume causes venous (curves shift up and to the left) by an increase in
pressure and right atrial pressure to increase. heart rate and inotropy and a decrease in afterload.
This reduces the pressure gradient for venous
return from the capillaries and will lead to a
new steady state where there is once again a because cardiac output, not stroke volume
balance between the flow that enters and leaves as in Figure 4.21, is the dependent variable.
the arteries. This steady state will be character- With a “normal” function curve, the cardiac
ized by increased venous volume and pressure, output is about 5 L/min at a right atrial pres-
and decreased arterial volume and pressure. sure of about 0 mm Hg. If cardiac performance
If the heart were suddenly stopped, the mean is enhanced by increasing heart rate or inot-
circulatory filling pressure would not be ropy or by decreasing afterload, it shifts the
appreciably different from before the systemic cardiac function curve up and to the left. At
vascular resistance was reduced because the the same right atrial pressure of 0 mm Hg,
decrease in arterial diameter (which increases the cardiac output will increase. Conversely,
arterial compliance) has little affect on overall a depressed cardiac function curve, as occurs
vascular compliance, which is overwhelmingly with decreased heart rate or inotropy or with
determined by venous compliance. increased afterload, will decrease the cardiac
output at any given right atrial pressure. How-
Cardiac Function Curves ever, the magnitude by which cardiac output
changes when cardiac performance is altered is
According to the Frank−Starling relationship, determined in large part by the state of systemic
an increase in right atrial pressure increases car- vascular function. Therefore, it is necessary
diac output. This relationship can be depicted to examine both cardiac and system vascular
using the same axis as used in systemic func- function at the same time.
tion curves in which cardiac output (depend-
ent variable) is plotted against right atrial Interactions between Cardiac
pressure (independent variable) (Fig. 5.20).
and Systemic Vascular
These curves are similar to the Frank−Starling
Function Curves
curves shown in Figure 4.21. There is no sin-
gle cardiac function curve, but rather a family By themselves, systemic vascular function and
of curves that depends on the inotropic state cardiac function curves provide an incomplete
and afterload (see Chapter 4). Changes in picture of overall cardiovascular dynamics;
heart rate also shift the cardiac function curve however, when coupled together, these curves

Klabunde_Chap05.indd 116 6/11/2011 11:10:17 AM


CHAPTER 5 • VASCULAR FUNCTION 117

15

Cardiac Output (L/min)


Cardiac
Stimulation D
Normal
Cardiac
C Function
10
¯ CV &
B ¯ SVR
5 A
¯ CV

0
0 10
PRA (mmHg)
■ FIGURE 5.21 Combined cardiac and systemic function curves: effects of exercise. Cardiac output
is plotted against right atrial pressure (PRA) to show the effects of altering both cardiac and systemic
function. Point A represents the normal operating point described by the intercept between the normal
cardiac and systemic function curves. Cardiac stimulation alone changes the intercept from point A to B.
Cardiac stimulation coupled with decreased venous compliance (CV) shifts the operating intercept to
point C. If systemic vascular resistance (SVR) also decreases, which is similar to what occurs during exer-
cise, the new intercept becomes point D.

can offer new understanding as to the way decrease in systemic vascular resistance, car-
cardiac and vascular function are coupled. diac output would be further enhanced (point
When the cardiac function and vascular D). These changes in venous compliance and
function curves are superimposed (Fig. 5.21), systemic vascular resistance, which occur
a unique intercept between a given cardiac during exercise, permit the cardiac output to
and a given vascular function curve (point A) increase. This example shows that for cardiac
exists. This intercept is the equilibrium point output to increase significantly during cardiac
that defines the relationship between cardiac stimulation, there must be some alteration in
and vascular function. The heart functions at vascular function so that venous return is aug-
this equilibrium until one or both curves shift. mented and right atrial pressure (ventricular
For example, if the sympathetic nerves to the filling) is maintained. Therefore, in the normal
heart are stimulated to increase heart rate heart, cardiac output is limited by factors that
and inotropy, only a small increase in cardiac determine vascular function.
output will occur, accompanied by a small In pathologic conditions such as heart fail-
decrease in right atrial pressure (point B). ure, cardiac function limits venous return. In
As previously discussed, cardiac stimulation heart failure, ventricular inotropy is dimin-
alone will not increase cardiac output appreci- ished, total blood volume is increased, and
ably if right atrial pressure becomes negative. systemic vascular resistance is increased (see
If at the same time, however, the venous com- Chapter 9).The former two lead to an increase
pliance is decreased by sympathetic activation in atrial and ventricular pressures and vol-
of venous vasculature, cardiac output will be umes (increased preload), which enables
greatly augmented (point C). If the decrease the Frank−Starling mechanism to partially
in venous compliance is accompanied by a compensate for the loss of inotropy. These

Klabunde_Chap05.indd 117 6/11/2011 11:10:17 AM


Cardiac Output (L/min)
10 Vol
Cv Cardiac
SVR Failure
5 A
C
B

0
0 10 20 30
PRA (mmHg)

Klabunde_Chap05.indd 118 6/11/2011 11:10:17 AM


CHAPTER 5 • VASCULAR FUNCTION 119

power, and it is directly related to • Arteries and veins are normally in


vessel length and blood viscosity. a partially constricted state (i.e.,
Vessel radius is the most important they possess vascular tone), which
factor for regulating resistance. is determined by the net effect of
• The parallel arrangement of vascular vasoconstrictor and vasodilator
beds in the body reduces overall influences acting upon the vessel.
resistance. Furthermore, because of • CVP is altered by changes in thoracic
this arrangement, a resistance change blood volume and venous compliance.
in one vascular bed has minimal Gravity, respiratory activity, and
influence on pressure and flow in other the pumping action of rhythmically
vascular beds. contracting skeletal muscle have
• Changes in large artery resistance important influences on CVP.
have little effect on total resistance • Cardiac output is strongly influenced
of a vascular bed because these by changes in systemic vascular
vessels normally comprise only a small function as described by cardiac and
percentage of the total resistance of systemic vascular function curves.
a vascular bed. In contrast, changes in In the normal heart, cardiac output
small artery and arteriolar resistances is limited by factors that determine
greatly affect total resistance. vascular function.

REVIEW QUESTIONS

For each question, choose the one best 2. A 17-year-old male patient who runs
answer: on the high school cross-country team
is found to have an arterial pressure of
1. Concerning different types of blood ves- 115/60 and a resting heart rate of
sels in a vascular network, 55 beats/minute. The most likely
a. Arterioles have the highest individual explanation for his elevated arterial pulse
resistance and, therefore, as a group pressure is
of vessels, have the greatest pressure a. Decreased mean arterial pressure.
drop. b. Elevated ventricular stroke volume.
b. Capillaries as a group of vessels con- c. Increased aortic compliance.
stitute the greatest resistance to flow d. Reduced systemic vascular resistance.
within an organ.
c. Capillaries and venules are the pri-
mary site for fluid exchange.
d. Large arteries are the most important
vessels for blood flow and pressure
regulation.

Klabunde_Chap05.indd 119 6/11/2011 11:10:19 AM


120 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

3. A patient who has coronary artery resistance in the kidney is unchanged,


disease is treated with a drug that blood flow to the right kidney will
reduces heart rate by 10% without decrease by what amount?
changing stroke volume. Furthermore, a. 50%
the drug is found to decrease mean b. <20%
arterial pressure by 10%. Assume that c. 8−fold
central venous pressure remains at 0 mm d. 16−fold
Hg. This drug
a. Decreases systemic vascular resistance 7. A patient in the Emergency Department
by 10%. with traumatic injuries from an automo-
b. Does not alter cardiac output. bile accident suddenly shows a fall in arte-
c. Does not alter systemic vascular rial pressure accompanied by an increase
resistance. in central venous pressure. These hemo-
d. Reduces pressure by dilating the dynamic changes could be explained by
systemic vasculature. a. A sudden fall in cardiac output.
b. Increased systemic venous
4. Which of the following will increase compliance.
blood flow to the greatest extent in c. Loss of blood volume.
a single isolated blood vessel that is d. Sympathetic activation.
perfused with blood in vitro at a con-
stant perfusion pressure? 8. Venous return to the right atrium is
a. Decreasing the blood temperature by a. Decreased as cardiac output increases.
10°C b. Decreased by sympathetic activation
b. Increasing perfusion pressure by 100% of veins.
c. Increasing blood viscosity by 100% c. Increased during a forced expiration
d. Increasing the vessel diameter by 50% against a closed glottis.
d. Increased during inspiration.
5. If cardiac output is 4500 mL/min, mean
arterial pressure is 94 mm Hg, and right 9. Mean circulatory filling pressure is
atrial pressure is 4 mm Hg, systemic vas- increased by
cular resistance (in peripheral resistance
units, PRU; mm Hg/mL · min−1) is a. Decreased venous compliance.
b. Increased systemic vascular resistance.
a. 0.02 c. Decreased blood volume.
b. 20 d. Increased cardiac output.
c. 50
d. 4.05 × 105 10. In a normal heart, cardiac output and
right atrial pressure are both increased by
6. A patient recently diagnosed with
hypertension is found to have a stenotic a. Decreased blood volume.
(narrowed) right renal artery. The inter- b. Decreased systemic vascular
nal diameter is reduced by 50%. Assum- resistance.
ing that renal artery resistance is 1% of c. Increased heart rate.
total renal resistance and that vascular d. Increased venous compliance.

Klabunde_Chap05.indd 120 6/11/2011 11:10:21 AM


CHAPTER 5 • VASCULAR FUNCTION 121

1. The correct answer is "c" because these 4. The correct answer is "d" because a
vessels are the most permeable to fluid. 50% increase in diameter will increase
Choice "a" is incorrect because capil- flow by about fivefold because flow is
laries, not arterioles, have the highest proportional to radius (or diameter) to
individual resistance because of their the fourth power in a single vessel seg-
small diameter. Choice "b" is incorrect ment (assuming that the pressure gradi-
because the large number of parallel ent does not change appreciably). Choice
capillaries reduces their overall resis- "a" is incorrect because decreasing tem-
tance as a group of vessels. Choice "d" is perature increases blood viscosity, which
incorrect because the small arteries and decreases flow. Choice "b" is incorrect
arterioles are the primary sites for pres- because increasing perfusion pressure by
sure and flow regulation. 100% increases flow by twofold. Choice
2. A pathological decrease in aortic com- "c" is incorrect because flow is inversely
pliance, which would increase pulse related to blood viscosity.
pressure, is very unlikely in this young, 5. The correct answer is "a" because sys-
healthy adult. Therefore, the correct temic vascular resistance equals arte-
answer most likely involves increased rial minus venous pressure (mm Hg)
stroke volume, and for this reason choice divided by cardiac output (mUmin).
"b" is correct. Stroke volume would be 6. The correct answer is "b" because the
elevated because of the low resting heart renal artery is the distributing artery to
rate and perhaps more forceful ventricu- the kidney, and therefore is in series
lar contractions because of exercise con- with the kidney. Although decreasing the
ditioning. Choice "a" is incorrect because renal artery diameter by 50% increases its
aortic compliance increases at lower aor- resistance 16-fold, the total renal resis-
tic pressures and volumes; therefore, this tance increases only about 15% because
would decrease pulse pressure. Choice the renal artery resistance is about
"c" is incorrect because increased aortic 1% of total renal resistance. Therefore,
compliance decreases pulse pressure. flow will decrease (assuming no change
Choice "d" is incorrect because reduced in perfusion pressure) about 13%
systemic vascular resistance has no effect because F = AP/R and R is increased by a
on pulse pressure except if mean arterial factor of 1.15. With increased perfusion
pressure is reduced, which would then pressure because of the hypertension, the
decrease pulse pressure. reduction in flow would be <13%.
3. The correct answer is "c." Choices "a" 7. The correct answer is "a" because a fall
and "b" are incorrect because reduc- in cardiac output causes arterial pres-
ing heart rate by 10% without changing sure to fall and blood to back up into the
stroke volume decreases cardiac output venous circulation, which increases cen-
by 10%. Because mean arterial pres- tral venous pressure. Choices "b" and "c"
sure is also reduced by 10% and mean are incorrect because increased systemic
arterial pressure equals cardiac output venous compliance and decreased blood
times systemic vascular resistance (when volume reduce central venous pressure,
central venous pressure is zero), sys- cardiac output, and arterial pressure.
temic vascular resistance is not changed. Choice "d" is incorrect because general-
Choice "d" is incorrect because systemic ized sympathetic activation would raise
vascular resistance changes if the sys- arterial pressure by increasing systemic
temic vasculature dilates. vascular resistance and cardiac output.

Klabunde_Chap05.indd 121 6/11/2011 11:10:21 AM


122 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

8. Choice "d" is correct because inspiration slope of the systemic function curve,
reduces intrapleural pressure, which but not its x-intercept. Choice "c" is
expands the right atrium, lowers its incorrect because a decrease in blood
pressure, and thereby enhances venous volume causes a parallel shift in the sys-
return. Choice "a" is incorrect because temic function curve to the left, which
an increase in cardiac output must decreases mean circulatory filling pres-
increase venous return because the cir- sure. Choice "d" is incorrect because
culatory system is closed. Choice "b" is mean circulatory filling pressure, by
incorrect because decreased sympathetic definition, is the intravascular pressure
activation of the veins causes them to when cardiac output is zero, and there-
relax, which increases their compli- fore it is independent of cardiac output.
ance. This reduces preload on the heart, 10. The correct answer is "b" because a
which leads to a reduction in cardiac decrease in systemic vascular resistance
output and venous return. Choice "c" increases the slope of the systemic func-
is incorrect because a Valsalva maneu- tion curve, which increases cardiac
ver increases intrapleural pressure, output and right atrial pressure. Choices
compresses the vena cava, and reduces "a" and "d" are incorrect because
venous return. decreased blood volume and increased
9. Choice "a" is correct because decreased venous compliance decrease right atrial
venous compliance shifts the systemic pressure and cardiac output by causing
function curve to the right, which a leftward parallel shift in the systemic
increases the mean circulatory filling function curve. Choice "c" is incorrect
pressure (value of the x-intercept). because increased heart rate increases
Choice "b" is incorrect because changes cardiac output a small amount and
in systemic vascular resistance alter the decreases right atrial pressure.

PROBLEM 5-1 Substituting the relative resistances given in


Under constant flow conditions, LlP oc LlR this problem, we obtain
(from Equation 5-l). Furthermore, Roc 1/r4
(from Equation 5-6). Therefore, ilP oc 1/r4 • 1
RT =1+--=3
Using this relationship, we find that decreas- 1 1
-+-
ing diameter (or radius, which is proportion- 4 4
al to diameter) by 50% (to 1/2 its original
PROBLEM 5-3
radius) increases LlP by a factor of 16 (recip-
From Equation 5-10, we know that
rocal of l/2 to the fourth power). Therefore,
the new pressure gradient along the length
of vessel will be 32 mm Hg (2 mm Hg x 16). SVR = (MAP - CVP)
PROBLEM 5-2 co
In this problem, the two smaller daughter
arterioles (~) are parallel with each other Because CVP is zero, this equation simplifies
and in series with the parent arteriole(~). to
Therefore, the total resistance (~) can be
found by the following equation: SVR= MAP
co
1
RT =Rp + 1 1
-+-
Ro Ro

Klabunde_Chap05.indd 122 6/11/2011 11:10:21 AM


CHAPTER 5 • VASCULAR FUNCTION 123

In this problem, CO is increased by 30% and remainder of the resistance elements (RX),
MAP is decreased by 10%: so that RT = RL + RX. Normally, RL = 0.01(RT)
and RX = 0.99(RT) because RL is 1% of RT, and
0.9 MAP therefore RT = 0.01(RT) + 0.99(RT) = 1(RT).
SVR = = 0.69
1.3 CO Decreasing the vessel diameter by 50%
Therefore, SVR is decreased by 31% (0.69 increases RL by a factor of 16 because R ∝
SVR is the equivalent of a 31% decrease), and 1/r4. Therefore, the resistance of the stenotic
the drug is a vasodilator. Note: In solving this vessel will be 16 times its normal resistance,
problem, MAP and CO cannot be multiplied so that RL = 16(0.01)RT, or RL = 0.16(RT). We
by their percentage change. can now say that RT = 0.16(RT) + 0.99(RT).
Therefore, RT = 1.15(RT), which means that
CASE 5-1 total coronary resistance increases by only
The total coronary resistance (RT) equals 15% (1.15 - 1.00) × 100] when the resistance
the sum of the series resistance elements. of the left main coronary artery increases
Therefore, the left main coronary artery 1500% (16−fold increase).
resistance (RL) would be in series with the

SUGGESTED RESOURCES Folkow B, Neil E. Circulation. New York: Oxford


Belloni FL. Teaching the principles of hemodynamics. University Press, 1971.
Am J Physiol 1999;277 (Adv Physiol Educ Guyton AC, Jones CE, Coleman TG. Circulatory
1999;22:S187–S202. Physiology: Cardiac Output and its Regulation.
Berne RM, Levy MN. Cardiovascular Physiology. 2nd Ed. Philadelphia: W.B. Saunders, 1973.
8th Ed. Philadelphia: Mosby, 2001. Rhoades RA, Bell DR. Medical Physiology: Principles for
Burton, AC. Physiology and Biophysics of the Clinical Medicine. 3rd Ed. Philadelphia: Lippincott
Circulation. 2nd Ed. Chicago: Year Book Medical Williams & Wilkins, 2009.
Publishers, 1972.

Klabunde_Chap05.indd 123 6/11/2011 11:10:24 AM


NEUROHUMORAL 0

6
:I:
>
CONTROL OF THE HEART 'tl
-1
m
;o
AND CIRCULATION
Understanding the concepts presented in this chapter will enable the student to:
1. Describe the origin and distribution of sympathetic and parasympathetic
nerves to the heart and circulation.
2. Know the location and function of alpha- and beta-adrenoceptors and
muscarinic receptors in the heart and blood vessels.
3. Describe the location and afferent connections from the carotid sinus, aortic
arch, and cardiopulmonary baroreceptors to the medulla oblongata.
4. Describe how carotid sinus baroreceptors respond to changes in arterial pres-
sure (mean pressure and pulse pressure), and explain how changes in barore-
ceptor activity affect sympathetic and parasympathetic outflow to the heart
and circulation.
5. Describe (a) the location of peripheral and central chemoreceptors; (b) the way
they respond to hypoxemia, hypercapnia, and acidosis; and (c) the effects of
their stimulation on autonomic control of the heart and circulation.
6. List the factors that stimulate the release of catecholamines, renin, aldosterone,
atrial natriuretic peptide, and vasopressin.
7. Describe how sympathetic nerves, circulating catecholamines, angiotensin II,
aldosterone, atrial natriuretic peptide, and vasopressin interact to regulate
arterial blood pressure.

INTRODUCTION chemoreceptors and osmoreceptors) also interact


with regions within the brain that control neuro-
Autonomic nerves and circulating hormones humoral status. The sensors work together with
serve as important mechanisms for regulating the neurohumoral mechanisms to ensure that
cardiac and vascular function. These mechanisms arterial blood pressure is adequate for perfusing
are controlled by sensors that monitor blood organs. Although the following sections describe
pressure (baroreceptors), blood volume (volume several individual neurohumoral mechanisms,
receptors), blood chemistry (chemoreceptors), note that these mechanisms interact together to
and plasma osmolarity (osmoreceptors). Periph- ensure cardiovascular homeostasis.
eral sensors such as baroreceptors are found in
arteries, veins, and cardiac chambers. They have AUTONOMIC NEURAL CONTROL
afferent nerve fibers that travel to the central
nervous system, where their activity is monitored
Autonomic Innervation of the
and compared against a "set point" for arterial
Heart and Vasculature
pressure. Deviations from the set point result in
selective activation or deactivation of neurohu- Autonomic regulation of cardiovascular func-
moral efferent control systems. Sensors located tion is controlled by the central nervous sys-
within the central nervous system (e.g., central tem. The medulla oblongata located within
124

Klabunde_Chap06.indd 124 6/11/2011 11:15:19 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 125

the brainstem, the hypothalamus, and the the NTS project to medullary regions containing
cortical regions work together to regulate cell bodies of parasympathetic (vagal) nerves.
autonomic function (Fig. 6.1). Regions Therefore, increased activity of the NTS enhances
within the medulla contain the cell bodies for vagal efferent nerve activity and inhibits sym-
the parasympathetic (vagal) and sympathetic pathetic nerve efferent activity. The NTS also
efferent nerves that control the heart and vas- sends fibers to the hypothalamus and receives
culature. The hypothalamus (in particular, input from the hypothalamus. Sensors within
the paraventricular nucleus and dorsal medial the hypothalamus that monitor blood tempera-
nucleus) plays an integrative role by modulat- ture (thermoreceptors) send fibers to medullary
ing medullary neuronal activity, for example, regions to modulate sympathetic outflow to the
during exercise or when the body needs to cutaneous circulation.
adjust blood flow to the skin to regulate body
temperature. Higher centers, including the
PARASYMPATHETIC INNERVATION
cortex and limbic and midbrain structures,
connect with the hypothalamus and medulla. The parasympathetic vagal fibers innervating
The higher centers can alter cardiovascular the heart originate from cell bodies located
function during times of emotional stress within the medulla of the brainstem (see
(e.g., caused by fear and anxiety). Figs. 6.1 and 6.2). These cell bodies are found
The central nervous system receives sensory in collections of neurons called the dorsal
(afferent) input from peripheral sensors and from vagal nucleus (DVN) and nucleus ambiguus
sensors within the brain. Afferent fibers from (NA). Increased activity of these nuclei
peripheral baroreceptors and chemoreceptors, reduces sinoatrial (SA) nodal firing (negative
as well as respiratory stretch receptors, enter the chronotropy) and slows AV nodal conduc-
medulla at the nucleus tractus solitarius (NTS) tion (negative dromotropy). It is important
(see Fig. 6.2). Inhibitory interneurons from to note that under normal resting conditions,
cells within the NTS project to other medullary these neurons are tonically active, thereby
regions containing cell bodies of sympathetic producing what is termed “vagal tone” on
nerves. In addition, excitatory interneurons from the heart, resulting in resting heart rates sig-
nificantly below the intrinsic firing rate of the
SA nodal pacemaker. Afferent nerves, particu-
Cerebral larly from peripheral baroreceptors that enter
Cortex
the medulla through the NTS, modulate the
activity of these vagal neurons. Excitatory
interneurons from the NTS, which normally
are excited by tonic baroreceptor activity,
stimulate vagal activity.
Efferent vagal fibers (also referred to as
preganglionic fibers) exit the medulla as the
Hypothalamus tenth cranial nerve (see Fig. 6.3) and travel
to the heart within the left and right vagus
Pons nerves. Branches from these nerves inner-
Medulla vate specific regions within the heart such
Spinal as the SA and atrioventricular (AV) nodes,
Cord conduction pathways, atrial myocytes, and
■ FIGURE 6.1 Regions of the central nervous the coronary vasculature. The preganglionic
system involved in cardiovascular regulation. The efferent fibers synapse within or near the tar-
primary site of cardiovascular regulation resides get tissue and form small ganglia, from which
in the medulla; the hypothalamus serves as an
short postganglionic fibers innervate specific
integrative region for coordinating cardiovascular
responses. Higher centers such as the cortex influ- tissue sites. The right vagus is usually the
ence cardiovascular function. primary vagal branch that innervates the SA

Klabunde_Chap06.indd 125 6/11/2011 11:15:22 AM


126 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Higher Centers

Hypothalamus


Sympathetic Vagal
– NTS
(RVLM) (DVN, NA) +
Medulla

+
Receptor
– Afferents
+

Blood Vessels Heart


■ FIGURE 6.2 Schematic representation of autonomic sympathetic and vagal interconnections within
the central nervous system. Receptor afferent nerve fibers (e.g., from baroreceptors) enter the medulla
at the nucleus tractus solitarius (NTS), which projects inhibitory interneurons to the sympathetic neurons
in the rostral ventrolateral medulla (RVLM) and excitatory fibers to the vagal neurons in the dorsal vagal
nucleus (DVN) and nucleus ambiguus (NA). The medulla receives input from the hypothalamus and higher
brain centers. Sympathetic activation (+) of blood vessels and the heart causes smooth muscle contrac-
tion (vasoconstriction), increased heart rate (positive chronotropy), increased conduction velocity within
the heart (positive dromotropy), and increased contractility (positive inotropy). Vagal activation of the
heart decreases (−) chronotropy, dromotropy, and inotropy.

node, whereas the left vagus primarily inner- Direct vasodilation by parasympathetic
vates the AV node. This can be demonstrated activation in some tissues (e.g., genitalia erec-
experimentally by electrically stimulating the tile tissue) is achieved through the release
right vagus nerve, which causes bradycardia of acetylcholine (Ach), which binds to mus-
(or SA nodal arrest) with little change in AV carinic receptors on the vascular endothelium
nodal conduction, as evidenced by a rela- to cause vasodilation through the subsequent
tively small increase in the P-R interval of the formation of nitric oxide (see Chapter 3).
electrocardiogram. Left vagal stimulation, in Parasympathetic stimulation causes indirect
contrast, usually results in a pronounced AV vasodilation in some organs (e.g., gastrointes-
nodal block (see Chapter 2), with relatively tinal circulation) by stimulating nonvascular
little decrease in heart rate. However, these tissue to produce vasodilator substances such
responses to vagal stimulation can be mark- as bradykinin, which then binds to vascular
edly different between individuals because of receptors to cause vasodilation. Note that any
crossover of the left and right vagal efferent existing parasympathetic nerves primarily serve
nerves. to regulate blood flow within specific organs and
Some efferent parasympathetic fibers inner- do not play a significant role in the regulation of
vate blood vessels in specific organs in which systemic vascular resistance and arterial blood
they directly or indirectly cause vasodilation. pressure.

Klabunde_Chap06.indd 126 6/11/2011 11:15:22 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 127

SYMPATHETIC INNERVATION of sympathetic denervation on the heart rate


are relatively small because the heart is under
The sympathetic adrenergic control of the
a high level of vagal tone and relatively weak
heart and vasculature originates from neurons
sympathetic tone. In contrast, sympathetic
found within the medulla, the most important
vascular tone is relatively high in most organ
of which are located in the rostral ventrolat-
circulations; therefore, sudden removal of sym-
eral medulla (RVLM). Increased activity of
pathetic tone produces significant vasodilation
these neurons produces cardiac stimulation
and hypotension.
and systemic vasoconstriction. Sympathetic
Axons from sympathetic neurons leave the
neurons within the RVLM have spontane-
medulla, travel down the spinal cord and syn-
ous action potential activity, which results in
apse within the intermediolateral cell column
tonic stimulation of the heart and vasculature.
of the spinal cord, and then exit at specific
Therefore, acute sympathetic denervation of
thoracolumbar levels (T1-L2) (Fig. 6.3).
the heart and systemic blood vessels usually
These preganglionic fibers (short compared
results in cardiac slowing and systemic vaso-
to preganglionic parasympathetic fibers) then
dilation. At low resting heart rates, the effects

Cranial Nerve X
(vagus) A

Cervical
B
T1
Heart C

Thoracic
D

T12
Lumbar
Blood Prevertebral Paravertebral
Vessels Ganglia Ganglia

■ FIGURE 6.3 Organization of sympathetic and vagal innervation of the heart and circulation. The tenth
cranial nerve (vagus; parasympathetic) arises from the brainstem. Preganglionic fibers (solid red line, A)
travel to the heart, where they synapse with cell bodies of short postganglionic fibers that innervate the
heart. Preganglionic sympathetic nerves (solid black lines) arise from thoracic (T1−T12) and lumbar seg-
ments of the spinal cord. Some of these fibers (B) enter the paravertebral ganglia (sympathetic chain) on
both sides of the spinal cord, and travel within the ganglia to synapse above (B) or below their entry level,
or at their level of entry (C). Postganglionic fibers (dotted black lines) from the cervical ganglia primarily
innervate the heart, whereas those from thoracic ganglia travel to blood vessels and to the heart. Pregan-
glionic fibers from lower thoracic and upper lumbar segments generally synapse in prevertebral ganglia
(D), from which postganglionic fibers travel to blood vessels.

Klabunde_Chap06.indd 127 6/11/2011 11:15:22 AM


128 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

synapse within sympathetic paravertebral sympathetic vasoconstrictor effects on the


ganglia (cervical, stellate, and thoracolumbar coronary vessels.
sympathetic chain) located on either side of Sympathetic activation of resistance vessels
the spinal cord, or they synapse within pre- significantly contributes to the vascular tone
vertebral ganglia located within the abdomen in many organs. This can be demonstrated
(celiac, superior mesenteric, and inferior mes- by abruptly removing sympathetic influ-
enteric ganglia) (Fig. 6.3). Some fibers also ences (e.g., by blocking α-adrenoceptors
travel to the adrenal glands where they syn- with drugs). When this is done, blood flow
apse. Postganglionic sympathetic fibers (long increases, the amount of which depends
compared to postganglionic parasympathetic upon the degree of sympathetic tone and
fibers) travel to target organs where they the strength of local autoregulatory mecha-
innervate arteries and veins; capillaries are not nisms that will attempt to maintain constant
innervated. Small branches of these efferent blood flow (see Chapter 7). For example, if
nerves are found in the adventitia (outer) layer α-adrenoceptors in the forearm circulation
of blood vessels. Varicosities, which are small are blocked pharmacologically, blood flow
enlargements along the sympathetic nerve fib- increases two- or threefold. Over time, how-
ers, provide the site of neurotransmitter release. ever, intrinsic autoregulatory mechanisms
Postganglionic sympathetic fibers traveling restore normal tone and blood flow.
to the heart innervate the SA and AV nodes, the As described further in Chapter 7, the
conduction system, and cardiac myocytes, as vascular response to sympathetic activation
well as the coronary vasculature. Sympathetic differs among organs. Nevertheless, general-
activation increases chronotropy, dromotropy, ized sympathetic activation of the circulation
and inotropy (see Table 6-1). In blood vessels, increases arterial pressure and reduces organ
sympathetic activation directly constricts both perfusion throughout the body except in the
resistance and capacitance vessels, thereby heart and brain.
increasing systemic vascular resistance (and
arterial blood pressure) and decreasing venous
RECIPROCAL SYMPATHETIC
capacitance (which increases venous pressure)
AND VAGAL ACTIVITY
(see Table 6-1). As described in Chapter 7,
sympathetic activation of the heart leads to Normally, there is reciprocal activation of the
paradoxical coronary vasodilation because medullary sympathetic RVLM and the nuclei
increased cardiac activity produces metabolic controlling vagal outflow. An example of this
coronary vasodilation that overrides the direct reciprocity occurs when a person stands up

TABLE 6-1 EFFECTS OF SYMPATHETIC AND PARASYMPATHETIC STIMULATION ON


CARDIAC AND VASCULAR FUNCTION
SYMPATHETIC PARASYMPATHETIC

Heart
Chronotropy (rate) +++ −−−
Inotropy (contractility) +++ −1
Dromotropy (conduction ++ −−−
velocity)
Vessels (Vasoconstriction)
Resistance (arteries, arterioles) +++ −2
Capacitance (veins, venules) +++ 0
Relative magnitude of responses (+, increase; −, decrease; 0, no response) indicated by number of + or − signs.
1
More pronounced in atria than ventricles.
2
Vasodilator effects only in specific organs such as genitalia.

Klabunde_Chap06.indd 128 6/11/2011 11:15:23 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 129

and arterial blood pressure falls. Baroreceptor


Sympathetic Vagus
reflexes (discussed later in this chapter) cause M2

the RVLM to increase sympathetic outflow to NE ACh
stimulate the heart (increase heart rate and – 2

inotropy) and to constrict the systemic vascu- NE ACh


lature. These cardiac and vascular responses
help to restore normal arterial pressure. As
sympathetic neurons in the RVLM are being 2 1 1 M2
activated, parasympathetic vagal activity orig-
inating from the DVN and nucleus ambiguus
is decreased. This is important because with- Chronotropy
Dromotropy
out removal of vagal influences on the heart, + Inotropy –
the ability of enhanced sympathetic activity
to increase heart rate is impaired. The reason Heart
for this is that vagal influences are dominant over
■ FIGURE 6.4 Adrenergic and muscarinic recep-
sympathetic influences in the heart.
tors in the heart. Norepinephrine (NE) released
Regions within the hypothalamus can inte- from sympathetic nerve terminals binds to
grate and coordinate cardiovascular responses postjunctional adrenoceptors (order of functional
by providing input to medullary centers. Stud- importance: β1 > β2 > α1) to increase (+) inotropy,
chronotropy, and dromotropy. Prejunctional
ies have shown that electrical stimulation of α2−adrenoceptors serve as a feedback mechanism
dorsomedial hypothalamus produces auto- to inhibit NE release. Parasympathetic (vagal)
nomic responses that mimic those that occur nerves release acetylcholine (ACh), which binds to
during exercise, or the flight-or-fight response. postjunctional M2 receptors to decrease (−) inot-
ropy, chronotropy, and dromotropy. ACh also binds
These coordinated responses include sympa- to prejunctional muscarinic receptors (M2) on
thetic-mediated tachycardia, increased inotropy, sympathetic nerve terminals to inhibit NE release.
catecholamine release, and systemic vasocon-
striction. These are brought about by hypotha-
lamic activation of sympathetic neurons within however, they are normally less important than
the RVLM and inhibition of vagal nuclei. β1-adrenoceptors. Beta-adrenoceptors are cou-
Input from higher cortical regions can pled to the Gs-protein/cAMP signal transduc-
alter autonomic function as well. For exam- tion pathway as described in Chapter 3. There
ple, sudden fear or emotion can sometimes are also postjunctional α1-adrenoceptors
cause vagal activation leading to bradycardia, located in cardiac tissue that bind to norepi-
withdrawal of sympathetic vascular tone, and nephrine, which activates the Gq-protein/IP3
fainting (vasovagal syncope). Fear and anxi- pathway to stimulate the heart (see Chapter 3).
ety can lead to sympathetic activation that Released norepinephrine can also bind to pre-
causes tachycardia, increased inotropy, and junctional α2-adrenoceptors located on the
hypertension. Chronic sympathetic activa- sympathetic nerve terminal. These receptors
tion induced by long-term emotional stress inhibit norepinephrine release through a nega-
can result in sustained hypertension, cardiac tive feedback mechanism.
hypertrophy, and arrhythmias. Activation of postganglionic vagal fibers
causes the release of the neurotransmitter
ACh. In the heart, this neurotransmitter binds
CARDIAC AND VASCULAR AUTONOMIC
to muscarinic receptors (M2) principally
RECEPTORS
in nodal tissue, and in atrial myocardium
Activation of sympathetic efferent nerves to the (Fig. 6.4). These receptors are coupled to the
heart releases the neurotransmitter norepineph- Gi-protein/cAMP signal transduction pathway
rine that binds primarily to β1-adrenoceptors (see Chapter 3), which decreases chronotropy,
located in nodal tissue, conducting tissues, dromotropy, and inotropy (more so in the atria
and myocardium (see Fig. 6.4). There are also than in the ventricles). Released ACh can also
postjunctional β2-adrenoceptors in the heart; bind to prejunctional M2 muscarinic receptors

Klabunde_Chap06.indd 129 6/11/2011 11:15:23 AM


130 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

found on nearby sympathetic adrenergic vasoconstriction. To observe this β2-adrenoceptor-


nerve terminals, which inhibits their release induced vasodilation experimentally, one can
of norepinephrine. stimulate vascular sympathetic nerves in the
In blood vessels, norepinephrine released presence of complete α-adrenoceptor blockade.
by sympathetic adrenergic nerves preferen- Normally, this small β2-receptor-mediated vaso-
tially binds to postjunctional α1-adrenoceptors dilator effect of norepinephrine is completely
to cause smooth muscle contraction and vaso- overwhelmed by simultaneous α-adrenoceptor
constriction (see Fig. 6.5). Similar responses activation, leading to vasoconstriction.
occur when norepinephrine binds to postjunc- Although there is relatively little or no
tional α2-adrenoreceptors located primarily parasympathetic innervation of most blood
on small arteries and arterioles, although vessels in the body, M2 muscarinic receptors
postjunctional α1-adrenoceptors are generally on coronary arteries can respond to vagal acti-
the more important α-adrenoceptor subtype vation in the heart by dilating, and similar
in most vessels. These α-adrenoceptors are receptors in genital erectile tissue respond by
coupled to the Gq-protein/IP3 signal trans- dilating to ACh release by parasympathetic
duction pathway as described in Chapter 3. nerves (Fig. 6.5).
In addition, norepinephrine can bind to
prejunctional α2-adrenoreceptors, which acts
Baroreceptor Feedback
as a negative feedback mechanism for modu-
Regulation of Arterial Pressure
lating norepinephrine release.
Blood vessels possess postjunctional As described above, sympathetic nerves play
β2-adrenoceptors in addition to α-adrenoceptors. an important role in regulating systemic vas-
Activation of postjunctional β2-adrenoceptors by cular resistance and cardiac function, and
norepinephrine (and, more importantly, by cir- therefore arterial blood pressure. But, how
culating epinephrine) causes vasodilation in the does the body control the systemic vascular
absence of opposing α-adrenoceptor-mediated resistance and cardiac output to establish and
maintain an arterial blood pressure to ensure
Sympathetic Para- adequate organ perfusion?
sympathetic
Arterial blood pressure is regulated through
NE ACh negative feedback systems incorporating pres-
– 2
sure sensors (i.e., baroreceptors) found in
NE ACh strategic locations within the cardiovascular
system. Arterial baroreceptors are found in
the carotid sinus (at the bifurcation of external
2 1 M2
2 and internal carotids) and in the aortic arch
(Fig. 6.6). The sinus nerve (nerve of Hering), a
branch of the glossopharyngeal nerve (cranial
Vascular nerve IX), innervates the carotid sinus. Affer-
+ Tone –
ent fibers from the carotid sinus travel in the
glossopharyngeal nerve up to the brainstem,
Blood Vessel where they synapse at the NTS. As already
■ FIGURE 6.5 Adrenergic and muscarinic described, the NTS modulates the activity of
receptors in blood vessels. Norepinephrine (NE) sympathetic neurons within the RVLM and
released from sympathetic nerve terminals medullary vagal nuclei. The aortic arch baro-
binds to postjunctional adrenoceptors (order of
receptors are innervated by the aortic nerve,
functional importance: α1 > α2 > β2). NE binding to
postjunctional α-adrenoceptors causes increased which then combines with the vagus nerve
(+) vascular tone (vasoconstriction), whereas (cranial nerve X) before traveling to the NTS.
binding to β2-adrenoceptors causes decreased The arterial baroreceptors respond to the
(−) vascular tone (vasodilation). In a few specific
organs (e.g., genitalia), ACh released by parasym-
stretching of the vessel walls produced by
pathetic nerves binds to vascular M2 receptors to increases in arterial blood pressure (Fig. 6.7).
produce endothelial-dependent vasodilation. Increased arterial pressure increases the firing

Klabunde_Chap06.indd 130 6/11/2011 11:15:23 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 131

Glossopharyngeal Nerve sinus respond to pressures ranging from about


(Cranial Nerve IX)
60 to 180 mm Hg. Therefore, if arterial blood
pressure decreases from normal, it lowers
Sinus Nerve
the firing rate of the carotid sinus barorecep-
R. Internal L. Internal tors; conversely, increased arterial pressure
Carotid Carotid
increases receptor firing.
R. External L. External
Carotid Carotid Baroreceptors are sensitive to the rate of pres-
sure change and to a steady or mean pressure.
At a given mean arterial pressure, decreasing
Carotid
Sinus Vagus Nerve the arterial pulse pressure decreases firing rate.
Receptors (Cranial Nerve X) This is important during conditions such as
hemorrhagic shock in which pulse pressure (as
well as mean pressure) decreases because of the
decline in stroke volume caused by decreased
Ascending ventricular preload and increased heart rate.
Aorta Therefore, reduced pulse pressure reinforces
Aortic Arch the baroreceptor reflex when mean arterial
Receptors pressure falls. The curve representing the fre-
quency of baroreceptor firing in Figure 6.7 is
■ FIGURE 6.6 Location and innervation of arterial
baroreceptors. Carotid sinus receptors are located
the integrated receptor firing at a given pulse
on the internal carotid artery just above the junc- pressure. At reduced pulse pressures, the curve
tion with the external carotid artery. These recep- shifts to the right, thereby decreasing the firing
tors are innervated by the sinus nerve of Hering, at any given mean arterial pressure.
which joins the glossopharyngeal nerve (cranial
nerve IX) before traveling up to the medulla. Affer-
Maximal carotid sinus sensitivity (the
ent nerves from the aortic arch receptors join the point of greatest slope of the response curve
vagus nerve (cranial nerve X), which then travel to in Fig. 6.7) occurs near the “set point” of nor-
the medulla. R, right; L, left. mal mean arterial pressures (∼95 mm Hg in
adults). Therefore, small deviations from this
rate of individual receptors and nerves. Each set point elicit large changes in barorecep-
individual receptor has its own threshold and tor firing frequency. This set point, and the
sensitivity to changes in pressure; therefore, entire receptor response curve, is not fixed.
additional receptors are recruited as pressure Chronic shifts in this curve can occur during
increases. Overall, the receptors of the carotid hypertension, heart failure, and other disease

100
Carotid Sinus Arterial Pressure Pulse
Maximal
Integrated Sensitivity
Mean
Receptor 50 Normal
Firing Rate Pulse
(% max) Pressure Reduce Receptor
Pulse Firing
Pressure
0
0 100 200
Mean Arterial Pressure
(mmHg)

■ FIGURE 6.7 Effects of arterial pressure on integrated carotid sinus firing rate. Left panel: The thresh-
old for receptor activation occurs at mean arterial pressures of about 60 mm Hg; maximal firing occurs
at about 180 mm Hg. Maximal receptor sensitivity occurs at normal mean arterial pressures. The receptor
firing−response curve shifts to the right with decreased pulse pressures; therefore, a decrease in pulse
pressure at a given mean pressure decreases firing. Right panel: Single receptor firing in response to pulsa-
tile pressure. Receptors fire more rapidly when arterial pressure is rapidly increasing during cardiac systole.

Klabunde_Chap06.indd 131 6/11/2011 11:15:23 AM


132 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

states. In hypertension, for example, the curve blood pooling below the heart, particularly
shifts to the right, thereby reducing the firing in the legs (see Chapter 5). This decreases
rate at any given mean arterial pressure. This venous return, central venous pressure,
resetting of the baroreceptor response can and ventricular preload, leading to a fall
occur at the level of the receptors themselves in cardiac output and arterial blood pres-
as well as in the brainstem. In arteriosclerosis, sure. Decreased stretching of baroreceptors
the carotid arteries at the region of the carotid results in decreased baroreceptor firing and
sinus become less compliant, and therefore decreased NTS activity. Nuclei within the
they stretch less in response to changes in RVLM respond by increasing sympathetic
arterial blood pressure—this decreases their outflow, which increases systemic vascular
sensitivity. During exercise, medullary and resistance (vasoconstriction) and cardiac
hypothalamic control centers can modulate output (increased heart rate and inotropy).
autonomic efferent responses at a given level Decreased vagal outflow from the medulla
of baroreceptor firing, thereby resetting arte- contributes to the elevation in heart rate.
rial pressure to a higher level. Note that baroreceptor firing normally exerts
Receptors located within the aortic arch a tonic inhibitory influence on sympathetic outflow
function similarly to carotid sinus receptors; from the medulla. Therefore, hypotension and
however, they have a higher threshold pres- decreased baroreceptor firing disinhibits sym-
sure for firing and are less sensitive than the pathetic outflow (i.e., it increases sympathetic
carotid sinus receptors. Therefore, the aortic activity) from the medulla. The combined
arch baroreceptors serve as secondary baro- effects on systemic vascular resistance and
receptors, with the carotid sinus receptors cardiac output increases arterial blood pres-
normally being the dominant arterial barore- sure back toward its set point.
ceptor. The carotid sinus reflex can be activated
To understand how the baroreceptor by rubbing the neck over the carotid sinus
reflex operates, consider the events that (i.e., carotid sinus massage). This mechani-
occur in response to a decrease in arterial cal stimulation of the receptors increases their
pressure (mean, pulse, or both) when a per- firing, which leads to decreased sympathetic
son suddenly stands up (Fig. 6.8). When and increased parasympathetic outflow from
upright posture is suddenly assumed from the medulla. This action is sometimes used to
the supine position, gravity causes venous abort certain types of arrhythmias by activat-
ing the vagus efferents to the heart.
Another example of the operation of the
Decreased Decreased baroreceptor reflex is when a Valsalva maneu-
Arterial Receptor ver is performed, which is sometimes used to
Pressure Firing
assess autonomic reflex control of cardiovas-
cular function in humans. It is performed by
+ + having the subject conduct a maximal, forced
CO SVR expiration against a closed glottis and main-
taining this for at least 10 seconds. Contrac-
+ + Sympathetic
CNS
tion of the thoracic cage compresses the lungs
Parasympathetic and causes a large increase in intrapleural
pressure (the pressure measured between
■ FIGURE 6.8 Baroreceptor feedback loop.
the lining of the thorax and the lungs—see
A sudden decrease in arterial pressure, as occurs
when a person suddenly stands up from a supine Fig. 5.16), which compresses the vessels
position, decreases baroreceptor firing, activating within the thoracic. Aortic compression
sympathetic nerves and inhibiting parasympathetic results in a transient rise in aortic pressure
(vagal) nerves. This change in autonomic balance
(Phase I of Fig. 6.9). This results in a reflex
increases (+) cardiac output (CO) and systemic vas-
cular resistance (SVR), which helps to restore nor- bradycardia caused by baroreceptor activa-
mal arterial pressure. CNS, central nervous system. tion. Because the thoracic vena cava also

Klabunde_Chap06.indd 132 6/11/2011 11:15:24 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 133

Aortic
Pressure
Phases: I II III IV

Heart
Rate

Valsalva

■ FIGURE 6.9 Baroreceptors responses during a Valsalva maneuver. During Phase I, which occurs at the
beginning of the forced expiration, aortic pressure increases (due to aortic compression) and heart rate
decreases reflexively. Aortic pressure falls during Phase II because compression of thoracic veins reduces
venous return and cardiac output; reflex tachycardia occurs. Phase III begins when normal respiration
resumes, and is characterized by a small transient fall in aortic pressure (because of removal of aortic
compression) and a small increase in heart rate. Aortic pressure increases (and heart rate reflexively
decreases) during Phase IV because resumption of normal cardiac output occurs while systemic vascular
resistance is elevated from sympathetic activation that occurred during Phase II.

becomes compressed, venous return to the caused by an increase in venous return can
heart is compromised, causing cardiac out- under some conditions increase heart rate via
put and aortic pressure to fall (Phase II). As medullary activation of sympathetic efferent
aortic pressure falls, the baroreceptor reflex activity to the SA node. This response, which
increases heart rate. A decrease in stroke vol- is called the Bainbridge reflex, increases heart
ume accounts for the fall in pulse pressure. rate when the initial heart rate is low.
After several seconds, arterial pressure (both An increase in blood volume and venous
mean and pulse pressure) is reduced, and pressure stimulates other types of cardiopul-
heart rate is elevated. When the subject begins monary receptors to decrease antidiuretic
breathing again, the sudden loss of compres- hormone (ADH, vasopressin) release by the
sion on the aorta causes a small, transient posterior pituitary. Decreased circulating
dip in arterial pressure and a further reflex ADH causes diuresis, which leads to a fall in
increase in heart rate (Phase III). When com- blood volume and venous pressure. If blood
pression of the vena cava is removed, venous volume is lost as a result of dehydration or
return suddenly increases, causing a rapid rise hemorrhage, these receptors will increase
in cardiac output several seconds later, which ADH release so that the kidneys excrete less
leads to a transient increase in arterial pres- water.
sure (Phase IV). Arterial pressure overshoots Unmyelinated vagal afferents are found
during Phase IV because the systemic vascu- throughout the atria and ventricles. Receptors
lar resistance is increased by sympathetic acti- associated with these vagal afferents respond
vation that occurred during Phase II owing to to stretch such that the firing rate of these
the baroreceptor reflex. Heart rate reflexively receptors is enhanced with increased atrial
decreases during Phase IV in response to the and ventricular pressures. The effects of these
transient elevation in arterial pressure. receptors on sympathetic and vagal outflow
In addition to arterial baroreceptors, stretch are similar to those on the arterial barore-
receptors are located at the venoatrial junctions ceptors. Depending upon the circumstances,
of the heart (cardiopulmonary receptors) and however, these receptors can either oppose
respond to atrial filling and contraction. These or reinforce arterial baroreceptor function.
tonically active receptors are innervated by In heart failure, atrial and ventricular filling
myelinated vagal afferents. Increased stretch pressures are increased, whereas arterial

Klabunde_Chap06.indd 133 6/11/2011 11:15:24 AM


134 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

pressure is decreased. Under this condition, The carotid bodies increase their firing in
increased firing by the AV receptors opposes response to a fall in arterial P0 2 (hypoxemia)
the decreased firing by arterial baroreceptors. or to an increase in arterial PC0 2 (hypercap-
During hemorrhage, cardiac chamber pres- nia) and hydrogen ion concentration (acido-
sures and arterial pressures are both reduced. sis). The threshold P0 2 for activation is about
This causes the atrioventricular receptors and 80 mm Hg (normal arterial P0 2 is about 95
the arterial baroreceptors to decrease their fir- mm Hg). Any elevation of PC0 2 above its
ing rates and therefore reinforce each other. normal value of 40 mm Hg, or a decrease in
pH below 7.4, also increases receptor firing.
PROBLEM 6·1 In addition, carotid body firing can be stimu-
How do the carotid sinus baroreceptors lated by reduced carotid body perfusion, as
respond to occlusion of both common occurs during hypotension associated with
carotid arteries? What are the cardio- circulatory shock. This response to reduced
vascular responses to bilateral carotid perfusion can occur without changes in arte-
occlusion? How would these responses rial P0 2 , PC0 2 , and pH. The mechanism may
be altered by bilateral vagotomy? involve cellular hypoxia resulting from inad-
How would these responses be altered equate oxygen delivery to the carotid bod-
by the pharmacologic blockade of ies (i.e., "stagnant hypoxia"). Another set of
p-adrenoceptors? peripheral chemoreceptors, the aortic bodies,
are located on the aortic arch, and they func-
tion similarly to the carotid bodies. Their
Chemoreceptors
afferent connections to the NTS travel with
Chemoreceptors are specialized cells located vagal afferent fibers.
on arteries (peripheral chemoreceptors) and Central chemoreceptors are found in
within the medulla (central chemoreceptors) medullary regions that control cardiovascu-
that monitor blood P0 2 (partial pressure of lar and respiratory activity. These receptors
oxygen), PC0 2 (partial pressure of carbon increase their firing in response to hypercap-
dioxide), or pH (log H+ concentration). Their nia and acidosis but not directly in response
primary function is to regulate respiratory to hypoxia. Carbon dioxide diffusing from
activity to maintain arterial blood P0 2 , PC0 2, the blood into the cerebrospinal fluid forms
and pH within a narrow physiologic range. hydrogen ion by the bicarbonate buffer sys-
Chemoreceptor activity, however, affects tem, and it is the hydrogen ion rather than the
cardiovascular function either directly by carbon dioxide that stimulates receptor firing.
influencing medullary cardiovascular cent- If a subject breathes a gas mixture contain-
ers or indirectly through altered pulmonary ing 10% instead of 21% oxygen, chemorecep-
stretch receptor activity. Impaired respiratory tor activation (primarily peripheral) increases
gas exchange, hypoxic environments, cerebral respiratory activity and stimulates sympathetic
ischemia, and circulatory shock, for example, activity to the heart and systemic vasculature,
increase chemoreceptor activity, leading to causing arterial blood pressure to increase. If,
enhanced sympathetic outflow to the heart however, respiratory rate and depth are not
and vasculature by activating neurons in the allowed to change, the sympathetic-mediated
RVLM. pressor response is accompanied by brady-
The peripheral chemoreceptors are cardia resulting from vagal activation of the
found in two locations. Small carotid bod- heart. This demonstrates that the tachycardia
ies are associated with the external carotid normally found during hypoxemia is second-
arteries near their bifurcation with the inter- ary to respiratory stimulation and activation
nal carotids. Afferent nerve fibers from the of pulmonary stretch receptors. Cardiovas-
carotid body receptors join with the sinus cular responses to hypercapnia and acido-
nerve before entering the glossopharyngeal sis likewise depend in part upon respiratory
nerve to synapse in the RTS in the medulla. responses.

Klabunde_Chap06.indd 134 6/11/2011 11:15:24 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 135

Other Autonomic Reflexes mediated by vagus nerve afferents and


Affecting the Heart efferents. This reflex is sometimes stimu-
and Circulation lated when dye or other chemical agents are
injected into coronary arteries during coro-
In addition to the baroreceptor and chemo- nary arteriography. Ventricular ischemia,
receptor reflexes already described, several particularly caused by right coronary artery
other reflexes affect cardiovascular function. occlusion, can also trigger this reflex.
1. Ischemic brain reflexes. Insufficient blood 4. Pulmonary and muscle stretch receptors.
flow to the brain (cerebral ischemia), which Lung inflation activates stretch receptors
occurs during severe hypotension (a mean located in the airways and respiratory
arterial pressure <60 mm Hg), or when muscles that inhibit medullary sympa-
there is cerebral vascular occlusion, causes thetic centers and cause arterial pressure to
intense sympathetic activation and con- fall; heart rate increases reflexively. These
striction of the systemic circulation. Mean receptors contribute to the normal cyclical
arterial pressure can rise to well over changes in heart rate and arterial pressure
200 mm Hg during severe cerebral associated with respiratory activity. Limb
ischemia. This can be thought of as a final muscles and tendons also possess recep-
effort by the body to restore perfusion to the tors that sense tension and length changes.
brain. An increase in intracranial pressure, Passive or active movement of joints can
which can occur following hemorrhagic stimulate sympathetic activity to the heart
stroke or brain trauma, can cause ischemia and circulation and help to reinforce car-
within the brainstem. This elicits a strong, diovascular responses to exercise.
sympathetic-mediated pressor response 5. Temperature reflexes. Changes in envi-
(Cushing reflex), often accompanied by ronmental temperature sensed by cold and
baroreceptor-mediated bradycardia. warm thermoreceptors in the skin can lead
2. Pain reflexes. Chest pain associated with to reflex changes in cutaneous blood flow
myocardial ischemia (insufficient coro- and sweating. Similarly, changes in core tem-
nary blood flow) or myocardial infarction perature, sensed by thermoreceptors located
can cause generalized sympathetic acti- in the hypothalamus, produce changes in
vation, leading to elevated arterial pres- sympathetic activity to the skin circulation.
sure, tachycardia, and increased sweating For example, a decrease in either skin surface
(diaphoresis). If cardiac output decreases temperature or hypothalamic blood temper-
significantly because of the ischemic ature leads to cutaneous vasoconstriction.
injury, arterial pressure may fall despite the
enhanced sympathetic activity. Deep pain HUMORAL CONTROL
produced by trauma or visceral distension
can produce hypotension (i.e., circulatory In addition to autonomic nerves, many circulat-
shock) caused by enhanced parasympa- ing factors (humoral substances) affect cardiac
thetic and decreased sympathetic activity. and vascular function. Some of these humoral
Another example of a pain reflex is the factors directly influence the heart and blood ves-
cold pressor response. If a person’s hand sels, whereas others indirectly alter cardiovascu-
or foot is submerged into ice-cold water, lar function through changes in blood volume.
arterial pressure increases as a result of Major humoral factors include circulating cat-
sympathetic activation. This test is some- echolamines, the renin-angiotensin-aldosterone
times used clinically to evaluate autonomic system, atrial natriuretic peptide, and anti-
function and vascular reactivity in patients. diuretic hormone (vasopressin). Although not
3. Bezold-Jarisch reflex. This reflex is triggered addressed in this chapter, note that many other
by stimulation of specific types of chemore- hormones and circulating substances (e.g., thy-
ceptors within the heart and coronary arteries roxin, estrogen, insulin, and growth hormone)
and produces bradycardia and hypotension have direct or indirect cardiovascular effects.

Klabunde_Chap06.indd 135 6/11/2011 11:15:25 AM


136 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Circulating Catecholamines the affinity of epinephrine for β-adrenoceptors


is much greater than for α-adrenoceptors. The
Circulating catecholamines originate from relative receptor affinities explain why, at low
two sources. The adrenal medulla releases cat- plasma concentrations, epinephrine binds pref-
echolamines (80% epinephrine, 20% norepi- erentially to β-adrenoceptors. Therefore, at low
nephrine) when preganglionic sympathetic to moderate circulating levels of epinephrine,
nerves innervating this tissue are activated. heart rate, inotropy, and dromotropy are stim-
This occurs during times of stress (e.g., exer- ulated (primarily β1-adrenoceptor mediated).
cise, heart failure, blood loss, emotional stress, Epinephrine at low concentrations binds to
excitement, or pain). Sympathetic nerves β2-adrenoceptors located on small arteries and
innervating blood vessels are another source arterioles (particularly in skeletal muscle) and
of circulating catecholamines, principally causes vasodilation.
norepinephrine. Normally, most of the nor- If a low dose of epinephrine is injected
epinephrine released by sympathetic nerves is intravenously while systemic hemodynamics
taken back up by the nerves and metabolized are monitored, heart rate (and cardiac out-
(some is taken up by extraneuronal tissues). put) will increase, systemic vascular resist-
A small amount of released norepinephrine, ance will fall, but mean arterial pressure will
however, diffuses into the blood and circu- change very little (Fig. 6.10). At high plasma
lates throughout the body. At times of high concentrations, the cardiovascular actions of
levels of sympathetic nerve activation, the epinephrine are different because epineph-
amount of norepinephrine spilling over into rine binds to α-adrenoceptors as well as to
the blood can increase dramatically. β-adrenoceptors. Increasing concentrations
Circulating epinephrine has several direct car- of epinephrine result in further cardiac stimu-
diovascular actions that depend upon the relative lation along with α-adrenoceptor-mediated
distribution of adrenergic receptors in different activation of vascular smooth muscle lead-
organs and the relative affinities of the different ing to vasoconstriction. This increases arte-
receptors for epinephrine. Epinephrine binds rial blood pressure (pressor response) owing
to β1-, β2-, α1-, and α2-adrenoceptors; however,

180 180 Norepinephrine


Epinephrine
mean
mean
140
Arterial 140
Pressure
100 100
(mmHg)
60 60
120 120
Heart
Rate 100 100
(beats/min)
80 80

60 60
Time (min) Time (min)

■ FIGURE 6.10 Effects of intravenous administration of epinephrine and norepinephrine on arterial pres-
sure and heart rate. A low dose of epinephrine (left panel) increases heart rate and arterial pulse pressure
(it increases systolic and decreases diastolic pressure) with little change in mean arterial pressure. These
changes occur because low concentrations of epinephrine preferentially bind to cardiac β1-adrenoceptors
(produces cardiac stimulation) and vascular β2-adrenoceptors (produces systemic vasodilation). Mean
pressure does not change very much because the increase in cardiac output is offset by the decrease in
systemic vascular resistance. Norepinephrine (right panel) increases mean arterial pressure and arterial
pulse pressure; heart rate transiently increases (β1-adrenoceptor stimulation) and then decreases owing to
baroreceptor reflex activation of vagal efferents to the heart. Mean arterial pressure rises because norepi-
nephrine binds to vascular α1-adrenoceptors, which increases systemic vascular resistance.

Klabunde_Chap06.indd 136 6/11/2011 11:15:25 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 137

to both an increase in cardiac output and the development and use of many different
an increase in systemic vascular resistance. types of a.- and P-adrenoceptor antagonists to
However, even at very high circulating con- modulate the effects of circulating catechola-
centrations of epinephrine, the systemic vas- mines as well as the norepinephrine released
cular resistance does not increase very much by sympathetic nerves.
above normal, or may still be reduced, because
the vasoconstrictor actions epinephrine act- PROBLEM 6-2
ing through the a.-adrenoceptors is attenu- How would the changes in arterial
ated by the epinephrine that is still bound to pressure and heart rate shown
the P2-adrenoceptors. If the P2-adrenoceptors in Figure 6.10 be different if
are blocked pharmacologically, then high P,-adrenoceptors were blocked before the
concentrations of epinephrine produce very administration of low-dose epinephrine?
large increases in systemic vascular resistance
because of the removal of the P2-adrenoceptor
vasodilator influence.
PROBLEM 6-3
Circulating norepinephrine affects the
heart and systemic vasculature by binding to How would the norepinephrine-induced
P1-, P2-, 0.1-, and 0.2-adrenoceptors; however, changes in arterial pressure and heart rate
the affinity of norepinephrine for P2- and shown in Figure 6.10 be different in the
CX.2-adrenoceptors is relatively weak. Therefore, presence of bilateral cervical vagotomy?
the predominant affects of norepinephrine are
mediated through P1- and 0.1-adrenoceptors. Renin-Angiotensin-Aldosterone
If norepinephrine is injected intravenously,
System
it causes an increase in mean arterial blood
pressure (systemic vasoconstriction) and The renin-angiotensin-aldosterone system plays
pulse pressure (owing to increased stroke an important role in regulating blood volume,
volume) and a paradoxical decrease in heart cardiac and vascular function, and arterial blood
rate after an initial transient increase in heart pressure. Although the pathways for renin and
rate (Fig. 6.10). The transient increase in angiotensin formation have been found in a
heart rate is due to norepinephrine binding number of tissues, the most important site
to P1-adrenoceptors in the SA node, whereas for renin formation and subsequent forma-
the secondary bradycardia is due to a baro- tion of circulating angiotensin is the kidney.
receptor reflex (vagal mediated), which is in Sympathetic stimulation of the kidneys (via
response to the increase in arterial pressure. P1-adrenoceptors), renal artery hypotension,
High levels of circulating catecholamines, and decreased sodium delivery to the distal
caused by a catecholamine-secreting adrenal tubules (usually caused by reduced glomeru-
tumor (pheochromocytoma), causes tachycar- lar filtration rate secondary to reduced renal
dia, arrhythmias, and severe hypertension (sys- perfusion) stimulate the release of renin into
tolic arterial pressures can exceed 200 mm Hg). the circulation. The renin is formed within,
Other actions of circulating catechola- and released from, juxtaglomerular cells
mines include (1) stimulation of renin release associated with afferent and efferent arterioles
with subsequent elevation of angiotensin II of renal glomeruli (see Chapter 7 for details),
(All) and aldosterone, and (2) cardiac and which are adjacent to the macula densa cells
vascular smooth muscle hypertrophy and of distal tubule segments that sense sodium
remodeling. These actions of catecholamines, chloride concentrations in the distal tubule.
in addition to the hemodynamic and cardiac Together, these components are referred to as
actions already described, make them a fre- the juxtaglomerular apparatus.
quent therapeutic target for the treatment of Renin is an enzyme that acts upon angio-
hypertension, heart failure, coronary artery tensinogen, a circulating substrate syn-
disease, and arrhythmias. This has led to thesized and released by the liver, which

Klabunde_Chap06.indd 137 6/11/2011 11:15:25 AM


138 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

undergoes proteolytic cleavage to form the kidneys to increase fluid retention and
the decapeptide angiotensin I. Vascular blood volume.
endothelium, particularly in the lungs, has 5. Stimulates thirst centers within the brain,
an enzyme, angiotensin-converting enzyme which can lead to an increase in blood
(ACE), that cleaves off two amino acids to volume.
form the octapeptide, angiotensin II. 6. Stimulates cardiac and vascular hypertrophy.
Angiotensin II has several important
Angiotensin II is continuously produced under
functions that are mediated by specific
basal conditions, and this production can
angiotensin II receptors (AT1) (Fig. 6.11).
change under different physiologic conditions.
Angiotensin II
For example, when a person exercises, circulat-
1. Constricts resistance vessels, thereby increas- ing levels of angiotensin II increase. An increase
ing systemic vascular resistance and arterial in renin release during exercise probably results
pressure. from sympathetic stimulation of the kidneys.
2. Enhances sympathetic adrenergic activ- Changes in body posture likewise alter circulat-
ity by facilitating norepinephrine release ing AII levels, which are increased when a per-
from sympathetic nerve endings, inhibit- son stands. As with exercise, this results from
ing norepinephrine reuptake by nerve end- sympathetic activation. Dehydration and loss
ings, and by binding to AT1 receptors in the of blood volume (hypovolemia) stimulate renin
RVLM, which increases sympathetic effer- release and angiotensin II formation in response
ent activity. to renal artery hypotension, decreased glomeru-
3. Acts upon the adrenal cortex to release lar filtration rate, and sympathetic activation.
aldosterone, which in turn acts upon the Several cardiovascular disease states are
kidneys to increase sodium and fluid reten- associated with changes in circulating angio-
tion, thereby increasing blood volume. tensin II and aldosterone. For example, second-
4. Stimulates the release of vasopressin from ary hypertension caused by renal artery stenosis
the posterior pituitary, which acts upon is associated with increased renin release and

Sympathetic
Stimulation Angiotensinogen

Hypotension Renin
Sodium AI
Delivery Kidney
ACE

Cardiac & Adrenal


Vascular AII Cortex
Hypertrophy
Sympathetic Aldosterone
Activation
ADH
Systemic Release Thirst
Vasoconstriction Renal
Arterial Cardiac Blood Sodium & Fluid
Pressure Output Volume Retention

■ FIGURE 6.11 Formation of angiotensin II and its effects on renal, vascular, and cardiac function. Renin is
released by the kidneys in response to sympathetic stimulation, hypotension, and decreased sodium deliv-
ery to distal tubules. Renin acts upon angiotensinogen to form angiotensin I (AI), which is converted to
angiotensin II (AII) by angiotensin−converting enzyme (ACE). AII has several important actions: stimulates
aldosterone release, which increases renal sodium reabsorption; directly stimulates renal sodium reab-
sorption; stimulates thirst; stimulates release of antidiuretic hormone (ADH); produces systemic vaso-
constriction; activates the sympathetic nervous system; and causes cardiac and vascular smooth muscle
hypertrophy. The overall systemic effect of increased AII is increased blood volume, venous pressure, and
arterial pressure.

Klabunde_Chap06.indd 138 6/11/2011 11:15:25 AM


Atrial distension
Sympathetic Degradation
stimulation NEP
Angiotensin II ANP
Endothelin Aldosterone
SVR
CVP Angiotensin II

CO Renin
GFR
R
Release

Arterial Blood
Pressure Volume Natriuresis
Diuresis

Klabunde_Chap06.indd 139 6/11/2011 11:15:26 AM


140 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

for the renin-angiotensin-aldosterone system. ANP for this is that NEP inhibition, by elevating
decreases aldosterone release by the adrenal ANP, reinforces the effects of ACE inhibition.
cortex; increases glomerular filtration rate; Brain-type natriuretic peptide (BNP), a
produces natriuresis and diuresis (potassium 32-amino acid peptide hormone related to
sparing); and decreases renin release, thereby ANP, is synthesized and released by the ventri-
decreasing angiotensin II. These actions cles in response to pressure and volume over-
reduce blood volume, which leads to a fall in load, particularly during heart failure. BNP
central venous pressure, cardiac output, and appears to have actions that are similar to those
arterial blood pressure. Chronic elevations of of ANP. Circulating BNP is now used clinically
ANP appear to decrease arterial blood pres- as a sensitive biomarker for heart failure.
sure primarily by decreasing systemic vascu-
lar resistance. Vasopressin (Antidiuretic
The mechanism of systemic vasodilation
Hormone)
may involve ANP receptor-mediated eleva-
tions in vascular smooth muscle cGMP (ANP Vasopressin (arginine vasopressin, AVP; anti-
activates particulate guanylyl cyclase). ANP diuretic hormone, ADH) is a nonapeptide
also attenuates sympathetic vascular tone. hormone released from the posterior pitui-
This latter mechanism may involve ANP acting tary (Fig. 6.13). AVP has two principal sites
upon sites within the central nervous system of action: the kidneys and blood vessels. The
as well as through inhibition of norepineph- most important physiologic action of AVP is
rine release by sympathetic nerve terminals. that it increases water reabsorption by the
A new class of drugs that are neutral kidneys by increasing water permeability in
endopeptidase (NEP) inhibitors is useful the collecting duct, thereby permitting the
in treating acute heart failure. By inhibiting formation of concentrated urine. This anti-
NEP, the enzyme responsible for the degra- diuretic property of AVP, which acts through
dation of ANP, these drugs elevate plasma renal V2 receptors, increases blood volume
levels of ANP. NEP inhibition is particularly and arterial blood pressure. This hormone
effective in some forms of heart failure when also constricts arterial blood vessels through
combined with an ACE inhibitor. The reason V1 vascular receptors; however, the normal

Angiotensin II
Hyperosmolarity
Decreased atrial receptor firing
Sympathetic stimulation
Pituitary
Vasopressin
Renal Fluid
Vasoconstriction
Reabsorption

Increased Increased
Arterial Pressure Blood Volume
■ FIGURE 6.13 Cardiovascular and renal effects of arginine vasopressin (AVP). AVP release from the
posterior pituitary is stimulated by angiotensin II, hyperosmolarity, decreased atrial receptor firing (usually
in response to hypovolemia), and sympathetic activation. The primary action of AVP is on the kidney to
increase water reabsorption (antidiuretic effect), which increases blood volume. AVP also has direct vaso-
constrictor actions at high concentrations. Increased arterial pressure is the overall effect of increased AVP.

Klabunde_Chap06.indd 140 6/11/2011 11:15:26 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 141

physiologic concentrations of AVP are below In summary, the importance of AVP in


its vasoactive range. Studies have shown, nev- cardiovascular regulation is primarily through
ertheless, that in severe hypovolemic shock, its effects on volume regulation, which in
when AVP release is very high, AVP contrib- turn affects ventricular preload and cardiac
utes to the compensatory increase in systemic output through the Frank-Starling relationship.
vascular resistance. This vasoconstrictor Increased AVP, by increasing blood volume,
property of AVP is sometimes utilized in the increases cardiac output and arterial pressure.
treatment of circulatory shock; AVP is admin- The vasoconstrictor effects of AVP are probably
istered to increase systemic vascular resist- important only when AVP levels are very high,
ance and therefore arterial pressure. as occurs during severe hypovolemia.
Several mechanisms regulate the release of
AVP. Specialized stretch receptors within the
atrial walls and large veins (cardiopulmonary INTEGRATION OF
baroreceptors) entering the atria decrease their NEUROHUMORAL MECHANISMS
firing rate when atrial pressure falls (as occurs
with hypovolemia). Afferents from these recep- Autonomic and humoral influences are neces-
tors synapse within the hypothalamus, which sary to maintain a normal arterial blood pres-
is the site of AVP synthesis. AVP is transported sure under the different conditions in which
from the hypothalamus via axons to the pos- the human body functions. Neurohumoral
terior pituitary, from where it is secreted into mechanisms enable the body to adjust to
the circulation. Atrial receptor firing normally changes in body posture, physical activity, or
inhibits the release of AVP. With hypovolemia environmental conditions. The neurohumoral
and decreased central venous pressure, the mechanisms act through changes in systemic
decreased firing of atrial stretch receptors vascular resistance, venous compliance, blood
leads to an increase in AVP release. AVP release volume, and cardiac function, and through
is also stimulated by enhanced sympathetic these actions, they can effectively regulate arte-
activity accompanying decreased arterial rial blood pressure (Table 6-2). Although each
baroreceptor activity during hypotension. An mechanism has independent cardiovascular
important mechanism regulating AVP release actions, it is important to understand that each
involves hypothalamic osmoreceptors, which mechanism also has complex interactions with
sense extracellular osmolarity. When osmolar- other control mechanisms that serve to rein-
ity rises, as occurs during dehydration, AVP force or inhibit the actions of the other control
release is stimulated, which increases water mechanisms. For example, activation of sym-
retention by the kidneys. Finally, angiotensin pathetic nerves either directly or indirectly
II receptors (AT1) located within the hypo- increases circulating angiotensin II, aldoster-
thalamus regulate AVP release; an increase in one, adrenal catecholamines, and arginine vas-
angiotensin II stimulates AVP release. opressin, which act together to increase blood
Heart failure causes a paradoxical increase volume, cardiac output, and arterial pressure.
in AVP. The increased blood volume and atrial These humoral changes are accompanied by
pressure associated with heart failure suggest an increase in ANP, which acts as a counter-
that AVP secretion should be inhibited, but it regulatory system to limit the effects of the
is not. It may be that sympathetic and renin– other neurohumoral mechanisms.
angiotensin system activation in heart failure Finally, it is important to note that some
override the volume and low-pressure cardio- neurohumoral effects are rapid (e.g., auto-
vascular receptors (as well as the osmoregula- nomic nerves and catecholamine effects on
tion of AVP) and cause the increase in AVP cardiac output and arterial pressure), whereas
secretion. This increase in AVP during heart others may take several hours or days because
failure may contribute to the increased sys- changes in blood volume must occur before
temic vascular resistance and to renal reten- alterations in cardiac output and arterial pres-
tion of fluid. sure can be fully expressed.

Klabunde_Chap06.indd 141 6/11/2011 11:15:27 AM


142 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

TABLE 6-2 EFFECTS OF NEUROHUMORAL ACTIVATION ON BLOOD VOLUME, CARDIAC


OUTPUT, AND ARTERIAL PRESSURE

..!.~~~.~.~.~~~ ........................................ .L~~~~~.~.?..~.~~~...... .L:~~.~!~:..?..~~~~:!: ...... .L~.~~~.~~~~.~.~~.~~~.~~...........


.. ~~.~.~~.~.~~·~·i·~ ·~:~~~~~.~...................
Vagal activity
l.!..................................l.!......................................l.!...............................................
:- : .J.. : .J..

: ~:(~~~!~~i:~:~ ::~~~~~~~:~:(~~::::::::::::rY::::::::::::::::::::::::::::::::rt.:::::::::::::::::::::::::::::::::::::r~!:~ ::::::::::::::::::::::::::::::::::::::::::


.. ~~~~~~.:.~:~~..!.1..................................L!..................................L!......................................L!...............................................
. ~~~.~.~.~~.~~~.:.....................................L!..................................L!......................................L!...............................................
..~~:.~~.1 ~.~~~~~.~~.~!.~ .!?.~.~.~~.~~.......... .L~ ..................................L~...................................... L~...............................................
..

. ~~~~.~~~.:. ~~.~.~.r?.:.::.~.i.~...................L!..................................L!......................................L!...............................................
t = increase; .J.. =decrease.
'Dependent upon plasma epinephrine concentration.

SUMMARY OF IMPORTANT CONCEPTS

• Autonomic regulation of the heart fibers to target tissues in the heart and
and vasculature is primarily controlled blood vessels.
by special regions within the medulla • Sympathetic activation increases
oblongata of the brainstem that heart rate, inotropy, and
contain the cell bodies of sympathetic dromotropy through the release
and parasympathetic (vagal) efferent of norepinephrine, which binds
nerves. primarily to postjunctional cardiac
• The hypothalamus plays an integrative P1-adrenoceptors. Norepinephrine
role by modulating medullary neuronal released by sympathetic nerves
activity (e.g., during exercise). constricts blood vessels by
• Sensory nerves from peripheral binding primarily to postjunctional
baroreceptors (e.g., carotid sinus a,-adrenoceptors.
baroreceptors) synapse within the • Parasympathetic activation decreases
medulla at the NTS, which modulates heart rate, inotropy, and dromotropy,
the activity of the sympathetic and and it produces vasodilation in
vagal neurons within the medulla. specific organs through the release
• Preganglionic parasympathetic efferent of ACh, which binds to postjunctional
nerves exit the medulla as the tenth muscarinic (M 2 ) receptors.
cranial nerve and travel to the heart • Baroreceptors respond to stretch
within the left and right vagus nerves. induced by an increase in pressure
Preganglionic fibers synapse within or volume. Arterial baroreceptor
ganglia located within the heart; activity (e.g., carotid sinus and aortic
short postganglionic fibers innervate arch receptors) tonically inhibits
the myocardial tissue. Preganglionic sympathetic outflow to the heart
sympathetic efferent nerves exit from and blood vessels, and it tonically
the spinal cord and synapse within stimulates vagal outflow to the heart.
paravertebral or prevertebral ganglia Decreased arterial pressure, therefore,
before sending out postganglionic decreases the firing of arterial

Klabunde_Chap06.indd 142 6/11/2011 11:15:27 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 143

baroreceptors, which leads to reflex IJ2 -adrenoceptors) or increases (via


activation of sympathetic influences vascular a,- and ~-adrenoceptors)
acting on the heart and blood vessels systemic vascular resistance,
and withdrawal of the vagal activity to depending upon the plasma
the heart. concentration.
• Peripheral chemoreceptors (e.g., • The renin-angiotensin-aldosterone
carotid bodies) and central system plays a major role in regulating
chemoreceptors (e.g., medullary renal excretion of sodium and water.
chemoreceptors) respond to decreased The overall systemic effect of increased
P0 2 and pH or increased PC0 2 of the angiotensin II is increased blood
blood. Their primary function is to volume, venous pressure, and arterial
regulate respiratory activity, although pressure.
chemoreceptor activation generally • Atrial natriuretic peptide (ANP), which
leads to activation of the sympathetic is released by the atria primarily in
nervous system to the vasculature, response to atrial stretch, functions as
which increases arterial pressure. a counterregulatory mechanism for the
• Reflexes triggered by changes in renin-angiotensin-aldosterone system.
blood volume, cerebral and myocardial Therefore, increased ANP reduces
ischemia, pain, pulmonary activity, blood volume, venous pressure, and
muscle and joint movement, and arterial pressure.
temperature alter cardiac and vascular • Arginine vasopressin (AVP; antidiuretic
function. hormone), which is released by the
• Sympathetic activation of the posterior pituitary when the body
adrenal medulla stimulates needs to reduce renal loss of water,
the release of catecholamines, enhances blood volume and increases
principally epinephrine. This arterial and venous pressures. At high
hormone produces cardiac plasma concentrations, AVP constricts
stimulation (via P,-adrenoceptors), resistance vessels.
and it either decreases (via vascular

REVIEW QUESTIONS

For each question, choose the one best b. Constricts blood vessels by binding to
answer: a 1-adrenoceptors.
c. Inhibits its own release by binding to
1. The cell bodies for the preganglionic prejunctional ~ 2-adrenoceptors.
vagal efferents innervating the heart are d. Decreases renin release in the
found in which region of the brain? kidneys.
a. Cortex
b. Hypothalamus 3. Stimulating efferent fibers of the right
c. Medulla vagus nerve
d. Nucleus tractus solitarius a. Decreases systemic vascular
resistance.
2. Norepinephrine released by sympathetic b. Increases atrial inotropy.
nerves c. Increases heart rate.
a. Binds preferentially to ~2-adrenoceptors d. Releases acetylcholine, which binds to
on cardiac myocytes. M 2 receptors.

Klabunde_Chap06.indd 143 6/11/2011 11:15:29 AM


144 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

4. A sudden increase in carotid artery 8. A 27-year-old female patient with severe


pressure hypertension is found to have bilateral
a. Decreases carotid sinus baroreceptor renal artery stenosis caused by fibro-
firing rate. muscular dysplasia of the renal arteries
b. Increases sympathetic efferent nerve resulting in elevated levels of circulating
activity to systemic circulation. renin. One mechanism contributing to
c. Increases vagal efferent activity to the her hypertension is
heart. a. Increased blood volume.
d. Results in reflex tachycardia. b. Increased circulating atrial natriuretic
peptide.
5. Which of the following can cause c. Increased sodium loss by the kidneys.
tachycardia? d. Inhibition of aldosterone release.
a. Increased arterial pulse pressure
b. Increased blood PCO2 9. A hospitalized patient with acute decom-
c. Increased firing of carotid sinus pensated heart failure is given a drug
baroreceptors that increases circulating atrial natriu-
d. Vasovagal reflex retic peptide by inhibiting its metabo-
lism. What beneficial effects would this
6. Infusion of a high dose of epinephrine treatment have in this patient?
following pharmacologic blockade of a. Decrease blood volume by promoting
β-adrenoceptors will sodium loss by the kidneys
a. Decrease mean arterial pressure. b. Increase blood pressure by constricting
b. Have no significant cardiovascular the arterial and venous vasculature
effects. c. Increase cardiac output by increasing
c. Increase heart rate. preload
d. Increase systemic vascular d. Stimulate the release of aldosterone
resistance. from the adrenal cortex

7. In an experimental protocol, intravenous 10. Following an automobile accident that


infusion of acetylcholine was found resulted in significant hemorrhage, a
to decrease mean arterial pressure and 48-year-old male patient is admitted to the
increase heart rate. These results can Emergency Department in critical condition
best be explained by with an arterial pressure of 65/45 mm Hg
and a heart rate of 140 beats/min. Fluid
a. Direct action of acetylcholine on resuscitation was augmented by administra-
muscarinic receptors at the sinoatrial tion of arginine vasopressin. The potential
node. benefit of adding vasopressin during resus-
b. Increased firing of carotid sinus baro- citation is derived from its ability to
receptors.
c. Reflex activation of sympathetic a. Augment sympathetic activity.
nerves. b. Increase systemic vascular resistance.
d. Reflex systemic vasodilation. c. Produce renal fluid loss (diuresis).
d. Stimulate the release of renin.

Klabunde_Chap06.indd 144 6/11/2011 11:15:31 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 145

1. The correct answer is "c" because this arterial pressure by withdrawing sympa-
region of the brainstem contains cell thetic tone on the systemic vasculature.
bodies for both sympathetic and para- 5. The correct answer is "b" because
sympathetic neurons; choices "a" and increased blood PC0 2 stimulates chemo-
"b" are therefore incorrect. Choice "d" receptors, which activate the sympathetic
is incorrect because the nucleus tractus nervous system to constrict the systemic
solitarius is the region in the medulla vasculature and raise arterial pressure.
that receives afferent fibers from periph- Choice "a" is incorrect because increased
eral sensors (e.g., baroreceptors) and arterial pulse pressure stimulates arterial
then sends excitatory or inhibitory fibers baroreceptors, which leads to vagal acti-
to sympathetic and parasympathetic vation of the heart. Choice "c" is incor-
neurons within the medulla. rect because increased carotid sinus firing
2. The correct answer is "b" because nor- (usually caused by elevated arterial pres-
epinephrine binds to <X 1-adrenoceptors sure) causes a reflex decrease in heart
located on vascular smooth muscle to rate brought about by vagal activation
stimulate vasoconstriction. Choice "a" and sympathetic withdrawal. Choice "d"
is incorrect because norepinephrine is incorrect because the vasovagal reflex
preferentially binds to ~ 1 -adrenoceptors causes vagal activation and bradycardia.
in the heart. Choice "c" is incorrect 6. The correct answer is "d" because a high
because prejunctional ~ 2-adrenoceptors dose of epinephrine binds to both ~2-
facilitate norepinephrine release and <X1-adrenoceptors on blood vessels.
(prejunctional <X2-adrenoceptors inhibit Therefore, if the ~ 2-adrenoceptors (which
release). Choice "d" is incorrect because produce vasodilation) are blocked, the
norepinephrine stimulates renin release <X1-adrenoceptors can produce vasocon-
through ~ 1 -adrenoceptors. striction unopposed by the ~ 2-adreno-
3. The correct answer is "d" because the ceptors. Choice "a" is incorrect because
vagus nerve is parasympathetic choliner- the unopposed <X-adrenoceptor activation
gic and therefore releases acetylcholine. increases arterial pressure. Choice "b"
Choice "a" is incorrect because efferent is incorrect because epinephrine binds
right vagal stimulation primarily affects to <X as well as ~-adrenoceptors. Choice
the sinoatrial node and has no signifi- "c" is incorrect because epinephrine-
cant direct effects on the systemic vas- induced increased heart rate is mediated
culature. Choice "b" is incorrect because primarily by ~-adrenoceptors (which are
vagal stimulation decreases atrial blocked), and systemic vasoconstriction
inotropy. Choice "c" is incorrect because will increase arterial pressure and cause
right vagal stimulation reduces heart a reflex decrease in heart rate.
rate by decreasing the slope of Phase 4 7. The correct answer is "c" because ace-
of the pacemaker action potential. tylcholine dilates blood vessels, which
4. The correct answer is "c" because lowers arterial pressure and causes a
increased carotid artery pressure stimu- baroreceptor-mediated increase in heart
lates the firing of carotid sinus barore- rate brought about by sympathetic acti-
ceptors (therefore, choice "a" is incor- vation. Choice "a" is incorrect because
rect), which leads to a reflex activation stimulation of muscarinic receptors on
of vagal efferents to slow the heart rate the sinoatrial node induces bradycar-
(therefore, choice "d" is incorrect). dia. Choice "b" is incorrect because the
Choice "b" is incorrect because the baro- hypotension causes decreased carotid
receptor reflex would attempt to reduce sinus firing. Choice "d" is incorrect

Klabunde_Chap06.indd 145 6/11/2011 11:15:31 AM


146 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

because reflex systemic vasodilation can accumulation of fluid that can cause
occur only if arterial pressure is elevated pulmonary and systemic edema. Choices
and baroreceptor firing increases. "b," "c," and "d" are incorrect because
8. The correct answer is "a" because atrial natriuretic peptide dilates vessels,
increased renin leads to increased angio- reduces preload and cardiac output, and
tensin II and aldosterone (therefore, decreases aldosterone release.
choice "d" is incorrect), both of which 10. The correct answer is "b" because
act on the kidney to increase sodium vasopressin constricts blood vessels
reabsorption and blood volume (there- directly through V1 receptors, and not
fore, choice "c" is incorrect). Choice "b" through augmentation of sympathetic
is incorrect because although circulating activity which will actually decline as
atrial natriuretic peptide is increased, pressure is elevated during vasopressin
this hormone counteracts the pressure- administration (therefore, choice "a"
elevating mechanisms of angiotensin II. is incorrect). Choice "c" is incorrect
9. The correct answer is "a" because atrial because vasopressin has an antidiuretic
natriuretic peptide produces natriuresis effect. Choice "d" is incorrect because
and diuresis, both of which are ben- circulating renin would decline as
eficial to the acutely decompensated pressure is elevated during vasopressin
heart failure patient who has excessive administration.

PROBLEM 6-1 (a.1-adrenoceptor mediated); however, the


The common carotid arteries are below the pressor response would be blunted significantly
carotid sinus baroreceptors. Therefore, occlu- because cardiac stimulation would be blocked.
sion of both carotid arteries reduces pressure
PROBLEM 6-2
within the carotid sinuses. This decreases
~ 1 -adrenoceptor activation is primarily
their firing, leading to increased sympa-
responsible for the tachycardia and increased
thetic and decreased vagal outflow from the
cardiac output produced by epinephrine.
medulla. This results in systemic vasocon-
Blocking ~ 1 -adrenoceptors would signifi-
striction, cardiac stimulation, and a rise in
cantly blunt the cardiac responses. Epineph-
arterial pressure.
rine at low plasma concentrations also binds
Bilateral vagotomy enhances the response
to vascular ~2-adrenoceptors to cause vasodi-
described above because as arterial pressure
lation; therefore, arterial pressure would fall
rises during carotid occlusion, the aortic
during infusion of a low dose of epinephrine
arch baroreceptors, which are innervated by
in the presence of ~ 1 -adrenoceptor blockade
the vagus nerve, increase their firing. This
because the large decrease in systemic vas-
partially counteracts the effects of decreased
cular resistance would not be offset by an
carotid sinus firing. Bilateral vagotomy
increase in cardiac output.
removes this influence of the aortic arch
baroreceptors. PROBLEM 6-3
Blockade of ~-adrenoceptors would prevent Bilateral cervical vagotomy would prevent
the sympathetic-mediated increases in heart vagal slowing of the heart and denervate the
rate and inotropy (although some withdrawal aortic arch baroreceptors. Heart rate (and
of vagal tone may still result in a small increase inotropy) would increase owing to norepi-
in heart rate). The pressor response would still nephrine binding to ~ 1 -adrenoceptors on the
occur because of systemic vasoconstriction heart that is now unopposed by the vagus.

Klabunde_Chap06.indd 146 6/11/2011 11:15:31 AM


CHAPTER 6 • NEUROHUMORAL CONTROL OF THE HEART AND CIRCULATION 147

This, along with aortic arch denervation, causes sodium and fluid retention by the
would enhance the pressor response of kidneys and an increase in blood volume,
norepinephrine. which increases cardiac output. Increased
vasopressin (stimulated by angiotensin II)
CASE 6-1 contributes to the increase in blood volume.
Bilateral renal artery stenosis reduces the Increased angiotensin II increases systemic
pressure within the afferent arterioles, vascular resistance by binding to vascular
which causes release of renin. This, in turn, AT1 receptors and by enhancement of sympa-
increases circulating angiotensin II, which thetic activity. These changes in cardiac out-
stimulates aldosterone release. Activation put and systemic vascular resistance lead to a
of the renin-angiotensin-aldosterone system hypertensive state.

SUGGESTED RESOURCES Rhoades RA, Bell, DR. Medical Physiology: Principles


Berne RM, Levy MN. Cardiovascular Physiology. 8th for Clinical Medicine. 3rd Ed. Philadelphia:
Ed. Philadelphia: Mosby, 2001. Lippincott Williams & Wilkins, 2009.
Guyenet PG. The sympathetic control of blood pressure. Touyz CB, Dominiczak AF, Webb RC, Johns DB.
Nature Reviews Neuroscience 2006;7:335–346. Angiotensin receptors: signaling, vascular patho-
Melo LG, Pang SC, Ackermann U. Atrial natriuretic physiology, and interactions with ceramide. Am
peptide: regulator of chronic arterial blood pressure. J Physiol 2001;281:H2337–H2365.
News Physiol Sci 2000;15:143–149.
Mendolowitz D. Advances in parasympathetic control
of heart rate and cardiac function. News Physiol Sci
1999;14:155–161.

Klabunde_Chap06.indd 147 6/11/2011 11:15:32 AM


(')

7
:I:
>
'tl
-1
m
~
ORGAN BLOOD FLOW
Understanding the concepts presented in this chapter will enable the student to:
1. Describe the distribution of cardiac output among major organs when a person
is at rest.
2. Describe how various tissue and endothelial factors influence tissue blood flow.
3. Explain how extravascular compression alters blood flow in the heart and con-
tracting skeletal muscle .

4. Define autoregulation of blood flow, reactive hyperemia, and active (functional)
hyperemia and describe their mechanisms in different organs.
5. Compare and contrast autonomic control of blood flow in major vascular beds
of the body.
6. Describe the specialized vascular anatomy and function in the following organs:
brain, heart, intestines and liver, skin, kidneys, and lungs.

INTRODUCTION Table 7-l summarizes the distribution of


cardiac output when a person is at rest. Most
This chapter describes the blood flow to dif- of the cardiac output (-80%) goes to the gas-
ferent organs of the body. The first part of the trointestinal tract, kidneys, skeletal muscle,
chapter emphasizes local regulatory mecha- heart, and brain, although these organs make
nisms by which organs regulate their own up <50% of the body mass. This relative dis-
blood flow to meet the metabolic and func- tribution of cardiac output, however, changes
tional requirements of the organ. The second greatly depending on environmental condi-
part of the chapter examines blood flow in spe- tions and the state of physical activity. For
cific organs of the body. example, in a hot, humid environment, the rel-
ative blood flow to the skin increases substan-
DISTRIBUTION OF CARDIAC tially as the body attempts to maintain its core
OUTPUT temperature by losing heat to the environment.
When a person exercises, the increased cardiac
We have previously seen that arterial pressure output primarily goes to the active skeletal
is generated as the heart pumps blood into the muscles, heart, and skin (see Chapter 9); at
systemic circulation. This arterial pressure the same time, blood flow decreases to the gas-
serves as the driving force for blood flow to trointestinal and renal circulations. Another
all the organ systems. The relative distribu- example of change in cardiac output distribu-
tion of blood flow to the organs is regulated tion occurs following a meal, when blood flow
by the vascular resistance of the individual to the gastrointestinal circulation increases.
organs, which is influenced by extrinsic (neu- Instead of one "normal" blood flow for an
rohumoral) and intrinsic (local regulatory) organ, there is a range of blood flows. Basal
mechanisms as summarized in Chapter 5, flow refers to the flow that is measured under
Figure 5.12. basal conditions (i.e., when a person is in a

148

Klabunde_Chap07.indd 148 6/10/2011 10:57:42 PM


CHAPTER 7 • ORGAN BLOOD FLOW 149

TABLE 7-1 BLOOD FLOW IN MAJOR ORGANS OF THE BODY


PERCENT BODY PERCENT CARDIAC NORMAL FLOW MAXIMAL FLOW
ORGAN WEIGHT OUTPUT AT REST (mL/min PER 100 g) (mL/min PER 100 g)

Heart 0.5 5 80 400


Brain 2 14 55 150
Skeletal muscle 40 18 3 60
Skin 3 4 10 150
Stomach, intestine, 6 23 30 250
liver, spleen, pan-
creas
Kidneys 0.5 20 400 600
Other 48 16 — —
Normal and maximal flows are approximate values for the whole organ. Many organs (e.g., brain, muscle, kidney, and intes-
tine) have considerable heterogeneity of flow within the organ depending on the type of tissue or region of organ being
perfused. The liver receives blood flow from the gastrointestinal venous drainage as well as from the hepatic artery (only
hepatic artery flow is included in this table). “Other” includes reproductive organs, bone, fat, and connective tissue.

fasted, resting state and at normal environ- constant pressure from a reservoir containing
mental conditions of temperature and humid- oxygenated blood, and then electrically stimu-
ity). The ratio of basal flow to maximal flow lated to induce muscle contractions, the blood
is a measure of the vascular tone, which is the flow increases. The increase in blood flow occurs
degree of vascular constriction (see Chapter 5). in the absence of neurohumoral influences and
The lower the basal flow relative to the maxi- therefore is a local or intrinsic mechanism.
mal flow, the higher the vascular tone. The The mechanisms responsible for local regu-
difference between basal flow and maximal lation originate from within the blood vessels
flow represents the flow capacity or vasodila- (e.g., endothelial factors, myogenic mecha-
tor reserve for the organ. Most organs have nisms) and from the surrounding tissue (i.e.,
a relatively large vasodilator reserve, whereas tissue factors), many of which are related to
others, such as the kidneys, have a relatively tissue metabolism or other biochemical path-
small vasodilator reserve (see Table 7-1). ways (e.g., arachidonic acid metabolites and
The changes that occur in organ blood flow bradykinin). Mechanical factors (e.g., com-
under different conditions depend on the inter- pressive forces during muscle contraction)
play between neurohumoral and local regulatory can also influence vascular resistance and
mechanisms that govern vascular resistance. thereby alter blood flow.
The neurohumoral mechanisms were discussed
in Chapter 6. The following sections focus on Tissue Factors
the local regulatory mechanisms that affect vas- Tissue factors are substances produced by the
cular resistance and organ blood flow. tissue surrounding blood vessels (Fig. 7.1).
These substances act on the blood vessel to
LOCAL REGULATION OF produce either relaxation or contraction of
BLOOD FLOW the smooth muscle, thereby altering resistance
and blood flow. In some cases, these sub-
Tissues and organs have the ability to regu- stances indirectly act on the vascular smooth
late, to a varying degree, their own blood flow. muscle by affecting endothelial function or
This intrinsic ability to regulate blood flow by altering the release of norepinephrine by
is termed “local regulation” and can occur in sympathetic nerves. Some of these vasoactive
the complete absence of any extrinsic neuro- substances are tissue metabolites that are prod-
humoral influences. For example, if a mus- ucts of cellular metabolism or activity (e.g.,
cle is removed from the body, perfused under adenosine, CO2, H+, K+, lactate). In addition,

Klabunde_Chap07.indd 149 6/10/2011 10:57:44 PM


150 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Paracrine-
Releasing Cell Cellular
Metabolism

¯ PO 2
Histamine Ado
Paracrine –
Bradykinin Vasodilator PO4

Hormones
Prostaglandins Substances Lactate
CO2
± – K+
H+
Arteriole
■ FIGURE 7.1 Vasoactive substances derived from tissue cells around arterioles. Increased tissue metabo-
lism leads to formation of metabolites that dilate (−) nearby arterioles. Increased oxygen consumption
decreases the tissue partial pressure of oxygen (PO2), which dilates arterioles. Some cells release locally
acting, paracrine hormones (or their precursors), which can either constrict (+) or dilate (−) arterioles.
Ado, adenosine; PO4−, inorganic phosphate; CO2, carbon dioxide; K+, potassium ion; H+, hydrogen ion.

different cell types surrounding blood vessels Their relative importance depends on the tis-
can release vasoactive substances referred to sue in which they are formed as well as differ-
as local, paracrine hormones (e.g., histamine, ent conditions that might cause their release.
bradykinin, and prostaglandins). A paracrine
hormone is a substance released by one cell 1. Adenosine is a potent vasodilator in
that acts on another nearby cell by diffusing most organs (although adenosine constricts
through the interstitial fluid. This is in con- renal vessels). It is formed by the action of
trast to endocrine hormones that circulate 5′-nucleotidase, an enzyme that dephos-
in the blood to reach distant target cells or phorylates adenosine monophosphate
autocrine substances that affect the same cell (AMP). The AMP is derived from hydroly-
from which they are released. sis of intracellular adenosine triphosphate
Increases or decreases in metabolism alter (ATP) and adenosine diphosphate (ADP).
the release of some of these vasoactive sub- Adenosine formation increases during
stances; thus, metabolic activity is closely hypoxia and increased oxygen consump-
coupled to blood flow in most organs of the tion, both of which lead to increased
body. For example, an increase in tissue ATP hydrolysis. Small amounts of ATP
metabolism, as occurs during muscle contrac- hydrolysis can lead to large increases in
tion or during changes in neuronal activity in adenosine formation because intracellular
the brain, leads to an increase in blood flow. concentrations of ATP are about a 1000-
Extensive evidence shows that the actively fold greater than adenosine concentrations.
metabolizing cells surrounding arterioles Experimental evidence supports the idea
release vasoactive substances that cause vaso- that adenosine formation is a particularly
dilation. This is termed the metabolic theory important mechanism for regulating coro-
of blood flow regulation. These vasoactive nary blood flow when myocardial oxygen
substances, which are linked to tissue metab- consumption increases or during hypoxic
olism, ensure that the tissue is adequately conditions.
supplied with oxygen and that products of 2. Inorganic phosphate is released by the
metabolism (e.g., CO2, H+, lactic acid) are hydrolysis of adenine nucleotides (ATP,
removed. Several substances have been impli- ADP, and AMP). Inorganic phosphate
cated in metabolic regulation of blood flow. may have some vasodilatory activity in

Klabunde_Chap07.indd 150 6/10/2011 10:57:44 PM


CHAPTER 7 • ORGAN BLOOD FLOW 151

contracting skeletal muscle, but its vasodilation may be direct (inadequate O2


importance is far less than that of adeno- to sustain smooth muscle contraction) or
sine, potassium, and nitric oxide in regu- indirect via the production of vasodilator
lating skeletal muscle blood flow. metabolites (e.g., adenosine, lactic acid,
3. Carbon dioxide formation increases during H+). Although hypoxia causes vasodilation
states of increased oxidative metabolism. in nearly all vascular beds, there is a nota-
CO2 concentrations in the tissue and vascu- ble exception—it causes vasoconstriction
lature can also increase when blood flow is in the pulmonary circulation.
reduced, which reduces the washout of CO2. 7. Osmolarity changes in the blood and in
As a gas, CO2 readily diffuses from parenchy- the tissue interstitium have been impli-
mal cells to the vascular smooth muscle of cated in local blood flow regulation. It is
blood vessels, where it causes vasodilation. well known that intra-arterial infusions
Considerable evidence indicates that CO2 of hyperosmolar solutions can produce
plays a significant role in regulating cerebral vasodilation. The molecules making up the
blood flow through the formation of H+. hyperosmolar solution need not be vasoac-
4. Hydrogen ion increases through the bicar- tive. Tissue ischemia and increased meta-
bonate buffer system when CO2 increases. bolic activity raise the osmolarity of the
Hydrogen ion also increases during states of tissue interstitial fluid and venous blood.
increased anaerobic metabolism (e.g., during Therefore, it has been suggested that non-
ischemia or hypoxia) when acid metabolites specific changes in osmolarity may play a
such as lactic acid are produced. Increased role in the regulation of blood flow.
H+ causes local vasodilation, particularly in
the cerebral circulation. Several tissue factors involved in regulating
5. Potassium ion is released by contracting blood flow are not directly coupled to tissue
cardiac and skeletal muscle. Muscle contrac- metabolism. These include paracrine hor-
tion is initiated by membrane depolariza- mones such as histamine, bradykinin, and
tion, which results from a cellular influx of products of arachidonic acid (eicosanoids).
Na+ and an efflux of K+. Normally, the Na+/ Histamine, released by tissue mast cells in
K+-ATPase pump is able to restore the ionic response to injury, inflammation, and aller-
gradients (see Chapter 2); however, the gic responses, causes arteriolar vasodilation,
pump does not keep up with rapid depolari- venous constriction in some vascular beds,
zations (i.e., there is a time lag) during mus- and increased capillary permeability. Both H1
cle contractions, and a small amount of K+ and H2 histamine receptors are involved in
accumulates in the extracellular space. Small the vascular effects of histamine. Bradykinin
increases in extracellular K+ around blood is formed from the action of kallikrein (a pro-
vessels cause hyperpolarization of the vascu- teolytic enzyme) acting on alpha2-globulin
lar smooth muscle cells, possibly by stimu- (kininogen), which is found in blood and
lating the electrogenic Na+/K+-ATPase pump tissues. Like histamine, bradykinin is a pow-
and increasing K+ conductance through erful dilator of arterioles. It acts on vascular
potassium channels. Hyperpolarization leads bradykinin receptors, which stimulate nitric
to smooth muscle relaxation. Potassium ion oxide formation by the vascular endothelium,
appears to play a role in causing the increase thereby producing vasodilation. In addition,
in blood flow in contracting skeletal muscle. bradykinin stimulates prostacyclin forma-
6. Oxygen levels within the blood, ves- tion, which produces vasodilation. One of
sel wall, and surrounding tissue are also the enzymes responsible for breaking down
important in local regulation of blood flow. bradykinin is angiotensin-converting enzyme
Decreased tissue partial pressure of oxygen (ACE) (see Chapter 6, Fig. 6.11). Therefore,
(PO2) resulting from reduced oxygen sup- drugs that inhibit ACE not only decrease
ply or increased oxygen utilization by tis- angiotensin II but also increase bradykinin,
sues causes vasodilation. Hypoxia-induced which is believed to be partly responsible for

Klabunde_Chap07.indd 151 6/10/2011 10:57:44 PM


152 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

the vasodilation accompanying ACE inhibi- vasodilators. In contrast, endothelin-1 is a


tion. Some arachidonic acid metabolites such powerful vasoconstrictor.
as prostaglandin E2 (PGE2) are vasodilators, Nitric oxide appears to be the most
whereas other eicosanoids such as PGF2α, important in terms of regulating blood flow
thromboxanes, and leukotrienes are generally under normal physiologic conditions. Nitric
vasoconstrictors. Drugs that block the forma- oxide is synthesized in the endothelium by
tion of these eicosanoids (e.g., cyclooxyge- the action of a nitric oxide synthase (NOS)
nase inhibitors such as aspirin or ibuprofen) enzyme on the amino acid, L-arginine. Nitric
alter vascular control by these substances. oxide diffuses from the endothelial cell to
the smooth muscle cells where it binds to
Endothelial Factors and activates intracellular guanylyl cyclase
to form cGMP, which leads to smooth muscle
The vascular endothelium serves an important relaxation (see Chapter 3). If nitric oxide syn-
paracrine role in the regulation of smooth mus- thesis is inhibited pharmacologically using
cle tone and organ blood flow. As described in NOS inhibitors, vasoconstriction occurs in
Chapter 3, the vascular endothelium produces most vascular beds. This demonstrates that
vasoactive substances that have significant there normally is a basal release of nitric
effects on vascular smooth muscle. Circulat- oxide that inhibits vascular tone; therefore,
ing (endocrine) and paracrine hormones, blocking nitric oxide formation leads to an
shearing forces, hypoxia, and many different increase in tone.
drugs can stimulate the formation and release Nitric oxide is involved in what is termed
of endothelial substances (Fig. 7.2). Among flow-dependent vasodilation. Experimental
their many actions, two of these substances, studies have shown that an increase in vessel
nitric oxide and prostacyclin, are powerful flow (actually an increase in shearing forces
acting on the vascular endothelium) stimu-
Circulating Hormones
Paracrine Hormones
lates endothelial nitric oxide production,
Shearing Forces which causes vasodilation. Flow-dependent
Hypoxia
vasodilation is particularly important as a
mechanism for increasing coronary blood
L-arg
Endothelial Cell flow when cardiac activity and metabolism
NOS are increased. Impaired nitric oxide synthesis
NO
AA or decreased bioavailability, as occurs during
coronary artery disease, limits the ability of
ECE
coronary blood flow to increase when cardiac
NO EDHF ET-1 PGI2 activity and oxygen demand are increased.
Other disorders such as hypertension, cer-
– – + – ebrovascular disease, and diabetes are asso-
Vascular ciated with impaired endothelial control of
CONTRACTION Smooth
Muscle vascular function as well.
Another endothelial factor is endothelial-
■ FIGURE 7.2 Endothelial-derived vasoactive derived hyperpolarizing factor (EDHF). Some
factors. Nitric oxide (NO) formed by nitric oxide substances (e.g., acetylcholine, bradykinin)
synthase (NOS) acting on L-arginine (L-arg), that stimulate nitric oxide production stimulate
endothelial-derived hyperpolarizing factor (EDHF),
EDHF as well. The identity of this factor is not
and prostacyclin (PGI2) derived from arachidonic
acid (AA) inhibit (−) smooth muscle contraction and known for certain, but its release causes smooth
cause vasodilation. Endothelin-1 (ET-1) formed by muscle hyperpolarization and relaxation.
endothelin-converting enzyme (ECE) causes smooth Prostacyclin (PGI2) is formed from arachi-
muscle contraction (+). The formation and release of
donic acid and the cyclooxygenase enzyme
these substances are influenced by circulating and
paracrine hormones, shearing forces acting on the within endothelial cells. This paracrine
endothelium, hypoxia, and many different drugs. substance is a potent vasodilator in addition to

Klabunde_Chap07.indd 152 6/10/2011 10:57:44 PM


CHAPTER 7 • ORGAN BLOOD FLOW 153

serving as an inhibitor of platelet aggregation. phosphorylation of myosin light chains, and


PGI2 synthesis is stimulated by adenosine and contraction (see Chapter 3).
nitric oxide, as well as by many other sub- Myogenic behavior has been observed in
stances, and therefore can play a secondary many different vascular beds, although its
role to the vasodilation produced by other relative functional significance differs among
substances. It causes vasodilation by activat- organs. It is difficult to evaluate myogenic
ing smooth muscle adenylyl cyclase, which mechanisms in vivo because changes in pres-
increases cAMP (see Chapter 3). sure are usually associated with changes in
Endothelin-1 (ET-1) is a potent vasoconstric- flow that trigger metabolic mechanisms, which
tor substance that is synthesized from an intra- usually dominate over myogenic mechanisms.
cellular precursor by endothelin-converting For example, increasing venous pressure to a
enzyme (ECE) found on the endothelial cell vascular bed should activate myogenic mecha-
membrane. ET-1 binds to ETA receptors on nisms to produce vasoconstriction because
smooth muscle cells, which are coupled to Gq- elevated venous pressures are transmitted back
proteins (see Chapter 3). ET-1 can also bind to the precapillary resistance vessels; however,
to a second type of receptor (ETB) located on the reduction in blood flow associated with the
the vascular endothelium that stimulates nitric increase in venous pressure (which reduces
oxide and prostacyclin synthesis and release, perfusion pressure) activates tissue meta-
which act as negative feedback mechanisms to bolic mechanisms that cause vasodilation. In
counteract the ETA-mediated vasoconstrictor most organs, conducting such an experiment
effects of ET-1. usually results in vasodilation because the
ET-1 formation and release by endothelial metabolic vasodilator response overrides the
cells is stimulated by angiotensin II, vasopres- myogenic vasoconstrictor response, if present.
sin (antidiuretic hormone, ADH), thrombin,
cytokines, reactive oxygen species, and shear-
Extravascular Compression
ing forces acting on the vascular endothelium.
ET-1 release is inhibited by nitric oxide, as well Mechanical compressive forces can affect vas-
as by prostacyclin and atrial natriuretic peptide. cular resistance and blood flow within organs.
Some forms of hypertension (e.g., pulmonary Sometimes this occurs during normal physi-
artery hypertension) appear to involve ET-1 ologic conditions; at other times, compressive
and are treated with ET-1 receptor blockers. forces can be the result of pathologic mecha-
nisms. The pressure that distends the wall of a
blood vessel is the transmural pressure (inside
Smooth Muscle (Myogenic)
minus outside pressure). Therefore, if the
Mechanisms
pressure outside of the vessel increases, then
Myogenic mechanisms originate within the the transmural pressure decreases. At very
smooth muscle of blood vessels, particu- high extravascular pressures, a vessel can com-
larly in small arteries and arterioles. When pletely collapse. Therefore, veins, which have a
the lumen of a blood vessel is suddenly relatively low intravascular pressure, are more
expanded, as occurs when intravascular likely to collapse when extravascular pressure
pressure is suddenly increased, the smooth is elevated; however, arteries can also become
muscle responds by contracting in order to significantly compressed when extravascular
restore the vessel diameter and resistance. pressure is elevated to very high levels.
Conversely, a reduction in intravascular Several examples of mechanical compres-
pressure results in smooth muscle relaxa- sion affecting organ blood flow exist. During
tion and vasodilation. Electrophysiologic cardiac systole or skeletal muscle contraction
studies have shown that vascular smooth (particularly tetanic contractions), vascu-
muscle cells depolarize when stretched, lar resistance is greatly increased and blood
leading to calcium entry into the cell (pri- flow is impeded by mechanical compression.
marily through L-type calcium channels), Lung inflation and deflation alter pulmonary

Klabunde_Chap07.indd 153 6/10/2011 10:57:44 PM


154 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

vascular transmural pressures (see Fig. 5.16) When perfusion pressure (arterial − venous
and thereby have substantial effects on pul- pressure; PA − PV) initially decreases, blood flow
monary vascular resistance. Excessive disten- (F) falls because of the following relationship
sion of the gastrointestinal tract, as occurs between pressure, flow, and resistance (R):
during intestinal obstruction, can increase
vascular resistance in the wall of the intestine (PA - PV )
F=
to a point where tissues become ischemic. R
Blood vessels in organs such as the brain or
kidneys, which are surrounded by a rigid cra- If resistance remains unchanged, the reduction
nium or capsule, are particularly susceptible in flow will be proportionate to the reduction
to increases in extravascular pressure that in perfusion pressure; however, in most organs
occur with edema, vascular hemorrhage (e.g., of the body, resistance does not remain constant
cerebral stroke), or the growth of a tumor. when perfusion pressure is decreased. The reduc-
tions in flow and perfusion pressure are thought
to activate metabolic and myogenic mechanisms
Autoregulation of Blood Flow that cause arteriolar vasodilation and a fall in
Autoregulation is the intrinsic ability of an organ resistance (R). As resistance decreases, blood
to maintain a constant blood flow despite changes flow increases despite the presence of a reduced
in perfusion pressure. For example, if perfusion perfusion pressure. This autoregulatory response
pressure is decreased to an organ by partial is shown in the left panel of Figure 7.3. For exam-
occlusion of the artery supplying the organ, ple, if perfusion pressure is reduced from 100 to
blood flow will initially fall, then return toward 70 mm Hg, it causes flow to decrease initially by
normal levels over the next few minutes. This approximately 30%. Over the next few minutes,
autoregulatory response occurs in isolated, however, flow begins to increase back toward
perfused organs, which are not subject to neu- control as the organ blood flow is autoregulated
ral or humoral influences. Therefore, it is a (red lines). Blood flow increases because vascu-
local or intrinsic response of the organ. lar resistance falls as the resistance vessels dilate.

No Autoregulation
Resistance
A Autoregulatory
Autoregulation Range
500
Blood Flow

Flow
(ml/min) B
300 B A
No Autoregulation
Pressure 100
(mm Hg)
70
0 2 4 6 0 100 200
Time (min) Perfusion Pressure (mm Hg)
■ FIGURE 7.3 Autoregulation of blood flow. The left panel shows that decreasing perfusion pressure from
100 to 70 mm Hg at point A results in a transient decrease in flow. If no autoregulation occurs, resistance
remains unchanged and flow remains decreased. With autoregulation (red line), the initial fall in pressure
and flow are followed by a decrease in vascular resistance, which causes flow to increase to a new steady-
state level despite the reduced perfusion pressure (point B). The right panel shows steady-state, autoreg-
ulatory flows plotted against different perfusion pressures. Points A and B represent the control flow and
autoregulatory steady-state flow, respectively, from the left panel. The autoregulatory range is the range
of pressures over which flow shows little change. Below or above the autoregulatory range, flow changes
are approximately proportional to the changes in perfusion pressure. The autoregulatory range as well as
the flatness of the autoregulatory response curve varies among organs.

Klabunde_Chap07.indd 154 6/10/2011 10:57:44 PM


CHAPTER 7 • ORGAN BLOOD FLOW 155

If the perfusion pressure to an organ is hypotension caused by blood loss, despite


increased and decreased over a wide range of baroreceptor reflexes that lead to constriction
pressures and the steady-state autoregulatory of much of the systemic vasculature, blood
flow response is measured, then the relation- flow to the brain and myocardium will not
ship between steady-state flow and perfusion decline appreciably (unless the arterial pres-
pressure can be plotted as shown in the right sure falls below the autoregulatory range).
panel of Figure 7.3. There is a range of pres- This is because of the strong capacity of these
sures (autoregulatory range) over which organs to autoregulate and their ability to
flow changes relatively little despite a large escape sympathetic vasoconstrictor influ-
change in perfusion pressure. The "flatness" ences. The autoregulatory response helps to
of the autoregulation curve varies considerably ensure that these critical organs have an ade-
among organs; the flatter the relationship, the quate blood flow and oxygen delivery even in
better the autoregulation. Coronary; cerebral, the presence of systemic hypotension.
and renal circulations show a high degree of Other situations occur in which systemic
autoregulation, whereas skeletal muscle and arterial pressure does not change, but in
gastrointestinal circulations show only a mod- which autoregulation is very important nev-
erate degree of autoregulation. The cutaneous ertheless. Autoregulation can occur when
circulation displays virtually no autoregulation. a distributing artery to an organ (e.g., coro-
Autoregulation has limits even in organs nary artery) becomes partially occluded. This
that display a high degree of autoregulation. arterial stenosis increases resistance and the
When the perfusion pressure falls below 60 pressure drop along the vessel length. This
to 70 mm Hg in the cerebral and coronary reduces pressure in small distal arteries and
circulations, the resistance vessels become arterioles, which are the primary vessels for
maximally dilated and their ability to autoreg- regulating blood flow within an organ. These
ulate is lost. Furthermore, at very high per- resistance vessels dilate in response to the
fusion pressures (-170 mm Hg in Fig. 7.3), reduced pressure and blood flow caused by
the upper limit of the autoregulatory range the upstream stenosis. This autoregulatory
is reached and the vessels undergo no fur- response helps to maintain normal blood flow
ther constriction with increases in perfusion in the presence of upstream stenosis, and it
pressure; therefore, flow increases as pressure is particularly important in organs such as
increases. The autoregulatory response can the brain and heart that depend on a steady
be modulated by neurohumoral influences delivery of oxygen to maintain normal organ
and disease states. For example, sympathetic function.
stimulation and chronic hypertension can
shift the cerebral autoregulatory range to the PROBLEM 7-1
right as described later in this chapter.
An experiment was conducted using
Autoregulation may involve both meta-
an isolated perfused organ (e.g.,
bolic and myogenic mechanisms. If the per-
intestinal segment) in which arterial
fusion pressure to an organ is reduced, the
and venous pressures were controlled
initial fall in blood flow leads to a fall in tis-
while blood flow was measured. When
sue P0 2 and the accumulation of vasodila-
venous pressure was suddenly raised
tor metabolites. These changes cause the
from 0 to 15 mm Hg while arterial
resistance vessels to dilate in an attempt to
pressure was maintained at 100 mm
restore normal flow. A reduction in perfusion
Hg, flow decreased by 25%. Calculate
pressure may also be sensed by the smooth
the percentage change that occurred
muscle in resistance vessels, which responds
in vascular resistance in response to
by relaxing (myogenic response), leading to
venous pressure elevation. Discuss the
an increase in flow.
involvement of metabolic and myogenic
Under what conditions does autoregu-
mechanisms in this response.
lation occur, and why is it important? In

Klabunde_Chap07.indd 155 6/10/2011 10:57:45 PM


156 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Reactive and Active Hyperemia the vascular bed and its metabolic activity.
For example, in the beating heart (high meta-
Reactive hyperemia is the transient increase in bolic activity), maximal reactive hyperemic
organ blood flow that occurs following a brief responses are seen with coronary occlusions
period of ischemia, usually produced by tem- of <1 minute, whereas in resting skeletal mus-
porary arterial occlusion. Figure 7.4 shows cle (low metabolic activity), several minutes
the effects of a 2-minute arterial occlusion of ischemia are necessary to elicit a maximal
on blood flow. During the occlusion period, vasodilator response. Myogenic mechanisms
blood flow goes to zero. When the occlusion may also contribute to reactive hyperemia
is released, blood flow rapidly increases above in some tissues because arterial occlusion
normal levels (hyperemia) that lasts for sev- decreases the pressure in arterioles, which can
eral minutes. In most tissues, experiments lead to myogenic-mediated vasodilation.
have suggested that the hyperemia occurs Several examples of reactive hyperemia
because during the occlusion period, tissue exist. The application of a tourniquet to a limb,
hypoxia and a buildup of vasoactive metabo- and then its removal, results in reactive hyper-
lites relax the smooth muscle of precapillary emia. During surgery, arterial vessels are often
resistance vessels. When the occlusion is clamped for a period of time; release of the
released and perfusion pressure is restored, arterial clamp results in reactive hyperemia.
flow becomes elevated because of the reduced Transient coronary artery occlusions (e.g., cor-
vascular resistance. During the hyperemia, onary vasospasm) result in subsequent reactive
oxygen becomes replenished and vasodilator hyperemia within the myocardium supplied by
metabolites are washed out of the tissue, caus- the coronary vessel.
ing the resistance vessels to regain their nor- Active hyperemia is the increase in organ
mal vascular tone and thereby return flow to blood flow that is associated with increased
normal levels. The longer the period of occlu- metabolic activity of an organ or tissue. With
sion, the greater the metabolic stimulus for increased metabolic activity, vascular resist-
vasodilation, leading to increases in peak flow ance decreases owing to vasodilation and
and duration of hyperemia. Maximal vasodi- vascular recruitment (particularly in skeletal
lation, as indicated by a maximal peak hyper- muscle). Active hyperemia occurs during mus-
emic flow, may occur following <1 minute of cle contraction (also termed exercise or func-
complete arterial occlusion, or it may require tional hyperemia), increased cardiac activity,
several minutes of occlusion depending on increased mental activity, and increased gas-
trointestinal activity during food absorption.
In Figure 7.5, the left panel shows the
Reactive Hyperemia
Excess Flow
effects of increasing tissue metabolism for
2 minutes on mean blood flow in a rhythmi-
cally contracting skeletal muscle. Within sec-
Flow onds of initiating contraction and the increase
No
Flow in metabolic activity, blood flow increases. The
vasodilation is thought to be caused by a com-
bination of tissue hypoxia and the generation
of vasodilator metabolites such as potassium
ion, carbon dioxide, nitric oxide, and adeno-
0 2 4 6 sine. This increased blood flow (i.e., hyper-
Time (min) emia) is maintained throughout the period of
■ FIGURE 7.4 Reactive hyperemia. Arterial occlu- increased metabolic activity and then subsides
sion (no flow) for 2 minutes followed by reperfu- after contractions cease and normal metabo-
sion results in a transient increase in blood flow
lism is restored. The amplitude of the active
(reactive hyperemia). The magnitude and duration
of the reactive hyperemia are directly related to hyperemia is closely related to the increase in
the duration of ischemia. metabolic activity (e.g., oxygen consumption)

Klabunde_Chap07.indd 156 6/10/2011 10:57:46 PM


CHAPTER 7 • ORGAN BLOOD FLOW 157

Steady-State Flow
Active or Functional
Hyperemia
Flow

Increased
Metabolism

0 2 4 6 8 Metabolic Activity
Time (min)
■ FIGURE 7.5 Active hyperemia. The left panel shows that increasing tissue metabolism for 2 minutes
transiently increases blood flow (active or functional hyperemia). The right panel shows that the steady-
state increase in blood flow during active hyperemia is directly related to the increase in metabolic activity
until the vessels become maximally dilated and flow can no longer increase.

as shown in the right panel. At high levels of arteries and veins. Regulatory mechanisms exist
metabolic activity, the vasculature becomes to ensure that adequate oxygen is delivered to
maximally dilated, resulting in a maximal the myocardium. Coronary artery disease or
increase in blood flow. Active hyperemia is the failure of regulatory mechanisms can lead
important because it increases oxygen deliv- to insufficient oxygen delivery to the myocar-
ery to tissues at a time of increased oxygen dium, which will impair cardiac function.
demand. Furthermore, the increased blood
flow enhances the removal of metabolic waste CORONARY VASCULAR ANATOMY
products from the tissue.
The vasodilatory capacity during active The two major branches of the coronary cir-
hyperemia differs considerably among organs. culation are the left main and right main
In skeletal muscle, blood flow can increase coronary arteries (Fig. 7.6). These vessels
more than 20- to 50-fold during exercise, arise from coronary ostia, which are small
depending on the type of muscle. Cerebral openings in the wall of the ascending aorta
blood flow, in contrast, increases no more just distal to the aortic valve. The left main
than twofold at maximal metabolic activity. coronary artery is relatively short in length
The reason for this difference is that resting (∼1 cm). After coursing behind the pulmonary
skeletal muscle has a high degree of vascular artery trunk, it divides into the left anterior
tone in contrast to the cerebral circulation, descending artery, which travels along the
which has a relatively low degree of vascu- interventricular groove on the anterior surface
lar tone because of its higher metabolic rate of the heart, and the circumflex artery, which
under basal conditions. travels posteriorly along the groove between
the left atrium and ventricle. These branches of
the left coronary artery supply blood primar-
SPECIAL CIRCULATIONS
ily to the left ventricle and atrium. The right
main coronary artery travels between the
Coronary Circulation
right atrium and ventricle (left atrioventricu-
In order to supply sufficient oxygen to support lar groove) toward the posterior regions of the
the high oxidative metabolism of the beating heart. This vessel and its branches serve the
heart, there must be an extensive network of right ventricle and atrium, and in most individ-
vessels that provide blood flow throughout the uals, the inferoposterior region of the left ven-
myocardium. These vessels are the coronary tricle. Significant variation is possible among

Klabunde_Chap07.indd 157 6/10/2011 10:57:46 PM


158 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

SVC

Ao Left Main
Coronary

PA Circumflex
Right
Coronary Left Anterior
Descending

Left
Ventricle
Right
Ventricle

IVC
■ FIGURE 7.6 Anterior view of the heart showing the major coronary arteries. The left main artery arises
from the aorta (Ao) just distal to the aortic valve, travels behind the pulmonary artery (PA), and then
branches into the circumflex artery (courses along the left atrioventricular groove) and left anterior
descending artery (courses along the interventricular groove), both of which primarily supply blood to
the left ventricle. The right coronary artery arises from the aorta and travels between the right atrium and
ventricle toward the posterior regions of the heart to supply the right ventricle and atrium and the infer-
oposterior wall of the left ventricle. SVC, superior vena cava; IVC, inferior vena cava.

individuals in the anatomical arrangement and Coronary veins are located adjacent to
distribution of flow by the coronary vessels. coronary arteries. These veins drain into the
The major coronary arteries lie on the coronary sinus located on the posterior aspect
epicardial surface of the heart and serve as of the heart. Blood flow from the coronary
low-resistance distribution vessels. These epi- sinus empties into the right atrium. Some
cardial arteries give off smaller branches that drainage also occurs directly into the cardiac
dive into the myocardium and become the chambers through the anterior cardiac veins
microvascular resistance vessels that regulate and thebesian vessels.
coronary blood flow. The resistance vessels
REGULATION OF CORONARY
give rise to a dense capillary network so that
BLOOD FLOW
each cardiac myocyte is closely associated
with several capillaries. The high capillary- When flow is measured within an epicardial
to-fiber density ensures short diffusion dis- coronary artery, it is found to decrease during
tances to maximize oxygen transport into the cardiac systole and increase during diastole
cells and removal of metabolic waste products (Fig. 7.7). Therefore, most of the blood flow
(e.g., CO2, H+) (see Chapter 8). to the myocardium occurs during diastole. The

Klabunde_Chap07.indd 158 6/10/2011 10:57:46 PM


CHAPTER 7 • ORGAN BLOOD FLOW 159

Diastole Systole Diastole


120
Aortic
Pressure
(mm Hg)
80
200

Coronary
Flow
(ml/min/100g)

0
0 1.0
Time (sec)
■ FIGURE 7.7 Pulsatile nature of coronary blood flow measured in the left coronary artery. Flow is lower
during systole because of mechanical compression of intramuscular coronary vessels. Flow is maximal
early in diastole as the heart is relaxing, and then it falls as aortic pressure declines.

reason that coronary flow is influenced by the This is not a problem when the coronary
cardiac cycle is that during systole, the contrac- arteries are normal, because they dilate with
tion of the myocardium compresses the micro- increased heart rate and metabolism; however,
vasculature within the ventricular wall, thereby if the coronaries are diseased and their vasodi-
increasing resistance and decreasing flow. Dur- lator reserve is limited, increases in heart rate
ing systole, blood flow is reduced to the great- can limit coronary flow and lead to myocardial
est extent within the innermost regions of the ischemia and anginal pain.
ventricular wall (i.e., in the subendocardium) The mechanical forces affecting coronary
because this is where the compressive forces flow are greatest within the left ventricle
are greatest. (This results in the subendocar- because this chamber develops pressures that
dial regions being more susceptible to ischemic are severalfold greater than those developed
injury when coronary artery disease or reduced by the right ventricle (see Chapter 4). The
aortic pressure is present.) As the ventricle right ventricle and, to a lesser extent, the atria
begins to relax in early diastole, the compres- show some effects of contraction and relaxa-
sive forces are removed and blood flow is per- tion on blood flow within their musculature,
mitted to increase. Blood flow reaches a peak but it is much less apparent than that observed
in early diastole and then falls passively as the in the left ventricle.
aortic pressure falls toward its diastolic value. Mean coronary blood flow (averaged over
Therefore, it is the aortic pressure during dias- several cardiac cycles) can range from 80 mL/
tole that is most crucial for perfusing the coro- min per 100 g of tissue at resting heart rates
naries. This explains why increases in heart rate to over 400 mL/min per 100 g during exercise
can reduce coronary perfusion. At high heart (see Table 7-1). Therefore, the coronary vas-
rates, the length of diastole is greatly shortened, culature normally has a relatively high vasodi-
which reduces the time for coronary perfusion. lator reserve capacity.

Klabunde_Chap07.indd 159 6/10/2011 10:57:47 PM


160 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Coronary blood flow is primarily regulated oxygen consumption of the beating heart
by changes in tissue metabolism. Adenosine (see Chapter 4) and the fact that the heart
has been shown to be important in dilating relies on oxidative metabolism (see Chapter
the coronary vessels when the myocardium 3), coronary blood flow (oxygen delivery) and
becomes hypoxic or when cardiac metabolism the metabolic activity of the heart need to be
increases during increased cardiac work. tightly coupled. This is all the more important
Experimental studies have shown that inhib- because, as discussed in Chapter 4, the beat-
iting adenosine formation, enhancing its ing heart extracts more than half of the oxy-
breakdown to inosine, or blocking vascular gen from the arterial blood; therefore, there
adenosine receptors impairs coronary vaso- is relatively little oxygen extraction reserve.
dilation under these conditions. In addition, In coronary artery disease, chronic narrowing
nitric oxide has been shown to be important of the vessels or impaired vascular function
in coronary vessels, particularly in producing reduces maximal coronary blood flow (i.e.,
flow-dependent vasodilation. Finally, there is there is reduced vasodilator reserve). When
also some evidence that prostaglandins play a this occurs, coronary flow fails to increase
role in regulating coronary blood flow. adequately as myocardial oxygen demands
Coronary vessels are innervated by both increase (Fig. 7.8). This leads to cardiac
sympathetic and parasympathetic nerves. hypoxia and impaired contractile function.
Unlike most other vascular beds, activation The relationship between coronary blood
of sympathetic nerves to the heart causes flow and the metabolic demand of the heart is
only transient coronary vasoconstriction often discussed in terms of the myocardial oxy-
(α-adrenoceptor mediated) followed by vaso- gen supply/demand ratio. The oxygen supply
dilation. The vasodilation occurs because is the amount of oxygen delivered per minute
sympathetic activation of the heart also to the myocardium in the arterial blood (mL
increases heart rate and inotropy through O2/min), which is the product of the coronary
β-adrenoceptors, which leads to enhanced blood flow (mL blood/min) and arterial oxy-
production of vasodilator metabolites that gen content (mL O2/mL blood). The oxygen
inhibit the vasoconstrictor response and cause demand of the heart is the myocardial oxygen
vasodilation. This is termed functional sym- consumption, which is the product of coro-
patholysis. If β-adrenoceptors are blocked nary blood flow and the difference between
experimentally, sympathetic stimulation of the arterial and venous oxygen contents
the heart causes coronary vasoconstriction.
Parasympathetic stimulation of the heart (i.e.,
Normal
vagal nerve activation) elicits modest coro- Coronaries
nary vasodilation owing to the direct effects of
Oxygen Deficit
released acetylcholine on the coronaries. How-
Blood
ever, if parasympathetic activation of the heart
Flow
results in a significant decrease in myocardial
oxygen demand, local metabolic mechanisms Diseased
increase coronary vascular tone (i.e., cause Coronaries
vasoconstriction). Therefore, parasympathetic
activation of the heart generally results in a Myocardial Oxygen Consumption
decrease in coronary blood flow, although the
■ FIGURE 7.8 Relationship between coronary
direct effect of parasympathetic stimulation of
blood flow and myocardial oxygen consumption.
the coronary vessels is vasodilation. Coronary blood flow increases as myocardial
oxygen consumption increases. However, if the
coronary vessels are diseased and have increased
INSUFFICIENT CORONARY BLOOD FLOW resistance owing to stenosis (red line), blood flow
(and therefore oxygen delivery) will be limited at
Coronary blood flow is crucial for the nor- higher oxygen consumptions, leading to an oxy-
mal function of the heart. Because of the high gen deficit and myocardial hypoxia.

Klabunde_Chap07.indd 160 6/10/2011 10:57:47 PM


CHAPTER 7 • ORGAN BLOOD FLOW 161

(see Equation 4-3). A decrease in the oxygen the process of angiogenesis, which causes
supply/demand ratio causes tissue hypoxia, new blood vessels to form. Collateralization
which can result in chest pain (angina pecto- increases myocardial blood supply by increas-
ris). This can occur by a decrease in oxygen ing the number of parallel vessels, thereby
supply (decreased coronary blood flow or arte- reducing vascular resistance within the myo-
rial oxygen content), an increase in myocardial cardium. This helps to supply blood flow to
oxygen consumption, or a combination of the ischemic regions caused by vascular stenosis or
two. One of the therapeutic goals for people thrombosis.
who have coronary artery disease and anginal
pain is to increase the oxygen supply/demand CASE 7-1
ratio either by improving coronary flow (e.g., A patient with known coronary artery
coronary bypass grafts or coronary stent place- disease (stenosis of multiple vessels) is
ment) or by decreasing myocardial oxygen also hypertensive. Explain why blood
consumption by reducing heart rate, inotropy; pressure-lowering drugs that produce
preload, and afterload (see Chapter 4). reflex tachycardia should be not be used
Both structural and functional changes in such a patient.
occur when coronary arteries become dis-
eased. Atherosclerotic processes decrease the
lumen diameter, causing stenosis. This com- Cerebral Circulation
monly occurs in the large epicardial arteries,
although the disease also afflicts small vessels. The brain is a highly oxidative organ that
The large coronary arteries ordinarily repre- consumes almost 20% of resting total-body
sent only a very small fraction of total coro- oxygen consumption. To deliver adequate
nary vascular resistance. Therefore, stenosis oxygen, the cerebral blood flow needs to be
in these vessels needs to exceed a 60% to 70% relatively high, about 50 to 60 mUmin per
reduction in lumen diameter (i.e., exceed the 100 g tissue weight (see Table 7-1). Although
critical stenosis) to have significant effects on the brain represents only about 2% of body
resting blood flow and maximal flow capacity weight, it receives approximately 14% of the
(see Chapter 5) . cardiac output.
In addition to narrowing the lumen and
MAJOR ARTERIES SUPPLYING THE BRAIN
increasing resistance to flow, atherosclero-
sis causes endothelial damage and dysfunc- The brain circulation is supplied by four prin-
tion. This leads to reduced nitric oxide and cipal arteries: the left and right carotid arter-
prostacyclin formation, which can precipitate ies and the left and right vertebral arteries
coronary vasospasm and thrombus formation, (Fig. 7.9). The vertebral arteries join together
leading to increased vascular resistance and on the ventral surface of the pons to form
decreased flow. Loss of these endothelial fac- the basilar artery, which then travels up the
tors impairs vasodilation, which decreases the brainstem to join the carotid arteries through
vasodilator reserve capacity. When coronary interconnecting arteries, forming the Circle
flow is compromised by coronary artery dis- of Willis. Arterial vessels originating from
ease either at rest or during times of increased the vertebral and basilar arteries as well as the
metabolic demand (e.g., during exercise), Circle of Willis distribute blood flow to differ-
the myocardium becomes hypoxic, which ent regions of the brain. This interconnecting
can impair mechanical function, precipitate network of arterial vessels at the brainstem
arrhythmias, and produce angina. provides a safety mechanism for cerebral perfu-
When coronary oxygen delivery is lim- sion. If, for example, a carotid artery becomes
ited by disease, collateral vessels can play an partly occluded and flow is reduced through
important adjunct role in supplying oxygen to that artery; increased flow through the other
the heart. Conditions of chronic stress (e.g., interconnecting arteries can help improve per-
chronic hypoxia or exercise training) stimulate fusion of the affected portion of the brain.

Klabunde_Chap07.indd 161 6/10/2011 10:57:47 PM


162 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

however, because the cerebral circulation is


located within a rigid cranium, changes in
R. Internal L. Internal intracranial pressure (ICP) can have signifi-
Carotid Circle Carotid cant effects on cerebral perfusion (Fig. 7.10).
of
Willis ICP is the pressure found in the fluid-filled
space between the rigid cranium and the brain
tissue. For example, cerebral vascular hemor-
rhage, brain edema caused by cerebral trauma,
or tumor growth can increase ICP, which can
lead to vascular compression and reduced cer-
ebral blood flow. The venous vessels are most
Basilar susceptible to compression because of their
low intravascular pressure, and thinner, com-
pliant walls. Because ICP is normally greater
than the venous pressure outside the cranium
and the venous vessels can easily collapse,
the effective perfusion pressure of the brain is
not the mean arterial pressure (MAP) minus
Vertebral central venous pressure, but rather the MAP
■ FIGURE 7.9 Major cerebral arteries perfusing the minus the ICP. ICP normally ranges from 0
brain. This view is of the ventral surface of the brain to 10 mm Hg; however, if it becomes elevated
and brainstem. The carotid and vertebral arteries
(e.g., 20 mm Hg or greater), and especially if
are the major source of cerebral blood flow and
are interconnected through the Circle of Willis and there is systemic hypotension, the effective
basilar artery. Smaller branches from these vessels cerebral perfusion pressure (CPP) and blood
perfuse different brain regions. L, left; R, right. flow can be significantly reduced.
Like the coronary circulation, the cer-
REGULATION OF CEREBRAL BLOOD FLOW
ebral blood flow is tightly coupled to oxy-
Like most other organs, cerebral blood flow is gen consumption. Therefore, cerebral blood
determined by its perfusion pressure (arterial− flow increases (active or functional hyper-
venous pressure) and its vascular resistance; emia) when neuronal activity and oxygen

Rigid Cranium ICP increased by:


• intracranial bleeding
Artery
MAP • cerebral edema
• tumor
 ICP
CVP Increased ICP:
Vein • collapses veins
• decreases effective CPP
CPP = MAP – ICP reduces blood flow
■ FIGURE 7.10 Effects of intracranial pressure (ICP) on cerebral blood flow. ICP is the pressure within the
rigid cranium (gray area of figure). Increased ICP decreases transmural pressure (inside minus outside
pressure) of blood vessels (particularly veins), which can cause vascular collapse, increased resistance,
and decreased blood flow. Therefore, the effective cerebral perfusion pressure (CPP) is mean arterial pres-
sure (MAP) minus ICP. CVP, central venous pressure.

Klabunde_Chap07.indd 162 6/10/2011 10:57:48 PM


CHAPTER 7 • ORGAN BLOOD FLOW 163

consumption are increased. Changes in tissue metabolism and blood flow. Increased
neuronal activity in specific brain regions lead oxidative metabolism increases carbon diox-
to increases in blood flow to those regions. ide production, which causes vasodilation. It
The brain shows excellent autoregulation is thought that the carbon dioxide diffuses into
between MAPs of about 60 and 130 mm Hg the cerebrospinal fluid, where hydrogen ion is
(Fig. 7.11). This is important because cerebral formed by the action of carbonic anhydrase;
function relies on a steady supply of oxygen the hydrogen ion then causes vasodilation. In
and cannot afford to be subjected to a reduc- addition, carbon dioxide and hydrogen ion
tion in flow caused by a fall in arterial pres- increase when perfusion is reduced because
sure. If MAP falls below 60 mm Hg, cerebral of impaired washout of carbon dioxide. Aden-
perfusion becomes impaired, which results in osine, nitric oxide, potassium ion, and myo-
depressed neuronal function, mental confu- genic mechanisms have also been implicated
sion, and loss of consciousness. When arte- in the local regulation of cerebral blood flow.
rial pressure is above the autoregulatory range Cerebral blood flow is strongly influenced
(e.g., in a hypertensive crisis), blood flow and by the partial pressure of carbon dioxide and,
pressures within the cerebral microcirculation to a lesser extent, oxygen in the arterial blood
increase. This may cause endothelial and vas- (Fig. 7.12). Cerebral blood flow is highly sen-
cular damage, disruption of the blood–brain sitive to small changes in arterial partial pres-
barrier, and hemorrhagic stroke. With chronic sure of CO2 (PCO2) from its normal value of
hypertension, the autoregulatory curve shifts about 40 mm Hg, with increased PCO2 (hyper-
to the right (see Fig. 7.11), which helps to pro- capnia) causing pronounced vasodilation and
tect the brain at higher arterial pressures. How- decreased PCO2 (hypocapnia) causing vaso-
ever, this rightward shift then makes the brain constriction. Hydrogen ion appears to be
more susceptible to reduced perfusion when responsible for the changes in vascular resist-
arterial pressure falls below the lower end of ance when changes occur in arterial PCO2.
the rightward-shifted autoregulatory range. The importance of CO2 in regulating cerebral
Local metabolic mechanisms play a domi- blood flow can be demonstrated when a per-
nant role in the control of cerebral blood flow. son hyperventilates, which decreases arterial
Considerable evidence indicates that changes PCO2. When this occurs, a person becomes
in carbon dioxide are important for coupling “light headed” as the reduced PCO2 causes
cerebral blood flow to decrease. Severe arte-
rial hypoxia (hypoxemia) increases cerebral
blood flow. Arterial PO2 is normally about 95
to 100 mm Hg. If the PO2 falls below 50 mm
Blood Flow

Normal
Hg (severe arterial hypoxia), it elicits a strong
vasodilator response in the brain, which
helps to maintain oxygen delivery despite
the reduction in arterial oxygen content. As
Chronic Hypertension described in Chapter 6, decreased arterial
Acute Sympathetic Stimulation PO2 and increased PCO2 stimulate chemore-
ceptors, which activate sympathetic efferents
to the systemic vasculature to cause vaso-
0 100 200 constriction; however, the direct effects of
Perfusion Pressure (mm Hg) hypoxia and hypercapnia override the weak
■ FIGURE 7.11 Autoregulation of cerebral blood effects of sympathetic activation in the brain
flow. Cerebral blood flow shows excellent autoreg- so that cerebral vasodilation occurs and oxy-
ulation between MAPs of 60 and 130 mm Hg. gen delivery is enhanced.
The autoregulatory curve shifts to the right with
Although sympathetic nerves innervate
chronic hypertension or acute sympathetic activa-
tion. This shift helps to protect the brain from the larger cerebral vessels, activation of these nerves
damaging effects of elevated pressure. has relatively little influence on cerebral blood

Klabunde_Chap07.indd 163 6/10/2011 10:57:48 PM


164 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

100

Cerebral Blood Flow


PO2 PCO2
(ml/min/100g)
50
Normal Arterial Values
PO2 @ 95 mm Hg
PCO2 @ 40 mm Hg
0
0 50 100
Arterial Blood Partial Pressure
(mm Hg)
■ FIGURE 7.12 Effects of arterial partial pressure of oxygen and carbon dioxide on cerebral blood flow. An
arterial partial pressure of oxygen (PO2) of <50 mm Hg (normal value is about 95 mm Hg) causes cerebral
vasodilation and increased flow. A reduction in arterial partial pressure of carbon dioxide (PCO2) below
its normal value of 40 mm Hg decreases flow, whereas PCO2 values >40 mm Hg increase flow. Therefore,
cerebral blood flow is more sensitive to changes from normal arterial PCO2 values than from normal arte-
rial PO2 values.

flow. Maximal sympathetic activation increases acetylcholine, produce localized vasodila-


cerebral vascular resistance by no more than tion. Other nerves appear to release the local
20% to 30%, in contrast to an approximately vasodilators calcitonin gene-related peptide
500% increase occurring in skeletal muscle. (CGRP) and substance P. Sympathetic adr-
The reason, in part, for the weak sympathetic energic nerves can release neuropeptide-Y
response by the cerebral vasculature is that (NPY) in addition to norepinephrine, which
metabolic mechanisms are dominant in regu- causes localized vasoconstriction. Vascular
lating flow; therefore, functional sympatholy- and neuronal sources of endothelin-1 can also
sis occurs during sympathetic activation. This produce vasoconstriction within the brain.
is crucial to preserve normal brain function;
otherwise, every time a person stands up or Skeletal Muscle Circulation
exercises, both of which cause sympathetic
activation, cerebral perfusion would decrease. The primary function of skeletal muscle is to
Therefore, baroreceptor reflexes have little contract and generate mechanical forces to
influence on cerebral blood flow. Sympathetic provide support to the skeleton and produce
activation shifts the autoregulatory curve to movement of joints. This mechanical activ-
the right, similar to what occurs with chronic ity consumes large amounts of energy and
hypertension. therefore requires delivery of considerable
In recent years, we have learned that neu- amounts of oxygen and substrates, as well
ropeptides originating in the brain signifi- as the efficient removal of metabolic waste
cantly influence cerebral vascular tone, and products. Both oxygen delivery and metabolic
they may be involved in producing head- waste removal functions are performed by the
aches (e.g., migraine and cluster headaches) circulation.
and cerebral vascular vasospasm during
MICROVASCULAR ORGANIZATION
strokes. Parasympathetic cholinergic fibers
IN SKELETAL MUSCLE
innervating the cerebral vasculature release
nitric oxide and vasoactive intestinal poly- The circulation within skeletal muscle is
peptide (VIP). These substances, along with highly organized (Fig. 7.13). Arterioles give

Klabunde_Chap07.indd 164 6/10/2011 10:57:48 PM


CHAPTER 7 • ORGAN BLOOD FLOW 165

Arteriole

Arteriole
Capillaries Muscle Fiber
(C/F ratio = 2-3)
(20-40m)

■ FIGURE 7.13 Microvascular organization in skeletal muscle. Long, parallel muscle fibers are each sur-
rounded by multiple, parallel capillaries that arise from arterioles. As shown in the cross section, there are
typically 2 to 3 capillaries per muscle fiber (C/F ratio), although that varies depending on the muscle type.
Arrows represent direction of flow.

rise to capillaries that generally run parallel be perfused, which increases the number of
to the muscle fibers. Because a given region of flowing capillaries around each muscle fiber
muscle may be served by multiple arterioles, (termed capillary recruitment). This anatom-
the direction of flow in some capillaries may ical arrangement of capillaries and the abil-
be opposite to the direction of flow in nearby ity to recruit capillaries decreases diffusion
capillaries. Each muscle fiber, which can be distances, leading to an efficient exchange of
20 to 40 µm in diameter, is surrounded by gases and molecules between the blood and
three to four capillaries. Because more than the myocytes, particularly under conditions
one muscle fiber may share an adjacent capil- of high oxygen demand.
lary, the overall capillary-to-fiber ratio is 2 to
MUSCLE BLOOD FLOW AT REST
3, depending on the type of muscle. Muscle
AND DURING CONTRACTION
fibers that have a high oxidative capacity gen-
erally have a higher capillary-to-fiber ratio In resting humans, almost 20% of cardiac out-
than fibers that have a low oxidative capac- put is delivered to skeletal muscle. This large
ity, but a high anaerobic (glycolytic) capacity. cardiac output to muscle occurs not because
Muscles with a higher oxidative capacity and blood flow is exceptionally high in resting
a greater number of capillaries generally have muscle, but because skeletal muscle makes up
a higher maximal flow capacity. about 40% of the body mass. In the resting,
When the muscle is not contracting, rela- noncontracting state, muscle blood flow is about
tively little oxygen is required and only about 3 mL/min per 100 g. This resting flow is much
one-fourth of the capillaries are perfused. In less than that found in organs such as the brain
contrast, during muscle contraction and active and kidneys, in which “resting” flows are about
hyperemia, all the anatomical capillaries may 55 and 400 mL/min per 100 g, respectively.

Klabunde_Chap07.indd 165 6/10/2011 10:57:49 PM


166 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

When muscles contract during exercise, REGULATION OF SKELETAL MUSCLE


blood flow can increase more than 20-fold. BLOOD FLOW
If muscle contraction is occurring during The precise mechanisms responsible for dilat-
whole-body exercise (e.g., running), more ing skeletal muscle vasculature during contrac-
than 80% of cardiac output can be directed to tion are not clearly understood, although many
the contracting muscles. Therefore, skeletal potential vasodilator candidates have been
muscle has a very large flow reserve (or capac- identified. These include increases in intersti-
ity) relative to its blood flow at rest, indicat- tial K+ during muscle contraction, formation of
ing that the vasculature in resting muscle has adenosine (particularly during ischemic con-
a high degree of tone (see Table 7-1). This tractions), increased H+ production, endothe-
resting tone is brought about by the interplay lial and skeletal muscle-derived nitric oxide
between vasoconstrictor (e.g., sympathetic and prostaglandins, and ATP release from red
adrenergic and myogenic influences) and vas- blood cells. Other candidates, although less
odilator influences (e.g., nitric oxide produc- likely, are CO2, increased interstitial and blood
tion, and tissue metabolites). In the resting osmolarity, and inorganic phosphate. It is very
state, the vasoconstrictor influences domi- likely that multiple factors play a role and at
nate, whereas during muscle contraction, different times during the flow response to
vasodilator influences dominate to increase muscle contraction. A nonchemical mecha-
oxygen delivery to the contracting muscle fib- nism that is very important in facilitating
ers and remove metabolic waste products that blood flow during coordinated contractions
accumulate. Vasodilation of resistance vessels, of groups of muscles (as occurs during nor-
particularly the small terminal arterioles, not mal physical activity such as running) is the
only increases muscle blood flow, but also skeletal muscle pump (see Chapter 5). Regard-
increases the number of flowing capillaries. In less of the mechanisms involved in producing
the past, some have hypothesized that muscle active hyperemia, the outcome is that there
capillary recruitment results from relaxation is a close correlation between the increase in
of precapillary sphincters; however, there is oxygen consumption and the increase in blood
little or no direct evidence for their existence. flow during muscle contraction.
Instead, capillary recruitment appears to be a Skeletal muscle vasculature is innervated
consequence of altered distribution of micro- primarily by sympathetic adrenergic fib-
vascular pressures brought about by arteriolar ers. The norepinephrine released by these
dilation. fibers binds to α-adrenoceptors and causes
The blood flow response to skeletal muscle vasoconstriction. Under resting conditions,
contraction depends on the type of contrac- a significant portion of the vascular tone is
tion. With rhythmic or phasic contraction of generated by sympathetic activity, so that if
muscle (Fig. 7.14, top panel), as occurs during a resting muscle is suddenly denervated or
normal locomotory activity, mean blood flow the α-adrenoceptors are blocked pharma-
increases during the period of muscle activity. cologically by a drug such as phentolamine,
However, if blood flow is measured without blood flow will transiently increase two- to
filtering or averaging the flow signal, the flow threefold until local regulatory mechanisms
is found to be phasic—flow decreases dur- reestablish a new steady-state flow. Activation
ing contraction and increases during relaxa- of the sympathetic adrenergic nervous sys-
tion phases of the muscle activity because of tem (e.g., baroreceptor reflex in response to
mechanical compression of the vessels. In hypovolemia) can dramatically reduce blood
contrast, a sustained muscle contraction (e.g., flow in resting muscle. When this reduction
lifting and holding a heavy weight) decreases in blood flow occurs, the muscle extracts
mean blood flow during the period of contrac- more oxygen (the arterial–venous oxygen
tion, followed by a postcontraction hyperemic difference increases) and activates anaerobic
response when the contraction ceases (see pathways for ATP production. However,
Fig. 7.14, bottom panel).

Klabunde_Chap07.indd 166 6/10/2011 10:57:49 PM


CHAPTER 7 • ORGAN BLOOD FLOW 167

Phasic
Contractions
Muscle Blood Flow

0 20 40 60 80
Time (sec)

Sustained
Contraction
Muscle Blood Flow

0 10 20 30 40
Time (sec)
■ FIGURE 7.14 Skeletal muscle active hyperemia following phasic and sustained (tetanic) contractions.
The top panel shows that phasic contractions cause flow to decrease during contraction and increase dur-
ing relaxation, although the net effect is an increase in flow during contraction. When contractions cease,
a further increase in flow occurs because mechanical compression of the vasculature is removed. The
bottom panel shows that sustained, tetanic contractions generate high intramuscular forces that com-
press the vasculature and reduce flow. When contraction ceases, a large hyperemia follows.

prolonged hypoperfusion of muscle caused cholinergic innervation of skeletal muscle


by intense sympathetic activation eventually resistance vessels. The neurotransmitter for
leads to vasodilator mechanisms dominating these fibers is acetylcholine, which binds to
over the sympathetic vasoconstriction, lead- muscarinic receptors to produce vasodilation.
ing to sympathetic escape and partial restora- This branch of the autonomic nervous sys-
tion of blood flow. tem has little or no influence on blood flow
Evidence exists, at least in nonprimate spe- under resting conditions; however, activation
cies such as cats and dogs, for sympathetic of these fibers in anticipation of exercise and

Klabunde_Chap07.indd 167 6/10/2011 10:57:49 PM


168 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

during exercise can contribute to the increase blood in the venous plexus is also responsi-
in blood flow associated with exercise. There ble for skin coloration in lightly pigmented
is no convincing evidence, however, for simi- individuals. In the skin of the nose, lips, ears,
lar active, neurogenic vasodilator mechanisms palms, toes, sole of the feet, and fingers (espe-
existing in humans. cially the fingertips), blood flows directly to the
venous plexus from the small subcutaneous
Cutaneous Circulation arteries through special interconnecting vessels
called arteriovenous (AV) anastomoses.
The nutrient and oxygen requirements of the The resistance vessels supplying the subepi-
skin are quite low relative to other organs; dermal capillary loops and the AV anastomoses
therefore, cutaneous blood flow does not pri- are richly innervated by sympathetic adrener-
marily serve a metabolic support role. Instead, gic fibers. Constriction of these vessels during
the primary role of blood flow to the skin is to sympathetic activation decreases blood flow
allow heat to be exchanged between the blood through the capillary loops and the venous
and the environment to help regulate body tem- plexus. Although the AV anastomoses are
perature. Therefore, the cutaneous circulation almost exclusively controlled by sympathetic
is primarily under the control of hypothalamic influences, the resistance vessels respond to
thermoregulatory centers that adjust the sym- both metabolic influences and sympathetic
pathetic outflow to the cutaneous vasculature. influences and therefore demonstrate local reg-
ulatory phenomena such as reactive hyperemia
MICROVASCULAR ORGANIZATION
OF THE SKIN
and autoregulation. These local regulatory
responses, however, are relatively weak com-
The microvascular network that supplies skin pared to those observed in most other organs.
is unique among organs and varies depending
on the type of skin. Small arteries arising from
EFFECTS OF TEMPERATURE ON SKIN
the subcutaneous tissues give rise to arterioles BLOOD FLOW
that penetrate into the dermis and give rise to
capillaries that loop underneath the epidermis At normal body and ambient temperatures, sym-
(Fig. 7.15). Blood flows from these capillary pathetic adrenergic activity contributes to a high
loops into venules and then into an extensive, degree of vascular tone, and skin blood flow
interconnecting venous plexus, in which most represents about 4% of the cardiac output (see
of the cutaneous blood volume is found. The Table 7-1). In times of severe cold stress, skin

Epidermis

Capillary
Dermis

Vein
Venous Plexus
Subcutaneous AV anastomosis
Tissue
Artery
■ FIGURE 7.15 Microvascular anatomy of the cutaneous circulation. Arteries within the subcutaneous tis-
sue give rise to either arterioles that travel into the dermis and give rise to capillary loops or AV anasto-
moses that connect to a plexus of small veins in the subdermis. The venous plexus also receives blood
from the capillary loops. Sympathetic stimulation constricts the resistance vessels and AV anastomoses,
thereby decreasing dermal blood flow.

Klabunde_Chap07.indd 168 6/10/2011 10:57:49 PM


CHAPTER 7 • ORGAN BLOOD FLOW 169

blood flow may be reduced to <1% of cardiac exposure, alternating periods of dilation and
ouput, and during severe heat stress, skin blood constriction may occur (“hunting response”).
flow can approach 60% of the cardiac output. The mechanism for cold-induced vasodilation
If core temperature decreases, heat reten- is not clear, but it probably involves changes in
tion mechanisms are activated by the hypo- local control of blood vessels.
thalamus, leading to increased sympathetic
adrenergic outflow to the skin. This decreases VASCULAR RESPONSES TO TISSUE INJURY
cutaneous blood flow and reduces heat loss to Tissue injury from mechanical trauma, heat, or
the environment. If core temperature begins to chemicals releases paracrine substances such
rise (e.g., during physical exertion), heat loss as histamine and bradykinin, which increase
mechanisms are activated by the hypothala- blood flow and cause localized edema by
mus, which decrease sympathetic adrenergic increasing microvascular permeability. If the
outflow to the skin. This reduces vasocon- skin is firmly stroked with a blunt object, the
strictor tone, thereby causing cutaneous skin initially blanches owing to localized vaso-
vasodilation and increased blood flow. Vaso- constriction. This is followed within a minute
dilation resulting from withdrawal of sympa- by the formation of a red line that spreads away
thetic vasoconstrictor influences is referred from the site of injury (red flare); both the red
to as “passive vasodilation.” If core tempera- line and red flare are caused by an increase in
ture continues to rise, then “active vasodila- blood flow. Localized swelling (wheal forma-
tion” results from sympathetic cholinergic tion) may then follow, caused by increased
nerve activation and the neuronal co-release microvascular permeability and leakage of fluid
of vasodilator substances such as vasoactive into the interstitium. The red line, flare, and
intestinal polypeptide (VIP). There is also evi- wheal are called the triple response. Both par-
dence that substance P, histamine, prostaglan- acrine hormones and local axon reflexes are
dins, and nitric oxide may contribute to active believed to be involved in the triple response.
vasodilation. Vasodilation enables more warm The vasodilator neurotransmitter involved in
blood to circulate in the subepidermal layer of local axon reflexes has not been identified.
the skin so that more heat can be transferred
to the environment.
Local changes in skin temperature selec- Splanchnic Circulation
tively alter blood flow to the affected region. The splanchnic circulation includes blood
For example, if a heat source is placed on a flow to the gastrointestinal tract, spleen, pan-
small region of the skin on the back of the hand, creas, and liver. Blood flow to these combined
blood flow will increase only to the region that organs represents 20% to 25% of cardiac out-
is heated. This response appears to be medi- put (see Table 7-1). Three major arteries aris-
ated by local axon reflexes and local formation ing from the abdominal aorta supply blood to
of nitric oxide instead of by changes in sympa- the stomach, intestine, spleen, and liver—the
thetic discharge mediated by the hypothalamic celiac, superior mesenteric, and inferior mes-
thermoregulatory regions. Localized cool- enteric arteries. The following focuses on
ing produces vasoconstriction through local blood flow to the intestines and liver.
mechanisms that involve sympathetic adrener-
gic nerves and locally stimulated norepineph-
INTESTINAL CIRCULATION
rine release. If tissue is exposed to extreme
cold, a phenomenon called cold-induced vaso- Several branches arising from the supe-
dilation may occur following an initial vaso- rior mesenteric artery supply blood to the
constrictor response, especially if the exposed intestine. These and subsequent branches
body region is a hand, foot, or face. This phe- travel through the mesentery that supports
nomenon causes light-colored skin to appear the intestine. Small arterial branches enter
red, and it explains the rosy cheeks, ears, and the outer muscular wall of the intestine and
nose a person may exhibit when exposed to divide into several smaller orders of arteries
very cold air temperatures. With continued and arterioles, most of which enter into the

Klabunde_Chap07.indd 169 6/10/2011 10:57:49 PM


170 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

submucosa from which arterioles and capil- HEPATIC CIRCULATION


laries arise to supply blood to the intestinal
Venous blood leaving the gastrointestinal tract,
villi. Water and nutrients transported from the
spleen, and pancreas drains into the hepatic
intestinal lumen into the villi enter the blood
portal vein, which supplies approximately 75%
and are carried away by the portal venous cir-
of the hepatic blood flow. The remainder of the
culation.
hepatic blood flow is supplied by the hepatic
Intestinal blood flow is closely coupled to
artery, which is a branch of the celiac artery.
the primary function of the intestine, which
Note that in this arrangement, most of the liver
is the absorption of water, electrolytes, and
circulation is in series with the gastrointestinal,
nutrients from the intestinal lumen. There-
splenic, and pancreatic circulations. Therefore,
fore, intestinal blood flow increases when food
changes in blood flow in these vascular beds
is present within the intestine. In an adult
have a significant influence on hepatic flow.
human, blood flow to the intestine (superior
Terminal vessels from the hepatic portal
mesenteric artery) in the fasted state is about
vein and hepatic artery form sinusoids within
300 mL/min, and increases two- to threefold
the liver, which function as capillaries. The
following a meal. This functional (absorptive)
pressure within these sinusoids is very low,
hyperemia is stimulated by gastrointestinal
just a few mm Hg above central venous pres-
hormones such as gastrin and cholecysto-
sure. This is important because hepatic sinu-
kinin, as well as by glucose, amino acids, and
soids are very permeable (see Chapter 8).
fatty acids that are absorbed by the intestine.
Changes in central venous and hepatic venous
Evidence exists that submucosal arteriolar
pressure are almost completely transmitted to
vasodilation during functional hyperemia is
the sinusoids. Therefore, elevations in central
mediated by hyperosmolarity and nitric oxide.
venous pressure during right ventricular fail-
The intestinal circulation is strongly influ-
ure can cause substantial increases in sinusoid
enced by the activity of sympathetic adren-
pressure and fluid filtration, leading to hepatic
ergic nerves. Increased sympathetic activity
edema and accumulation of fluid within the
during exercise or in response to decreased
abdominal cavity (ascites).
baroreceptor firing (e.g., during hemorrhage
The liver circulation does not show classical
or standing) constricts both arterial resist-
autoregulation; however, decreases in hepatic
ance vessels and venous capacitance vessels.
portal flow result in reciprocal increases in
Because the intestinal circulation receives such
hepatic artery flow, and vice versa. Sympathetic
a large fraction of cardiac output, sympathetic
nerve activation constricts vessels derived from
stimulation of the intestine causes a substan-
both the hepatic portal system and hepatic
tial increase in total systemic vascular resist-
artery. The most important effect of sympathetic
ance. Additionally, the large blood volume
activation is on venous capacitance vessels,
contained within the venous vasculature is
which contain a significant fraction (∼15%) of
mobilized during sympathetic stimulation to
the venous blood volume in the body. The liver,
increase central venous pressure. The spleen
like the gastrointestinal circulation, functions
is also an important venous reservoir, and in
as an important venous reservoir.
some species (e.g., dogs), this organ stores
hemoconcentrated blood. Stressful conditions
Renal Circulation
in the dog (e.g., blood loss) can cause splenic
contraction, which can substantially increase Approximately 20% of the cardiac output
circulating blood volume and hematocrit. perfuses the kidneys although the kidneys
Parasympathetic activation of the intestine represent only about 0.4% of total body
increases motility and glandular secretions, weight. Renal blood flow, therefore, is about
which is associated with an increase in blood 400 mL/min per 100 g of tissue weight, which
flow. This may involve metabolic mechanisms is the highest of any major organ within the
or local paracrine influences such as the body (see Table 7-1). Only the pituitary and
formation of bradykinin and nitric oxide. carotid bodies have higher blood flows per

Klabunde_Chap07.indd 170 6/10/2011 10:57:49 PM


CHAPTER 7 • ORGAN BLOOD FLOW 171

unit tissue weight. Whereas blood flow in flow supplies the cortex, with the remainder
many organs is closely coupled to tissue oxi- supplying the medullary regions.
dative metabolism, this is not the case for
the kidneys, in which the blood flow greatly
RENAL VASCULAR ORGANIZATION
exceeds the need for oxygen delivery. The
very high blood flow results in a relatively low The vascular organization within the kidneys is
extraction of oxygen from the blood (about very different from most organs. The abdominal
1 to 2 mL O2/mL blood) despite the fact that aorta gives rise to renal arteries that distribute blood
renal oxygen consumption is high (∼5 mL O2/ flow to each kidney. The renal artery enters the
min per 100 g). The reason for renal blood kidney at the hilum and gives off several branches
flow being so high is that the primary func- (interlobar arteries) that travel in the kidney
tion of the kidneys is to filter blood and form toward the cortex. Subsequent branches (arcu-
urine. The kidney comprises three major ate and interlobular arteries) then form afferent
regions: the cortex (the outer layer that con- arterioles, which supply blood to each glomerulus
tains glomeruli for filtration), the medulla (Fig. 7.16). As the afferent arteriole enters the glo-
(the middle region that contains renal tubules merulus, it gives rise to a cluster of glomerular
and capillaries involved in concentrating the capillaries, from which fluid is filtered into Bow-
urine), and the hilum (the inner region where man capsule and into the renal proximal tubule.
the renal artery and vein, nerves, lymphatics, The glomerular capillaries then form an efferent
and ureter enter or leave the kidney). Because arteriole from which arise peritubular capillaries
most of the filtering takes place within the that surround the renal tubules. Efferent arterioles
cortex, about 90% of the total renal blood associated with juxtamedullary nephrons

Bowman’s
Capsule

Efferent Proximal
Arteriole Tubule

Glomerular
Capillaries

Afferent
Arteriole Peritubular
Capillaries
Interlobular
Artery

ArcuateArtery
■ FIGURE 7.16 Renal microvascular anatomy. Small vessels derived from branches of the renal artery form
arcuate arteries and interlobular arteries, which then become afferent arterioles that supply blood to the
glomerulus. As the afferent arteriole enters the glomerulus, it gives rise to a cluster of glomerular capillaries,
from which fluid is filtered into Bowman capsule and into the renal proximal tubule. The glomerular capillar-
ies then form an efferent arteriole from which arise peritubular capillaries that surround the renal tubules.

Klabunde_Chap07.indd 171 6/10/2011 10:57:49 PM


172 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

located in the inner cortex near the outer medulla in afferent and efferent arteriole resistance
give rise to very long capillaries (vasa recta) that affect not only blood flow, but also the hydro-
loop down deep within the medulla. The capil- static pressures within the glomerular and
laries are involved with countercurrent exchange peritubular capillaries. Glomerular capil-
and the maintenance of medullary osmotic gra- lary pressure, which is about 50 mm Hg, is
dients. Capillaries eventually form venules and much higher than that in capillaries found in
then veins, which join together to exit the kidney other organs. This high pressure drives fluid
as the renal vein. Therefore, within the kidney, a filtration (see Chapter 8). The peritubular
capillary bed (glomerular capillaries) is located capillary pressure, however, is low (about 10
between the two principal sites of resistance to 20 mm Hg). This is important because it
(afferent and efferent arterioles). Furthermore, a permits fluid reabsorption to limit water loss
second capillary bed (peritubular capillaries) is in and urine excretion. About 20% of the plasma
series with the glomerular capillaries and is sepa- entering the kidney is filtered. If significant
rated by the efferent arteriole. reabsorption did not occur, a high rate of
urine formation would rapidly lead to hypov-
RENAL HEMODYNAMICS
olemia and hypotension and an excessive loss
The vascular arrangement within the kid- of electrolytes. Figure 7.17 shows the effects
ney is very important for filtration and of afferent and efferent arteriole dilation and
reabsorption functions of the kidney. Changes constriction on blood flow and glomerular

P P
R*
A F

P P
R*
B F

P P
R*
C F

P P
R*
D F

AA GC EA PC
■ FIGURE 7.17 Effects of renal afferent and efferent arteriole resistances on blood flow and renal capillary
pressures. The following descriptions assume constant aortic pressure. Panel A. Decreased afferent arte-
riole (AA) resistance (R) increases glomerular capillary (GC) and peritubular capillary (PC) pressures (P)
and increases flow (F). Panel B. Increased AA resistance decreases GC and PC pressures and decreases
F. Panel C. Decreased efferent arteriole (EA) resistance decreases GC pressure, increases PC pressure,
and increases F. Panel D. Increased EA resistance increases GC pressure, decreases PC pressure, and
decreases F. *, arteriole undergoing resistance change.

Klabunde_Chap07.indd 172 6/10/2011 10:57:50 PM


CHAPTER 7 • ORGAN BLOOD FLOW 173

capillary pressure. Dilation of the afferent the juxtaglomerular apparatus is a group of


arteriole (panel A) increases distal pressures specialized cells of the distal tubule that lie
(glomerular capillaries, efferent arteriole, and adjacent to the afferent arteriole as the distal
peritubular capillaries), while increasing total tubule loops up back toward the glomerulus.
flow (assuming constant aortic pressure); These cells sense solute osmolarity, particu-
this causes increased glomerular filtration. larly sodium chloride. Some investigators have
If the afferent arteriole constricts (panel B), proposed that adenosine (which is a vasocon-
distal pressures, glomerular filtration, and strictor in the kidney), locally produced angi-
blood flow are reduced. If the efferent arteri- otensin II (a vasoconstrictor), or vasodilators
ole dilates (panel C), this increases total flow such as nitric oxide, PGE2, and prostacyclin
but reduces glomerular capillary pressure and are involved in tubuloglomerular feedback
filtration, while increasing peritubular capil- and autoregulation. Locally produced angio-
lary pressure. Efferent arteriole constriction tensin II strongly influences efferent arteriole
increases glomerular capillary pressure and tone. Thus, inhibition of angiotensin II forma-
glomerular filtration while reducing flow and tion by an ACE inhibitor dilates the efferent
peritubular capillary pressure (panel D). arteriole, which decreases glomerular capil-
lary pressure and reduces glomerular filtration
under some conditions (e.g., renal artery ste-
REGULATION OF RENAL BLOOD FLOW
nosis). Drugs that inhibit prostaglandin and
The renal circulation exhibits strong autoreg- prostacyclin biosynthesis (cyclooxygenase
ulation between arterial pressures of about inhibitors such as aspirin or ibuprofen) alter
80 to 180 mm Hg. Autoregulation of blood renal hemodynamics and may impair renal
flow is accompanied by autoregulation of glo- function, particularly with long-term use.
merular filtration so that filtration remains The renal circulation responds strongly
essentially unchanged over a wide range of to sympathetic adrenergic stimulation.
arterial pressures. For this to occur, glomeru- Under normal conditions, relatively little
lar capillary pressure must remain unchanged sympathetic tone on the renal vasculature
when arterial pressure changes. This takes occurs; however, with strenuous exercise or
place because the principal site for autoregula- in response to severe hemorrhage, increased
tion is the afferent arteriole. If arterial pressure renal sympathetic nerve activity can virtually
falls, the afferent arteriole dilates, which helps shut down renal blood flow. Because renal
to maintain the glomerular capillary pressure blood flow receives a relatively large fraction
and flow despite the fall in arterial pressure. of cardiac output and therefore contributes
Two mechanisms have been proposed significantly to systemic vascular resistance,
to explain renal autoregulation: myogenic renal vasoconstriction can serve an important
mechanisms and tubuloglomerular feedback. role in maintaining arterial pressure under
Myogenic mechanisms were described earlier these conditions; however, intense renal vaso-
in this chapter. Briefly, a reduction in afferent constriction seriously impairs renal perfusion
arteriole pressure is sensed by the vascular and function, and it can lead to renal failure.
smooth muscle, which responds by relaxing;
an increase in pressure induces smooth mus-
Pulmonary Circulation
cle contraction. The tubuloglomerular feed-
back mechanism is poorly understood, and Two separate circulations perfusing respiratory
the actual mediators have not been identified. structures exist: the pulmonary circulation,
It is believed, however, that changes in perfu- which is derived from the pulmonary artery
sion pressure alter glomerular filtration and and supplies blood flow to the alveoli for gas
therefore tubular flow and sodium delivery to exchange, and the bronchial circulation, which
the macula densa of the juxtaglomerular appa- is derived from the thoracic aorta and sup-
ratus, which then signals the afferent arteriole plies nutrient flow to the trachea and bronchial
to constrict or dilate. The macula densa of structures. The pulmonary circulation receives

Klabunde_Chap07.indd 173 6/10/2011 10:57:50 PM


174 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

all of the cardiac output of the right ventricle, Increased pulmonary vascular pressure
whereas the bronchial circulation receives about can have two adverse consequences. First,
1% of the left ventricular output. The following increased pulmonary artery pressure increases
focuses on the pulmonary circulation. the afterload on the right ventricle, which can
The pulmonary circulation is a low- impair ejection, and with chronic pressure
resistance, low-pressure, high-compliance vas- elevation, cause right ventricular failure. Sec-
cular bed. Although the pulmonary circulation ond, an increase in pulmonary capillary pres-
receives the same cardiac output as the sys- sure increases fluid filtration (see Chapter 8),
temic circulation, the pulmonary pressures are which can lead to pulmonary edema. Pulmo-
much lower. The pulmonary artery systolic and nary capillary pressures are ordinarily about
diastolic pressures are about 25 and 10 mm Hg, 10 mm Hg, which is less than half the value
respectively. The mean pulmonary artery pres- found in most other organs, and this low pres-
sure is therefore about 15 mm Hg. If we assume sure is necessary to ensure that excessive fluid
that the left atrial pressure averages 8 mm Hg, filtration from the pulmonary capillaries does
the perfusion pressure for the pulmonary circu- not occur under normal circumstances.
lation (mean pulmonary artery pressure minus Unlike other major organs, the concept of
left atrial pressure) is only about 7 mm Hg. blood flow autoregulation is not applicable to
This is considerably lower than the perfusion the pulmonary circulation because pulmonary
pressure for the systemic circulation (about 90 artery pressure is the dependent variable instead
mm Hg). Because the flow is the same, but the of flow. The reason for this is that the entire pul-
perfusion pressure is much lower in the pul- monary blood flow is determined by the right
monary circulation, the pulmonary vascular ventricular output, and therefore, pulmonary
resistance must be very low. In fact, pulmonary artery pressure changes as a function of this flow
vascular resistance is generally 10- to 15-fold and the pulmonary vascular resistance. Because
lower than systemic vascular resistance. The other organs of the body are in parallel with
reason for the much lower pulmonary vascular each other, changes in left ventricular output
resistance is that the vessels are larger in diam- do not necessarily change blood flow in a given
eter, shorter in length, and have many more organ except through changes in arterial pres-
parallel elements than the systemic circulation. sure that may occur. Therefore, in other organs,
Pulmonary vessels are also much more com- blood flow is the dependent variable because
pliant than systemic vessels. Because of this, an flow depends on perfusion pressure and organ
increase in right ventricular output does not vascular resistance. Instead of autoregulating
cause a proportionate increase in pulmonary blood flow, the pulmonary circulation autoregu-
artery pressure. The reason for this is that the lates pulmonary arterial pressure through pas-
pulmonary vessels passively distend as the pul- sive changes in resistance of highly compliant
monary artery pressure increases, which lowers vessels and through vessel recruitment.
their resistance. Increased pressure also recruits Because of their low pressures and high
additional pulmonary capillaries, which further compliance, pulmonary vascular diameters
reduces resistance. This high vascular compli- are strongly influenced by gravity and by
ance and ability to recruit capillaries are impor- changes in intrapleural pressure during res-
tant mechanisms for preventing pulmonary piration. When a person stands up, gravity
vascular pressures from rising too high when increases hydrostatic pressures within ves-
cardiac output increases (e.g., during exercise). sels located in the lower regions of the lungs,
If there were no change in pulmonary vascular which distends these vessels, decreases resist-
resistance, then increasing cardiac output five- ance, and increases blood flow to the lower
fold during exercise would cause mean pulmo- regions. In contrast, vessels located in the
nary artery pressure to increase from 15 to 43 upper regions of the lungs have reduced
mm Hg (assuming left atrial pressure remains intravascular pressures; this increases resist-
at 8 mm Hg), and the pulmonary artery systolic ance and reduces blood flow when a person
pressure would be even higher. is standing. Changes in intrapleural pressure

Klabunde_Chap07.indd 174 6/10/2011 10:57:50 PM


CHAPTER 7 • ORGAN BLOOD FLOW 175

during respiration (see Chapter 5) alter the to regional variations in ventilation, helps
transmural pressure that distends the vessels. to maintain normal ventilation–perfusion
For example, during normal inspiration, the ratios in the lung. Maintenance of normal
fall in intrapleural pressure increases vascular ventilation–perfusion ratios is important
transmural pressure, which distends extra- because high blood flow to hypoxic regions,
alveolar vessels (i.e., pulmonary arteries and for example, would decrease the overall oxy-
veins), decreases resistance, and increases gen content of the blood leaving the lungs.
regional flow. The opposite occurs during a Sympathetic adrenergic nerves innervate the
forced expiration, particularly against a high pulmonary vasculature, although their activation
resistance (e.g., Valsalva maneuver). Vessels has relatively weak effects on pulmonary vascu-
associated with the alveoli are compressed lar resistance and pulmonary artery pressure.
as the alveoli fill with air and enlarge during
inspiration. With very deep inspirations, this
Summary of Special Circulations
capillary compression can cause an increase
in overall pulmonary resistance. Perfusion pressure and vascular resistance
The primary purpose of the pulmonary cir- determine blood flow in organs. Under normal
culation is to perfuse alveoli for the exchange circumstances, the perfusion pressure remains
of blood gases. Gas exchange depends, in part, fairly constant owing to baroreceptor mecha-
on diffusion distances and the surface area nisms. Therefore, the primary means by which
available for exchange. The capillary–alveolar blood flow changes within an organ is by
arrangement is such that diffusion distances changes in vascular resistance, which is influ-
are minimized and surface area is maximized. enced by extrinsic factors (e.g., sympathetic
Pulmonary capillaries differ from their sys- nerves and hormones) and intrinsic factors
temic counterparts in that they form thin (e.g., tissue metabolites and endothelial-derived
interconnecting sheets around and between substances). Basal vascular tone is determined
adjacent alveoli, which greatly increase their by the net effect of the extrinsic and intrinsic
surface area and reduce diffusion distances. factors acting on the vasculature. Resistance
Unlike other organs, alveolar or arterial can either increase or decrease from the basal
hypoxia causes pulmonary vasoconstric- state by alterations in the relative contribu-
tion. The mechanism is not known; however, tion of extrinsic and intrinsic factors. Table 7-2
evidence suggests that endothelin, reactive summarizes the relative importance of sympa-
oxygen species, and intracellular calcium thetic and metabolic control mechanisms and
mobilization may be involved. This hypoxic the intrinsic autoregulatory capacity of several
vasoconstriction, especially in response major organ vascular beds.

TABLE 7-2 COMPARISON OF VASCULAR CONTROL MECHANISMS IN DIFFERENT


VASCULAR BEDS
CIRCULATORY BED SYMPATHETIC CONTROL METABOLIC CONTROL AUTOREGULATION
1
Coronary + +++ +++
Cerebral + +++ +++
Skeletal muscle ++ +++ ++
Cutaneous +++ + +
Intestinal +++ ++ ++
Renal ++ + +++
Pulmonary + +2 NA
+, weak; ++, moderate; +++, strong.
NA, not applicable because pressure is the dependent variable instead of flow as in other organs.
1
Sympathetic vasoconstriction in the coronaries is overridden by metabolic vasodilation during sympathetic activation of the heart.
2
Hypoxia causes vasoconstriction, the opposite of all other organs.

Klabunde_Chap07.indd 175 6/10/2011 10:57:50 PM


176 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

SUMMARY OF IMPORTANT CONCEPTS

• The relative distribution of blood flow in the coronary circulation and in


to organs is regulated by the vascular contracting skeletal muscle.
resistance of the individual organs, • Flow autoregulation is very important
which is determined by extrinsic in organs such as the heart, brain,
(neurohumoral) and intrinsic (local and kidneys; the gastrointestinal and
regulatory) mechanisms. skeletal muscle circulations show
• Important local mechanisms regulating moderate autoregulation.
organ blood flow include the following: • Blood flow is tightly coupled to
(1) tissue factors such as adenosine, oxidative metabolism particularly in
K•, 0 2 , C0 2 , and W; (2) paracrine coronary, cerebral, skeletal muscle, and
hormones such as bradykinin, gastrointestinal circulations; therefore, an
histamine, and prostaglandins; (3) increase in tissue oxygen consumption
endothelial factors such as nitric oxide, leads to an increase in blood flow
endothelin-1, and prostacyclin; and (4) (functional or active hyperemia).
myogenic mechanisms intrinsic to the
vascular smooth muscle. • Blood flow in the following organs
is moderately to strongly influenced
• The following local factors produce by sympathetic vasoconstrictor
vasodilation in most tissues: adenosine, mechanisms: resting skeletal muscle,
K+, W, C02, hypoxia, bradykinin, kidneys, gastrointestinal circulation,
histamine, PGE2 , prostacyclin, and nitric and skin (related to thermoregulation).
oxide. The following local factors produce
vasoconstriction: endothelin-1 and the • Vascular control mechanisms linked
myogenic response to vascular stretch. to oxidative metabolism (metabolic
mechanisms) are particularly strong in
• Mechanical compression of blood the heart, brain, and skeletal muscle.
vessels strongly influences blood flow

REVIEW QUESTIONS

For each question, choose the one best 2. If a coronary artery is occluded for
answer: 1 minute and then the occlusion is
released,
1. Two minutes after perfusion pressure to a. A period of active hyperemia follows.
a kidney is suddenly reduced from 100 b. Coronary flow increases because of
to 70 mm Hg, which of the following vasoconstriction occurring during the
will occur? ischemia.
a. Afferent arterioles will be dilated. c. Endothelial release of nitric oxide will
b. Renal blood flow will be reduced by contribute to the reactive hyperemia.
30%. d. Interstitial adenosine concentrations
c. Renal vascular resistance will be will increase and constrict coronary
increased. arterioles.
d. The kidney will become hypoxic.

Klabunde_Chap07.indd 176 6/10/2011 10:57:50 PM


CHAPTER 7 • ORGAN BLOOD FLOW 177

3. Which one of the following organ 9. Skeletal muscle circulation _____


circulations is most strongly constricted
during sympathetic activation resulting 10. A hypertensive patient is diagnosed
from a baroreceptor reflex when a person with bilateral renal artery stenosis. You
suddenly stands up? consider administering an ACE inhibi-
a. Brain tor to lower the blood pressure. By what
b. Heart mechanism might this drug adversely
c. Intestine affect renal glomerular filtration?
d. Skin a. Constriction of afferent arterioles
b. Constriction of efferent arterioles
Match the organs listed in questions 4 to 9 c. Dilation of afferent arterioles
with answers “a” through “i” below. Each d. Dilation of efferent arterioles
question may have more than one correct
answer. 11. A patient with anginal symptoms is
a. Blood flow is primarily regulated by diagnosed with coronary artery disease
CO2 and H+. and coronary vasospasm. Which of
b. Capillary beds found between two the following might be responsible for
in-series arterioles increased susceptibility to vasospasm?
c. Hypoxic vasoconstriction a. Diminished endothelial production of
d. Highest capillary pressure nitric oxide
e. Highest arterial–venous oxygen b. Diminished sympathetic tone on the
difference coronary vessels
f. Abundant arterial–venous anastomoses c. Ischemia-induced production of aden-
g. Largest organ mass osine
h. Receives most of its blood supply d. Reduced coronary endothelial pro-
directly from other organs duction of endothelin-1
i. Controlled primarily by hypothalamic
thermoregulatory centers 12. Intracranial pressure is steadily increas-
ing in a patient who has recently suffered
4. Skin circulation _____ a hemorrhagic stroke. You are concerned
about this finding because elevated
5. Renal circulation _____ intracranial pressure can result in
a. Decreased blood flow.
6. Coronary arteries _____ b. Decreased cerebral vascular
resistance.
7. Pulmonary circulation _____ c. Increased cerebral perfusion pressure.
d. Increased cerebral vascular
transmural pressure.
8. Cerebral circulation _____

Klabunde_Chap07.indd 177 6/10/2011 10:57:52 PM


178 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

1. The correct answer is "a" because in 7. The correct answer is "c."


response to a reduction in perfusion 8. The correct answer is "a."
pressure and blood flow, the kidney 9. The correct answer is "g."
undergoes autoregulation through dila- 10. The correct answer is "d" and the other
tion of the afferent arterioles. Choice "b" choices are incorrect because in renal
is incorrect. When the pressure is first artery stenosis, increased angiotensin
reduced, blood flow will fall by about II, which preferentially constricts the
30%, but after 2 minutes, the blood efferent arteriole, helps to maintain
flow will be near normal owing to the glomerular capillary pressure and filtra-
autoregulation. Choice "c" is incorrect tion despite the fall in renal perfusion
because afferent arteriolar vasodilation pressure. Therefore, decreasing angio-
reduces renal vascular resistance. Choice tensin II with an ACE inhibitor removes
"d" is incorrect because autoregulation, this constriction, which leads to a
by maintaining blood flow, protects the decrease in glomerular capillary pressure
kidney against ischemia and hypoxia. and glomerular filtration.
2. The correct answer is "c" because the 11. The correct answer is "a" because coro-
increase in flow (reactive hyperemia) nary artery disease is associated with
following release of the occlusion causes endothelial dysfunction and decreased
a flow-dependent release of nitric oxide nitric oxide production. The vasodila-
by the vascular endothelium, which tor actions of nitric oxide normally
further contributes to the increase in oppose vasoconstrictor mechanisms,
blood flow. Choice "a" is incorrect and therefore, reduced nitric oxide
because active hyperemia is associated enhances vasoconstrictor responses and
with increased tissue metabolic activity can increase susceptibility to vasospasm.
and not with postischemic hyperemia. Choice "b" is incorrect because dimin-
Choice "b" is incorrect because vasodila- ished sympathetic tone reduces vasocon-
tion occurs during ischemia. Choice "d" strictor influences on the vessels. Choice
is incorrect because increased interstitial "c" is incorrect because adenosine is a
adenosine dilates coronary arterioles. vasodilator and therefore opposes vaso-
3. The correct answer is "c." Choice "a" constriction. Choice "d" is incorrect
is incorrect because the brain responds because endothelin-1 is a vasoconstric-
little to sympathetic activation. Although tor and may contribute to vasospasm.
the coronary vasculature in the heart 12. The correct answer is "a" because
(choice "b") is capable of responding to increased intracranial pressure reduces
sympathetic activation, concurrent stim- cerebral blood flow by compressing
ulation of heart rate and inotropy lead vessels and increasing their resistance
to metabolic vasodilation. Choice "d" is (therefore, choice "b" is incorrect) and
incorrect because sympathetic control of decreases the effective perfusion pressure,
the skin circulation is primarily related to which can be approximated as the mean
thermoregulation; therefore, the barore- arterial pressure minus the intracranial
ceptor reflex associated with standing has pressure (therefore, choice "c" is incor-
little influence on cutaneous blood flow. rect). Choice "d" is incorrect because the
4. The correct answers are "f' and "i." increased intracranial pressure increases
5. The correct answers are "b" and "d." the pressure outside of the vessels so that
6. The correct answer is "e." the transmural pressure decreases.

Klabunde_Chap07.indd 178 6/10/2011 10:57:52 PM


CHAPTER 7 • ORGAN BLOOD FLOW 179

PROBLEM 7-1 in this experiment, we can conclude that the


The initial perfusion pressure was 100 mm myogenic (vasoconstrictor) mechanism was
Hg (mean arterial pressure minus venous dominant over the metabolic (vasodilator)
pressure). Elevating the venous pressure to mechanism. These results have been observed
15 mm Hg reduced the perfusion pressure to experimentally in organs such as the intestine.
85 mm Hg. According to the equation relating
blood flow, perfusion pressure, and vascular CASE 7-1
resistance (F = LlPIR), flow would decrease by It is important to control arterial pressure in
15% with a 15% decrease in perfusion pressure patients with coronary artery disease because
(assuming that resistance does not change). hypertension increases ventricular afterload
However, in this case, flow decreased by 25% and myocardial oxygen demand. However, it
indicating that resistance increased by 13.3% is important to lower arterial pressure using
(R = LlPIF = 0.85/0.75). The metabolic theory drugs that do not cause a reflex tachycar-
for autoregulation states that as perfusion pres- dia for two reasons. First, reflex tachycardia
sure and flow are reduced, an accumulation of (baroreceptor-mediated) increases myocardial
vasodilator metabolites decreases resistance oxygen demand and offsets the beneficial effects
in an attempt to restore flow; however, resist- of reducing afterload (see Chapter 4). Second,
ance did not decrease in this experiment. The tachycardia further impairs coronary perfu-
myogenic theory states that increased trans- sion because the duration of diastole relative
mural pressure causes vascular smooth muscle to systole decreases at elevated heart rates.
to contract, thereby increasing resistance and This reduces the time available for coronary
decreasing flow. Increasing venous pressure in perfusion during diastole, which is the time
this experiment increased the transmural pres- when the greatest amount of coronary perfu-
sure in arterioles, causing them to constrict and sion occurs. It is common in clinical practice to
increase their resistance. Therefore, increasing give a drug such as a ~-blocker to a patient with
venous pressure produces opposite and com- both coronary artery disease and hypertension
peting responses between these two mecha- because it lowers arterial pressure and prevents
nisms. Because vascular resistance increased reflex tachycardia.

SUGGESTED RESOURCES Joyner MJ, Halliwill JR. Sympathetic vasodilation in


Clifford PS, Hellsten Y. Vasodilatory mechanisms human limbs.] Physiol2000;526:471-480.
in contracting skeletal muscle. J Appl Physiol Kellogg DL. In vivo mechanisms of cutaneous vaso-
2004:393-403. dilation and vasoconstriction in humans dur-
Deussen A, Brand M, Pexa A, Weichselj. Metabolic ing thermoregulatory challenges. ] Appl Physiol
coronary flow regulation- Current concepts. Basic 2006;100: 1709-1718.
Res Cardio 2006;101:453-464. Lassen NA: Brain. In johnson PC, ed. Peripheral
Hill MA, Meininger GA, Davis MJ, Laher I. Therapeutic Circulation. New York: john Wiley&: Sons, 1978.
potential of pharmacologically targeting arteriolar myo- Rhoades RA, Bell, DR. Medical Physiology: Principles
genic tone. Trends Pharmacol Sci 2009;30:363-374. for Clinical Medicine. 3rd Ed. Philadelphia:
johnson PC. Autoregulation of blood flow. Circ Res Lippincott Williams&: Wilkins, 2009.
1986;59:483-495.

Klabunde_Chap07.indd 179 6/10/2011 10:57:53 PM


0

8
:I:
>
EXCHANGE FUNCTION OF 'tl
-1
m
;o
II:IE MICROCIRCULATION
Understanding the concepts presented in this chapter will enable the student to:
1. Describe the principal mechanisms by which gases, fluid, electrolytes, and
macromolecules move across the capillary endothelium.
2. Name three different types of capillaries; know the organs in which they are
found, and describe their differences in permeability to macromolecules and fluid.
3. Describe the factors that determine oxygen's rate of exchange between the

microcirculation and tissue.
4. Explain the relationship between oxygen content of blood, percent saturation,
and partial pressure of oxygen.
5. Describe the relationship between oxygen delivery to a tissue, oxygen extrac-
tion, and oxygen consumption.
6. Describe the mechanisms responsible for the movement of fluid across capillaries.
7. Describe the relationship between interstitial fluid volume, interstitial hydro-
static pressure, and interstitial compliance.
8. Describe how changes in capillary hydrostatic pressure, plasma oncotic pres-
sure, capillary permeability, and lymphatic function can lead to tissue edema.

INTRODUCTION 4. The microcirculation is where gases, circu-


lating substances (e.g., nutrients, hormones,
The microcirculation consists of small arteries, therapeutic drugs), metabolic wastes from
arterioles, capillaries, venules, small veins, and the tissues, fluid, and thermal energy are
small lymphatic vessels found within organs exchanged between the blood and tissues.
and tissues (see Chapter 5, Fig. 5.1) and has Within the microcirculation, capillaries are
the following important functions: quantitatively the most important site for
1. Small arteries and arterioles are the principal exchange because of their physical structure
sites of resistance within the systemic cir- (small volume-to-surface area ratio and thin
culation and therefore play a major role in walls), large number, and enormous surface area
the regulation of arterial blood pressure and available for exchange. This chapter focuses on
blood flow within organs (see Chapter 5). the exchange function of capillaries.
2. Venules and small veins have an important
capacitance function and therefore deter- MECHANISMS OF EXCHANGE
mine the distribution of blood volume
within the body. Fluid, electrolytes, gases, and small and large
3. The microcirculation allows passage of leu- molecular weight substances transverse the cap-
kocytes from the blood into the extravascular illary endothelium by several different mecha-
space, which is important in inflammation nisms: diffusion, bulk flow, vesicular transport,
and infection. and active transport (Fig. 8.1). Some substances
180

Klabunde_Chap08.indd 180 6/11/2011 10:04:10 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 181

Active
Diffusion Bulk Flow Vesicles Transport
Blood
Endothelial
Cell

Tissue
Interstitium
O2, CO2, H2O, Macro- Ions, small
lipid-soluble electrolytes, molecules molecules
substances small molecules

■ FIGURE 8.1 Mechanisms of exchange across the capillary endothelium. Lipid-soluble substances like
oxygen and carbon dioxide readily exchange across capillary endothelial cells by diffusion. Water and
electrolytes move across the endothelium primarily by bulk flow through intercellular clefts (“pores”).
Vesicular transport mechanisms move large molecules across the endothelium. Active transport mecha-
nisms move ions and other small molecules across the endothelium.

are primarily transported by one mechanism, The diffusion constant is a value that represents
whereas other substances are able to use more the ease with which a specific substance can
than one mechanism. This is determined by cross the capillary wall (or other barrier) by dif-
the physical and chemical characteristics of fusion. The higher the diffusion constant for a
the substance as well as the type of capillary specific substance, the greater its flux across
endothelium, which differs among organs. the barrier at a given concentration gradient.
The diffusion constant is determined by the
Diffusion physical and chemical structure of the barrier
as well as the physical and chemical charac-
Diffusion is the movement of a molecule from a high
teristics (e.g., size, electrical charge) of the
concentration to a low concentration. This mecha-
diffusing molecule. For example, the diffu-
nism of exchange is particularly important for
sion constant for oxygen (small and highly
gases (O2 and CO2) and other lipid-soluble sub-
lipophilic) across cell membranes (which are
stances (e.g., steroid hormones, anesthetics).
lipid bilayers) is very high compared to glu-
Fluid and electrolytes also are exchanged across
cose (large and hydrophilic).
the endothelium, in part, by diffusion.
Equation 8-1 indicates that the rate of dif-
The movement of a substance by diffusion
fusion is directly related to the concentration
is described by Fick’s first law of diffusion
difference, the diffusion constant, and the area
(Equation 8-1), in which the movement of
available for diffusion, and it is inversely related
a molecule per unit time (flux JS; moles/s)
to the diffusion distance. The diffusion distance
equals the diffusion constant (D) of the (ΔX) in Equation 8-1 is sometimes combined
barrier (e.g., capillary wall) multiplied by with the diffusion constant (D) and called the
the surface area (A) available for diffusion permeability coefficient (P). This simplifies
and the concentration gradient (ΔC/ΔX), Equation 8-1 to JS = PS(ΔC) in which S is the
which is the concentration difference across surface area available for exchange. The com-
the barrier (ΔC) divided by the diffusion bined value of the permeability coefficient
distance (ΔX). times the surface area has been calculated
ΔC for different substances in many organs and
Eq. 8-1 JS = DA
ΔX tissues; it is called the PS product.

Klabunde_Chap08.indd 181 6/11/2011 10:04:13 AM


182 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Bulk Flow for the translocation of macromolecules


(e.g. proteins) across capillary endothelium.
A second mechanism for exchange is bulk Compared to diffusion and bulk flow, vesicu-
flow. This mechanism is important for the lar transport plays a relatively minor role in
movement of water and small lipid-insoluble transcapillary exchange (except for macromol-
substances across capillaries. Bulk flow of fluid ecules). Evidence exists, however, that vesicles
and electrolytes, and of small molecules, occurs can sometimes fuse together, creating a chan-
through intercellular clefts between endothelial nel through a capillary endothelial cell, thereby
cells (see Fig. 8.1). These extracellular path- permitting bulk flow to occur across the cell.
ways are sometimes referred to as “pores.” Active transport is a fourth mechanism of
The physical structure of capillaries varies exchange. Some molecules (e.g., ions, glucose,
considerably among organs; these differences amino acids) are actively transported across
greatly affect exchange by bulk flow. Some cap- capillary endothelial cells; however, this is
illaries (e.g., skeletal muscle, skin, lung, and not normally thought of as a mechanism for
brain) have a very “tight” endothelium and exchange between plasma and interstitium, but
continuous basement membrane (termed con- rather as a mechanism for exchange between
tinuous capillaries), which reduces bulk flow an individual cell and its surrounding milieu.
across the capillary wall. In contrast, some vas-
cular beds have fenestrated capillaries (e.g.,
in exocrine glands, renal glomeruli, and intes- EXCHANGE OF OXYGEN
tinal mucosa), which have perforations (fenes- AND CARBON DIOXIDE
trae) in the endothelium, resulting in relatively
high permeability and bulk flow. Discontinu- Oxygen Diffusion
ous capillaries (found in the liver, spleen, and Oxygen diffuses from the blood to tissue cells
bone marrow) have large intercellular gaps, as to support mitochondrial respiration. The
well as gaps in the basement membrane, and lipid solubility of oxygen enables it to readily
therefore have the highest permeability. diffuse through tissues; however, the distance
Bulk flow follows Poiseuille’s equation for that oxygen is able to diffuse within a tissue is
hydrodynamic flow (see Chapter 5, Equa- limited by cellular utilization of oxygen. For
tion 5-7). Changes in pressure gradients (either example, as oxygen diffuses out of a capillary
hydrostatic or colloid osmotic) across a capil- into surrounding skeletal muscle cells, oxy-
lary alter fluid movement across the capillary. gen is consumed by the mitochondria. Con-
In addition, changes in the size and number of sequently, little oxygen diffuses all the way
“pores” or intercellular clefts alter exchange. through one cell to reach another. Therefore,
Pore size and path length are analogous to in tissues having a high demand for oxygen, it
vessel radius and length in Poiseuille’s equa- is essential that the capillary density is great
tion; they are major factors in the resistance to enough to provide short diffusion distances.
bulk flow across capillaries. In some organs, the Large amounts of oxygen diffuse across
number of perfused capillaries can be regulated. the capillaries not only because of their thin
As described in Chapter 7, the number of per- walls and high diffusion constant for oxy-
fused capillaries in contracting skeletal muscle, gen, but more importantly, because of their
for example, is greater than at rest. An increase large surface area available for diffusion. It
in perfused capillaries increases the surface area has been observed that significant amounts
available for fluid exchange and the net move- of oxygen also diffuse out of arterioles.
ment of fluid across capillaries by bulk flow. Some of this oxygen diffuses through arteri-
olar walls into the surrounding cells, and in
Vesicular and Active Transport
some cases, it diffuses from arterioles into
Vesicular transport is a third mechanism by the venules that often are found adjacent to
which exchange occurs between blood and tis- arterioles. Normally, systemic arterial blood
sue. This mechanism is particularly important is fully saturated with oxygen and has a PO2

Klabunde_Chap08.indd 182 6/11/2011 10:04:13 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 183

of about 95 mm Hg. Direct measurements of microvascular blood flow, thereby delivering


PO2 in small arterioles (20 to 80 μm diameter) more oxygen to the capillaries per unit time,
of some tissues reveal that the PO2 is only 25 which results in higher PO2 values in the cap-
to 35 mm Hg, which corresponds to a 30% illary blood. If vasodilation is accompanied by
to 60% loss of oxygen content of the blood. an increase in the number of flowing capillar-
Therefore, substantial amounts of oxygen ies (as occurs during skeletal muscle contrac-
can diffuse out of the blood before the blood tion), this increases the surface area available
reaches the capillaries; however, capillar- for oxygen diffusion and further enhances
ies are still the most important site for tissue oxygen transport into the tissue. For exam-
oxygenation because the relatively high cap- ple, if the cell shown in Figure 8.2 were sur-
illary density ensures that diffusion distances rounded by three capillaries instead of one,
between the blood and tissue cells are short. then there would be an increase in the rate of
Figure 8.2 illustrates the diffusion of oxy- oxygen diffusion into the cell, which would
gen from blood within a capillary, across the be necessary if the mitochondrial oxygen con-
capillary endothelium, and then into a cell. sumption increased significantly.
The PO2 is only slightly reduced just out-
side of the capillary (from 25 to 24 mm Hg) Oxygen Delivery and Extraction
because little oxygen is consumed as it dif-
fuses through the endothelial cell and into The previous discussion described oxygen dif-
the interstitial fluid surrounding the capillary. fusion from blood into tissue cells, and how
The oxygen in the interstitium then diffuses the PO2 gradient from the blood to the tissue
down a concentration gradient into nearby cell plays an important role in determining
cells. Because the mitochondria inside a cell the rate of diffusion. While the PO2 gradient
are consuming oxygen, the PO2 may be very
low inside the cytoplasm of the cell. Although
an intracellular PO2 of 5 mm Hg is shown in PO2 =
Figure 8.2, the value depends on where the Capillary 25 mmHg
PO2 is measured within the cell, the rate of
mitochondrial oxygen consumption, and
O2
the capillary blood PO2. Just inside the cell C = 25 - 5 mmHg
Interstitium 24 mmHg = 20 mmHg
membrane, the PO2 is much higher than at
the center of the cell; the lowest PO2 is found O2
within the mitochondria. Therefore, signifi- 5 mmHg
Cell
cant oxygen gradients exist within cells.
In Figure 8.2, the overall concentration
gradient driving oxygen diffusion into the
cell is 20 mm Hg. According to Fick’s first
law (Equation 8-1), the rate of oxygen dif- JO2 = DA ( C/ X)
fusion ( JO2) is proportionate to the concen- JO2 C
tration difference of oxygen (expressed as
■ FIGURE 8.2 Diffusion of oxygen (JO2) from capil-
PO2 difference) between the capillary blood laries into the tissue follows Fick’s first law of diffu-
and inside the cell, assuming a fixed diffu- sion. Because the diffusion constant (D), the area
sion constant, diffusion distance, and surface for exchange (A), and the diffusion distance (ΔX)
remain relatively constant in a single capillary, the
area. Therefore, increasing capillary blood
diffusion of oxygen is governed primarily by the dif-
PO2 (as occurs when a person breathes pure ference in partial pressure of oxygen (PO2) between
oxygen) or decreasing tissue PO2 (as occurs the blood and cells (ΔC), which is 20 mm Hg in this
with increased tissue oxygen consumption) illustration. Most of this PO2 gradient is between the
interstitium and cell when mitochondria are actively
increases the rate of oxygen diffusion into
consuming oxygen; there is only a small gradient
the tissue. Capillary PO2 is also increased by across the capillary endothelium. Increasing the
dilation of resistance vessels. This increases overall PO2 gradient increases the rate of diffusion.

Klabunde_Chap08.indd 183 6/11/2011 10:04:13 AM


184 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

determines the rate of oxygen diffusion, the at normal arterial PO2 values is approximately
total amount of oxygen that is available per 20 mL O2/100 mL blood (or, 20 vol %).
unit time for diffusion is determined by the The hemoglobin–oxygen dissociation
amount of hemoglobin-bound oxygen in the curve is sigmoidal in shape; therefore, small
blood and the rate of blood flow into the tissue. decreases in arterial PO2 from normal values
The amount of oxygen in the blood (oxygen do not significantly reduce the oxygen content
content) is determined by the PO2 of the blood, of the arterial blood. However, as the arterial
along with the amount of hemoglobin in the PO2 begins to fall below 80 mm Hg, and espe-
red cells and the hemoglobin binding affinity cially in the range of tissue PO2 values (20 to
for oxygen (Fig. 8.3). This relationship is called 40 mm Hg), the curve becomes very steep and
the hemoglobin–oxygen dissociation curve. At there is a large decrease in the amount of oxy-
normal arterial PO2 values (95 mm Hg), about gen bound to hemoglobin as PO2 decreases.
97% of the hemoglobin is bound to oxygen (97% At a PO2 of about 25 mm Hg, hemoglobin is
hemoglobin saturation; SaO2). If the blood con- only 50% saturated (P50 = 25 mm Hg). There-
tains 15 g of hemoglobin per 100 mL of blood fore, as blood flows into tissues, the relatively
(normal value), and a gram of hemoglobin can low PO2 in the tissue results in oxygen diffus-
bind to 1.34 mL oxygen, then 20.1 mL oxygen ing from the blood into the tissue. This lowers
(15 g/100 mL × 1.34 mL O2/g) will be bound to the blood PO2 and causes oxygen to dissoci-
hemoglobin in 100 mL of blood when 100% sat- ate from the hemoglobin so that it can diffuse
urated, and 19.5 mL O2/100 mL blood is bound into the tissue. Unloading of oxygen from
at 97% saturation. A small amount of oxygen hemoglobin can also be enhanced by factors
(~0.3 mL O2/100 mL blood) is dissolved in the that cause a rightward shift in the oxygen dis-
free water of the plasma and cells at normal sociation curve. For example, increased tem-
arterial PO2 values. Therefore, the total amount perature and PCO2, and decreased pH shift
of hemoglobin-bound and dissolved oxygen the curve to the right, which shifts the P50 to
the right. Therefore, at any given tissue PO2
100 20 value, a rightward shift causes more unload-
ing of oxygen from the hemoglobin because
CaO2 (ml O2/100 ml blood)

HbO2
80 16 of reduced binding affinity to oxygen. This
% HbO2 Saturation

is an important mechanism to increase tissue


60
Temp
12 oxygenation when the metabolic activity of
PCO2 a tissue increases (e.g., contracting muscle),
pH
40 8 which increases tissue temperature and CO2
production, and decreases pH.
20 4
The oxygen content of the arterial blood
Dissolved O2 (CaO2; mL O2/100 mL blood) multiplied by
0 0
0 20 40 60 80 100 600
the arterial blood flow (F; mL/min) represents
PO2 (mmHg) the oxygen delivery (DO2; mL O2/min) to the
tissue (Fig. 8.4).
■ FIGURE 8.3 Hemoglobin–oxygen dissociation
curve. Percent oxygen saturation of hemoglobin
DO2 = F ⋅ CaO2
(% HbO2) has a sigmoidal relationship with the par-
tial pressure of oxygen (PO2). In this example, 100%
saturation corresponds to an arterial blood oxygen Therefore, the oxygen delivery to a tissue is
content (CaO2) of about 20 mL O2 / 100 mL blood. determined by the arterial blood flow and the
This assumes that the hemoglobin concentration
is 15 g/100 mL blood and that 1.34 mL O2 bind to arterial oxygen content. Because arterial blood
each gram of hemoglobin. Note that the amount is normally near its maximal oxygen capac-
of dissolved oxygen is very small relative to the ity (>95% saturated), oxygen delivery to a
amount of oxygen bound to hemoglobin. The
tissue can only be enhanced by increasing
dissociation curve shifts to the right (decreased
hemoglobin affinity for oxygen) with increased blood flow. On the other hand, oxygen deliv-
temperature and PCO2, and decreased pH. ery can be reduced by decreasing either flow

Klabunde_Chap08.indd 184 6/11/2011 10:04:13 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 185

CaO2 CvO2
Arteriole Capillaries Venule

O2 Delivery: DO2 = F CaO2


O2 Consumption: = F (CaO2 CvO2 )

Venous O2 Content: CvO2 = CaO2

■ FIGURE 8.4 Model for oxygen delivery and balance in tissues. Oxygen .delivery (DO2) is the product of blood
flow (F) and the arterial oxygen content (CaO2). Oxygen consumption (V O2) is the product of flow and arte-
rial–venous oxygen difference (oxygen extraction; CaO2 − CvO2) according to the Fick principle.
. Rearranging
the equation shows that venous oxygen content (CvO2) depends on CaO2 minus the ratio of VO2 to F.

(e.g., ischemia) or arterial oxygen content 


VO2 = F (CaO2 − CvO2 )
(e.g., anemia, hypoxemia).
Oxygen delivery represents only what is If this equation is solved for the oxygen
available to the tissue, not what is utilized by extraction, then we see that oxygen extrac-
the tissue. As arterial blood enters the micro- tion is determined by the ratio of oxygen con-
circulation, and particularly the capillaries, sumption to blood flow.
oxygen diffuses from the blood into the tis-

VO
sues, and this reduces the oxygen content of (CaO2 − CvO2 ) = 2

the blood (see Fig. 8.4). The greater the oxy- F


gen consumption of the tissue, the greater the Therefore, oxygen extraction is increased if
amount of oxygen that diffuses from the blood. oxygen consumption increases or blood flow
Therefore, as the blood leaves the tissues, the decreases. Because the arterial oxygen content
venous blood has a lower oxygen content than normally does not change significantly, then
the arterial blood. For example, if 5 mL O2/100 increased extraction reduces the venous oxygen
mL blood were removed as the blood passes content. This is more clearly seen by solving the
through a tissue (i.e., oxygen extraction), previous equation for venous oxygen content:
then the venous blood oxygen content (CvO2)
will be 15 mL O2/100 mL blood if CaO2 is 20 
VO
mL O2/100 mL blood. Note, that as previously CvO2 = CaO2 − 2
F
described in Chapter 7, oxygen extraction dif-
fers among organs and depends on their oxy- Venous oxygen measurements are used for
gen consumption and blood flow. When the monitoring patients in intensive care settings,
oxygen extraction (CaO2 − CvO2; mL O2 /mL and therefore, the above relationship helps to
blood) is multiplied by the blood flow (F; mL / explain what can cause venous oxygen levels
min), this represents the amount of oxygen (usually measured as venous oxygen saturation
.
consumed by the tissue (VO2; mL O2 /min), or PO2) to fall. Venous oxygen saturation meas-
which is described by the Fick Principle (see ured in the pulmonary artery (SvO2) is nor-
Equation 4-3 and below). Note that oxygen mally about 75% and has a PO2 of about 40 mm
content needs to be expressed as mL O2 /mL Hg. If it is abnormally low, this can be caused
blood instead of mL O2 /100 mL blood when by elevated organ consumption by organs,
calculating values using the Fick Principle. reduced organ blood flow, or reduced arterial

Klabunde_Chap08.indd 185 6/11/2011 10:04:13 AM


186 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

oxygen content. In intensive care settings, if (i.e., blood) within the cardiac chambers and
arterial oxygen saturation is normal, reduced blood vessels of the body. The extravascular
Sv02 is usually caused by underperfusion of system is everything outside of the intravascu-
organs resulting from reduced cardiac output, lar compartment. The extravascular compart-
which causes greater extraction of oxygen from ment is made up of many subcompartments
the blood, thereby lowering venous oxygen such as the cellular, interstitial, and lymphatic
saturation and content. subcompartments and a specialized system
containing cerebrospinal fluid within the cen-
PROBLEM 8-1 tral nervous system.
An experiment is done on a human Fluid readily exchanges between the intra-
subject that measures the P0 2 of venous vascular and extravascular compartments.
blood leaving the forearm during Fluid leaves blood vessels (primarily capil-
reactive hyperemia following a period laries) and enters the tissue interstitium of
of ischemia. During the initial phase the extravascular compartment. This is called
of reactive hyperemia, venous P0 2 fluid filtration (Fig. 8.5). It is estimated that
is transiently lower than normal and about l% of the plasma is filtered into the
then becomes elevated. As blood flow interstitium in a typical organ. The intersti-
returns toward normal near the end of tial fluid is exchanged with the fluid found
the hyperemic response, the venous P0 2 within the subcompartments of the extracel-
also returns to its normal value. How lular compartment. It is crucial that a steady
would you explain these findings? state is achieved in which the same volume
of fluid that leaves the vasculature is returned
Carbon Dioxide Diffusion to the vasculature; otherwise, the extravascu-
lar compartment would swell with fluid (i.e.,
Carbon dioxide is a by-product of oxidative become edematous).
metabolism and must be removed from the There are two routes by which fluid is
tissue and transported to the lungs by the returned to the blood. First, fluid reabsorp-
blood. Uke oxygen, carbon dioxide is very tion returns most of the filtered fluid to the
lipid-soluble and readily diffuses from cells blood at the venular end of capillaries or at
into the blood. In fact, its diffusion constant is postcapillary venules (see Fig. 8.5). The rate
about 20 times greater than oxygen in aque- of reabsorption is less than filtration; there-
ous solutions. The removal of carbon diox- fore, a second mechanism is required to main-
ide from tissues is not diffusion-limited; its tain fluid balance. This second mechanism
removal depends primarily on the blood flow. involves lymphatic vessels. These specialized
Therefore, reduced tissue perfusion leads to an vessels, similar in size to venules, comprise
increase in tissue and venous PC0 2 • Increased an endothelium with intercellular gaps sur-
oxidative metabolism of a tissue (e.g., contract- rounded by a highly permeable basement
ing muscle) increases C02 production by cells, membrane. Terminal lymphatics end as blind
thereby increasing the concentration gradient sacs within the tissue. The terminal lymphatics
for C02 diffusion from the tissue to the blood take up the excess filtered fluid (including elec-
and increasing venous PC0 2 • The magnitude trolytes and macromolecules) and transport it
of the increase in venous P0 2 depends on the into larger lymphatics that leave the tissue. It is
relative increase in metabolism and blood flow. estimated that 5% to 10% of capillary filtration
is transported out of tissues by the lymphat-
TRANSCAPILLARY FLUID ics. The larger lymphatics have smooth muscle
EXCHANGE cells that undergo spontaneous vasomotion
that serves to "pump" the lymph. Vasomo-
The body is comprised of two basic fluid com- tion is spontaneous rhythmic contraction and
partments: intravascular and extravascular. relaxation of the lymphatic vessels. Evidence
The intravascular compartment contains fluid exists that as a lymphatic vessel fills with fluid,

Klabunde_Chap08.indd 186 6/11/2011 10:04:13 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 187

Capillary

Reabsorption

Filtration

Ly m ph Fl ow

Lymphatic

Filtration = Reabsorption + Lymph Flow

■ FIGURE 8.5 Capillary filtration, reabsorption, and lymph flow. Fluid filters out of the arteriolar end of
the capillary and into the interstitium. Most of this fluid is reabsorbed at the venular end of the capillary,
with the rest of the fluid entering terminal lymphatics to be carried away from the tissue and eventually
returned to the blood. Fluid exchange is in balance (i.e., at a steady state) when filtration equals reabsorp-
tion plus lymph flow.

the increased pressure stretches the vessel and fluid in the blood from the fluid within the
induces a myogenic contraction. Sympathetic interstitium. As described earlier, the tran-
nerves can modulate this vasomotion. Lym- scapillary movement of fluid can be described
phatic vessels contain one-way valves that by Poiseuille equation for hydrodynamic
direct lymph away from the tissue and eventu- flow (see Equation 5-7), or in more simpli-
ally back into the systemic circulation via the fied terms, it can be described by the general
thoracic duct and subclavian veins. Approxi- hydrodynamic equation (Equation 5-5) that
mately 2 to 4 L/d of lymph are returned to the relates flow (F), driving pressure (ΔP), and
circulation by this manner. resistance (R) (i.e., F = ΔP/R). In single cap-
In the steady state, the rate of fluid enter- illaries, a more common way to express this
ing the tissue interstitium by filtration is the hydrodynamic equation for transcapillary
same as that of the fluid leaving the tissue by fluid movement (fluid flux, J) is to substitute
capillary reabsorption and lymph flow. That hydraulic conductivity (Lp) for resistance,
is, filtration equals reabsorption plus lymph which are reciprocally related. Hydraulic con-
flow. When this balance is altered, the volume ductivity is related to the ease by which fluid
and pressure of fluid within the interstitium passes across the capillary wall. Fluid flux is
change. For example, if net filtration tran- the number of molecules of water (or volume)
siently increases and lymph flow does not per unit time that moves across the exchange
increase to the same extent, interstitial volume barrier; therefore, fluid flux can be expressed
and pressure will increase, causing edema. in similar units as flow. For a single capil-
Factors that cause edema are discussed in the lary, fluid flux equals the product of capillary
last section of this chapter. hydraulic conductivity and the net driving
force (i.e., J = Lp · NDF). The NDF combines
both those hydrostatic and oncotic pressures
Physical Mechanisms Governing
that drive fluid movement across the capillary
Fluid Exchange
wall.
The movement of fluid across a capillary is In an organ, fluid is moving across many
determined by several physical factors: the capillaries, and therefore the net fluid flux is
hydrostatic pressure, oncotic pressure, and related not only to the hydraulic conductiv-
physical nature of the barrier (i.e., the per- ity of single capillaries and to the net driving
meability of the capillary wall) separating the force, but also to the surface area available for

Klabunde_Chap08.indd 187 6/11/2011 10:04:14 AM


188 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

fluid exchange. When examining fluid flux have a much higher KF (i.e., permeability)
across capillaries within an organ, filtration than continuous capillaries. Furthermore,
constant (KF) and surface area (A) are sub- paracrine substances such as histamine,
stituted for hydraulic conductivity of a sin- bradykinin, and leukotrienes can significantly
gle capillary. With these substitutions, a new increase KF. The surface area (A) is primarily
expression relating net fluid flux is obtained related to the length, diameter, and number
(Equation 8-2): of vessels (capillaries and postcapillary ven-
ules) available for exchange. The surface area
Eq. 8-2 J = KF ⋅ A (NDF) is dynamic in vascular beds such as skeletal
muscle. In that tissue, the number of perfused
Equation 8-2 and Figure 8.6 show that net capillaries can increase severalfold during
fluid movement (net fluid flux, J) is directly exercise. In experimental studies using whole
related to the filtration constant (KF), the sur- organs, KF and A, which cannot be indepen-
face area available for fluid exchange (A), and dently measured, are combined and called the
the net driving force (NDF). At a given NDF capillary filtration coefficient (CFC).
(assuming that the NDF is not equal to zero), The direction of fluid movement (filtration
the amount of fluid filtered or reabsorbed per or reabsorption) in Equation 8-2 depends on
unit time is determined by the filtration con- whether the NDF is positive (filtration) or
stant and surface area available for exchange. negative (reabsorption). If the NDF is zero,
The filtration constant is determined by the no net fluid movement occurs even if KF and
physical properties of the barrier (i.e., size A are very large.
and number of “pores” and the thickness of As already mentioned, the NDF is deter-
the capillary barrier), and therefore, it rep- mined by hydrostatic and oncotic forces.
resents the permeability of the capillaries to Two hydrostatic and two oncotic pressures
fluid. Fenestrated capillaries, for example, affect transcapillary fluid exchange: capillary

H2 O

H2 O
H 2O
NDF H2 O

H2 O J
H2 O
H2 O H 2O

KF • A
J = KF • A (NDF)
■ FIGURE 8.6 Factors determining fluid movement. The rate of fluid movement (flux, J) across the capil-
lary endothelium, designated as water molecules in this figure, is determined by the net driving force
(NDF), the capillary filtration constant (KF), and the capillary surface area (A) available for exchange.

Klabunde_Chap08.indd 188 6/11/2011 10:04:14 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 189

hydrostatic pressure, tissue (interstitial) at the arteriolar end of the capillary where
hydrostatic pressure, capillary (plasma) capillary hydrostatic pressure is greatest.
oncotic pressure, and tissue (interstitial) The average capillary hydrostatic pressure
oncotic pressure (Fig. 8.7). These physical is determined by arterial and venous pres-
forces are sometimes referred to as Starling sures (PA and PV), and by the ratio of post-to-
forces in honor of Ernest Starling who pro- precapillary resistances (RV/RA). An increase
posed in 1896 that these forces govern cap- in either arterial or venous pressure increases
illary fluid exchange. The net hydrostatic capillary pressure; however, the effects of ele-
pressure driving fluid out of the capillary vations in venous pressure are much greater
(filtration) is the hydrostatic pressure inside than those of an equivalent elevation in
the capillary minus the interstitial hydrostatic arterial pressure. The reason for this is that
pressure (Pc − Pi). The net oncotic pressure postcapillary resistance is much lower than
drawing fluid into the capillary (reabsorption) precapillary resistance. In most organs, the
is the capillary plasma oncotic pressure minus postcapillary resistance is only 10% to 20%
the interstitial oncotic pressure (πc − πi). of the precapillary resistance; therefore, RV / RA
ranges from 0.1 to 0.2. If we assume that
CAPILLARY HYDROSTATIC PRESSURE RV / RA = 0.2, the following relationship (Equa-
tion 8-3) can be derived:
Capillary hydrostatic pressure (PC) drives
fluid out of the capillary, and it is highest at ⎛ RV ⎞ P + P
the arteriolar end of the capillary and lowest ⎜⎝ R ⎟⎠ A V
0.2 PA + PV
Eq. 8-3 PC = A
⇒ PC =
at the venular end. Depending on the organ, ⎛ R ⎞ 1.2
1+ ⎜
⎝ RA ⎟⎠
V
the pressure may drop along the length of the
capillary (axial or longitudinal pressure gradi-
The above equation assumes that PC repre-
ent) by 15 to 30 mm Hg owing to capillary
sents a point between two series resistances—
resistance. Because of this pressure gradient
an arterial or precapillary resistance (RA) and
along the capillary length, filtration is favored
a venous or postcapillary resistance (RV). It
also assumes that the flow that enters the cap-
illary and exits the capillary is the same (i.e.,
Capillary Interstitium there is conservation of flow). Therefore, on
the precapillary side, flow into the capillary
Pc Pi
can be expressed as: Fin = (PA − Pc)/R A. On the
c i postcapillary, the flow out of the capillary can
be expressed as: Fout = (Pc − PV)/R V. Assum-
ing that Fin equals Fout, solving for Pc results in
NDF = (Pc - P i) - ( c - i) Equation 8-3.
Equation 8-3 shows that increasing venous
Filtration: NDF > 0
Reabsorption: NDF < 0 pressure by 20 mm Hg increases mean capil-
lary pressure by 16.7 mm Hg when RV / RA =
■ FIGURE 8.7 Net driving force for fluid move- 0.2. In contrast, increasing arterial pressure
ment across capillaries. Hydrostatic and oncotic
pressures within the capillary (Pc, pc) and the tis- by 20 mm Hg increases mean capillary pres-
sue interstitium (Pi, pi) determine the net driving sure by only 3.3 mm Hg. The reason for this
force (NDF) for fluid movement out of the capil- difference is that the high precapillary resist-
lary (filtration) or into the capillary (reabsorption).
ance blunts the effects of increased arte-
The hydrostatic pressure difference favors filtra-
tion (red arrow) because Pc is greater than Pi. The rial pressure on the downstream capillaries.
oncotic pressure difference favors reabsorption Therefore, mean capillary hydrostatic pres-
(black arrow) because πc is greater than πi. The sure is more strongly influenced by changes in
oncotic pressure difference is multiplied by the
venous pressure than by changes in arterial pres-
reflection coefficient (σ), a factor that represents
the permeability of the capillary to the proteins sure. This has significant clinical implication.
responsible for generating the oncotic pressure. Conditions that increase venous pressure

Klabunde_Chap08.indd 189 6/11/2011 10:04:14 AM


190 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

(e.g., right ventricular failure, cirrhosis of the


liver, venous thrombosis) can lead to edema in

Interstitial Volume (Vi)


peripheral organs and tissues by significantly High Interstitial Compliance
increasing capillary hydrostatic pressure and (e.g., subcutaneous tissue)
capillary fluid filtration. This relationship also
shows that arteriolar vasodilation or venous Vi
C
constriction increases capillary hydrostatic Pi
pressure, which increases filtration.
Low Interstitial Compliance
TISSUE (INTERSTITIAL) HYDROSTATIC (e.g., brain)
PRESSURE

Tissue (interstitial) hydrostatic pressure (Pi) Interstitial Pressure (Pi)


is the pressure within the tissue interstitium ■ FIGURE 8.8 Effects of interstitial compliance on
that is exerted against the outside wall of the interstitial fluid volumes and pressures. Compli-
capillary and therefore opposes the capillary ance (C) is the change in interstitial volume (ΔVi)
hydrostatic pressure. In many tissues under divided by the change in interstitial pressure (ΔPi),
which is the slope of the relationship between
normal states of hydration, tissue hydro- volume and pressure. Low interstitial compliance
static pressure is subatmospheric by a few (e.g., brain tissue) causes large increases in inter-
millimeters of mercury (mm Hg), whereas stitial fluid pressure when interstitial fluid volume
in others it is slightly positive by a few mm increases, which can occur during cerebral edema
or hemorrhage within the brain. In contrast, tissue
Hg. Increased tissue fluid volume, as occurs with high interstitial compliance (e.g., subcutane-
during states of enhanced capillary fluid ous tissues), show relatively small increases in
filtration or lymphatic blockage, increases interstitial pressure as interstitial volume increases.
tissue hydrostatic pressure. In contrast,
dehydration reduces tissue fluid volume and organs, such as the brain and kidney, have a
hydrostatic pressure. low interstitial compliance. The reason is that
The effect of changes in interstitial fluid the tissue is surrounded by a rigid boney skull
volume on interstitial pressure is determined or capsule, respectively. Therefore, relatively
by interstitial compliance (C). This is defined small increases in interstitial volume can
as the change in interstitial fluid volume (ΔVi) lead to large increases in interstitial pressure.
divided by the change in interstitial fluid pres- A large increase in pressure can be very dam-
sure (ΔPi). Rearranging this relationship gives aging to the tissues and lead to cellular dys-
the following: function and death. In contrast, subcutaneous
tissues have a relatively high interstitial com-
ΔVi
ΔPi = pliance so that large increases in intersti-
C tial volume can occur with relatively small
Therefore, an increase in interstitial fluid vol- increases in interstitial pressure. Despite a rel-
ume increases interstitial fluid pressure, and atively high compliance at low interstitial fluid
the magnitude of the change varies inversely volumes, subcutaneous interstitial pressures
with the compliance of the interstitium. can still increase to high values at very high
Figure 8.8 is a graphical representation interstitial volumes during severe limb edema.
of the relationship between interstitial fluid
CAPILLARY PLASMA ONCOTIC PRESSURE
volume and pressure, and interstitial compli-
ance. The slope of the relationship between Capillary plasma oncotic pressure (πc) is the
interstitial volume and pressure is intersti- osmotic pressure within the capillary that
tial compliance. Note that the compliance is determined by the presence of proteins.
decreases at higher interstitial volumes, which Because this is an osmotic force within the
causes the pressure to increase disproportion- plasma, it opposes filtration and promotes
ately as volume increases. Some tissues and reabsorption. Because the capillary barrier is

Klabunde_Chap08.indd 190 6/11/2011 10:04:14 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 191

readily permeable to ions, the ions have no TISSUE (INTERSTITIAL) ONCOTIC


significant effect on osmotic pressure within PRESSURE
the capillary. Instead, the osmotic pressure is The tissue (or interstitial) oncotic pressure
principally determined by plasma proteins that (πi), a force that promotes filtration, is deter-
are relatively impermeable. Rather than being mined by the interstitial protein concentration
called “osmotic” pressure, this pressure is and the reflection coefficient of the capillary
referred to as the “oncotic” pressure or “colloid wall for those proteins. The protein concen-
osmotic” pressure because it is generated by tration is influenced, in part, by the amount
macromolecular colloids. Albumin, the most of fluid filtration into the interstitium. For
abundant plasma protein, generates about 70% example, increased capillary filtration into the
of the oncotic pressure; globulins and fibrino- interstitium decreases interstitial protein con-
gen generate the remainder of the oncotic pres- centration and reduces the oncotic pressure.
sure. The plasma oncotic pressure typically is This effect of filtration on protein concentra-
25 to 30 mm Hg. When capillaries are filtering tion serves as a mechanism to limit excessive
fluid, the oncotic pressure increases along the capillary filtration. The interstitial oncotic
length of the capillary, particularly in capillar- pressure, which is typically about 5 mm Hg,
ies having high filtration rates (e.g., renal glo- acts on the capillary fluid to enhance filtration
merular capillaries). This occurs because the and oppose reabsorption; therefore, when the
filtered fluid leaves behind proteins, increasing interstitial proteins are diluted and this pres-
the plasma protein concentration. sure falls, filtration is reduced. The interstitial
When oncotic pressure is determined, it is protein concentration is also determined by
measured across a semipermeable membrane, the capillary permeability to protein. If this is
that is, a membrane that is permeable to fluid increased, for example, by vascular damage or
and electrolytes but not permeable to large inflammation, then more proteins will be fil-
protein molecules. In most capillaries, how- tered with the fluid into the interstitium. An
ever, the endothelial barrier has a finite perme- increase in interstitial protein concentration
ability to proteins. The actual permeability to facilitates net filtration by reducing the net
proteins depends on the type of capillary and force for reabsorption.
on the nature of the proteins (size, shape, and
charge). Because of this finite permeability, the SUMMARY OF STARLING FORCES AND
effective oncotic pressure generated across the TRANSCAPILLARY FLUID MOVEMENT
capillary membrane is less than that calculated
from the protein concentration. The reflection Together, the hydrostatic and oncotic forces are
coefficient (σ) across a capillary wall repre- related to the NDF as shown in Equation 8-4
sents the effective oncotic pressure divided and in Figure 8.7. The net hydrostatic pres-
by the oncotic pressure measured with a true sure, which normally promotes filtration,
semipermeable membrane. If the capillary is is represented by (Pc − Pi). The net oncotic
impermeable to protein, σ = 1. If the capillary is pressure, which promotes reabsorption, is
freely permeable to protein, σ = 0. Continuous represented by (πc − πi), multiplied by the
capillaries have a high σ (>0.9), whereas dis- reflection coefficient (σ). This equation shows
continuous capillaries (e.g., liver and spleen), that the NDF is increased by increases in Pc
which are very “leaky” to proteins, have a rela- and πi and decreased by increases in Pi and πc.
tively low σ. In the latter case, plasma and tis- Eq. 8-4 NDF = (Pc − Pi ) − σ(π c − πi )
sue oncotic pressures may have a negligible
influence on the NDF. If the capillary endothe- If the above expression for NDF is incorpo-
lium becomes damaged by physical injury or rated into Equation 8-2, the following equa-
inflammation, the reflection coefficient may tion is derived, which is sometimes referred to
decrease significantly, which reduces the abil- as the Starling equation:
ity of plasma oncotic pressure to oppose filtra-
tion, thereby increasing net filtration. Eq. 8-5 J = KF i A ⎡⎣(Pc − Pi ) − σ(π c − πi )⎤⎦

Klabunde_Chap08.indd 191 6/11/2011 10:04:14 AM


192 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

The expression in brackets represents the NDF. Arteriole Venule


If the NDF is positive, filtration occurs (J > 0), Reabsorption
and if it is negative, reabsorption occurs (J < 0).
For a given NDF, the rate of fluid movement (J) Pc
is determined by the product of KF and A. Capillary
Filtration

Capillary Exchange Model 30


Capillary fluid exchange can be modeled as 20
shown in Figure 8.9. This model assumes that Pc
the following values remain constant along cap- 10
illary length: Pi = 1 mm Hg, πc = 25 mm Hg, πi = 0
6 mm Hg, and σ = 1. According to Equation 8-4, 10
if Pc is 30 mm Hg at the entrance to the capil-
5 Reab
lary and falls linearly to 15 mm Hg at the end NDF
of the capillary, the NDF changes from +10 at 0
the entrance of the capillary to −5 at the end of Filtr
-5
the capillary. Filtration occurs along most of the Capillary Length
length of the capillary wherever NDF is greater
■ FIGURE 8.9 Model of capillary fluid exchange.
than zero. Reabsorption occurs where NDF is
Assuming that Pi = 1, πc = 25, πi = 6 mm Hg, and
less than zero, which is near the venular end of σ = 1, and assuming that capillary hydrostatic pres-
the capillary. Net fluid movement is zero at the sure (Pc) at the beginning and end of the capillary
point along the capillary where NDF = 0. Exper- are 30 mm Hg and 15 mm Hg, respectively, the net
driving force [NDF = (Pc − Pi) − (πC − πi)] is greater
imental studies have shown that the hydraulic than zero along most of the length of the capillary,
conductivity in single capillaries increases sev- which causes filtration (Filtr) to occur. Near the
eralfold from the arteriolar to the venular end venular end of the capillary, the NDF is less than
of the capillary. Therefore, significant reabsorp- zero and reabsorption (Reab) occurs.
tion can still occur at the distal end of a capillary
when the NDF is only slightly negative. lymphatic flow also increases. The lymphat-
This model is highly simplified because it ics, therefore, along with the dynamic changes
assumes that Pi, πc, and πi remain constant, in Pc, Pi, πc, and πi help to maintain a proper
which does not occur in vivo. As fluid leaves state of interstitial hydration and thereby pre-
the arteriolar end of the capillary, πc increases, vent edema from occurring.
Pi increases, and πi decreases. These changes Finally, it is important to note that there
oppose the filtration. For most capillaries, the is considerable heterogeneity among capil-
fraction of fluid filtered from the capillary (fil- laries in terms of filtration and reabsorption.
tration fraction) is <1%, so Pi, πc, and πi do not Some capillaries may filter along most or all
change appreciably. Renal capillaries, however, of their length, whereas others may display
are different because the filtration fraction in reabsorption along most of their length. Fur-
these capillaries is very high (~20%), which thermore, this can change depending on the
leads to significant increases in plasma oncotic balance of hydrostatic and oncotic forces,
pressure. In nonrenal capillaries, if capillary which can vary under different physiological
permeability is increased, or if capillary hydro- and pathophysiological conditions. With arte-
static pressure is increased to high levels by riolar vasodilation or increased venous pres-
venous occlusion or heart failure, the increase sure, capillaries may filter along most or all of
in filtration can lead to significant changes in their length. Inflammation is accompanied by
Pi, πc, and πi in a manner that opposes and arteriolar vasodilation and increased capillary
therefore limits the net filtration of fluid. permeability, along with increased permeabil-
Lymphatics (not shown in Fig. 8.9) pick ity of small postcapillary venules, which can
up excess filtered fluid and transport it out become the major site of fluid filtration under
of the tissue. When net filtration increases, inflammatory conditions.

Klabunde_Chap08.indd 192 6/11/2011 10:04:15 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 193

PROBLEM 8·2 TABLE 8·1 CAUSES OF EDEMA


=
Given that Pc 22 mm Hg, P; = -3 mm Increased Capillary Pressure
= =
Hg, 1tc 26 mm Hg, 1t; 6 mm Hg, and Increased venous pressure
=
cr 0.9, answer the following questions: - Heart failure
a) What is the net driving force for - Increased blood volume
- Venous obstruction (thrombosis or
transcapillary fluid exchange?
compression)
b) Is filtration or reabsorption occurring? - Incompetent venous valves
c) If the product of KF and A is doubled, -Gravity
what will happen to the net rate of Increased arterial pressure
fluid movement across the capillary, - Hypertension
assuming that the net driving force Decreased arterial resistance
does not change? - Vasodilation (physiologic or
pharmacologic)
Increased Capillary Permeability
Vascular damage (e.g., burns, trauma)
EDEMA FORMATION Inflammation
Decreased Plasma Oncotic Pressure
When the fluid volume within the intersti- Reduced plasma proteins
tial compartment increases because filtration (e.g., malnutrition, burns, liver dysfunction)
exceeds the rate of capillary reabsorption plus Lymphatic Blockage (Lymphedema)
lymphatic flow, the interstitial compartment Tissue injury
increases in volume, leading to tissue swelling Inflammation of lymphatics
Lymphatic invasion by parasites
(i.e., edema). As already discussed, the change
(e.g., filariasis)
in interstitial pressure that results from an
increase in interstitial volume depends on the
compliance of the interstitial compartment. (e.g., sprained ankle, bee sting), which causes
Edema can damage organs and, in some the release of local paracrine substances (e.g.,
cases, cause death. For example, cerebral histamine, bradykinin, and leukotrienes) that
edema following brain trauma can lead to cel- increase capillary and venular permeability.
lular death because the increased interstitial Some of these substances (e.g., histamine)
pressure damages neurons and causes tissue also increase capillary pressure by dilating
ischemia by compressing blood vessels. Even arterioles and constricting venules.
in tissues that are relatively compliant, such as The treatment for edema involves modify-
skin and skeletal muscle, severe edema can lead ing one or more of the physical factors that
to tissue necrosis. Pulmonary edema can be life regulates fluid movement. For example, in
threatening because gas exchange is impaired. pulmonary or systemic edema secondary to
Table 8-l lists some of the many causes of heart failure, diuretics are given to the patient
edema. Every cause of edema can be related to to reduce blood volume and venous pressure,
one or more of the following: thereby reducing capillary hydrostatic pres-
sure. A patient suffering from ankle edema
• Increased capillary hydrostatic pressure
following an injury will be instructed to keep
• Increased capillary permeability
that foot elevated whenever possible to dimin-
• Decreased plasma oncotic pressure
ish the effects of gravity on capillary pressure
• Lymphatic obstruction
and to use a tight-fitting elastic stocking or
The most common cause of edema is elevated bandage around the ankle to increase tissue
capillary pressure, such as occurs during heart hydrostatic pressure (which opposes filtra-
failure or venous obstruction. Both conditions tion). Drugs (e.g., corticosteroids, antihis-
increase venous pressure, which is transmit- tamines) are sometimes used to block the
ted back to the capillaries, causing an increase release or action of paracrine substances that
in fluid filtration. Localized edema in tissues is increase capillary permeability following
commonly caused by injury or inflammation tissue injury or inflammation.

Klabunde_Chap08.indd 193 6/11/2011 10:04:15 AM


194 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

SUMMARY OF IMPORTANT CONCEPTS

• Diffusion is the primary mechanism for transcapillary fluid exchange, is


the exchange of gases (e.g., oxygen) determined by arterial and venous
and lipid-soluble substances across the pressures, and precapillary and
capillary barrier. The rate of diffusion postcapillary resistances.
is directly related to the concentration • Changes in venous pressure have a
difference of the molecule across the much greater quantitative influence
capillary wall. on capillary pressure than do similar
• Exchange of water and electrolytes changes in arterial pressure.
across capillaries (and postcapillary • Filtration occurs when the net driving
venules) occurs primarily by bulk flow force is greater than zero, which
through intercellular clefts ("pores") generally occurs at the arteriolar end
between endothelial cells. Bulk flow of the capillary. Reabsorption occurs
is governed by the same factors that when the net driving force is less than
determine the blood flow through zero, which generally occurs at the
vessels. venular end of the capillary where
• Movement of fluid across a capillary is capillary hydrostatic pressure is lower.
determined by hydrostatic and osmotic • An increase in tissue fluid volume
driving forces, the permeability of the (edema) occurs when the rate of fluid
capillary to fluid movement, and the filtration exceeds the sum of the rate of
surface area for exchange of fluid. fluid reabsorption and lymphatic flow.
• The net driving force that determines • Edema can be caused by increased
fluid movement is the net hydrostatic capillary hydrostatic pressure,
pressure difference across the capillary increased capillary permeability,
wall minus the opposing effective decreased plasma oncotic pressure, or
oncotic pressure difference across the lymphatic blockage.
capillary wall.
• Capillary hydrostatic pressure, which
plays a major role in regulating

REVIEW QUESTIONS

For each question, choose the one best 2. Which of the following can increase the
answer: rate of oxygen diffusion from blood to
tissue?
1. Which of the following mechanisms is a. Arteriolar vasodilation
most important quantitatively for the b. Decreased arteriolar P0 2
exchange of electrolytes across capillaries? c. Increased tissue P0 2
a. Bulk flow d. Decreased number of flowing capillaries
b. Diffusion
c. Osmosis
d. Vesicular transport

Klabunde_Chap08.indd 194 6/11/2011 10:04:15 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 195

3. A trauma patient in the hospital emer- 6. A malnourished, protein-deficient child


gency department is found to have a presents with abdominal swelling, which
venous oxygen saturation (SvO) of 50%; is determined to be caused by increased
arterial oxygen saturation (SaO) is 95%. fluid in the abdominal cavity (ascites).
The Sv02 value suggests that The ascites in this child is most likely
a. Cardiac output is low relative to the caused by
organ oxygen demand. a. Decreased blood volume.
b. Organ oxygen consumption is b. Decreased interstitial oncotic pres-
depressed. sure.
c. Tissue oxygen delivery is elevated. c. Increased capillary reabsorption of
d. Tissue oxygen extraction is reduced. fluid.
d. Reduced plasma oncotic pressure.
4. Net capillary fluid filtration is enhanced by
a. Decreased capillary plasma oncotic 7. A patient being treated for hypertension
pressure. with an arterial vasodilator develops
b. Decreased venous pressure. peripheral edema. The most likely cause
c. Increased precapillary resistance. of this edema is
d. Increased tissue hydrostatic pressure. a. Decreased capillary hydrostatic
pressure.
5. If capillary hydrostatic pressure= IS mm b. Decreased capillary filtration
Hg, capillary oncotic pressure = 28 mm constant.
Hg, tissue interstitial pressure= -5 mm c. Increased postcapillary/precapillary
Hg, and tissue oncotic pressure = 6 mm resistance ratio.
Hg (assume that a= I), these Starling d. Reduced venous pressure.
forces will result in
a. Net filtration.
b. Net reabsorption.
c. No net fluid movement.

1. The correct answer is "a" because 2. The correct answer is "a" because arte-
this is the mechanism by which fluid riolar vasodilation increases blood flow
and accompanying electrolytes move to the capillaries and increases capillary
through capillary intercellular junc- P0 2 , which increases the concentra-
tions. Choice "b" is incorrect because tion gradient for diffusion out of the
diffusion, although an important blood. Choice "b" is incorrect because
mechanism of exchange, is quan- decreased arteriolar P0 2 decreases capil-
titatively less important than bulk lary P0 2 and therefore the oxygen gradi-
flow. Furthermore, electrolytes are ent between the blood and tissue. Choice
charged ions and therefore do not dif- "c" is incorrect because increased tissue
fuse through membrane lipid bilayers. P0 2 decreases the concentration gradi-
Choice "c" is incorrect because osmo- ent for oxygen diffusion from the blood
sis concerns the movement of water. into the tissue. Choice "d" is incorrect
Choice "d" is incorrect because vesicu- because a decrease in the number of
lar transport is primarily for the trans- flowing capillaries decreases the surface
port of large macromolecules. area available for oxygen exchange.

Klabunde_Chap08.indd 195 6/11/2011 10:04:18 AM


196 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

3. The correct answer is "a" because when incorrect because the net driving force is
cardiac output is reduced, oxygen deliv- a negative value.
ery to organs is reduced (choice "c" is 6. The correct answer is "d" because
therefore incorrect) because of reduced protein deficiency leads to hypopro-
organ flow. If this flow reduction occurs teinemia, which reduces plasma oncotic
under conditions of normal organ oxygen pressure, thereby increasing net capil-
consumption, there will be an increase in lary fluid filtration. Choice "a" is incor-
oxygen extraction (choice "d" is there- rect because decreased blood volume
fore incorrect), which will reduce Sv02• caused by dehydration in this child
Choice "b" is incorrect because decreased would decrease capillary hydrostatic
oxygen consumption would increase pressure and fluid filtration. Choice "b"
Sv02• Note that the reduced Sv02 is not a is incorrect because decreased inter-
consequence of reduced Sa02 in this case stitial oncotic pressure opposes filtra-
because that value is within the normal tion. Choice "c" is incorrect because
range. increased capillary fluid reabsorption
4. The correct answer is "a" because capil- would decrease edema and ascites.
lary plasma oncotic pressure opposes fil- 7. The correct answer is "c" because
tration; therefore, decreasing this pressure increased postcapillary/precapillary
enhances filtration. Choices "b" and "c" resistance ratio caused by arterial (pre-
are incorrect because decreasing venous capillary) vasodilation increases capil-
pressure or increasing precapillary resis- lary hydrostatic pressure and fluid filtra-
tance reduces capillary hydrostatic pres- tion. Choice "a" is incorrect because a
sure, thereby decreasing filtration. Choice decreased capillary hydrostatic pressure
"d" is incorrect because increased tissue decreases fluid filtration. Choice "b" is
hydrostatic pressure opposes filtration. incorrect because a decreased capillary
5. The correct answer is "b" because the filtration constant decreases net filtra-
net driving force, calculated from the tion. Choice "d" is incorrect because a
given values, is -2 mm Hg, which causes reduced venous pressure decreases capil-
reabsorption. Choices "a" and "c" are lary pressure and filtration.

PROBLEM 8-1 toward the end of the hyperemic response,


During the period of ischemia, the P0 2 and the P0 2 will normalize.
oxygen content of the static blood within
PROBLEM 8-2
the microcirculation fall as oxygen diffuses
a) The net driving force, NDF = [(Pc- Pi)-
from the blood into the tissues. When blood
0"(7tc - 7t;) ]. Substituting the given values,
flow is restored, this oxygen-depleted blood
the NDF = 7 mm Hg.
is washed out of the tissue; a sample of this
blood will have reduced P0 2 and oxygen NDF=[22-(-3)]-0.9 [C26-6)]
content. During the phase of reactive hyper- =7mm Hg
emia, the increased blood flow and oxygen
delivery to the tissue is greater than what is b) Because the NDF is greater than zero, filtra-
needed to supply the oxidative metabolism tion is occurring.
of the tissue. If the ratio of oxygen delivery c) The net rate of fluid movement, ] = ~ · A
to oxygen consumption is increased above (NDF). Therefore, if the product of~ and
normal, then the venous oxygen content and A is doubled, then] (the filtration in this
P0 2 will increase. As the balance is restored problem) is doubled because the NDF "# 0.

Klabunde_Chap08.indd 196 6/11/2011 10:04:18 AM


CHAPTER 8 • EXCHANGE FUNCTION OF THE MICROCIRCULATION 197

SUGGESTED RESOURCES Circulation. New York: John Wiley & Sons, 1978.
Duling BR, Berne RM. Longitudinal gradients in periar- Michel CC, Curry RE. Microvascular permeability.
teriolar oxygen tension. A possible mechanism for Physiol Rev 1999;79:703–761.
the participation of oxygen in local regulation of Takahashi E, Sato K, Endoh H, Xu Z, Doi K. Direct
blood flow. Circ Res 1970;27:669–678. observation of radial intracellular PO2 gradients
Intaglietta M, Johnson PC. Principles of capil- in a single cardiomyocyte of the rat. Am J Physiol
lary exchange. In, Johnson PC, ed. Peripheral 1998;275:H225–H233.

Klabunde_Chap08.indd 197 6/11/2011 10:04:19 AM


CARDIOVASCULAR 0

9
:I:
>
INTEGRATION, ADAPTATION, "tl
-1
m
;:o
AND PATHO~I:IYSIOLOGY
Understanding the concepts presented in this chapter will enable the student to:
1. Describe the cardiac and vascular changes that occur during exercise, the
mechanisms responsible for those changes, and factors that can alter the
responses.
2. Describe how cardiovascular function is altered during pregnancy.
3. Describe the conditions that can lead to hypotension and the compensatory
mechanisms that are activated to restore arterial pressure.
4. Explain how positive feedback mechanisms can lead to irreversible shock and
death following severe hemorrhage.
5. Describe several different causes of hypertension and how it is treated.
6. Define systolic and diastolic ventricular failure and describe how these two
types of failure alter cardiac and vascular function at rest and during physical
exertion.
7. Describe the compensatory mechanisms that operate during heart failure.
8. Describe how heart valve stenosis and regurgitation affect cardiac function.

INTRODUCTION CARDIOVASCULAR RESPONSES


TO EXERCISE
Previous chapters emphasized physiologic con-
cepts concerning cardiac and vascular function The cardiovascular system must be able to
at the cellular and organ level. In addition, they respond to a wide range of demands placed
examined mechanisms, such as baroreceptors on it by the body. Previous chapters focused
and circulating hormones, that regulate over- on cardiovascular function in normal resting
all cardiovascular function. This chapter inte- states; however, physical activity is (or should
grates all the components of the cardiovascular be!) a normal, daily activity of humans. Physi-
system and shows how they work together to cal movement is associated with increases in
maintain normal perfusion of organs under the metabolic activity of contracting muscles.
conditions of increased organ demand for This increased metabolic activity is largely oxi-
blood flow (e.g., during exercise and preg- dative; therefore, the cardiovascular system
nancy) or during abnormal stressful condi- needs to increase blood flow and oxygen deliv-
tions such as hemorrhage. This chapter also ery to the contracting muscles.
examines changes that occur in cardiovascular The cardiovascular responses to physical
function during pathologic conditions such as activity are summarized in Table 9-1. If large
hypertension, heart failure, and valve disease. muscle groups are involved in the physical

198

Klabunde_Chap09.indd 198 6/11/2011 10:11:40 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 199

TABLE 9-1 SUMMARY OF occur as heart rate and inotropy increase


CARDIOVASCULAR (see Chapter 4). Therefore, all the cardio-
CHANGES DURING EXERCISE vascular changes occurring during physical
↑ Cardiac output activity ensure that active muscles are sup-
• ↑ heart rate (↑ sympathetic adrenergic plied with increased blood flow and oxygen
and ↓ parasympathetic activity) while maintaining normal, or even elevated,
• ↑ stroke volume (↑ CVP; ↑ inotropy; arterial pressures.
↑ lusitropy)
↑ Mean arterial pressure and pulse Mechanisms Involved in
pressure
• CO increases more than SVR Cardiovascular Response
decreases to Exercise
• ↑ stroke volume increases pulse
pressure
Four fundamental mechanisms are responsi-
ble for cardiovascular changes during physical
↑ Central venous pressure
activity: mechanical, metabolic, autonomic,
• Venous constriction (↑ sympathetic
adrenergic activity) and hormonal. When a person suddenly
• Muscle pump activity begins to run, cardiac output increases before
• Abdominothoracic pump metabolic and neurohumoral mechanisms are
↓ Systemic vascular resistance activated. This initial increase in cardiac out-
• Metabolic vasodilation in active muscle put results primarily from the skeletal mus-
and heart cle pump system, which enhances venous
• Cutaneous vasodilation (↓ sympathetic return and increases cardiac output by the
adrenergic activity) Frank-Starling mechanism. Within a few sec-
• Vasoconstriction in splanchnic, onds of the initiation of muscle contraction,
nonactive muscle, and renal circula-
metabolic mechanisms in the contracting
tion (↑ sympathetic adrenergic
activity) muscle dilate resistance vessels and increase
blood flow. At about the same time, changes
CVP, central venous pressure; CO, cardiac output; SVR,
systemic vascular resistance. begin to occur in the autonomic nervous sys-
tem (Fig. 9.1). Hypothalamic centers coor-
dinate a pattern of increased sympathetic
activity (e.g., running, bicycling), metabolic and decreased parasympathetic (vagal) out-
vasodilation (see Chapter 7) in these mus- flow from the medulla (see Chapter 6). This
cles causes a large fall in systemic vascular leads to an increase in heart rate, inotropy,
resistance. Ordinarily, this would cause arte- and lusitropy, which increases cardiac out-
rial pressure to fall; however, during physical put. Increased sympathetic efferent activity
activity, arterial pressure normally increases constricts resistance and capacitance vessels
because cardiac output increases at the same in the splanchnic circulation and nonactive
time that systemic vascular resistance begins muscles to help maintain arterial pressure
to fall. Furthermore, increased sympathetic and central venous pressure. In addition,
activity (see Chapter 6) leads to vasocon- during strenuous activity, sympathetic nerves
striction in the gastrointestinal tract, non- constrict the renal vasculature.
active muscles, and kidneys, which helps to Exercise activates several different hor-
limit the fall in systemic vascular resistance monal systems that affect cardiovascular
as well as shift blood flow to the active mus- function. Many of the hormonal systems
cles. Venous return to the heart is augmented are activated by the enhanced sympathetic
by venous constriction and by the skeletal activity. Because hormonal changes take
muscle and abdominothoracic pumps (see longer to occur, cardiovascular responses to
Chapter 5). Enhanced venous return enables these changes lag behind the direct effects
the cardiac output to increase by preventing of autonomic activation on the heart and
a fall in cardiac preload that would otherwise circulation.

Klabunde_Chap09.indd 199 6/11/2011 10:11:43 AM


200 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Central Hypothalamus Muscle and Joint


Command Afferents
Parasympathetic
Inhibition
Medulla Sympathetic
Activation

Heart Adrenals Blood Vessels


 Heart rate Catecholamine Arterial and venous
 Inotropy release constriction
 Lusitropy
■ FIGURE 9.1 Summary of adrenergic and cholinergic control mechanisms during exercise. The hypothala-
mus functions as an integrative center that receives information from the brain and muscle and joint recep-
tors, then modulates sympathetic and parasympathetic (vagal) outflow from the medulla. Sympathetic
nerves are activated leading to cardiac stimulation, arterial and venous constriction (not in active muscles),
and adrenal release of catecholamines; parasympathetic inhibition removes vagal tone on the heart.

Sympathetic nerves innervating the adre- Circulating norepinephrine constricts blood


nal medulla cause the secretion of epineph- vessels by binding preferentially to α1-
rine and lesser amounts of norepinephrine adrenoceptors in most organs. During exer-
into the blood (see Chapter 6). Plasma norep- cise, circulating levels of norepinephrine and
inephrine concentrations increase more than epinephrine can become very high so that the
10-fold during exercise. A large fraction of net effect on the vasculature is α-adrenocep-
this norepinephrine comes from sympathetic tor-mediated vasoconstriction, except in
nerves. Normally, most of the norepineph- those organs (e.g., heart and active skeletal
rine released by sympathetic nerves is taken muscle) in which metabolic mechanisms pro-
back up by the nerves (neuronal reuptake); duce vasodilation. It is important to note that
however, some of the norepinephrine can dif- vasoconstriction produced by sympathetic
fuse (“spillover”) into the capillary blood and nerves and circulating catecholamines does
enter the systemic circulation. This spillover not occur in the active skeletal muscle, coro-
is greatly enhanced when the level of sympa- nary circulation, or brain because blood flow
thetic activity is high in the body. The blood in these organs is primarily controlled by local
transports the epinephrine and norepineph- metabolic vasodilator mechanisms.
rine to the heart and other organs, where these Increased sympathetic activity stimulates
hormones act upon α- and β-adrenoceptors renal release of renin, which leads to the for-
to enhance cardiac function and either con- mation of angiotensin II. Increased angio-
strict or dilate blood vessels. In Chapter 6, tensin II increases renal sodium and water
we learned that epinephrine (at low concen- reabsorption by directly affecting renal func-
trations) binds to β2-adrenoceptors in skel- tion and by stimulating aldosterone secretion;
etal muscle, which causes vasodilation. At in addition, angiotensin II augments sympa-
high concentrations, epinephrine also binds thetic activity (see Chapter 6). Circulating
to postjunctional α1- and α2-adrenoceptors arginine vasopressin (antidiuretic hormone)
on blood vessels to cause vasoconstriction. also increases during exercise, most likely

Klabunde_Chap09.indd 200 6/11/2011 10:11:43 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 201

resulting from increased plasma osmolarity. and medullary autonomic control regions to
Although these hormonal changes promote enhance the sympathetic outflow to the heart
renal retention of sodium and water, espe- and systemic vasculature.
cially after prolonged periods of exercise, Arterial baroreceptor function is altered
blood volume often decreases during exercise during physical activity. Exercise normally is
(particularly in hot environments) because associated with a rise in both arterial pressure
of water loss through sweating and increased and heart rate. If arterial baroreceptor func-
respiratory exchange. tion were not modified, the increase in arterial
Two mechanisms operate to activate the pressure would result in a reflex bradycardia.
autonomic nervous system during exercise. Instead, the baroreceptor reflex is modified
One mechanism is referred to as “central (reset to a higher control point) by the central
command.” When physical activity is antici- nervous system (see Chapter 6).
pated or already under way, higher brain
centers (e.g., the cortex) relay information Steady-State Changes in
to hypothalamic centers to coordinate auto- Cardiovascular Function
nomic outflow to the cardiovascular system.
during Exercise
By this central command mechanism, antici-
pation of exercise can lead to autonomic Changes in cardiovascular function dur-
changes that increase cardiac output and ing physical activity depend upon the level
arterial pressure before exercise begins. This of physical exertion. If the level of physical
serves to prime the cardiovascular system exertion is expressed as workload, heart rate,
for exercise. A second mechanism involves cardiac output, and arterial pressure increase
muscle mechanoreceptors and chemorecep- in nearly direct proportion to the increase
tors. Once physical activity is underway, in workload (Fig. 9.2, panel A). In contrast,
these muscle receptors respond to changes in systemic vascular resistance falls as workload
muscle mechanical activity and tissue chemi- increases because of vasodilation in the active
cal environment (e.g., increased lactic acid), muscles. Ventricular stroke volume increases
and then relay that information to the central at low-to-moderate workloads and then pla-
nervous system via afferent fibers. This infor- teaus. Although not shown in Figure 9.2, the
mation is processed by the hypothalamus increase in stroke volume is responsible for an

A B
300 400 2000
Muscle
CO
300 1500
Percent Change

Percent Change

Percent Change

200 Skin
(Muscle)

HR
200 1000
100
SV 100 500
Renal
MAP Brain
0 0 0
SVR GI

Rest Moderate Heavy Rest Moderate Heavy

■ FIGURE 9.2 Systemic hemodynamic and organ blood flow responses at different levels of exercise
intensity. Panel A shows systemic hemodynamic changes. Systemic vascular resistance (SVR) decreases
because of vasodilation in active muscles; mean arterial pressure (MAP) increases because cardiac output
(CO) increases more than SVR decreases. CO and heart rate (HR) increase almost proportionately to the
increase in workload. Stroke volume (SV) plateaus at high heart rates. Panel B shows organ blood flow
changes. Muscle blood flow increases to very high levels because of active hyperemia; skin blood flow
increases because of the need to remove excess heat from the body. Sympathetic-mediated vasoconstric-
tion decreases gastrointestinal (GI) blood flow and renal blood flow. Brain blood flow changes very little.

Klabunde_Chap09.indd 201 6/11/2011 10:11:43 AM


202 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

increase in arterial pulse pressure that accom- within an individual, depending on the type
panies the increase in mean arterial pressure. of exercise and the environmental conditions.
Stroke volume may decline at very high Blood flow to major organs depends upon
workloads because ventricular filling time is the level of physical activity (Fig. 9.2, panel B).
reduced as heart rate increases. Decreased fill- During whole-body exercise (e.g., running),
ing time decreases ventricular filling (decreases the blood flow to the working muscles may
preload), which decreases stroke volume by increase more than 20-fold (see Chapter 7).
the Frank-Starling mechanism. This would At rest, muscle blood flow is about 20% of car-
prevent the heart from increasing cardiac out- diac output; this value may increase to 90%
put during physical activity if not for several during strenuous exercise. Coronary blood
mechanisms that work together to ensure flow can increase severalfold as the metabolic
that stroke volume is maintained and even demands of the myocardium increase and
increased as heart rate increases (Table 9-2). local regulatory mechanisms cause coronary
For example, during a physical activity such vasodilation. The need for increased blood
as running, enhanced venous return by the flow to active muscles and the coronary cir-
muscle pump and abdominothoracic pump culation would exceed the reserve capacity of
systems helps to maintain preload despite the heart to increase its output if not for blood
the increase in heart rate (see Chapter 5). flow being reduced to other organs. During
Furthermore, increased atrial and ventricular exercise, blood flow decreases to the splanch-
inotropy enhances ventricular stroke volume nic circulation (gastrointestinal, splenic, and
and ejection fraction, and increased lusitropy hepatic circulations) and nonactive skel-
helps to augment ventricular filling. When etal muscle as workload increases. This is
the heart rate approaches its maximal rate, the brought about primarily by increased sym-
effects of reduced filling time can predomi- pathetic nerve activity to these organs. With
nate over these compensatory mechanisms, very strenuous exercise, renal blood flow
thereby compromising ventricular filling and is also decreased by sympathetic-mediated
reducing stroke volume. The point at which vasoconstriction.
increased heart rate begins to decrease stroke Skin blood flow increases with increasing
volume varies considerably among individu- workloads, but it can then decrease at very
als because of age, health, and physical con- high workloads, especially in hot environ-
ditioning. Furthermore, this point can vary ments. Increases in cutaneous blood flow are
controlled by hypothalamic thermoregula-
tory centers (see Chapter 7). During physical
TABLE 9-2 MECHANISMS MAINTAINING
activity, increased blood temperature is sensed
STROKE VOLUME AT HIGH by thermoreceptors in the hypothalamus. To
HEART RATES DURING enhance heat loss through the skin, the hypo-
EXERCISE thalamus decreases sympathetic nerve activity
• Increased venous return promoted by the to cutaneous blood vessels, which increases
abdominothoracic and skeletal muscle skin blood flow. At the same time, activation
pumps maintains central venous pressure of sympathetic cholinergic nerves to the skin
and therefore ventricular preload. causes sweating.
• Venous constriction (decreased venous While cutaneous vasodilation is essential
compliance) maintains central venous
for thermoregulation during physical activ-
pressure.
• Increased atrial inotropy augments atrial
ity, this requirement must be balanced by the
filling of the ventricles. need to maintain arterial pressure. Cutaneous
• Increased ventricular inotropy decreases vasodilation contributes to the fall in systemic
end-systolic volume, which increases vascular resistance primarily brought about
stroke volume and ejection fraction. by vasodilation in active muscles. If increased
• Enhanced rate of ventricular relaxation cardiac output is unable to maintain arterial
(lusitropy) aids in filling.
pressure at very high workloads, baroreceptor

Klabunde_Chap09.indd 202 6/11/2011 10:11:43 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 203

mechanisms restore sympathetic tone to the system cannot operate to promote venous
skin and decrease its blood flow. Although return, and so, cardiac output increases rela-
this may help to preserve arterial pressure tively little. Furthermore, the abdominotho-
temporarily; reduced heat exchange through racic pump does not contribute to enhancing
the skin can lead to dangerous elevations in venous return, particularly if the subject
core temperature, resulting in organ damage holds his or her breath during the forceful
and loss of autonomic control. Heat stroke contraction, effectively performing a Valsalva
is a potentially lethal condition that occurs maneuver. Unlike dynamic exercise, static
when core temperatures rise above l05°F. exercise leads to a large increase in systemic
vascular resistance, particularly if a large mus-
CASE 9-1 cle mass is being contracted at maximal effort.
The increased systemic vascular resistance
A 45-year-old male patient with type 2
results from enhanced sympathetic adrener-
diabetes is diagnosed with autonomic
gic activity to the peripheral vasculature and
neuropathy, which impairs autonomic
from mechanical compression of the vascula-
function. He complains of becoming
ture in the contracting muscles. As a result,
weak and "light headed" when he
systolic arterial pressure may increase to over
performs physical work such as mowing
250 mm Hg during forceful isometric contrac-
the lawn. Explain how this patient's
tions, particularly those involving large mus-
autonomic dysfunction may account for
cle groups. This acute hypertensive state can
his inability to be engaged in normal
produce vascular damage (e.g., hemorrhagic
physical activities.
stroke) in susceptible individuals. In con-
trast, dynamic exercise leads to only modest
Factors Influencing increases in arterial pressure.
Cardiovascular Response Body posture also influences how the cardi-
ovascular system responds to exercise because
to Exercise
of the effects of gravity on venous return and
The cardiovascular changes associated with central venous pressure (see Chapter 5). When
physical activity are modified by many dif- a person exercises in the supine position (e.g.,
ferent factors. The level of activity; which is swimming), central venous pressure is higher
commonly expressed as work performed or than when the person is exercising in the
whole-body oxygen consumption, affects the upright position (e.g., running). In the resting
cardiac and vascular responses. Several other state before the physical activity begins, ven-
important factors influence cardiovascular tricular stroke volume is higher in the supine
responses at a given workload. position than in the upright position owing
The type of exercise significantly affects to increased right ventricular preload. Fur-
cardiovascular responses. The previous sec- thermore, the resting heart rate is lower in the
tion described the cardiovascular responses to supine position. When exercise commences
dynamic exercise such as running, walking, in the supine position, the stroke volume can-
bicycling, or swimming. Dynamic exercise not be increased appreciably by the Frank-
results in joint movement as muscles contract Starling mechanism because the high resting
rhythmically. In contrast, muscle contraction preload reduces the reserve capacity of the
without joint movement (isometric or static ventricle to increase its end-diastolic volume.
contraction) elicits a different cardiovascular Stroke volume still increases during exercise
response. An example of this activity would although not as much as when exercising
be trying to lift a very heavy weight at maxi- while standing because, in the supine posi-
mal effort (e.g., bench or leg press).This type tion, the increased stroke volume is resulting
of activity does not incorporate rhythmic primarily from increases in inotropy and ejec-
contraction of synergistic and antagonistic tion fraction with minimal contribution from
muscle groups; therefore, the muscle pump the Frank-Starling mechanism. Because heart

Klabunde_Chap09.indd 203 6/11/2011 10:11:43 AM


204 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

rate is initially lower in the supine position, Environmental conditions can significantly
the percent increase in heart rate is greater in alter cardiovascular responses to exercise.
the supine position, which compensates for High altitudes, for example, decrease maximal
the reduced ability to increase stroke volume. stroke volume and cardiac output. The reason
Overall, the change in cardiac output during for this is that arterial PO2 and oxygen content
exercise, which depends upon the fractional are reduced at higher elevations because of
increases in both stroke volume and heart decreased atmospheric pressure. This decreases
rate, is not appreciably different in the supine oxygen delivery to tissues, particularly to con-
versus standing position. tracting muscle (both skeletal and cardiac),
Physical conditioning permits a person to thereby resulting in insufficient oxygena-
achieve a higher cardiac output, whole-body tion at lower workloads. Myocardial hypoxia
oxygen consumption, and workload than decreases maximal inotropy, which results
a person who has a sedentary lifestyle. The in reduced stroke volume. Reduced oxygen
increased cardiac output capacity is a conse- delivery to exercising muscle reduces exercise
quence, in part, of increased ventricular and capacity in the muscle and results in increased
atrial responsiveness to inotropic stimulation production of lactic acid as the muscle switches
by sympathetic nerves. Conditioned individu- over to anaerobic metabolism in the absence of
als also have stronger, hypertrophied hearts, adequate oxygen; that is, the anaerobic thresh-
much like what happens to skeletal muscle old is reached at a lower workload.
in response to weight training. Coupled with Increased temperature and humidity affect
enhanced capacity for promoting venous cardiovascular responses during exercise by
return by the muscle pump system, these diverting a greater fraction of cardiac output
cardiac changes permit highly conditioned to the skin to enhance heat removal from
individuals to achieve ventricular ejection the body. This decreases the availability of
fractions that can exceed 90% during exer- blood flow for the contracting muscles. With
cise. In comparison, a sedentary individual elevated temperature and humidity, maximal
may not be able to increase ejection fraction cardiac output and oxygen consumption are
above 75%. reached at lower workloads, thereby reducing
In a conditioned individual, resting heart exercise capacity as well as endurance. Fur-
rate is lower and resting stroke volume is thermore, dehydration can accompany high
higher than in a sedentary person—resting temperatures. Dehydration reduces blood
cardiac output is not necessarily different. volume and central venous pressure, which
Because the maximal heart rate of a condi- attenuates the normal increase in cardiac out-
tioned individual is similar to that of a sed- put associated with exercise. This can lead
entary individual of the same age, the lower to a fall in arterial pressure and heat exhaus-
resting heart rates of a conditioned person tion. Signs of heat exhaustion include general
allow for a greater percent increase in heart fatigue, muscle weakness, nausea, and mental
rate during exercise. This greater capacity to confusion; it usually results from dehydration
increase heart rate, coupled with a greater and loss of sodium chloride associated with
capacity to enhance stroke volume, permits physical activity in a hot environment—core
a conditioned individual to achieve maximal temperature is not necessarily elevated.
cardiac outputs that can be 50% higher than Increased age reduces maximal exer-
those found in sedentary people. Another cise capacity. Maximal oxygen consump-
important distinction between a sedentary tion decreases about 40% between 20 and
and conditioned person is that for a given 70 years of age. There are many reasons for
workload, the conditioned person has a lower this decline. With increasing age, maximal
heart rate. Furthermore, a conditioned per- heart rate decreases. Maximal heart rate is
son is able to sustain higher workloads for a approximately 220 beats/min minus the age
longer duration and recover from the exercise of a person. Therefore, the maximal heart rate
much more rapidly. of a 70-year-old person is about 25% lower

Klabunde_Chap09.indd 204 6/11/2011 10:11:44 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 205

than the maximal heart rate of a 20-year-old +50


person. Increasing age also reduces maximal CO
stroke volume because of impaired ventricu- +25 SV

Percent Change
lar filling (decreased ventricular compliance) HR
and reduced inotropic responsiveness to sym- 0
pathetic stimulation. Together, these changes
MAP
reduce maximal cardiac output substantially. -25
Older individuals have reduced skeletal mus- SVR
cle mass as well as decreased maximal muscle -50
blood flow per unit weight of muscle. A reduc- First Second Third
Trimester Trimester Trimester
tion in vasodilatory capacity of resistance ves-
sels in skeletal muscle in older persons may be ■ FIGURE 9.3 Changes in maternal hemody-
related to reduced endothelial production or namics during pregnancy. Early in the course of
pregnancy, cardiac output (CO) increases because
bioavailability of nitric oxide and altered vascu- stroke volume (SV) increases owing to an increase
lar smooth muscle responsiveness to metabolic in blood volume; systemic vascular resistance
vasodilators. Although increasing age inevita- (SVR) and mean arterial pressure (MAP) decrease.
bly limits exercise capacity, exercise habits and Heart rate (HR) gradually increases throughout
pregnancy; SV declines as HR increases.
general health can significantly influence the
decline in maximal cardiac output with age.
Gender influences cardiovascular responses increases in stroke volume. By the third tri-
to exercise. Generally, males can reach and mester, however, stroke volume may be only
sustain significantly higher workloads and slightly elevated. At this stage of pregnancy,
maximal oxygen consumptions than can the increased cardiac output is sustained by
females. Maximal cardiac outputs are about an elevated heart rate, which may increase by
25% less in females, although the maximal 10 to 20 beats/min.
heart rates are similar. This difference is partly Cardiac output increases because blood
owing to increased skeletal muscle mass and volume (and therefore ventricular preload)
to increased cardiac mass in males. increases dramatically during pregnancy. By
Finally, cardiac disease can significantly week 6, blood volume may be increased by
limit exercise capacity. As described later in this 10%. By the end of the third trimester, blood
chapter, diseases that impair cardiac function volume may be increased by 50%. The increase
(e.g., heart failure) can limit the ability of the in blood volume is brought about by estrogen-
heart to increase cardiac output during physi- mediated activation of the renin-angiotensin-
cal activity. Arrhythmias, such as atrial fibril- aldosterone system, which increases sodium
lation or AV nodal block, can reduce exercise and water retention by the kidneys.
capacity by decreasing maximal cardiac output. Although cardiac output is elevated, mean
arterial pressure generally falls owing to a
MATERNAL CHANGES IN disproportionate decrease in systemic vascu-
CARDIOVASCULAR FUNCTION lar resistance. The fall in systemic vascular
DURING PREGNANCY resistance may be caused in part by hormonal
changes that dilate resistance vessels; however,
Pregnancy causes significant changes in the the major factor contributing to the reduced
cardiovascular system (Fig. 9.3). Increased resistance is the development of low-resistance
uterine mass and the developing fetus require uterine circulation, particularly in the later
large amounts of blood flow. To supply this stages of pregnancy. Diastolic pressure falls
flow, cardiac output increases by 30% to more than systolic pressure because of reduced
50% during the first and second trimesters systemic vascular resistance, so there is an
and then plateaus during the third trimes- increase in pulse pressure. Increased pulse
ter. In the first half of the pregnancy, the pressure results from the increase in stroke
cardiac output is primarily increased through volume during the first and second trimesters.

Klabunde_Chap09.indd 205 6/11/2011 10:11:44 AM


206 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Pregnancy significantly alters the cardio- vascular resistance, a decrease in either will
vascular responses to exercise. Because cardiac reduce arterial pressure (see Chapter 5).
output at rest is substantially elevated, there is A reduction in systemic vascular tone or
less capacity for it to increase during exercise. impaired vasoconstrictor responsiveness to
In addition, compression of the inferior vena baroreceptor reflexes can lead to hypoten-
cava caused by an elevated intra-abdominal sion. For example, septic shock (or Systemic
pressure, particularly during the third trimes- Inflammatory Response Syndrome, SIRS),
ter, limits venous return and thereby prevents which usually results from a bacterial infec-
stroke volume from increasing as it normally tion in the blood, causes a loss of vascular
would during exercise. Compression of the tone and hypotension. Septic shock is caused
inferior vena cava, especially in the supine by the release of bacterial endotoxins (e.g.,
position, can also diminish venous return at lipopolysaccharide) that activate the inflam-
rest, thereby reducing cardiac output and arte- matory cascade. This leads to the produc-
rial pressure (supine hypotensive syndrome). tion of cytokines (e.g., tumor necrosis factor,
interleukins) and excessive amounts of nitric
HYPOTENSION oxide, causing systemic vasodilation. Severe
allergic reactions can lead to anaphylactic
shock. Another cause of vasodilatory circula-
Causes of Hypotension
tory shock is damage to the spinal cord sym-
Hypotension is often defined clinically as pathetic tracts (neurogenic shock) resulting
a systolic arterial pressure <90 mm Hg, or in loss of vascular sympathetic tone. Systemic
a diastolic pressure <60 mm Hg. There are vascular resistance can also be decreased if
many causes of hypotension as summarized autonomic dysfunction occurs. For example,
in Figure 9.4. Because arterial pressure is in diabetic individuals having autonomic
the product of cardiac output and systemic neuropathy, baroreceptor-mediated reflex

Hypotension

Cardiac Systemic
¯ Output ¯ Vascular Resistance
• Circulatory Shock
- sepsis
- anaphylaxis
- neurogenic
Stroke Heart • Autonomic Dysfunction
¯ Volume ¯ Rate
• Arrhythmias
- sinus bradycardia
¯ Preload ¯ Inotropy - AV nodal block
- ventricular fibrillation
• Hypovolemia • Heart Failure
- hemorrhage • Cardiogenic Shock
- dehydration • Autonomic Dysfunction
• Volume Redistribution
- postural changes
- impaired venous return
• Arrhythmias
- atrial fibrillation
- tachycardia

■ FIGURE 9.4 Mechanisms and causes of hypotension. Ultimately, hypotension occurs because there is a
reduction in cardiac output, systemic vascular resistance, or both.

Klabunde_Chap09.indd 206 6/11/2011 10:11:44 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 207

vasoconstriction may be impaired, which can vasopressin. These hormone systems serve to
result in a fall in arterial pressure when the increase blood volume and reinforce the vaso-
person stands up (orthostatic hypotension) constriction caused by increased sympathetic
and when the person exercises. activity. Neurohumoral compensatory mecha-
Hypotension can also occur when cardiac nisms increase arterial pressure and thereby
output is reduced by a decrease in either heart help to maintain normal cerebral and coronary
rate or stroke volume. Ventricular rate can be perfusion at the expense of reduced blood flow
reduced by sinus bradycardia, which may to less essential organs. The following dis-
be caused by excessive vagal activation of the cussion specifically addresses compensatory
SA node. A vasovagal reflex can lower heart mechanisms in hypotension caused by hemor-
rate and arterial pressure sufficiently to cause rhage-induced hypovolemia.
syncope (see Chapter 6). Second- and third- The fall in blood volume during hemor-
degree AV nodal blockade (see Chapter 2) rhage reduces central venous pressure, which
reduce ventricular rate. Ventricular fibrilla- reduces cardiac filling and stroke volume
tion prevents coordinated ventricular beats so through the Frank-Starling mechanism. The
the effective ventricular rate is zero. fall in cardiac output causes the arterial pres-
Stroke volume can be reduced by decreases sure fall. The baroreceptor reflex is the first
in either inotropy or ventricular filling compensatory mechanism to become acti-
(preload) (see Chapter 4). Reduced inotropy vated in response to blood loss (Fig. 9.5).
occurs during heart failure (systolic failure) This reflex occurs within seconds of a fall in
or when autonomic dysfunction decreases arterial pressure. As described in Chapter 6,
sympathetic outflow to the heart. A sudden a reduction in mean arterial pressure and
loss of mechanical efficacy by the heart, as arterial pulse pressure decreases the firing
occurs following acute ischemic damage (e.g., of arterial baroreceptors. This activates the
myocardial infarction), is a frequent cause of sympathetic nervous system and inhibits
cardiogenic shock. Decreased preload can vagal influences to the heart, thereby increas-
be caused by several conditions: (1) hypov- ing heart rate and inotropy. It is important to
olemia, which results from blood loss (hem- note that cardiac stimulation alone does not
orrhage) or dehydration; (2) a redistribution lead to a significant increase in cardiac out-
of blood volume, as occurs when a person put. For cardiac output to increase, some
stands up (orthostatic hypotension; see mechanism must increase central venous
Chapter 5); (3) reduced venous return, which pressure and therefore filling pressure for the
can result from compression of the vena cava ventricles. This is accomplished, at least ini-
(e.g., supine hypotensive syndrome during tially following hemorrhage, by an increase in
pregnancy); and (4) tachyarrhythmias, such venous tone produced by sympathetic stimu-
as atrial fibrillation and ventricular tachycar- lation of the venous capacitance vessels. The
dia, which reduce ventricular filling. partially restored central venous pressure
increases stroke volume through the Frank-
Starling mechanism. The increased preload,
Compensatory Mechanisms
coupled with cardiac stimulation, attenuates
during Hypotension
the decline in cardiac output. The partially
When hypotension occurs, the body attempts to compensated cardiac output along with sys-
restore arterial pressure by activating neurohu- temic vasoconstriction causes the arterial
moral compensatory mechanisms (see Chap- pressure to increase toward its normal value.
ter 6). Initial, short-term mechanisms involve Although the baroreceptor reflex can
baroreceptor reflex activation of sympathetic respond quickly to a fall in arterial pressure
nerves, which constrict systemic vascular beds and provide initial compensation, the long-
and stimulate the heart. More slowly activated, term recovery of cardiovascular homeostasis
long-term compensatory mechanisms include requires activation of hormonal compensatory
the renin-angiotensin-aldosterone system and mechanisms to restore blood volume through

Klabunde_Chap09.indd 207 6/11/2011 10:11:44 AM


208 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Blood Loss

+ ¯ Central Venous
Pressure

¯ Stroke +
Volume

¯ Cardiac +
Output

+ ¯ Arterial
Pressure

¯ ¯ Baroreceptor
Systemic Vascular Firing
Resistance
¯
Sympathetic ¯ Parasympathetic
¯
Venous
Tone ¯
Heart Rate
¯ and
Contractility
■ FIGURE 9.5 Activation of baroreceptor mechanisms following acute blood loss (hemorrhage). Blood
loss reduces central venous pressure (cardiac preload), which decreases cardiac output and arterial pres-
sure. Reduced firing of arterial baroreceptors activates the sympathetic nervous system, which stimulates
cardiac function, and constricts resistance and capacitance vessels. These actions help to elevate (+) the
reduced central venous pressure, stroke volume, cardiac output, and arterial pressure, and thereby help to
restore arterial pressure.

renal mechanisms (Fig. 9.6). Some of these Working together, angiotensin II, aldosterone,
humoral systems also reinforce the barorecep- and vasopressin cause the kidneys to retain
tor reflex by causing cardiac stimulation and sodium and water, thereby increasing blood
vasoconstriction. volume, cardiac preload and cardiac output.
The renin-angiotensin-aldosterone system Increased vasopressin also stimulates thirst
is activated by increased renal sympathetic so that more fluid is ingested. The renal and
nerve activity and renal artery hypotension vascular responses to these hormones are fur-
via decreased sodium delivery to the macula ther enhanced by decreased secretion of atrial
densa, which releases renin leading to the natriuretic peptide by the atria, resulting from
formation of angiotensin II (see Chapter 6). decreased atrial stretch associated with the
Increased circulating angiotensin II constricts hypovolemic state.
the systemic vasculature directly by binding The vascular responses to angiotensin II
to AT1 receptors and indirectly by enhancing and vasopressin occur rapidly in response
sympathetic effects. Angiotensin II stimulates to increased plasma concentrations of these
aldosterone secretion. Vasopressin secre- vasoconstrictors. The renal effects of angioten-
tion is stimulated by reduced atrial stretch, sin II, aldosterone, and vasopressin, in contrast,
sympathetic stimulation, and angiotensin II. occur more slowly as decreased sodium and

Klabunde_Chap09.indd 208 6/11/2011 10:11:44 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 209

Blood Loss

Blood + Arterial + CO
Volume Pressure SVR
+
SVR +
Vasopressin

SVR Adrenal
Pituitary Kidney Medulla

Renal Na+ &


Angiotensin II Renin Catecholamines
H2O Retention
(Epi,NE)
Adrenal
Aldosterone
Cortex
■ FIGURE 9.6 Activation of humoral mechanisms following acute blood loss (hemorrhage). Decreased
arterial pressure activates the sympathetic nervous system (baroreceptor reflex), which stimulates cat-
echolamine release (Epi, epinephrine; NE, norepinephrine) from the adrenal medulla. This increases cardiac
output (CO) and systemic vascular resistance (SVR), which elevates (+) arterial pressure. Renin release
is stimulated by the enhanced sympathetic activity, increased circulating catecholamines, and hypoten-
sion; this leads to the formation of angiotensin II and aldosterone. Vasopressin release from the posterior
pituitary is stimulated by angiotensin II, reduced atrial pressure (not shown), and increased sympathetic
activity (not shown). These hormones act together to increase blood volume through their renal actions
(sodium and water retention), which elevates arterial pressure. Angiotensin II and vasopressin also elevate
arterial pressure by increasing SVR. These changes in systemic vascular resistance, blood volume, and
cardiac output thereby help to restore the arterial pressure.

water excretion gradually increases blood in the carotid body chemoreceptors, which
volume over several hours and days. results from reduced carotid body blood flow,
Enhanced sympathetic activity stimulates stimulates chemoreceptor firing. If cerebral
the adrenal medulla to release catecholamines perfusion becomes impaired and the brain
(epinephrine and norepinephrine). This becomes ischemic, intense sympathetic-
causes cardiac stimulation (β1-adrenoceptor mediated vasoconstriction of the systemic
mediated) and peripheral vasoconstriction vasculature will result.
(α-adrenoceptor mediated), and contributes Reduced arterial and venous pressures,
to the release of renin by the kidneys through coupled with a decrease in the post-to-
renal β-adrenoceptors. precapillary resistance ratio, decreases capil-
Other mechanisms besides the barorecep- lary hydrostatic pressures (see Chapter 8).
tor reflex and hormones have a compensa- This leads to enhanced capillary fluid reab-
tory role in hemorrhagic hypotension. Severe sorption. This mechanism can result in up to
hypotension can lead to activation of chem- 1 L/h of fluid being reabsorbed back into the
oreceptors (see Chapter 6). Low perfusion intravascular compartment, which can lead
pressures and reduced organ blood flow cause to a significant increase in blood volume and
increased production of lactic acid as organs arterial pressure after a few hours. Although
are required to switch over to anaerobic glyco- capillary fluid reabsorption increases intra-
lysis for the production of ATP. Acidosis stim- vascular volume and serves to increase arte-
ulates peripheral and central chemoreceptors, rial pressure, it also leads to a reduction in
leading to increased sympathetic activity to hematocrit and dilution of plasma proteins
the systemic vasculature. Stagnant hypoxia until new blood cells and plasma proteins

Klabunde_Chap09.indd 209 6/11/2011 10:11:44 AM


210 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

are synthesized. The reduced hematocrit person's blood volume is lost by hemorrhage,
decreases the oxygen-carrying capacity of the arterial pressure may begin to recover as com-
blood. Eventually; dilution of plasma proteins pensatory mechanisms are activated; how-
decreases plasma oncotic pressure sufficiently ever, the recovery may last only an hour or
to limit the amount of fluid reabsorption. two before arterial pressure once again falls,
Most of the compensatory responses causing death despite heroic interventions.
described above occur regardless of the cause This secondary fall in arterial pressure
of hypotension; however, the ability of the results from the activation of decompensatory
heart and vasculature to respond to a specific mechanisms. These decompensatory mecha-
compensatory mechanism may differ depend- nisms are positive feedback cycles, in con-
ing upon the cause of the hypotension. For trast to the negative feedback control offered
example, if hypotension is caused by cardio- by compensatory mechanisms. A negative
genic shock (a form of acute heart failure) sec- feedback mechanism attempts to restore a
ondary to a myocardial infarction, the heart controlled variable (in this case arterial pres-
will not be able to respond to sympathetic sure) to its normal value, whereas a positive
stimulation in the same manner as would a feedback mechanism causes the controlled
normal heart. As another example, vascu- variable to move even farther away from its
lar responsiveness to sympathetic-mediated control point.
vasoconstriction is significantly impaired in a In the case of severe hemorrhagic shock
person in septic shock. Finally, drugs that a and some other forms of hypotensive shock
person is taking for hypertension (e.g., beta- (e.g., cardiogenic and septic shock), several
blockers, alpha-blockers, ACE inhibitors) can potential positive feedback mechanisms can
interfere with neurohumoral compensatory lead to irreversible shock and death. These
responses to hypotension. mechanisms include cardiac depression, sym-
pathetic escape, metabolic acidosis, cerebral
CASE 9·2 ischemia, rheological factors, and systemic
A patient who is being aggressively inflammatory responses.
treated for severe hypertension with Figure 9. 7 illustrates how cardiac depres-
a diuretic, an angiotensin-converting sion and sympathetic escape can lead to
enzyme (ACE) inhibitor, and a decompensation in severe hemorrhage. If
calcium channel blocker is in a serious mean arterial pressure falls below 60 mm Hg,
automobile accident that causes coronary blood flow is insufficient to sup-
significant intra-abdominal bleeding. port the metabolic demands of the heart
How might these drugs affect the because this pressure is below the coro-
compensatory mechanisms that are nary autoregulatory range (see Chapter 7).
activated following hemorrhage? How Reduced coronary blood flow causes myocar-
might this alter the course of this dial hypoxia, which impairs cardiac contrac-
patient's recovery? tions (reduces inotropy). When this occurs,
stroke volume and cardiac output decrease,
causing additional decreases in arterial pres-
Decompensatory Mechanisms sure and coronary perfusion-a positive feed-
Following Severe and Prolonged back cycle. Also shown in Figure 9. 7 is the
effect of hypotension on organ blood flow.
Hypotension
Hypotension decreases organ blood flow by
Severe, prolonged hypotension can lead to decreasing perfusion pressure and through
irreversible shock and death. This occurs baroreceptor-mediated sympathetic activa-
when normal compensatory mechanisms (and tion that constricts resistance vessels. This
additional medical resuscitation) are unable reduced flow causes tissue hypoxia. The more
to restore arterial pressure to adequate levels hypoxic a tissue becomes and the longer it
in a timely manner. For example, if 40% of a remains hypoxic (especially under low flow

Klabunde_Chap09.indd 210 6/11/2011 10:11:45 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 211

Severe
Hemorrhage

¯ Cardiac ¯ Organ
Output Blood Flow

¯ Arterial Tissue
¯ Inotropy Hypoxia
Pressure

¯ Coronary Vasodilation
Perfusion (Sympathetic Escape)

■ FIGURE 9.7 Positive feedback decompensatory mechanisms triggered by severe hypotension. Impair-
ment of coronary perfusion leads to a loss in cardiac inotropy and an additional decrease in cardiac
output and pressure. Prolonged tissue ischemia (reduced blood flow) and hypoxia caused by hypoten-
sion and sympathetic vasoconstriction lead to vasodilation (sympathetic escape), which reduces systemic
vascular resistance and arterial pressure.

conditions), the greater the buildup of vaso- regulatory centers cease to function
dilator metabolites. These metabolites even- because of the lack of oxygen. This with-
tually override the sympathetic-mediated drawal of sympathetic tone causes arterial
vasoconstriction (sympathetic escape), and pressure to fall, which further reduces cer-
blood flow begins to increase within the ebral perfusion.
organ. When this sympathetic escape occurs • Reduced organ perfusion during hypoten-
within major organs of the body (e.g., skeletal sion and intense sympathetic vasocon-
muscle and gastrointestinal tract), systemic striction causes increased blood viscosity
vascular resistance falls. This reduces arterial within the microcirculation, microvascular
pressure and further reduces organ perfusion, plugging by leukocytes and platelets, and
which leads to further vasodilation and hypo- disseminated intravascular coagulation.
tension—a positive feedback cycle. Low-flow states within the microcircula-
Several other positive feedback cycles can tion cause red blood cells to adhere to
contribute to irreversible shock: each other, which increases the viscosity
of the blood. Furthermore, low-flow states
• Prolonged hypotension with accompany-
enhance leukocyte–endothelial adhe-
ing tissue hypoxia results in metabolic
sion and platelet–platelet adhesion. This
acidosis as organs begin to generate ATP
reduces organ perfusion even more and
by anaerobic pathways. Acidosis impairs
can lead to ischemic damage and stimula-
cardiac contraction and vascular smooth
tion of inflammatory processes, which can
muscle contraction, which decreases car-
further enhance metabolic acidosis and
diac output and systemic vascular resist-
impair cardiac and vascular function.
ance, thereby lowering arterial pressure
even more. In summary, the body responds to hypoten-
• Cerebral ischemia and hypoxia during sion by activating neurohumoral mechanisms
severe hypotension, although initially that serve as negative feedback, compensatory
causing strong sympathetic activation, mechanisms to restore arterial pressure. With
eventually results in depression of all severe hypotension, positive feedback control
autonomic outflow as the cardiovascular mechanisms may become operative. These

Klabunde_Chap09.indd 211 6/11/2011 10:11:45 AM


212 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

mechanisms counteract the compensatory HYPERTENSION


mechanisms and eventually lead to an addi-
tional reduction in arterial pressure. High blood pressure (hypertension) is a con-
dition that afflicts about one-third of Ameri-
can adults and is a leading cause of morbidity
Physiologic Basis for Therapeutic
and mortality. Hypertension is much more
Intervention
than a “cardiovascular disease” because it can
Treatment for hypotension depends upon damage other organs such as kidney, brain,
the underlying cause of the hypotension. and eye. One-third of hypertensive people
If hypotension is caused by hypovolemia are not aware of being hypertensive because
owing to hemorrhage or excessive fluid it is usually asymptomatic until the damaging
loss (e.g., dehydration), increasing blood effects of hypertension (such as stroke, myo-
volume by administration of blood or flu- cardial infarction, renal dysfunction, visual
ids becomes a treatment priority. Restoring disturbances, etc.) are observed.
blood volume increases preload on the heart The term “hypertension” is applied to ele-
and thereby increases cardiac output, which vations in diastolic or systolic pressures above
is reduced in hypovolemic states. Admin- normal values. Normal arterial pressure is
istration of fluids is occasionally accom- defined as a systolic pressure <120 mm Hg (but
panied by the administration of pressor >90 mm Hg) and diastolic pressure <80 mm Hg
agents. These drugs increase arterial pres- (but >60 mm Hg). Diastolic pressures of 80 to
sure by increasing systemic vascular resist- 89 mm Hg and systolic pressures of 120 to
ance (e.g., α-adrenoceptor agonists such as 139 mm Hg are considered prehypertension.
norepinephrine and phenylephrine; or vaso- Hypertension is defined as diastolic or systolic
pressin) or by stimulating cardiac function pressures ≥90 or 140 mm Hg, respectively.
(e.g., β-adrenoceptor agonists such as dobu- Both diastolic and systolic hypertension have
tamine). Treatment of hypotension caused been shown to be significant risk factors for
by cardiogenic shock can include drugs that causing other cardiovascular disorders such as
stimulate the heart (e.g., β-adrenoceptor stroke and myocardial infarction. Mean arte-
agonists such as dobutamine or dopamine, rial pressure is usually not discussed in the
or cAMP-dependent phosphodiesterase context of hypertension because it is not nor-
inhibitors such as milrinone that inhibit the mally measured in a patient.
degradation of cAMP); however, depending Chronic hypertension is caused by
upon the magnitude of the hypotension, increases in systemic vascular resistance and
either pressor or depressor agents may be cardiac output. The elevation in cardiac out-
used. Because the primary cause of hypo- put is normally caused by an increase in blood
tension in cardiogenic shock is impaired volume, which increases ventricular preload
cardiac function, drugs such as phosphodi- and stroke volume. It is important to note
esterase inhibitors that stimulate the heart that to sustain a hypertensive state it is neces-
and dilate arterial vessels can improve car- sary to increase blood volume through renal
diac function by enhancing inotropy and retention of sodium and water. Evidence for
decreasing afterload on the heart. Systemic this comes from studies showing that eleva-
vasodilators, however, cannot be used alone tions in arterial pressure produced by infus-
if the hypotension is severe, because arte- ing a vasoconstrictor drug for several days are
rial pressure may fall further. Hypotension not sustained because of pressure natriuresis
associated with septic shock results from in the kidneys. When renal artery pressure
systemic vasodilation and, in its later stages, is elevated by increasing systemic vascular
cardiac depression. Therefore, pressor resistance, the kidneys respond by increasing
agents are commonly used with this form of glomerular filtration and excretion of sodium
shock in addition to administration of fluid and water. The loss of sodium and water
and antibiotics. decreases blood volume and restores pressure

Klabunde_Chap09.indd 212 6/11/2011 10:11:45 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 213

after a day to two despite continued infusion diagnosed with hypertension (Table 9-3).
of the vasoconstrictor. Therefore, with normal This diagnosis is made after known causes of
renal function, an acute elevation in arterial hypertension (i.e., secondary hypertension)
pressure caused by increasing systemic vascu- are eliminated. Therefore, essential hyper-
lar resistance (or by stimulating the heart) is tension is a diagnosis by exclusion. Despite
compensated by a reduction in blood volume, many years of research, no unifying hypoth-
which restores the arterial pressure to normal. esis accounts for the pathogenesis of essential
Considerable evidence shows that in chronic hypertension. However, a natural progression
hypertension, the renal pressure natriuresis of this disease suggests that early elevations in
curve is shifted to the right so that a higher arte- blood volume and cardiac output might pre-
rial pressure is required to maintain sodium bal- cede and then initiate subsequent increases
ance. The elevated pressure is sustained by an in systemic vascular resistance. This has led
increase in blood volume. These changes in renal some investigators to suggest that the basic
handling of sodium and water can be brought underlying defect in hypertensive patients is
about by changes in sympathetic activity and an inability of the kidneys to adequately han-
hormones that affect renal function (e.g., angio- dle sodium. Increased sodium retention could
tensin II, aldosterone, vasopressin). In addition, account for the increase in blood volume.
altered kidney filtration and sodium balance Indeed, many excellent experimental studies
in renal disease can shift the pressure natriure- as well as clinical observations have shown
sis curve to the right, leading to an increase in that impaired renal natriuresis (sodium excre-
blood volume and sustained hypertension. tion) can lead to chronic hypertension.
Besides the renal involvement in hyperten-
sion, it is well known that vascular changes can
Essential (Primary) Hypertension
contribute to hypertensive states, especially in
Essential (or primary) hypertension accounts the presence of impaired renal function. For
for approximately 90% to 95% of patients example, essential hypertension is usually
associated with increased systemic vascular
resistance caused by a thickening of the walls of
TABLE 9-3 CAUSES OF HYPERTENSION resistance vessels and by a reduction in lumen
Essential hypertension (90%–95%) diameters. In some forms of hypertension, this
• Unknown causes is mediated by enhanced sympathetic activity
• Involves: or by increased circulating levels of angiotensin
- increased blood volume II, causing smooth muscle contraction and vas-
- increased systemic vascular resistance
(vascular disease)
cular hypertrophy. Experimental studies have
• Associated with: suggested that changes in vascular endothelial
- heredity function may cause these vascular changes.
- abnormal response to stress For example, in hypertensive patients, the vas-
- diabetes and obesity cular endothelium produces less nitric oxide.
- age, race, and socioeconomic status Nitric oxide, besides being a powerful vasodi-
Secondary hypertension (5%–10%) lator, inhibits vascular hypertrophy. Increased
• Renal artery stenosis endothelin-1 production may enhance vascu-
• Renal disease lar tone and induce hypertrophy. Evidence sug-
• Hyperaldosteronism (primary)
gests that hyperinsulinemia and hyperglycemia
• Pheochromocytoma (catecholamine-
secreting tumor)
in type 2 diabetes (non–insulin-dependent dia-
• Aortic coarctation betes) cause endothelial dysfunction through
• Pregnancy (preeclampsia) increased formation of reactive oxygen species
• Hyperthyroidism/hypothyroidism and decreased nitric oxide bioavailability, both
• Cushing syndrome (excessive glucocorti- of which may contribute to the abnormal vas-
coid secretion) cular function and hypertension often associ-
• Sleep apnea
ated with diabetes.

Klabunde_Chap09.indd 213 6/11/2011 10:11:45 AM


214 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Essential hypertension is related to hered- angiotensin II and aldosterone. Angiotensin II


ity, age, race, and socioeconomic status. The causes vasoconstriction by binding to vascu-
strong hereditary correlation may be related lar AT1 receptors and by augmenting sympa-
to genetic abnormalities in renal function thetic influences. Furthermore, angiotensin II
and neurohumoral control mechanisms. The along with aldosterone increases renal sodium
incidence of essential hypertension increases and water reabsorption. The net effect of the
with age, and people of African descent are renal actions is an increase in blood volume
more likely to develop hypertension than are that augments cardiac output by the Frank-
Caucasians. Hypertension is more prevalent Starling mechanism. In addition, chronic ele-
among lower socioeconomic groups. vation of angiotensin II promotes cardiac and
Some patients with essential hyperten- vascular hypertrophy. Therefore, hyperten-
sion are more strongly influenced by stressful sion caused by renal artery stenosis is asso-
conditions than are normotensive individu- ciated with increases in cardiac output and
als. Stress not only leads to acute elevations in systemic vascular resistance.
arterial pressure, but it can also lead to chronic Renal disease (e.g., diabetic nephropathy,
elevations in pressure. Stress activates the sym- glomerulonephritis) damages nephrons in the
pathetic nervous system, which increases car- kidney. When this occurs, the kidney cannot
diac output and systemic vascular resistance. excrete normal amounts of sodium, and the
Furthermore, stress causes the adrenal medulla pressure natriuresis curve shifts to the right,
to secrete more catecholamines (epinephrine which leads to sodium and water retention,
and norepinephrine) than normal. Sympathetic increased blood volume, and increased car-
activation increases circulating angiotensin II, diac output. Renal disease may increase the
aldosterone, and vasopressin, which together release of renin, leading to a renin-dependent
can increase systemic vascular resistance and, form of hypertension. The elevation in arte-
through their renal effects, increase sodium rial pressure secondary to renal disease can
and water retention. In addition, prolonged be viewed as an attempt by the kidney to
elevation of angiotensin II and catecholamines increase renal perfusion, thereby restoring
leads to vascular and cardiac hypertrophy. normal glomerular filtration and sodium
excretion.
Primary hyperaldosteronism is increased
Secondary Hypertension
secretion of aldosterone by an adrenal ade-
Secondary hypertension accounts for 5% noma or adrenal hyperplasia. This condition
to 10% of hypertensive cases. This form of causes renal retention of sodium and water,
hypertension has identifiable causes that often thereby increasing blood volume and arterial
can be remedied. Regardless of the underly- pressure. Aldosterone acts upon the distal
ing cause, arterial pressure becomes elevated convoluted tubule and cortical collecting duct
through an increase in cardiac output, an of the kidney to increase sodium reabsorption
increase in systemic vascular resistance, or in exchange for potassium and hydrogen ion,
both. When cardiac output is elevated, it is which are excreted in the urine. Plasma renin
often related to increased blood volume and levels generally are decreased as the body
neurohumoral activation of the heart. Several attempts to suppress the renin-angiotensin
causes of secondary hypertension are summa- system. In addition, hypokalemia is associ-
rized in Table 9-3 and discussed below. ated with the high levels of aldosterone.
Renal artery stenosis occurs when the renal A pheochromocytoma (a catecholamine-
artery becomes narrowed (stenotic) owing secreting tumor, usually in the adrenal
to atherosclerotic or fibromuscular lesions. medulla) can cause high levels of circulating
This reduces the pressure at the afferent arte- catecholamines (both epinephrine and
riole, which stimulates the release of renin norepinephrine). A pheochromocytoma is
by the kidney (see Chapter 6). Increased diagnosed by measuring plasma or urine
plasma renin activity increases circulating catecholamine levels and their metabolites

Klabunde_Chap09.indd 214 6/11/2011 10:11:46 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 215

(vanillylmandelic acid and metanephrine). Hyperthyroidism induces systemic vaso-


This condition leads to α-adrenoceptor- constriction, an increase in blood volume,
mediated systemic vasoconstriction and and increased cardiac activity, all of which
β1-adrenoceptor-mediated cardiac stimula- can lead to hypertension. It is less clear why
tion that can cause substantial elevations in some patients with hypothyroidism also
arterial pressure. Although arterial pressure develop hypertension, but it may be related
rises to very high levels, tachycardia still to decreased tissue metabolism reducing the
occurs because of the direct effects of the release of vasodilator metabolites, thereby
catecholamines on the heart and vasculature. producing vasoconstriction and increased
Excessive β1-adrenoceptor stimulation in the systemic vascular resistance.
heart often leads to arrhythmias in addition to Cushing syndrome, which results from
the hypertension. excessive glucocorticoid secretion, can lead to
Aortic coarctation is a narrowing of the hypertension. Glucocorticoids such as corti-
aortic arch usually just distal to the left sub- sol, which are secreted by the adrenal cortex,
clavian artery. It is a congenital defect that share some of the same physiologic proper-
obstructs aortic outflow, leading to elevated ties as aldosterone, a mineralocorticoid also
pressures proximal to the coarctation (i.e., ele- secreted by the adrenal cortex. Therefore,
vated arterial pressures in the head and arms). excessive glucocorticoids can lead to volume
Distal pressures, however, are not necessarily expansion and hypertension.
reduced as would be expected from the hemo- Sleep apnea is a disorder in which peo-
dynamics associated with a stenosis. The rea- ple repeatedly stop breathing for short peri-
son for this is that reduced systemic blood ods of time (10 to 30 seconds) during their
flow, and in particular reduced renal blood sleep; this can occur dozens of times per
flow, leads to an increase in the release of renin hour. Breathing is most commonly inter-
and an activation of the renin-angiotensin- rupted by airway obstruction, and less com-
aldosterone system. This in turn elevates monly by disorders of the central nervous
blood volume and arterial pressure. Although system. This condition is often associated
the aortic arch and carotid sinus barorecep- with obesity. Individuals suffering from
tors are exposed to higher-than-normal pres- sleep apnea have a higher incidence of
sures, the baroreceptor reflex is blunted owing hypertension. The mechanism of hyperten-
to structural changes in the walls of vessels sion may be related to sympathetic activa-
where the baroreceptors are located. Further- tion and hormonal changes associated with
more, baroreceptors become desensitized to repeated periods of apnea-induced hypoxia
chronic elevation in pressure and become and hypercapnia, and from stress associated
“reset” to the higher pressure. with the loss of sleep.
Preeclampsia is a type of hypertension that
occurs in about 5% of pregnancies during late
Physiologic Basis for Therapeutic
second and third trimesters. Preeclampsia
Intervention
differs from less severe forms of pregnancy-
induced hypertension (gestational hyperten- If a person has secondary hypertension, it is
sion) in that preeclampsia is associated with a sometimes possible to correct the underlying
loss of albumin in the urine because of renal cause. For example, renal artery stenosis can
damage, and pulmonary and systemic edema. be corrected by placing a wire stent within the
Preeclampsia is also associated with increased renal artery to maintain vessel patency; aortic
vascular responsiveness to vasoconstrictors, coarctation can be surgically corrected; a phe-
which can lead to vasospasm. It is unclear ochromocytoma can be removed. However,
why some women develop this condition dur- for the majority of people who have essential
ing pregnancy; however, it usually disappears hypertension, the cause is unknown, so it
after parturition unless an underlying hyper- cannot be targeted for correction. Therefore,
tensive condition exists. the therapeutic approach for these patients

Klabunde_Chap09.indd 215 6/11/2011 10:11:46 AM


216 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

involves modifying the factors that determine especially aerobic exercise, reduces arterial
arterial pressure by using drugs. pressure and has beneficial effects on vascular
Because hypertension results from an function.
increase in cardiac output and increased
systemic vascular resistance, these are the HEART FAILURE
two physiologic mechanisms that are tar-
geted in drug therapy. In most hyperten- Heart failure occurs when the heart is unable
sive patients, altered renal function causes to supply adequate blood flow and therefore
sodium and water retention. This increases oxygen delivery to peripheral tissues and
blood volume, cardiac output, and arte- organs, or to do so only at elevated filling pres-
rial pressure. Therefore, the most com- sures. Heart failure most commonly involves
mon treatment for hypertension is the use the left ventricle. Right ventricular failure,
of a diuretic to stimulate renal excretion of although sometimes found alone or in asso-
sodium and water. This reduces blood vol- ciation with pulmonary disease, more often
ume and arterial pressure very effectively occurs secondary to left ventricular failure.
in many patients. In addition to a diuretic, Mild heart failure is manifested as reduced
most hypertensive patients are given at least exercise capacity and the development of
one other drug. This is because decreasing shortness of breath during physical activity
blood volume with a diuretic leads to acti- (exertional dyspnea). In more severe forms of
vation of the renin-angiotensin-aldosterone heart failure, a patient may have virtually no
system, which counteracts the effects of capacity for physical exertion and will experi-
the diuretic. Therefore, these patients may ence dyspnea even while at rest. Furthermore,
be given an ACE inhibitor or angiotensin the patient will likely have significant pulmo-
receptor blocker (ARB) as well. nary or systemic edema.
In addition to using diuretics, cardiac out-
put can be reduced using beta-blockers and Causes of Heart Failure
the more cardioselective calcium channel
Heart failure can be caused by factors origi-
blockers (e.g., verapamil). Beta-blockers are
nating from the heart (i.e., intrinsic disease
particularly useful in patients who may have
or pathology) or from external factors that
excessive sympathetic stimulation caused by
place excessive demands upon the heart. The
emotional stress, and these drugs also inhibit
number-one cause of heart failure is coro-
sympathetic-mediated release of renin.
nary artery disease, which reduces coronary
In combination with a diuretic, some
blood flow and oxygen delivery to the myo-
hypertensive patients can be effectively
cardium, thereby causing myocardial hypoxia
treated with an α-adrenoceptor antagonist,
and impaired function. A related common
which dilates resistance vessels and reduces
cause of heart failure is myocardial infarc-
systemic vascular resistance. Other drugs that
tion. Infarcted tissue does not contribute to
reduce systemic vascular resistance include
the generation of mechanical activity, and
ACE inhibitors, ARBs, calcium channel block-
noninfarcted regions must compensate for
ers (especially dihydropyridines), and direct-
the loss of function. Over time, the additional
acting arterial dilators such as hydralazine.
demands placed upon the noninfarcted tissue
Although pharmacologic intervention is
can cause functional changes leading to fail-
an important therapeutic modality in treat-
ure. Other heart conditions that can lead to
ing hypertension, improved diet and exercise
failure include:
have been shown to be effective in reducing
arterial pressure in many patients. A proper, • Valvular disease and congenital defects,
balanced diet that includes sodium restriction which place increased demands upon the
can prevent the progression of, and in some heart
cases reverse, cardiovascular changes asso- • Cardiomyopathies (intrinsic diseases of the
ciated with hypertension. Regular exercise, myocardium) of known origin (e.g., bacterial

Klabunde_Chap09.indd 216 6/11/2011 10:11:46 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 217

or viral; alcohol-induced) or unknown origin Frank-Starling mechanism to help maintain


(idiopathic) stroke volume despite the loss of inotropy.
• Infective or noninfective myocarditis (inflam- If preload did not undergo a compensatory
mation of the myocardium) increase, the decline in stroke volume would
• Chronic arrhythmias be even greater for a given loss of inotropy.
As systolic failure progresses, the ability of
External factors precipitating heart failure
the heart to compensate by the Frank-Starling
include increased afterload (pressure load;
mechanism becomes exhausted as sarcomeres
e.g., uncontrolled hypertension), increased
stretch to their maximal length. Furthermore,
stroke volume (volume load; e.g., arterial–
with chronic systolic failure, the ventricle
venous shunts), and increased body demands
anatomically remodels by dilating. This is
(high output failure; e.g., thyrotoxicosis,
achieved by new sarcomeres being added in
pregnancy).
series to existing sarcomeres. Increased wall
circumference with the addition of new sar-
Systolic versus Diastolic comere units prevents the individual sarcom-
Dysfunction eres from overstretching in the presence of
Heart failure can result from impaired ability elevated filling pressures and volumes. The
of the heart muscle to contract (systolic fail- dilated ventricle has increased compliance so
ure) or impaired filling of the heart (diastolic that it can accommodate large end-diastolic
failure). Systolic failure is caused by changes volumes without excessive increases in end-
in cellular signal transduction mechanisms diastolic pressure (see Fig. 4.5).
and excitation–contraction coupling that The effects of a loss of inotropy on stroke
impair inotropy (see Chapter 3). Functionally, volume, end-diastolic volume, and end-
this causes a downward shift in the Frank- systolic volume can be depicted using ventric-
Starling curve (Fig. 9.8). This decreases ular pressure–volume loops (Fig. 9.9, panel
stroke volume and causes a compensatory rise A) (the concept of pressure–volume loops was
in preload (clinically assessed as increased developed in Chapter 4; see Fig. 4.4). Systolic
ventricular end-diastolic pressure or volume, failure decreases the slope of the end-systolic
or increased pulmonary capillary wedge pres- pressure–volume relationship, which occurs
sure). Increased preload is an important com- because of reduced inotropy. Because of this
pensatory mechanism because it activates the change, at any given ventricular volume, less
pressure can be generated during systole, and
100 therefore, less volume can be ejected. This
leads to an increase in end-systolic volume.
The pressure–volume loop also shows that
A the end-diastolic volume increases (com-
Stroke pensatory increase in preload). Ventricular
Volume 50 preload increases because as the heart loses
(mL) B its ability to eject blood, more blood remains
in the ventricle at the end of ejection. This
results in the ventricle filling to a larger end-
0 diastolic volume as venous return enters
0 10 20 30
the ventricle. Increased ventricular filling
LVEDP (mm Hg) is enhanced by ventricular remodeling that
■ FIGURE 9.8 Effects of systolic failure on left enlarges the chamber size (ventricular dila-
ventricular Frank-Starling curves. Systolic fail- tion) and increases compliance. This permits
ure depresses the Frank-Starling curve, which larger end-diastolic volumes with smaller
decreases stroke volume and leads to an increase
increases in end-diastolic pressure, although
in ventricular preload (LVEDP, left ventricular end-
diastolic pressure). Point A, control point; point B, this pressure can still rise to levels that lead
systolic failure. to blood backing up into the left atrium and

Klabunde_Chap09.indd 217 6/11/2011 10:11:46 AM


218 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Systolic Failure
200
A Loss of
Inotropy

LV
Pressure 100
(mm Hg)

0
0 100 200
LV Volume (mL)

Diastolic Failure
200
B

LV Decreased
Pressure 100 Compliance
(mm Hg)

0
0 100 200
LV Volume (mL)

Systolic & Diastolic Failure


200
C

LV
Pressure 100
(mm Hg)

0
0 100 200
LV Volume (mL)
■ FIGURE 9.9 Effects of systolic, diastolic, and combined failure on left ventricular pressure–volume loops.
Panel A shows that systolic failure (loss of inotropy) decreases the slope of the end-systolic pressure–
volume relationship and increases end-systolic volume. This causes a secondary increase in end-diastolic
volume, which is augmented under chronic conditions by ventricular dilation that shifts the passive filling
curve down and to the right. The net effect is that stroke volume and ejection fraction decrease. Panel B
shows that diastolic failure increases the slope of the end-diastolic pressure–volume relationship (pas-
sive filling curve) because of reduced ventricular compliance caused by either hypertrophy or decreased
lusitropy. This reduces the end-diastolic volume and increases end-diastolic pressure. End-systolic volume
may decrease slightly as a result of reduced afterload. The net effect is reduced stroke volume; ejection
fraction may or may not change. Panel C shows that combined systolic and diastolic failure reduces end-
diastolic volume and increases end-systolic volume so that stroke volume is greatly reduced; end-diastolic
pressure may become very high.

Klabunde_Chap09.indd 218 6/11/2011 10:11:46 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 219

pulmonary vasculature, leading to pulmonary Increased ventricular end-diastolic pres-


edema. The increase in end-diastolic volume, sure, which can exceed 30 mm Hg in left
however, is not as great as the increase in end- ventricular failure, can have serious clinical
systolic volume. Therefore, the net effect is a consequences because left atrial and pul-
decrease in stroke volume (decreased width monary capillary pressures rise. Pulmonary
of the pressure–volume loop). Because stroke edema can occur when the left ventricular
volume decreases and end-diastolic volume end-diastolic pressure exceeds 20 mm Hg. If
increases, a substantial reduction in ejec- the right ventricle is in diastolic failure, the
tion fraction occurs. Ejection fraction (stroke increase in end-diastolic pressure is reflected
volume divided by end-diastolic volume) is back into the right atrium and systemic
normally >55%, but it can fall below 20% in venous vasculature. This can lead to periph-
severe systolic failure. eral edema and abdominal ascites.
The second type of heart failure is diastolic It is not uncommon in chronic heart fail-
failure, which is caused by impaired ventricu- ure to have a combination of both systolic
lar filling. Diastolic failure can be caused by and diastolic dysfunction to varying degrees
either decreased ventricular compliance (e.g., (Fig. 9.9, panel C). With both systolic and
as occurs with ventricular hypertrophy; see diastolic dysfunction, the slope of the end-
Chapter 4) or impaired relaxation (decreased systolic pressure–volume relationship is
lusitropy; see Chapter 3). Ventricular hyper- decreased, and the slope of the passive fill-
trophy most commonly is caused by chronic, ing curve is increased. This causes a dramatic
uncontrolled hypertension, which results in reduction in stroke volume because end-sys-
a thickening of the ventricular wall as new tolic volume is increased and end-diastolic
sarcomeres are added in parallel to exist- volume is decreased. This combination of
ing sarcomeres. The hypertrophy enables systolic and diastolic dysfunction can lead to
the heart to contract more forcefully against high end-diastolic pressures that can cause
the higher pressure in the aorta and helps pulmonary congestion and edema.
to normalize wall stress (see Equation 4-2).
Therefore, a hypertrophied heart may exhibit
Systemic Compensatory
a leftward shift in the end-systolic pressure-
Mechanisms in Heart Failure
volume relationship (not shown in Fig. 9.9,
panel B). Other causes of diastolic failure Heart failure, whether systolic or diastolic in
include hypertrophic cardiomyopathy, a dis- nature, leads to a reduction in stroke volume
ease resulting from a genetic defect that alters and cardiac output. In the absence of compen-
myocardial structure. Normal age-related satory mechanisms, a fall in cardiac output
changes to cardiac structure can make the has two effects on pressure: decreased arterial
ventricle less compliant, leading to impaired pressure and increased central venous pressure
ventricular filling in the elderly population. (see Fig. 5.18). These changes activate neuro-
Reduced ventricular compliance, whether humoral mechanisms that attempt to restore
of anatomic or physiologic origin, shifts the cardiac output and arterial pressure (Fig. 9.10).
ventricular end-diastolic pressure–volume In response to an acute reduction in car-
relationship (i.e., passive filling curve) up diac output and arterial pressure, decreased
and to the left (Fig. 9.9, panel B). This results firing of arterial baroreceptors activates the
in less ventricular filling (decreased end- sympathetic adrenergic nerves to the heart
diastolic volume) and a greater end-diastolic and vasculature. The baroreceptor reflex
pressure. Stroke volume, therefore, decreases. responds mainly to acute changes in arterial
Depending upon the relative change in stroke pressure and therefore cannot be responsible
volume and end-diastolic volume, ejection for maintaining the increased sympathetic
fraction may or may not change. For this rea- drive when hypotension accompanies chronic
son, reduced ejection fraction is useful only as heart failure. In addition, not all patients in
an indicator of systolic failure. chronic heart failure are hypotensive. It is not

Klabunde_Chap09.indd 219 6/11/2011 10:11:46 AM


220 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Ventricular
Failure

– Cardiac +
Output

+ Arterial
Pressure

Sympathetic Blood
Systemic Angiotensin II Volume Venous Pulmonary Edema
Vascular Pressure Systemic Edema
Aldosterone Venous
Resistance
Vasopressin Tone
– – –
Atrial
Natriuretic
Peptide

■ FIGURE 9.10 Summary of neurohumoral changes associated with heart failure. Activation of the sym-
pathetic nervous system, the renin-angiotensin-aldosterone system, and vasopressin cause an increase
in systemic vascular resistance, blood volume, and central venous pressure. Although increased central
venous pressure helps to elevate (+) cardiac output by the Frank-Starling mechanism, it can also lead to
pulmonary and systemic edema. The increased systemic vascular resistance, although helping to elevate
arterial pressure, can depress (−) cardiac output further because of increased afterload. Increased atrial
natriuretic peptide counterregulates the other hormonal systems.

clear what drives the characteristic increase in spillover of norepinephrine into the circula-
sympathetic activity in chronic heart failure, tion from highly activated sympathetic nerves.
although humoral changes and cardiac stretch These changes in neurohumoral status
receptors may be involved, along with barore- constrict resistance vessels, which causes an
ceptor resetting. increase in systemic vascular resistance to
Important humoral changes occur dur- help maintain arterial pressure. Venous capac-
ing heart failure to help compensate for the itance vessels constrict as well. This increased
reduction in cardiac output. Arterial hypo- venous tone contributes to the increase in
tension, along with sympathetic activation, venous pressure. Angiotensin II and aldoster-
stimulates renin release, leading to the for- one, along with vasopressin, increase blood
mation of angiotensin II and aldosterone. volume by increasing renal reabsorption of
Vasopressin (antidiuretic hormone) release sodium and water, which further increases
from the posterior pituitary is also stimulated. venous pressure. The increased venous pres-
Increased vasopressin release seems para- sure increases cardiac preload and helps to
doxical because right atrial pressure is often maintain stroke volume through the Frank-
elevated in heart failure, which should inhibit Starling mechanism. Increased right atrial
the release of vasopressin (see Chapter 6). It pressure stimulates the synthesis and release
may be that vasopressin release is stimulated of atrial natriuretic peptide to counterregu-
in heart failure by sympathetic activation and late the renin-angiotensin-aldosterone sys-
increased angiotensin II. Circulating catecho- tem. These neurohumoral responses function
lamines (norepinephrine and epinephrine) as compensatory mechanisms, but they can
are also elevated in heart failure because of aggravate heart failure by increasing ventricu-
sympathetic stimulation of the adrenals and lar afterload (which depresses stroke volume)

Klabunde_Chap09.indd 220 6/11/2011 10:11:46 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 221

and increasing venous pressures and cardiac can increase cardiac output by only 50%, com-
preload to the point at which pulmonary or pared to a 221% increase in the normal person.
systemic congestion and edema occur. The The reduced cardiac output is a consequence
volume and afterload increases also increase of the inability of the left ventricle to augment
oxygen demand by the heart, which can fur- stroke volume as well as a lower maximal heart
ther exacerbate ventricular failure over time. rate (exercise intolerance limits the heart rate
increase). The CHF patient has a significant
Exercise Limitations Imposed reduction in arterial pressure during exercise
in contrast to the normal person’s increase in
by Heart Failure
arterial pressure. Arterial pressure falls because
Heart failure can severely limit exercise the increase in cardiac output is not sufficient
capacity. In early or mild stages of heart fail- to maintain arterial pressure as the systemic
ure, cardiac output and arterial pressure may vascular resistance falls during exercise. The
be normal at rest because of compensatory maximal whole-body oxygen consumption is
mechanisms. When the person in heart fail- greatly reduced in the CHF patient because
ure begins to perform physical work, how- reduced perfusion of the active muscles lim-
ever, the maximal workload is reduced, and its oxygen delivery and therefore the oxygen
he or she experiences fatigue and dyspnea at consumption of the muscles. The CHF patient
less than normal maximal workloads. experiences substantial fatigue and dyspnea
A comparison of exercise responses in a during exertion, which limits the patient’s abil-
normal person and in a heart failure patient is ity to sustain the physical activity.
shown in Table 9-4. In this example, the degree Some of the neurohumoral compensatory
of heart failure is moderate to severe. At rest, mechanisms that operate to maintain resting
the person with congestive heart failure (CHF) cardiac output in heart failure contribute to
has reduced cardiac output (decreased 29%) limiting exercise capacity. The chronic increase
caused by a 38% decrease in stroke volume. in sympathetic activity to the heart down-
Mean arterial pressure is slightly decreased, regulates β1-adrenoceptors, which reduces the
and resting heart rate is elevated. Whole-body heart’s chronotropic and inotropic responses
oxygen consumption is normal at rest, but the to acute sympathetic activation during exer-
reduced cardiac output results in an increase in cise. Increased sympathetic activity (and
the arterial–venous oxygen difference as more possibly circulating vasoconstrictors) to the
oxygen is extracted from the blood because skeletal muscle vasculature limits the degree
organ blood flow is reduced. At a maximally of vasodilation during muscle contraction.
tolerated exercise workload, the CHF patient This limits oxygen delivery to the working

TABLE 9-4 COMPARISON OF CARDIOVASCULAR FUNCTION IN A NORMAL PERSON


AND A PATIENT WITH MODERATE-TO-SEVERE CHF AT REST AND AT
MAXIMAL (MAX) EXERCISE
Hr (Beats/ CaO2–CvO2 (Ml
Co (L/Min) Min) Sv (Ml) Map (Mm Hg) Vo2 (Ml O2/Min) O2/100 Ml)

Normal 5.6 70 80 95 220 4.0


(Rest)
Normal 18.0 170 106 120 2500 13.9
(Max)
CHF (Rest) 4.0 80 50 90 220 5.5
CHF (Max) 6.0 120 50 85 780 13.0
CO, cardiac output; HR, heart rate; SV, stroke volume; MAP, mean arterial pressure; VO2, whole-body oxygen consumption;
CaO2–CvO2, arterial–venous oxygen difference. VO2 is calculated from the product of CO and CaO2–CvO2, after the units for
CO are converted to mL/min and the units for CaO2–CvO2 are converted to mL O2/mL blood.

Klabunde_Chap09.indd 221 6/11/2011 10:11:46 AM


222 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

muscle and leads to increased oxygen extrac- The third approach is to use drugs that
tion (increased arterial-venous oxygen differ- stimulate ventricular inotropy. A commonly
ence), enhanced lactic acid production (and a used drug is digoxin, which inhibits the
lower anaerobic threshold), and muscle fatigue Na+JK+-ATPase and thereby increases intra-
at lower workloads. The increase in blood cellular calcium (see Chapter 2). This drug,
volume, although helping to maintain stroke however, has not been shown to reduce
volume at rest through the Frank-Starling mortality associated with heart failure.
mechanism, decreases the reserve capacity of Drugs that stimulate ~ 1 -adrenoceptors (e.g.,
the heart to increase preload during exercise. dobutamine) or inhibit cAMP-dependent
phosphodiesterase (e.g., milrinone) are
sometimes used as inotropic agents (see
Physiologic Basis for Therapeutic
Chapter 3). With the exception of digoxin,
Intervention
inotropic drugs are used only in acute
Therapeutic goals in the pharmacologic treat- heart failure and end-stage failure because
ment of heart failure include (1) reducing the their long-term use has been shown to be
clinical symptoms of edema and dyspnea; (2) deleterious to the heart.
improving cardiovascular function to enhance The fourth therapeutic approach involves
organ perfusion and increase exercise capac- using beta-blockers. Although this might
ity; and (3) reducing mortality. seem counterintuitive, many clinical tri-
Four pharmacologic approaches are taken als have clearly demonstrated the efficacy of
to achieve these goals. The first approach is some beta-blockers (e.g., carvedilol and meto-
to reduce venous pressure to decrease edema prolol). The mechanism of their efficacy is not
and help relieve the patient of dyspnea. Diu- clear, but it is known that long-term sympa-
retics are routinely used to reduce blood vol- thetic activation of the heart is deleterious.
ume by increasing renal excretion of sodium Therefore, beta-blockers probably work by
and water. Drugs that dilate the venous vascu- reducing the deleterious actions of long-term
lature (e.g., ACE inhibitors) also can reduce sympathetic activation. Beta-blockers (as well
venous pressure. judicious use of these drugs as ACE inhibitors) provide long-term benefit
to decrease blood volume and venous pres- through ventricular remodeling (e.g., reduc-
sure does not significantly reduce stroke ing ventricular hypertrophy or dilation).
volume because the Frank-Starling curve Furthermore, ~-blockers significantly reduce
associated with systolic failure is relatively mortality in heart failure.
flat at left ventricular end-diastolic pressures It should be noted that vasodilators, ino-
above 15 mm Hg (see Fig. 9.8). tropic drugs, and ~-blockers are nearly always
The second approach is to use drugs that used in combination with a diuretic.
reduce afterload on the ventricle by dilating
the systemic vasculature. Drugs such as ACE CASE 9-3
inhibitors and ARBs have proven to be use-
A patient is diagnosed with dilated
ful in this regard for patients with chronic
cardiomyopathy. The echocardiogram
heart failure. Decreasing the afterload on the
shows substantial left ventricular
ventricle can significantly enhance stroke
dilation (end-diastolic volume is
volume and ejection fraction, which sec-
240 ml) and an ejection fraction
ondarily reduces ventricular end-diastolic
of 20%; the arterial pressure is
volume (preload). Because arterial vasodila-
115/70 mm Hg. Calculate the stroke
tors enhance cardiac output in heart failure
volume and end-systolic volume. How
patients, the reduction in systemic vascular
would combined therapy with an ACE
resistance does not usually lead to an unac-
inhibitor and diuretic alter ventricular
ceptable fall in arterial pressure. Vasodilators
volumes, ejection fraction, and arterial
also have the benefit of decreasing myocardial
pressure?
oxygen demand.

Klabunde_Chap09.indd 222 6/11/2011 10:11:46 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 223

VALVE DISEASE compensatory responses include systemic


vasoconstriction, increased blood volume,
Normal valve function as described in Chapter 4 and increased heart rate and inotropy. Cardiac
is characterized as having (1) low pressure remodeling involves hypertrophy or dilation,
gradients across the valve as blood flows depending on the valve defect. When these
through the orifice and (2) unidirectional compensatory mechanisms fail to maintain
flow. These normal features are altered when cardiac output and arterial pressure within
heart valves function abnormally. When this normal limits (termed “decompensation”),
occurs, net ventricular outflow can decrease, the patient develops symptoms of heart fail-
leading to a fall in cardiac output and clinical ure as described in the previous section.
signs of heart failure. The following discussion examines cardiac
There are two general categories of valve changes during valve disease in the absence of
defects: stenosis and insufficiency. Valve significant heart failure at rest, therefore rep-
stenosis results from a narrowing of the resenting compensated conditions.
valve orifice. Fibrosis, often accompanied
by calcification, causes the valve leaflets Valve Stenosis
to thicken so that they cannot open fully,
which decreases cross-sectional area of the Stenosis can occur at either an outflow
open orifice. Furthermore, the valve cusps valve (aortic or pulmonic valve) or inflow
can fuse together, which prevents them valve (mitral or tricuspid valve). Stenosis
from fully opening. Congenital valve defects increases the resistance to flow across the
can also produce stenosis. Valve regurgita- valve, which causes a high pressure gradi-
tion (insufficiency) occurs when the valve ent across the valve. The pressure gradient
leaflets do not completely seal when the across a valve is the pressure difference on
valve is closed; this causes blood to flow either side of the leaflets as blood is flowing
backward (regurgitate) into the proximal through the valve. For the aortic valve, the
chamber. Both of these valve defects alter pressure gradient is the left ventricular pres-
intracardiac pressures and volumes during sure minus the aortic pressure; for the mitral
the cardiac cycle. valve, the pressure gradient is the left atrial
Valve defects produce murmurs that can pressure minus the left ventricular pressure.
be heard with a stethoscope. A murmur is a In normal valves, the pressure gradient is
rumbling or rasping sound caused by vibra- only a few mm Hg when blood is flowing
tions generated by the abnormal movement of across the open valve.
blood within or between cardiac chambers, or The following equation is the general
by turbulent flow within the pulmonary artery hemodynamic expression that relates pressure
or aorta just distal to the outflow valve. If a gradient (ΔP), flow (F), and resistance (R)
murmur is heard during systole between the under laminar, nonturbulent flow conditions:
first (S1) and second (S2) heart sounds, it is
termed a systolic murmur. If it is heard dur- ΔP = F ⋅ R
ing diastole (between S2 and S1), it is termed A reduced valve orifice increases the resist-
a diastolic murmur. The sound intensifies ance to flow across the valve because resist-
with increasing flow and turbulence across ance is inversely related to the radius (r) of
the valve. the valve orifice to fourth power (equiva-
The following sections describe pres- lent to valve orifice area [A] to the second
sure and volume changes that occur during power because A = π r2) (see Chapter 5).
valve stenosis and regurgitation. Because Therefore, the above equation can also be
valve disease is generally a chronic problem, expressed as:
neurohumoral activation and cardiac remod-
eling occur in an attempt to maintain normal F
ΔP ∝
cardiac output and arterial pressure. These A2

Klabunde_Chap09.indd 223 6/11/2011 10:11:47 AM


224 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Using the above relationship, if the valve ori- (Fig. 9.11, left panel). This leads to a large
fice area is reduced by 75%, the valve resist- pressure gradient across the valve during
ance is increased 16-fold, which increases the ejection, the magnitude of which depends
pressure gradient 16-fold if flow through the on the degree of stenosis and the flow across
valve remains unchanged. In reality, the for- the valve. Increased flow velocity through
mation of turbulence increases the pressure the stenotic valve causes turbulence and
gradient across the valve even further. Tur- a systolic murmur. In moderate-to-severe
bulence occurs because a reduced orifice area aortic stenosis, the aortic pressure may be
leads to an increase in the velocity of blood reduced because ventricular stroke volume
flow across the valve. Because flow (F) equals (and cardiac output) is reduced. The degree
the product of velocity (V) and area (A), the of hypotension depends on the ability of
velocity equals flow divided by area (V = F/A). neurohumoral mechanisms to increase blood
Therefore, if flow remains unchanged, a 75% volume and systemic vascular resistance.
reduction in area causes a fourfold increase Because ejection is impeded by the increase
in velocity, which increases turbulence and in ventricular afterload, more blood remains
produces a murmur. In summary, at a given in the heart after ejection, which leads to an
flow across a valve, a reduction in valve ori- increase in left atrial volume and pressure.
fice area increases the pressure gradient across Changes in left ventricular pressure–
the valve that is required to drive the flow, volume loops with moderate aortic steno-
increases the velocity of flow, and increases sis are shown in Figure 9.11 (right panel).
the turbulence. Because left ventricular emptying is impaired
by the increased afterload (see Chapter 4),
the stroke volume is reduced, which leads
AORTIC VALVE STENOSIS
to an increase in end-systolic volume. With
In aortic valve stenosis, left ventricular pres- chronic aortic stenosis, the left ventricle
sure is increased above normal during sys- hypertrophies. This decreases ventricular
tole to eject blood across the narrowed valve compliance, elevates end-diastolic pressure,

Aortic
LV Pressure (mm Hg)

S1 S2 S1
Pressure (mm Hg)

Stenosis
200 200
LVP
Pressure Normal
Gradient Loop

AP
100 100
LAP

0 0
Time 0 100 200
LV Volume (mL)
■ FIGURE 9.11 Changes in cardiac pressures and volumes associated with chronic aortic valve stenosis in
the absence of systolic failure. The left panel shows that during ventricular ejection, left ventricular pres-
sure (LVP) exceeds aortic pressure (AP) (the gray area represents the pressure gradient generated by the
stenosis); a systolic murmur is present between S1 and S2, and left atrial pressure (LAP) is elevated. Aortic
pressure may be reduced because of decreased stroke volume. The right panel shows the effects of aortic
valve stenosis (red loop) on the left ventricular (LV) pressure–volume loop. The end-systolic volume is
increased, with little or no change in end-diastolic volume; therefore, stroke volume is decreased. Ven-
tricular hypertrophy reduces ventricular compliance, which elevates end-diastolic pressure at any given
end-diastolic volume.

Klabunde_Chap09.indd 224 6/11/2011 10:11:47 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 225

and may impair filling (i.e., produces diastolic filling pressure (Fig. 9.12, left panel). During
dysfunction). This is shown in the pressure– ventricular filling, turbulence caused by the
volume loop as an elevated and steeper filling narrowed mitral valve causes a diastolic mur-
curve (see Figs. 4.5 and 9.9). Whether end- mur. In moderate-to-severe mitral stenosis,
diastolic volume is increased or decreased reduced ventricular filling causes a reduction
depends on the changes in compliance and in ventricular preload (both end-diastolic vol-
filling pressure. Recall from Chapter 4 that ume and pressure decrease) (Fig. 9.12, right
an acute increase in afterload, which initially panel). This leads to a decrease in stroke
leads to an increase in end-systolic volume, volume (width of pressure–volume loop)
usually causes a secondary increase in end- through the Frank-Starling mechanism, and
diastolic volume that helps to preserve stroke a fall in cardiac output and aortic pressure.
volume. But in chronic aortic stenosis, this Reduced afterload (particularly if aortic pres-
secondary increase in preload often will not sure falls) enables the end-systolic volume to
occur because of the reduced ventricular decrease slightly, but not enough to overcome
compliance. the decline in end-diastolic volume. These
In summary, aortic valve stenosis is char- changes will be influenced by neurohumoral
acterized by a large pressure gradient across activation, which increases blood volume,
the aortic valve during systole, a systolic ejec- systemic vascular resistance, cardiac inotropy,
tion murmur, reduced stroke volume, ven- and heart rate.
tricular hypertrophy (reduced compliance), In summary, mitral valve stenosis impairs
increased left ventricular filling pressure, and ventricular filling, which reduces preload and
increased left atrial and pulmonary vascular therefore stroke volume. A diastolic murmur
pressures. is present, and left atrial and pulmonary vas-
cular pressures are elevated.
MITRAL VALVE STENOSIS
PULMONIC AND TRICUSPID VALVE
Mitral valve stenosis increases the pressure gra-
STENOSIS
dient across the mitral valve during ventricular
filling, which leads to an increase in left atrial Pulmonic stenosis produces changes to the
pressure and a reduction in left ventricular right side of the heart that are analogous to

200 200
LV Pressure (mm Hg)
Pressure (mm Hg)

Normal
S1 S2 S1 Loop Mitral
Stenosis
AP
100 100
Pressure
LVP LAP Gradient

0 0
Time 0 100 200
LV Volume (mL)
■ FIGURE 9.12 Changes in cardiac pressures and volumes associated with chronic mitral valve stenosis in
the absence of systolic failure. The left panel shows that during ventricular filling, left atrial pressure (LAP)
exceeds left ventricular pressure (LVP) (the gray area represents the pressure gradient generated by the
stenosis); a diastolic murmur is present between S2 and S1. Aortic pressure (AP) is reduced by severe mitral
stenosis because of decreased cardiac output. The right panel shows the effects of mitral valve stenosis
(red loop) on the left ventricular (LV) pressure–volume loop. End-diastolic volume is reduced because of
impaired ventricular filling, and end-systolic volume may be slightly reduced because of reduced after-
load; therefore, stroke volume is reduced.

Klabunde_Chap09.indd 225 6/11/2011 10:11:47 AM


226 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

those produced on the left side of the heart which permits blood to flow backward
by aortic stenosis. Stenosis of the pulmonic (regurgitate) across the valve. Aortic or
valve results in a pressure gradient across pulmonary insufficiency most commonly
that valve during right ventricular ejec- occurs through disease processes that alter
tion, as well as a systolic murmur. Reduced valve structure. Mitral and tricuspid valve
right ventricular stroke volume decreases regurgitation can occur following rupture of
left ventricular filling and stroke volume, the chordae tendineae, following ischemic
which leads to activation of neurohumoral damage to the papillary muscles, in response
compensatory mechanisms. The right ven- to infective or degenerative disease of the
tricle hypertrophies, which contributes to valve tissue, or when the ventricles are path-
elevated filling pressures that are transmit- ologically dilated (e.g., as occurs in dilated
ted back into the right atrium and systemic cardiomyopathy).
venous circulation.
Tricuspid stenosis impairs right ventricular AORTIC VALVE REGURGITATION
filling and stroke volume, and elevates right
Aortic valve regurgitation causes blood to enter
atrial and systemic venous pressures. Because
right ventricular output is reduced, left ven- the left ventricle from the aorta (backward
tricular stroke volume is also diminished, flow) during the time that the valve would
which can trigger compensatory neurohu- normally be closed. Because blood leaves the
moral mechanisms. As with mitral stenosis, aorta by two pathways (back into the ventricle
there is a diastolic murmur. as well as down the aorta), the aortic pressure
falls more rapidly than usual during diastole,
thereby reducing aortic diastolic pressure
Valve Regurgitation
(Fig. 9.13, left panel). Ventricular (and aortic)
Valvular insufficiency can occur with out- peak systolic pressures are increased because
flow valves (aortic or pulmonic) or inflow there is an increase in stroke volume into the
valves (mitral or tricuspid). In this condi- aorta because of increased ventricular filling.
tion, the valve does not close completely, The increased systolic pressure and decreased

200 200
LV Pressure (mm Hg)

Aortic
Pressure (mm Hg)

S1 S2 S1 Regurgitation
¯ Norma l
Aortic
AP Puls e Loop
100 Pressure 100
LAP LVP

0 0
Time 0 100 200
LV Volume (mL)
■ FIGURE 9.13 Changes in cardiac pressures and volumes associated with chronic aortic valve regur-
gitation in the absence of systolic failure. The left panel shows that during ventricular relaxation, blood
flows backwards from the aorta into the ventricle, causing a more rapid fall in aortic pressure (AP), which
decreases diastolic pressure and increases aortic pulse pressure; left atrial pressure (LAP) increases
because of blood backing up into the atrium as left ventricular end-diastolic volume and pressure
increase. An increase in ventricular stroke volume (because of increased filling) leads to an increase in
peak ventricular and aortic pressures; a diastolic murmur is present between S2 and S1. The right panel
shows the effects of aortic valve regurgitation (red loop) on the left ventricular (LV) pressure–volume
loop. End-diastolic volume and stroke volume are greatly increased, and there are no true isovolumetric
phases because blood flows across the valve whenever there is a pressure difference across the valve.

Klabunde_Chap09.indd 226 6/11/2011 10:11:47 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 227

diastolic pressure increase the aortic pulse volumes may be 120 mL. If half of that stroke
pressure. The regurgitation, which takes place volume flows backward into the ventricle
as the ventricle relaxes and fills, causes a dias- (regurgitant fraction = 0.5), then the net out-
tolic murmur, which is louder early in diastole ward stroke volume will be 60 mL, which is
(decrescendo murmur). smaller than normal.
Because of the backward flow of blood In summary, aortic valve insufficiency is
from the aorta into the left ventricle, there characterized by an increase in aortic pulse
is no true phase of isovolumetric relaxation pressure, a diastolic murmur, increased stroke
(see Fig. 9.13, right panel). Instead, the left volume but reduced net aortic flow, ventricu-
ventricle begins to fill with blood from the lar dilation, no true isovolumetric phases,
aorta before the mitral valve opens. Once increased ventricular filling pressure, and
the mitral valve opens, ventricular filling increased left atrial and pulmonary vascular
occurs from the left atrium; however, blood pressures.
continues to flow from the aorta into the
ventricle throughout diastole because aortic
MITRAL VALVE REGURGITATION
pressure is higher than ventricular pressure
during diastole. This greatly enhances ven- In mitral valve regurgitation, blood flows
tricular filling (end-diastolic volume), which backward into the left atrium as the left
activates the Frank-Starling mechanism to ventricle contracts. This leads to a large
increase the force of contraction and stroke increase in the v wave of the left atrial pres-
volume as shown by the increased width of sure tracing (Fig. 9.14, left panel) and the
the pressure–volume loop. With chronic generation of a systolic murmur that spans
aortic regurgitation, the ventricle remod- briefly beyond S2. Ventricular systolic and
els by dilating, which increases compliance. aortic pressures decrease if the net ejec-
This helps the ventricle to accommodate the tion of blood into the aorta is significantly
large increase in volume without excessive reduced.
increases in end-diastolic pressure. As long There are several important changes in the
as the ventricle is not in failure, normal end- left ventricular pressure–volume loop in the
systolic volumes can be sustained; however, presence of mitral insufficiency (Fig. 9.14,
the end-systolic volume increases when the right panel). First, there is no true isovolumet-
ventricle goes into systolic failure. Because ric phase at the beginning of systole. As soon
the aortic valve never completely closes, as the ventricle begins to contract and develop
blood will always be moving across the valve pressure, blood begins to flow across the mitral
depending on the aortic and left ventricular valve and back into the left atrium. Mitral
pressure difference. Consequently, there is no regurgitation reduces the afterload on the left
true isovolumetric phase at the beginning of ventricle (total outflow resistance is reduced),
diastole or systole. When the ventricle first which causes stroke volume to be larger and
begins to contract, blood continues to enter end-systolic volume to be smaller than nor-
the ventricle from the aorta until the ventric- mal; however, end-systolic volume increases if
ular pressure exceeds the aortic pressure. It the heart goes into systolic failure in response
is important to note that the stroke volume, to chronic mitral regurgitation. Because the
calculated from the difference between the mitral valve is never completely closed, blood
end-diastolic and end-systolic volumes, is flows back into the left atrium as long as intra-
increased. However, the net stroke volume ventricular pressure is greater than left atrial
into the aorta (net forward flow in the aorta) pressure; therefore, there is no true phase of
is lower than normal. For example, assume isovolumetric relaxation. During diastole,
that stroke volume is normally 70 mL. During the elevated pressure within the left atrium is
aortic regurgitation, the stroke volume calcu- transmitted to the left ventricle during filling
lated from the end-diastolic and end-systolic so that left ventricular end-diastolic pressure

Klabunde_Chap09.indd 227 6/11/2011 10:11:47 AM


228 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

200 200

LV Pressure (mm Hg)


Mitral
Pressure (mm Hg)

Norma l Regurgitation
S1 S2 S1
Loop

AP
100 100
Tall
v-wave LVP

LAP
0 0
Time 0 100 200
LV Volume (mL)
■ FIGURE 9.14 Changes in cardiac pressures and volumes associated with chronic mitral valve regurgita-
tion in the absence of systolic failure. The left panel shows that during ventricular contraction, the left
ventricle ejects blood back into the left atrium as well as into the aorta, thereby increasing left atrial pres-
sure (LAP), particularly the v wave. The aortic pressure (AP) and left ventricular pressure (LAP) may fall
in response to a reduction in the net volume of blood ejected into the aorta; a systolic murmur is present
between S1 and just beyond S2. The right panel shows the effects of mitral valve regurgitation (red loop)
on the left ventricular (LV) pressure–volume loop. End-systolic volume is reduced because of decreased
outflow resistance (afterload); end-diastolic volume is increased because increased left atrial pressure
increases ventricular filling; stroke volume is greatly enhanced. There are no true isovolumetric phases
because blood flows across the valve whenever there is a pressure difference across the valve.

and volume increase. In chronic mitral regur- analogous to those produced on the left side
gitation, volume overload causes the ventri- of the heart by aortic regurgitation. Regur-
cle to undergo dilation, thereby increasing its gitation across the pulmonic valve leads to
compliance. This dilation would cause wall increased pulmonary artery pulse pressure,
stress (afterload) to increase if it were not for increased right ventricular end-diastolic
the reduced outflow resistance that tends to volume and pressure, and a diastolic mur-
decrease afterload during ejection. The net mur. There is no true isovolumetric phase
effect of these changes is that the width of during right ventricular systole and dias-
the pressure–volume loop (stroke volume) is tole. Because the right ventricle becomes
increased; however, ejection into the aorta is volume overloaded, it responds by dilating,
reduced by the regurgitant fraction. and right atrial and systemic venous pres-
In summary, mitral regurgitation is charac- sures increase.
terized by a tall v wave, a systolic murmur, Tricuspid regurgitation causes a tall
increased stroke volume but reduced net v wave in the right atrial pressure tracing, an
ventricular outflow into the aorta, ventricu- overall increase in right atrial volume and
lar dilation, no true isovolumetric phases, systemic venous pressures, and a systolic
increased ventricular filling pressures, and murmur. Right ventricular stroke volume
increased left atrial and pulmonary vascular is increased, but ejection into the pulmo-
pressures. nary artery may be reduced because of the
large volume of blood ejected into the right
atrium during ventricular systole. Reduced
PULMONIC AND TRICUSPID VALVE
ejection into the pulmonary artery decreases
REGURGITATION
left ventricular filling and stroke volume,
Pulmonic regurgitation produces changes leading to activation of neurohumoral com-
to the right side of the heart that are pensatory mechanisms.

Klabunde_Chap09.indd 228 6/11/2011 10:11:48 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 229

SUMMARY OF IMPORTANT CONCEPTS

• Dynamic exercise such as running positive feedback mechanism lead to


is associated with a large fall in irreversible shock and death.
systemic vascular resistance as active • Hypertension results from increased
muscles dilate. To maintain (and cardiac output, usually caused by
elevate) arterial pressure, sympathetic increased blood volume, and from
activation and circulating hormones increased systemic vascular resistance.
increase cardiac output and constrict Essential hypertension is of unknown
blood vessels in other major organs of origin, whereas secondary hypertension
the body; the abdominothoracic and results from identifiable causes such as
skeletal muscle pumps promote venous renal disease, excessive sympathetic
return to maintain adequate cardiac activation, or abnormal levels of
preload conditions. hormones.
• Cardiovascular responses to exercise • Heart failure occurs when the heart is
are significantly influenced by the unable to supply adequate blood flow
type of exercise (dynamic versus to organs, or when it is able to do so
static), body posture, physical only at elevated filling pressures. It may
conditioning, environmental factors, involve systolic dysfunction (depressed
age, gender, and the presence of ventricular inotropy) or diastolic
heart disease. dysfunction (impaired filling).
• Pregnancy is associated with an • The body compensates for heart failure
increase in blood volume and cardiac by activating the sympathetic nervous
output, and a decrease in systemic system, ren in-angiotensin-aldosterone
vascular resistance and mean arterial system, and other circulating
pressure; heart rate gradually increases hormones. These compensatory
during pregnancy. mechanisms increase systemic vascular
• Arterial hypotension can result from resistance, stimulate the heart, and
mechanisms that reduce cardiac output increase blood volume.
or cause systemic vasodilation. • Structural defects in heart valves result
• Compensatory negative feedback in valve stenosis and/or regurgitation,
mechanisms, triggered by hypotension, which affect pressure and volume
help to restore arterial pressure. relationships within the heart during
Baroreceptor and renal mechanisms systole and diastole, and lead to
play a prominent role. Failure of these reduced cardiac output and elevated
mechanisms and the expression of venous pressures.

REVIEW QUESTIONS

For each question, choose the one best b. Sympathetic-mediated vasoconstric-


answer: tion occurs in the skin.
c. Systemic vascular resistance increases
1. During a moderate level of whole-body owing to sympathetic activation.
exercise (e.g., running), d. Vagal influences on the sinoatrial
node are inhibited.
a. Arterial pulse pressure decreases
owing to the elevated heart rate.

Klabunde_Chap09.indd 229 6/11/2011 10:11:48 AM


230 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

2. One important reason why stroke vol- 6. Long-term recovery of cardiovascular


ume is able to increase during running homeostasis following moderate hemor-
exercise is that rhage involves
a. Central venous pressure decreases. a. Aldosterone inhibition of renin
b. Heart rate increases. release.
c. The rate of ventricular relaxation b. Enhanced renal loss (excretion) of
decreases. sodium.
d. Venous return is enhanced by the c. Increased capillary fluid filtration.
muscle pump system. d. Vasopressin-mediated water reabsorp-
tion by the kidneys.
3. In an exercise study, the subject’s resting
heart rate and left ventricular stroke 7. A trauma patient is admitted the Emer-
volume were 70 beats/min and 80 mL/ gency Department following massive
beat, respectively. While the subject was blood loss. The bleeding is controlled,
walking rapidly on a treadmill, the heart and resuscitation with fluids and pres-
rate and stroke volume increased to sor agents elevates the mean arterial
140 beats/min and 100 mL/beat, respec- pressure to 60 mm Hg. Despite addi-
tively; ejection fraction increased from tional efforts to raise arterial pressure,
60% to 75%. The subject’s mean arterial the pressure begins to fall after 2 hours,
pressure increased from 90 mm Hg at and the patient dies. Which of the fol-
rest to 110 mm Hg during exercise. One lowing most likely contributed to the
can conclude that cardiovascular collapse in this patient?
a. Cardiac output doubled. a. Excessive fluid sodium and water
b. Compared to rest, the cardiac output retention by the kidneys
increased proportionately more dur- b. Increased capillary fluid reabsorption
ing exercise than systemic vascular c. Myocardial depression by metabolic
resistance decreased. acidosis
c. Ventricular end-diastolic volume d. Sympathetic-mediated vasoconstric-
increased. tion
d. The increase in mean arterial pressure
during exercise indicates that sys- 8. A 43-year-old female patient consistently
temic vascular resistance increased. has arterial pressure values of about
150/105 mm Hg. This patient’s hyperten-
4. During the second trimester of preg- sion could be the result of
nancy, a. Diminished secretion of aldosterone.
a. Systemic vascular resistance is b. Excessive renal excretion of sodium.
increased. c. Suppressed release of renin.
b. Heart rate is decreased. d. Thyroid disorder.
c. Cardiac output is decreased.
d. Blood volume is increased. 9. The echocardiogram report that you
receive for your patient indicates that he
5. The baroreceptor reflex in hemorrhagic has left ventricular diastolic dysfunction.
shock Which of the following is usually associ-
ated with this condition?
a. Decreases venous compliance.
b. Decreases systemic vascular resist- a. Increased ventricular compliance
ance. b. Elevated end-diastolic pressure
c. Increases vagal tone on the SA node. c. Decreased end-systolic volume
d. Stimulates angiotensin II release from d. Large decrease in ejection fraction
the kidneys.

Klabunde_Chap09.indd 230 6/11/2011 10:11:50 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 231

10. Compared to the maximal exercise 12. A patient is diagnosed with moderately
responses of a normal subject, a patient severe aortic valve regurgitation. In the
with moderate-to-severe heart failure absence of ventricular failure, which of
during maximal exercise will have a the following changes is associated with
a. Lower arterial pressure. this valve defect?
b. Lower arterial-venous oxygen a. Aortic diastolic pressure is
extraction. increased.
c. Higher ejection fraction. b. Aortic systolic pressure is decreased.
d. Similar maximal oxygen consumption. c. Left ventricular stroke volume into
the aorta is increased.
11. A patient with a history of mild hyperten- d. Left ventricular preload is
sion has recently been diagnosed with left decreased.
ventricular systolic dysfunction associated
with dilated cardiomyopathy. In addition 13. A patient is diagnosed with mitral
to a diuretic, the patient is also prescribed valve stenosis with no evidence of
a mixed arterial-venous dilator (e.g., an systolic dysfunction. This patient will
ACE inhibitor). The rationale for adding likely have
the vasodilator is that it will increase a. A systolic murmur.
a. Stroke volume by increasing preload. b. Elevated left atrial pressure.
b. Ventricular afterload by reducing c. Increased left ventricular end-diastolic
preload. pressure.
c. Ventricular ejection fraction by d. Reduced left ventricular ejection
increasing stroke volume. fraction.
d. Ventricular end-systolic volume by
increasing stroke volume.

l. The correct answer is "d" because heart venous pressure would decrease stroke
rate is increased during exercise through volume. Choice "b" is incorrect because
activation of sympathetic adrenergic an increase in heart rate, with no other
nerves and inhibition of vagal (parasym- compensatory changes, decreases stroke
pathetic) nerves on the sinoatrial node. volume. Choice "c" is incorrect because
Choice "a" is incorrect because arterial the rate of ventricular relaxation (lus-
pulse pressure increases during moder- itropy) increases during exercise owing
ate exercise because of the increase in to sympathetic influences, which aids
stroke volume. Choice "b" is incorrect ventricular filling and enhances stroke
because cutaneous vasodilation occurs volume.
during exercise to facilitate heat loss 3. The correct answer is "b" because arteri-
from the body. Choice "c" is incorrect al pressure increased; therefore, cardiac
because systemic vascular resistance falls output must have increased more than
owing to vasodilation in the active skel- systemic vascular resistance decreased
etal muscle. because mean arterial pressure is
2. The correct answer is "d" because the approximately the product of cardiac
muscle pump system facilitates venous output and systemic vascular resistance.
return, which maintains or elevates ven- Choice "a" is incorrect because cardiac
tricular filling pressures. Choice "a" is output (the product of heart rate and
incorrect because a decrease in central stroke volume) increased from 5.6 to

Klabunde_Chap09.indd 231 6/11/2011 10:11:50 AM


232 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

14.0 L/min (i.e., it more than doubled). a compensatory mechanism following


Choice “c” is incorrect because stroke hemorrhage.
volume increased by 25% (from 80 to 7. The correct answer is “c” because
100 mL/beat), and the ejection fraction reduced oxygen delivery to the periph-
increased by 25% (from 60% to 75%). eral organs stimulates anaerobic metabo-
Therefore, end-diastolic volume could lism, leading to metabolic acidosis,
not have changed because ejection which impairs cardiac contraction.
fraction equals stroke volume divided Choices “a,” “b,” and “d” are incorrect
by end-diastolic volume. Choice “d” is because these are normal compensatory
incorrect because the percent change mechanisms that help to maintain arte-
in cardiac output is much greater than rial pressure following hemorrhage.
the percent change in arterial pressure; 8. The correct answer is “d” because either
the systemic vascular resistance can be hypothyroidism or hyperthyroidism can
approximated from the arterial pressure cause hypertension. Choices “a,” “b,”
divided by the cardiac output. and “c” are incorrect because each of
4. The correct answer is “d” because these can decrease blood volume, which
activation of the renin-angiotensin- would decrease arterial pressure.
aldosterone system during pregnancy 9. The correct answer is “b” because dia-
increases blood volume. Choice “a” stolic dysfunction caused by decreased
is incorrect because systemic vascular ventricular compliance (choice “a” is
resistance decreases during pregnancy therefore incorrect) leads to an elevated
owing to the developing uterine circula- end-diastolic pressure at any given end-
tion. Choices “b” and “c” are incorrect diastolic volume. Choice “c” is incorrect
because heart rate and cardiac output because changes in end-systolic volume
increase during pregnancy. are normally associated with changes in
5. The correct answer is “a” because the systolic function. Choice “d” is incor-
baroreceptor reflex activates sympathetic rect because ejection fraction does not
adrenergic nerves that constrict arte- necessarily change much with diastolic
rial and venous vessels. Choice “b” is dysfunction because reduced stroke vol-
incorrect because sympathetic activation ume is usually associated with reduced
increases systemic vascular resistance. end-diastolic volume.
Choice “c” is incorrect because sympa- 10. The correct answer is “a” because car-
thetic activation is accompanied by with- diac output is unable to increase suf-
drawal of vagal tone on the heart. Choice ficiently to maintain arterial pressure as
“d” is incorrect because renin, not angio- systemic vascular resistance falls during
tensin II, is released from the kidneys. exercise. Choice “b” is incorrect because
6. The correct answer is “d” because reduced organ perfusion increases oxy-
long-term recovery from hypovolemia gen extraction from the arterial blood.
requires renal retention of water, which Choice “c” is incorrect because impaired
is partially regulated by vasopres- inotropic responses during exercise
sin. Choice “a” is incorrect because reduce ejection fraction. Choice “d”
increased renin release and subsequent is incorrect because the heart failure
formation of angiotensin II and aldo- patient achieves lower maximal oxygen
sterone contribute to renal reabsorp- consumption because maximal cardiac
tion of sodium and water. Choice “b” is output is reduced.
incorrect because sodium reabsorption, 11. The correct answer is “c” because reduc-
not loss, is enhanced following hemor- ing afterload increases stroke volume
rhage. Choice “c” is incorrect because and reduces ventricular end-diastolic
increased capillary fluid filtration would volume; these changes enhance ejection
decrease blood volume and not serve as fraction. Choice “a” is incorrect because

Klabunde_Chap09.indd 232 6/11/2011 10:11:50 AM


CHAPTER 9 • CARDIOVASCULAR INTEGRATION, ADAPTATION, AND PATHOPHYSIOLOGY 233

the mixed vasodilator decreases pre- diastole, which decreases the diastolic
load as well as afterload. Choice "b" is pressure.
incorrect because afterload is decreased. 13. The correct answer is "b" because with
Choice "d" is incorrect because reducing mitral stenosis, blood has difficulty
afterload leads to a decrease in end-sys- flowing from the left atrium into the
tolic volume, which increases in stroke left ventricle. This leads to blood back-
volume. ing up into the left atrium and increas-
12. The correct answer is "c" because ven- ing its pressure. Choice "a" is incorrect
tricular filling (preload) is increased because turbulence occurs as blood
(choice "d" is therefore incorrect) flows across the narrowed valve during
because blood flows from the aorta diastole, thereby producing a diastolic
back into the ventricle during diastole murmur. Choice "c" is incorrect because
in aortic regurgitation; this increases left ventricular filling can be impaired,
the volume of blood ejected into the which decreases its end-diastolic volume
aorta, which increases aortic systolic and pressure. Choice "d" is incorrect
pressure (choice "b" is therefore incor- because reduced ventricular filling is
rect). Choice "a" is incorrect because accompanied by a reduced stroke vol-
the retrograde flow causes aortic ume; therefore, ejection fraction does
pressure to fall more rapidly during not change much.

CASE 9-1 perfusion, causing fatigue. Decreased cere-


Autonomic neuropathy affects the function bral perfusion caused by hypotension can
of most organ systems of the body because lead to dizziness, visual disturbances, and
autonomic nerves play a vital role in regu- syncope.
lating normal function. In the cardiovascu-
CASE 9-2
lar system, autonomic nerves, particularly
Recovery from hemorrhage partly involves
sympathetic adrenergic nerves, regulate
arterial and venous constriction, cardiac
arterial pressure through their actions on
stimulation, and renal retention of sodium
the heart and vasculature. Patients with type
and water. The diuretic would counter the
2 diabetes who have impaired autonomic
normal renal compensatory mechanisms of
control of the cardiovascular system may
sodium and water retention. The ACE inhibi-
have abnormal responses to exercise because
tor would reduce the formation of circulating
heart rate and inotropy may not increase
angiotensin II that normally plays an impor-
normally, and sympathetic stimulation
tant compensatory role through constricting
of the arterial and venous system may be
blood vessels and increasing blood volume by
impaired. This loss of sympathetic control
enhancing renal reabsorption of sodium and
may result in a fall in arterial pressure dur-
water. The calcium channel blocker, depend-
ing exercise owing to a greater-than-normal
ing upon its class, would depress cardiac
reduction in systemic vascular resistance, a
function and cause systemic vasodilation,
decrease in central venous pressure owing to
both of which would counteract normal com-
loss of venous tone, and a reduction in car-
pensatory responses to hemorrhage. These
diac output caused by smaller-than-normal
drugs, therefore, would impair and prolong
increases in heart rate and stroke volume.
the recovery process following hemorrhage.
Hypotension during exercise impairs muscle

Klabunde_Chap09.indd 233 6/11/2011 10:11:50 AM


234 CARDIOVASCULAR PHYSIOLOGY CONCEPTS

Fortunately, many of these drugs have rela- of the diuretic on the kidney and also cause
tively short half-lives so that their effects dilation of resistance and capacitance ves-
diminish within several hours. sels. These actions would further decrease
end-diastolic pressure by decreasing venous
CASE 9-3
pressure, and would reduce the afterload.
Given that the ejection fraction is 20% and
This latter effect enhances stroke volume
the end-diastolic volume is 240 mL, the
by decreasing the end-systolic volume and
stroke volume is 48 mL/beat using the fol-
increasing the cardiac output. The increased
lowing relationship: stroke volume = ejec-
stroke volume and decreased end-diastolic
tion fraction × end-diastolic volume. The
volume would cause the ejection fraction
end-systolic volume is the end-diastolic vol-
to increase. Although the ACE inhibitor
ume minus the stroke volume, which equals
would decrease systemic vascular resistance,
192 mL. The administration of a diuretic
the increased cardiac output might prevent
would decrease the end-diastolic volume by
arterial pressure from falling, or at least
decreasing blood volume. The ACE inhibi-
partially offset the pressure-lowering effect
tor, by reducing circulating angiotensin II
of systemic vasodilation.
and aldosterone, would reinforce the effects

SUGGESTED RESOURCES Laughlin MH, Korthius RJ, Duncker DJ, Bache RJ.
Chapman AB, Abraham WT, Zamudio S, et al. Temporal Control of blood flow to cardiac and skeletal muscle
relationships between hormonal and hemodynamic during exercise. In: Rowell LB, Shepherd JT, eds.
changes in early human pregnancy. Kidney Int Handbook of Physiology; Exercise: Regulation and
1998;54:2056–2063. Integration of Multiple Systems. New York: Oxford
Chobanian AV, Bakris GL, Black HR, et al. Joint University Press, 1996.
National Committee on prevention, detection, evalu- Lilly LS. Pathophysiology of Heart Disease. 5th Ed.
ation, and treatment of high blood pressure: the JNC Philadelphia: Lippincott Williams & Wilkins, 2011.
7 report. JAMA 2003;289:2560–2572. Rowell LB, O’Leary DS, Kellogg DL: Integration of
Hall JE. The kidney, hypertension, and obesity. cardiovascular control systems in dynamic exer-
Hypertension 2003;41:625–633. cise In: Rowell LB, Shepherd JT, eds. Handbook of
Janicki JS, Sheriff DD, Robotham JL, Wise RA. Cardiac Physiology; Exercise: Regulation and Integration
output during exercise: contributions of the cardiac, of Multiple Systems. New York: Oxford University
circulatory, and respiratory systems. In: Rowell Press, 1996.
LB, Shepherd JT, eds. Handbook of Physiology; Wei JY. Age and the cardiovascular system. N Engl J
Exercise: Regulation and Integration of Multiple Med 1992;327:1735–1739.
Systems. New York: Oxford University Press, 1996.

Klabunde_Chap09.indd 234 6/11/2011 10:11:51 AM


INDEX

Page numbers in italics denote figures; those followed by a t denote tables.

A Aortic valve regurgitation, 226-227


Arachidonic acid metabolites, 152
"a-wave" 63 Arcuate artery, 171, 171
Abdolllln'othoracic pump, 110-112, 111 Arginine vasopressin (AVP), 133, 140, 140-141, 142t, 143
ACE (angiotensin-converting enzyme), 138, 151, 173 Arms, ECG leads, 32-34, 32-35
ACE inhibitors, 139, 216, 222 Arrhythmia, 25, 28, 217, 229
Acetylcholine, 19-20, 53 Arterial baroreceptors, 130-131, 131, 142, 201
Acidosis, 134, 211 Arterial blood pressure, 97-100, 130, 131
Actin, 43 aortic pulse pressure, 9S-100, 99-100
Action potentials, 16-18, 16-21, 20-21, 20t, 36 diastolic pressure, 97
abnormal, 21, 21 mean arterial pressure (MAP), 97-98, 98, 162,
conduction, 21-22, 21-25, 23t, 24--25 162-163, 199, 199t, 212
nonpacemaker, 17, 17-18 regulation, 106-107
pacemaker, 18, 1S-19 systolic pressure, 97
Activation gate, 15 Arterial dilation, 113
Active hyperemia Arterial hypoxia, 175
organ blood flow, 156-157, 157, 162 Arterial pressure, 5-6, 6, 142t, 212
skeletal muscle, 166, 167 Arterial vasodilators, 222
Active tension, 71-72 Arteries
Active transport, 182 anatomy, 93-95, 94, 94t
Active vasodilation, 169 cerebral, 161-162, 162
Adenosine, 53, 150, 160 coronary, 157-158, 158
Adenosine diphosphate (ADP), 45, 150 renal, 171
Adenosine monophosphate (AMP), 150 Arterioles, 94, 94t, 95
Adenosine triphosphate (ATP), 43, 150 Arteriosclerosis, 132
Adenylyl cyclase, 46, 47, 52, 53 Arteriovenous (AV) anastomoses, 168
ADH (antidiuretic hormone), 5, 133, 140, 140-141 AT 1 receptor blockers, 139, 216, 222
ADP (adenosine diphosphate), 45, 150 Atherosclerosis, 161
Adrenoceptors, 82, 128, 166, 200 ATP (adenosine triphosphate), 43, 150
Adventitia, 50, 50 ATP-dependent Ca• pump, 13
Afferent arterioles, 171, 172 ATPase,13
Afferent fibers, 125 Atrial contraction, 75
Afferent nerves, 125 Atrial fibrillation, 28, 28
Afterdepolarizations, 21, 21, 29 Atrial flutter, 28, 28
Afterload, 77, 88 "Atrial kick", 64
defined, 77 Atrial natriuretic peptide (ANP), 5, 139-140, 142t, 143
fiber shortening, 77-79 Atrial systole, 62-64
force-velocity relationship, 77-78, 7S-79 Atrioventricular node (AV node), 22, 22-23, 25, 62, 125, 126
Frank-Starling curve, 79, 79 Augmented limb leads, 33
pressure-volume loops, 79-80 Autocrine substances, 150
stroke volume, 77 Autonomic innervation, 61-62
Age, exercise and, 204 Autonomic nerve activity, 6
Albumin, 191 Autonomic neural control, 124--135, 141-143
Aldosterone, 137, 142t, 214 baroreceptor feedback regulation, 130-134, 131,
Alveolar hypoxia, 175 132, 133
AMP (adenosine monophosphate), 150 cardiac and vascular autonomic receptors, 129-130,
Angina pectoris, 161 129-130
Angiogenesis, 161 chemoreceptors, 134
Angiotensin-converting enzyme (ACE), 138, 151, 173 exercise and, 201
Angiotensin II, 53, 137,138, 13S-139, 142t, 143, 208,214 parasympathetic innervation, 20, 125, 125-126, 126, 127,
Angiotensin receptor blocker (ARB), 139, 216, 222 160, 164, 170
Angiotensinogen, 137 reciprocal sympathetic and vagal activity, 12S-129
ANP (atrial natriuretic peptide), 5, 139-140, 142t, 143 sympathetic innervation, 20, 127, 127-128, 128t, 160, 164
Anrep effect, 82 Autonomic reflexes, 135
Antiarrhythmic drugs, 24, 25 Autoregulation
Antidiuretic hormone (ADH), 5, 133, 140, 140-141 cerebral blood flow, 163, 163
Antihistamines, 193 organ blood flow, 154, 154-155, 176
Aorta, 3, 61, 61, 93-97, 94, 94t, 97 renal circulation, 173
Aortic arch, 130, 131 AV (arteriovenous) anastomoses, 168
Aortic bodies, 134 AV nodal blockade, 126
Aortic coarctation, 215 AV node (atrioventricular node), 22, 22-23, 25, 62, 125, 126
Aortic compliance, aortic pulse pressure, 199, 100 AV node blockage, 24, 24-25
Aortic pulse pressure, 9S-100, 99-100, 118 AV valves, cardiac cycle, 62-66
Aortic stenosis, 224--225 AVP (vasopressin), 133, 140, 140-141, 142t, 143
Aortic valve, 61, 61, 65, 67 Axial reference system, 33, 33
235

Klabunde_Index.indd 235 6/10/2011 11:41:06 PM


236 INDEX

binding to TN-C, 47–48


B cardiac excitation-contraction coupling and, 43–45, 44, 45, 45t
Bainbridge reflex, 133 cardiac ion channels and currents, 14t
Baroreceptor feedback regulation, 130–134, 131, 132, 133 entry into myocytes, 46, 46
Baroreceptor reflexes, 129, 207 regulation of relaxation and, 48–49, 55
Baroreceptors, 6, 142 release by the sarcoplasmic reticulum, 46–47, 47
arterial baroreceptors, 130–131, 131, 142–143, 201 “trigger” calcium, 44
feedback regulation, 130–134, 131, 132, 133 uptake by sarcoplasmic reticulum, 48
hypotension, 207–208, 208 Calcium channel blockers, 17, 19, 20, 216
Basal flow, 148–149 Calcium channels, 15–18
Basilar artery, 161, 162 Calcium-release channels, 44
Beta-adrenoceptor agonists, 82, 212 Calmodulin, 52
Beta-blockers, 216, 222 cAMP (cyclic adenosine monophosphate), 46, 52, 55, 212, 222
Bezod-Jarisch reflex, 135 Capillaries, 93, 94t, 95
Blood flow, 2 (see also Cardiac output; Organ blood flow) edema, 193, 193t
hemodynamics, 100–105, 102–105 mean blood pressure in, 96
local regulation, 149, 151, 163 physical structure, 182
resistance, 100–105 renal, 171–172
Blood loss, cardiac function and, 207–210, 208, 209 transcapillary fluid exchange, 186–192, 187–190, 192, 194
Blood pressure Capillary filtration coefficient (CFC), 188
arterial, 97–100, 118 Capillary fluid exchange model, 192, 192
diastolic pressure, 97 Capillary hydrostatic pressure, 189–190, 194
distribution in vascular system, 95–97, 96 Capillary plasma oncotic pressure, 190–191
hypotension, 110 Capillary recruitment, 165
mean arterial pressure (MAP), 96, 97–98, 98, 118, Carbon dioxide
162–163, 199, 199t, 212 blood flow regulation and, 151
sphygmomanometer, 97, 98 cerebral blood flow and, 163
systolic pressure, 97 removal from tissues, 186
venous, 113–118 Cardiac cycle, 62–67, 63, 67, 68, 87
Blood pressure cuff, 97 altered pressure and volume changes during, 67
Blood vessels (see also entries under Vascular) cardiac output, 67–83, 70–76, 78–82, 84
anatomy, 93–95, 94, 94t diagram, 62, 63
arterial blood pressure, 97–100 intracardiac pressures, 66–67, 67
function, 5 phases, 18–19, 62–66
hemodynamics, 100–105, 102–105 ventricular pressure-volume relationship, 67, 69
pressure and volume, 95–97, 96 Cardiac function, 60–87
radius, 100–102, 102, 105 autonomic neural control, 124–135, 141–143
structure, 49–55, 50–53 humoral control, 135–141
systemic vascular function curves, 113–116, 114–115 regulation, 5–6, 124–143
types, 94 Cardiac function curves, 114, 116–118, 116–118
vascular tone, 107, 107 Cardiac glycosides, 48
venous compliance, 75, 108 Cardiac index, 68
venous return and cardiac output, 113–118 Cardiac ischemia, ECG and, 35
Blood viscosity, 95, 100–102 Cardiac myocytes (see Myocytes)
Blood volume, 96, 96, 107–109 Cardiac output, 87, 119
central venous pressure and, 107–109, 108 central venous pressure and, 98
gravity, 109, 109–110 distribution, 148–149, 149t
neurohumoral activation and, 142t equation, 67
BNP (brain-type natriuretic peptide), 140 exercise and, 199, 199t
Bowditch effect, 82 heart rate, 67, 69
Bowman’s capsule, 171 measurement, 69
Bradycardia, 20, 24, 207 neurohumoral activation and, 142t
causes, 28, 29, 126 pregnancy and, 205, 205, 206
sinus bradycardia, 28 regulation, 69–84, 70–76, 78–82, 84
Bradykinin, 150, 151, 176 to skeletal muscle, 165
Brain units, 68
arteries, 161–162, 162 venous return and, 113
blood flow, 149t, 157, 161–164, 162–164, 175t, 176 Cardiogenic shock, 210, 212
edema, 193 Cardiomyopathy, 216
intracranial pressure, 162, 162 Cardiopulmonary receptors, 133
mean arterial pressure, 162–163 Cardiovascular disease (see also Coronary artery disease;
Brain-type natriuretic peptide (BNP), 140 Hypertension; Hypotension)
Bulk flow, 182, 194 angina pectoris, 161
Bundle branch blocks, 24, 27 arrhythmia, 25, 28, 216, 229
Bundle branches, 22, 23 coronary artery disease, 87, 161
Bundle of His, 22, 23, 29 heart failure, 49, 133, 207, 216–217, 217–218, 219–222,
220, 221t, 229
stenosis, 76, 105, 161
C systolic failure, 82
valve disease, 65, 84
“c-wave”, 65 Cardiovascular function
Cadiopulmonary receptors, 133 exercise and, 198–205, 199t, 200, 201, 202t, 229
Calcitonin gene-related peptide (CGRP), 164 hypertension, 138, 212–216, 213t, 229
Calcium hypotension, 110, 206, 206–212, 208–209, 211, 229

Klabunde_Index.indd 236 6/10/2011 11:41:06 PM


INDEX 237

pain and, 135 Corticosteroids, 193


pregnancy and, 205, 205–206, 229 Cranial nerve IX, 130, 131
regulation, 5–6, 36 Cranial nerve X, 130, 131
Cardiovascular system (see Blood vessels; Cardiac cycle; Critical stenosis, 105
Cardiovascular disease; Cardiovascular function; Cushing reflex, 135
Heart; Myocytes; Vascular system) Cushing’s syndrome, 215
Carotid arteries, 130, 131, 161, 162 Cutaneous blood flow, 149t, 168, 168–169, 175t, 202
Carotid bodies, 134 Cutaneous vasodilation, 202
Carotid sinus baroreceptors, 130, 131, 142 Cyclic adenosine monophosphate (cAMP), 46, 52, 55, 212, 222
Carotid sinus massage, 132 Cyclic guanosine monophosphate (cGMP), 53
Carvedilol, 222 Cyclo-oxygenase inhibitors, 173
Catecholamines, 23, 136, 136–137
Caveolae, 50, 51
Cell membrane, concentration gradients across, 10, 10–11, 36 D
Cell membrane potentials, 36
action potentials, 16–18, 16–21, 20–21, 20t, 36 Decompensatory mechanisms, hypotension, 210–212
ion channels, 14–16, 14t, 15 Delayed afterdepolarization, 21, 21
ionic gradients, 12–14, 13 Dense bands, 50, 51
resting membrane potentials, 10, 10–12, 36 Dense bodies, 50, 51
Central chemoreceptors, 134 Depolarization, 17, 29
Central command, 201 Diabetes, kidneys and, 213
Central nervous system, 125, 125, 126 (see also Neurohumoral Diabetic nephropathy, 214
control) Diaphoresis, 135
Central venous pressure, 107–113, 108–112, 199t Diastole, 62, 65, 158, 159
Cerebral blood flow, 149t, 157, 161–164, 162–164, 175t, 176 Diastolic failure, 217, 218, 219
Cerebral edema, 193 Diastolic murmur, 223
Cerebral hypoxia, 211 Diastolic pressure, 97, 212
Cerebral ischemia, 135, 211 Diffusion, 181, 194
CFC (capillary filtration coefficient), 188 Diffusion constant, 181
cGMP (cyclic guanosine monophosphate), 53 Digoxin, 82, 222
CGRP (calcitonin gene-related peptide), 164 Diltiazem, 17
Chemical gradient, 10 Discontinuous capillaries, 182
Chemoreceptors, 134, 143 Diuretics, 222
Chest leads (ECG), 34–35, 35, 36 Dobutamine, 82, 212, 222
CHF (congestive heart failure), 221, 221t Dopamine, 82, 212
Chordae tendineae, 61, 61 Dorsal vagal nucleus (DVN), 125
Chronic heart failure, 219, 222 Double product, 85
Chronotropy, 19, 62, 125, 126, 128t dP/dt max, 64
Circle of Willis, 161, 162 Driving pressure, 187
Circulating catecholamines, 136, 136–137 Dromotropy, 62
Circulatory system (see Cardiovascular system; Microcirculation) autonomic nervous system, 125, 126, 128t
Circumflex artery, 157, 158 Dynamic exercise, 203, 229
Cluster headaches, 164 Dyspnea, exertional, 216
Cold-induced vasodilation, 169
Cold pressor response, 135
Collateralization, 161 E
Colloid osmotic pressure, 191
Compensatory mechanisms Early afterdepolarization, 21, 21
heart failure, 219–221, 220, 229 ECG (see Electrocardiogram)
hypotension, 207–210 Ectopic foci, 18
Compliance Edema, 193, 193t
aortic, 94 EDHF (endothelial-derived hyperpolarizing factor), 152
defined, 98 EDPVR (end-diastolic pressure-volume relationship), 67
venous, 75, 108, 108, 109 EDV (end-diastolic volume), 64, 70–71, 87
ventricular, 69–71, 70 Effective refractory period (ERP), 17
Compliance curves, veins, 108, 108 Efferent arteriole, 171, 171, 172
Concentration gradient, 181 Efferent parasympathetic fibers, 126
Conduction, abnormal, 24 Efferent vagal fibers, 125
Conduction system, within the heart, 21–22, 21–23 Eicosanoids, 151
Conduction velocity, regulation, 23–24, 23t Einthoven triangle, 33, 33
Congestive heart failure (CHF), 221, 221t Ejection fraction, 66, 81, 87
Continuous capillaries, 182 Ejection murmurs, 65
Contractility (see Inotropy) Electrical activity of the heart, 9–36 (see also
Contraction Electrocardiogram)
excitation-contraction coupling, 43–45, 44–45, 45t abnormal, 24
isovolumetric, 64–65 action potentials, 16–18, 16–21, 20–21, 20t, 36
regulation, 5, 45–47, 45–48, 56 cell membrane potentials, 36
vascular smooth muscle, 51–54, 52–53, 56 conduction, 21–22, 21–23
Coronary arteries, 157, 157, 158 conduction velocity, 23–24, 23t
Coronary artery disease, 87, 161 ion channels, 14–16, 14t, 15
Coronary blood flow, 1–5, 2, 3, 149t, 157–161, 158–160, ionic gradients, 12–14, 13
175t, 176, 202 reentry, 24–25, 24–25
Coronary ostia, 157 resting membrane potentials, 10, 10–12, 36
Coronary sinus, 158 Electrical defibrillation, 29
Coronary veins, 158 Electrical vectors, 29, 30–32, 31

Klabunde_Index.indd 237 6/10/2011 11:41:06 PM


238 INDEX

Electrocardiogram (ECG), 26–35 Glomerular capillaries, 171, 171


interpretation, 28–32, 28, 30, 31 Glomerulonephritis, 214
leads, placing, 32–35, 32–35 Glossopharyngeal nerve, 130, 131, 134
myocardial ischemia and infarction, 35 Glucocorticoids, 215
tracing details, 26, 26–27, 27t Gq-proteins, 47
Electrolytes, 2 Gravity, central venous pressure and, 109, 109–110
End-diastolic pressure-volume relationship (EDPVR), 67 Gs-protein (stimulatory G-protein), 46, 52
End-diastolic volume (EDV), 64, 70–71, 87 Guanylyl cyclase, 53, 53–54, 58
End-systolic pressure-volume relationship (ESPVR), 67
End-systolic volume (ESV), 65
Endocrine hormones, 150 H
Endothelial cells, 54, 54–55, 58
Endothelial-derived hyperpolarizing factor (EDHF), 152 h-gate, 15, 15, 16
Endothelial factors, organ blood flow and, 152, 152–153, 176 Heart, 2, 3, 4–5 (see also Electrical activity of the heart; entries
Endothelin-1 (ET-1), 54, 55, 153, 176 under Cardiac; Myocytes)
Epinephrine, 83, 84, 130, 136, 136–137, 200 anatomy, 2–3, 60–61, 61, 157–158, 158
Equilibrium potentials, 10, 10–11 autonomic neural control, 124–135, 141–143
ERP (effective refractory period), 17, 17–18 blood flow, 2–5, 4, 149t, 157–161, 158–160, 175t, 176, 202
ESPVR (end-systolic pressure-volume relationship), 67 cell structure, 41–49, 57
Essential (primary) hypertension, 213–214, 213t, 215 conduction system, 21–22, 21–23
ESV (end-systolic volume), 65 excitation-contraction coupling, 43–45, 44–45, 45t
ET-1 (endothelin-1), 54, 55, 153, 176 hormone synthesis by, 5
Exchange of gases, 1–2 humoral control, 135–141
Excitation-contraction coupling, 43–45, 44–45, 45t intracardiac pressures, 66–67, 67
Excitatory interneurons, 125 myocardial oxygen consumption, 84–87, 88
Exercise pumping action, 4–5
cardiovascular response to, 198–205, 199t, 200, regulation of contraction, 5, 45–47, 45–48
201, 202t, 229 regulation of relaxation, 48–49
exertional dyspnea, 216 ventricular pressure-volume relationship, 67, 68
heart failure, limitations on, 221–222, 221t Heart disease (see Cardiovascular disease; Coronary artery
Exercise hyperemia, 156 disease)
Exertional dyspnea, 216 Heart failure, 49, 133, 207, 216–217, 217–218, 219–222, 220,
External carotid artery, 131 221t, 229
Extravascular compression, organ blood flow and, 153–154 brain-type natriuretic peptide (BNP) for, 140
causes, 216–217
chronic, 219, 222
F compensatory mechanisms, 219–221, 220, 229
congestive heart failure (CHF), 221, 221t
“Fast response” action potentials, 17 diastolic failure, 217, 218, 219
Fast sodium channels, 15, 15, 16, 23–24 exercise limitations and, 221–222
Fenestrated capillaries, 182 neutral endopeptidase (NEP) inhibitors, 140
Fick principle, 85, 88 systolic failure, 217
Fick’s first law, 181, 183 systolic vs. diastolic dysfunction, 217, 218, 219
Fight-or-fight response, 129 treatment, 222
Filtration, 188, 194 vasopressin and, 141
Filtration constant, 188 Heart rate, 4, 5, 75
First-degree AV nodal block, 28, 28 cardiac output, 67, 69
First heart sound, 64 stroke volume and, 69
Flow-dependent vasodilation, 152 units, 68
Fluid exchange, transcapillary, 186–192, 187–190, 192, 194 Heart sounds, 63, 64–66, 87
Fluid filtration, 2, 186, 187 Heat exhaustion, 204
Fluid flux, 187 Heat stroke, 203
Fluid reabsorption, 186, 187, 194 Hematocrit, 101
Force-velocity relationship, 77–78, 78–79, 81, 81 Hemodynamics, 100–105, 102–105
Fourth heart sound, 64 Hemorrhage, cardiac function and, 207–210, 208, 209
Frank, Otto, 74 Hemorrhagic hypotension, 207–210, 208, 209
Frank-Starling curves, 74–75, 74, 217 Hemorrhagic shock, 131, 210
afterload, 79, 79 Hepatic artery, 170
inotropy, 81, 81 Hepatic circulation, 3, 3
systolic failure and, 217 Hepatic portal vein, 170
Frank-Starling mechanism, 74–75, 74–75, 83, 87, 110–113, 199 Histamine, 150, 152, 176
Functional hyperemia, 156, 162 Hormones
Functional sympatholysis, 160 arterial pressure and, 6
“Funny” (pacemaker) currents, 19 sympathetic stimulation by, 199
synthesis by heart, 5
Humoral control, 135–141
G atrial natriuretic peptide, 5, 139–140
circulating catecholamines, 136, 136–137
Gap junctions, 21, 22, 41, 51 hypotension, 207–208, 208
Gas exchange renin-angiotensin-aldosterone system, 137–139, 138,
diffusion, 181, 194 143, 208
pulmonary circulation, 175 vasopressin, 133, 140, 140–141, 142t, 143
Gastrointestinal system, blood flow, 149t, 169–170, 175t, 176 Hydraulic conductivity, 187
Gender, exercise and, 205 Hydrogen ion, blood flow regulation and, 151
Gi-protein (inhibitory G-protein), 20, 46 Hydrostatic pressure, 190

Klabunde_Index.indd 238 6/10/2011 11:41:07 PM


INDEX 239

Hyperaldosteronism, primary, 139, 214 Isotonic contraction, 77, 79


Hypercapnia, 134 Isovolumetric contraction, 64–65
Hyperemia, organ blood flow, 156, 156–157, 157, 162, 166, 167 Isovolumetric relaxation, 65–66
Hyperkalemia, 20
Hyperpolarization, 12, 18
Hypertension, 212–216, 229 J
causes, 212–213, 213t
essential (primary), 213–214, 213t, 215 Jugular pulse, 63, 66
hyperthyroidism and, 215 Juxtaglomerular cells, 137
kidneys and, 212, 213 Juxtamedullary nephrons, 172
preeclampsia, 215
secondary, 213t, 214–215
treatment, 215–216 K
Hyperthyroidism, 20, 215
Hyperventilation, 175 K+, concentration gradient across cell membrane, 10, 10–11, 35
Hypotension, 110, 206, 206–212, 208–209, 211, 229 K+ channels, 17
causes, 206, 206–207 KACh channel, 20
compensatory mechanisms during, 207–210 Kallikrein, 151
decompensatory mechanisms, 210–212, 211 Kidneys
hemorrhagic, 207–210, 208, 209 blood flow, 149t, 170–173, 171, 172, 175t, 202
treatment, 212 disease, 214
Hypothalamus, 142, 202 humoral mechanisms acting on, 5
Hypothyroidism, 20, 215 hypertension and, 212, 213
Hypoxemia, 134, 163 interdependence of circulatory and organ function, 5
Hypoxia, 175, 211 pressure natriuresis, 212
Hypoxic vasoconstriction, 175 Kininogen, 151

I L
If (pacemaker current), 19 L-type calcium channels, 17–21, 44, 46, 51, 52, 57
Inactivation gate, 15 Large arteries, 94, 94, 94t, 119
Incisura, 65 Leads (ECG), placing, 32–35, 32–35
Inferior vena cava, 61, 95 Left atrium, 2, 3, 4, 61, 61
Inflow resistance, 76 Left axis deviation, 34
Inhibitory G-protein (Gi-protein), 20, 46 Left ventricle (LV), 2, 3, 4, 4, 61, 61
Initial repolarization, 17 Length-dependent activation, 74
Inorganic phosphate, blood flow regulation and, 150–151 Length-independent activation, 80, 82
Inositol triphosphate (IP3), 47 Length-tension relationship, 71–73, 71–74, 80, 80
Inotropic drugs, 222 Leukotrienes, 152
Inotropy, 62, 76, 126, 128, 128t Limb leads (ECG), 32–34, 32–34, 36
antonomic nervous system and, 126, 128t Liver, blood circulation, 3, 3, 170
defined, 80 Local axon reflexes, 169
factors influencing, 82, 82 Lungs
force-velocity relationship, 81, 81 blood flow, 173–175, 175t
Frank-Starling curves, 81, 81 edema, 193
mechanisms, 82–83 Lusitropy, regulation, 48–49, 58
negative inotropy, 62 Lymphatic vessels, 3, 186–187, 187, 194
pressure-volume loops, 81–82, 82
regulation, 45–47, 45–48, 57
stroke volume, 80 M
Instantaneous mean vector, 30
Intercalated disks, 22, 41 m-gate, 15, 15–16
Interlobar arteries, 171 Mean aortic pressure, 97
Interlobular artery, 171, 171 Mean arterial pressure (MAP), 96, 97–98, 98, 118, 162–163,
Internal carotid artery, 131 199, 199t, 212
Interstitial hydrostatic pressure, 190 Mean circulatory filling pressure, 114
Interstitial oncotic pressure, 191 Mean electrical axis, 29–30, 29–31
Intestinal blood flow, 149t, 170, 175t Mean electrical vectors, 29, 30
Intima, 49, 50 Mean pulmonary artery pressure, 174
Intracardiac shunts, 65 Medulla oblongata, 125, 125, 127
Intracranial pressure, 162, 162 Membrane potentials, 10, 10–12
Intrapleural pressure, 111, 174–175 Metabolic acidosis, 211
Ion channels, 14–16, 14t, 15 Metabolic theory of blood flow regulation, 150
Ionic conductance, 11–12 Metabolism, myocyte, 49
Ionic gradients, 12–14, 13 Metanephrine, 215
Ionic pumps, 13, 13–14 Microcirculation, 180
IP3 (inositol triphosphate), 47 active transport, 182
IP3 pathways, 53, 53–54 bulk flow, 182, 194
Ischemia, 34–35, 211 diffusion, 181, 194
Ischemic brain reflex, 135 edema, 193, 193t, 194
Isoelectric period, 26 exchange of oxygen and carbon dioxide, 182–186, 183–185
Isoelectric voltage, 30 filtration, 188, 194
Isometric contraction, 77, 78 transcapillary fluid exchange, 186–192, 187–190, 192, 194
Isoproterenol, 82 vesicular transport, 182

Klabunde_Index.indd 239 6/10/2011 11:41:07 PM


240 INDEX

Migraine headaches, 164 Organ blood flow, 3, 3, 148–176


Milrinone, 82, 212, 222 autoregulation, 154, 154–155, 176
Mitral stenosis, 225 basal flow, 148–149
Mitral valve, 61, 61 cardiac output, distribution of, 148–149, 149t
Mitral valve regurgitation, 227–228, 228 cerebral, 149t, 157, 161–164, 162–164, 175t, 176
Muscarinic receptors, 19, 46, 126, 129, 129 coronary, 149t, 157–161, 158–160, 175t, 176, 202
Muscle and joint stretch receptors, 135 cutaneous, 149t, 168, 168–169, 175t, 202
Muscles, skeletal muscle circulation, 149t, 164–168, 167, endothelial factors, 152, 152–153
175t, 176 gastrointestinal system, 149t, 169–170, 175t, 176
Myocardial infarction, ECG and, 35 hepatic, 3, 3, 170
Myocardial oxygen consumption, 84–87, 160 hyperemia, 156, 156–157, 157, 162, 166, 167
determining, 85 local regulation, 149–157, 176
factors influencing, 86–87, 86t mechanical compression and, 153–154, 176
Fick principle, 85 myogenic mechanisms, 153
Myocarditis, 217 pancreas, 149t, 170
Myocytes physical activity, 202
calcium entry into, 46, 46 pulmonary, 173–175, 175t
excitation-contraction coupling, 43–45, 44–45, 45t renal, 149t, 170–173, 171, 172, 175t, 202
length-tension relationship, 71–73, 71–74, 80, 80 skeletal muscle circulation, 149t, 164–168, 167, 175t, 176
metabolism, 49 splanchnic, 169–170, 202
regulation of calcium efflux by, 48 tissue factors and, 149–152, 150
regulation of contraction, 45–47, 45–48 vasodilator reserve, 149
regulation of relaxation, 48–49 Orthostatic hypotension, 110
resting membrane potentials (Em), 10, 10–12, 35 Osmolarity, blood flow regulation and, 151
structure, 41–43, 42–43, 55 Osmotic pressure, 191
Myofilaments, 42, 43 Overdrive suppression, 18
Myogenic mechanisms, for organ blood flow regulation, Oxygen
51–52, 153 blood flow regulation and, 151
Myosin, 42 delivery and extraction, 183–186, 184, 185
Myosin ATPase (myosin adenosine triphosphatase), 43 diffusion, 182–183, 183
Myosin light chain kinase, 52 Oxygen supply/demand ratio, 160
Myosin light chains, 48, 52

P
N
P wave, 26, 27, 27t, 62
Na+, concentration gradient across cell membrane, 10, 10–11, 35 P-R interval, 26, 27t, 27
Na+ channels, 17 Pacemaker action potentials, 18, 18–20, 20, 36
Na+/K+-ATPase, 13 Pacemaker current (If), 19
NDF (Net driving force), 188, 188–189, 189, 191–192, 194 Pain, cardiovascular function and, 135, 136
Negative chronotropy, 19, 62 Papillary muscles, 61
Negative dromotropy, 62 Paracrine hormones, 150, 176
Negative feedback, 6 Parasympathetic innervation, 20, 61–62, 125, 125–126, 126,
Negative inotropy, 62 127, 128t, 142
NEP inhibitors (neutral endopeptidase inhibitors), 140 brain, 164
Nernst potential, 11 heart, 160
Nerve of Hering, 130 intestine, 170
Nerves, autonomic, 61–62 Paravertebral ganglia, 127, 128
Net driving force (NDF), 188, 188–189, 189, 191–192, 194 Passive tension, 71, 71
Net electrochemical force, 11 PCO2, 134, 186
Net oncotic pressure, 189 Peripheral baroreceptors, 142
Neurohumoral control Peripheral chemoreceptors, 134, 143
autonomic control, 124–135, 141–143 Peritubular capillaries, 171, 171
humoral control, 135–141 Permeability coefficient, 181
integration of mechanisms, 141–142 PGE2 (Prostaglandin E2), 152
Neuropeptide-Y (NPY), 164 PGF2α, 152
Neutral endopeptidase (NEP) inhibitors, 140 PGI2 (Prostacyclin), 53, 152, 173
Nitric oxide, 53–54, 54, 126, 151, 152, 160, 213 pH, 134
Nitric oxide (NO)-cGMP system, 53 Phase 0 depolarization, 18
Nitric oxide synthase (NOS), inhibitors, 152 Pheochromocytoma, 137, 214–215
Nonpacemaker action potentials, 17, 17–18, 36 Phosphodiesterase inhibitors, 212
Norepinephrine, 53, 129, 130, 136, 136, 137, 200 Phospholamban, 48, 49
NOS inhibitors (Nitric oxide synthase inhibitors), 152 Phospholipase C, 47, 52
NPY (Neuropeptide-Y), 164 Phosphorylation, 46
Nucleus ambiguous (NA), 125 Physical activity, cardiovascular response to, 198–205, 199t,
Nucleus tractus solitarius (NTS), 125, 126 200, 201, 202t, 229
Physical conditioning, exercise and, 204
Plasma, viscosity, 101
O Plateau phase, 17
PO2, 134, 163, 183
“Oncotic” pressure, 191 Poiseuille’s equation, 101, 105, 182
Organ (see also Organ blood flow) Polycythemia, 101
parallel arrangement, 104, 119 Pores, 182, 194
perfusion pressure, 154 Positive chronotropy, 19
vascular arrangement, 3, 3, 104–105, 105, 119 Positive feedback cycle, 210

Klabunde_Index.indd 240 6/10/2011 11:41:07 PM


INDEX 241

Post-capillary resistance, 189 Regulation (see also Neurohumoral control)


Post-capillary venules, 95, 96, 194 conduction velocity, 23–24, 23t
Postganglionic fibers, 125, 128 of contraction, 45–47, 45–48
Postural hypotension, 110 Relative refractory period, 18
Potassium, cardiac ion channels and currents, 14t Relaxation, isovolumetric, 65–66
Potassium ion, blood flow regulation and, 151 Renal artery stenosis, 173, 214, 215
Precapillary resistance, 189 Renal blood flow, 149t, 170–173, 171, 172, 175t, 202
Preeclampsia, 215 Renal disease, 214
Preganglionic fibers, 125 Renin, 137, 143
Preganglionic parasympathetic efferent nerves, 142 Renin-angiotensin-aldosterone system, 137–139, 138,
Pregnancy 143, 208, 216
cardiovascular function and, 205, 205–206, 229 Repolarization, 17, 19, 27, 32
preeclampsia, 215 Resistance, blood flow, 100–102, 102
Preload, 47 Resistance vessels, 95, 96, 128, 158, 168
defined, 69 Respiration, venous return and, 110–112, 111
left ventricular preload, 76 Respiratory pump (see Abdominothoracic pump)
length-tension relationship, 71–73, 71–74 Respiratory system, pulmonary blood flow, 173–175
right ventricular preload, 75, 76 Resting heart rate, 127
stroke volume, 74–75, 74–75 Resting membrane potential (Em), 10, 10–12, 36
ventricular compliance, 69–71, 70 Rhythm strip, 26, 28
ventricular preload, 75–76, 76 Right atrium, 2, 2, 4, 4, 60–61, 61
Premature depolarization, 29 Right axis deviation, 34
Pressure (see also Arterial blood pressure; Venous blood Right venticular preload, 75, 76
pressure) Right ventricle, 2, 2, 4, 4, 61, 61
intracardiac pressures, 66–67, 67 Rostral ventrolateral medulla (RVLM), 127
perfusion pressure, 154, 175 Ryanodine receptors, 44
ventricular pressure-volume relationship,
67, 68
Pressure natriuresis, 212 S
Pressure pulse, 97
Pressure-rate product, 85 SA nodal arrest, 126
Pressure-volume loops, 67 SA node (sinoatrial node), 19–20, 20, 20t, 62, 125
afterload and, 79, 80, 79–80 Sarcolemma, 44
heart failure and, 217, 218, 219 ion channels, 14–16, 14t, 15
inotropy and, 81–82, 82 ion pumps and exchanges, 12–14, 13
Prevertebral ganglia, 127, 128 Sarcomeres, 42, 42
Primary (essential) hypertension, 213–214, 213t, 215 length, 69, 70–71
Primary hyperaldosteronism, 139, 214 sliding filament theory, 44, 45
Prostacyclin (PGI2), 53, 152, 173 Sarcoplasmic reticulum, 43–44, 44, 45
Prostaglandin E2 (PGE2), 152 Second-degree AV nodal block, 28, 29
Prostaglandins, 150, 173, 176 Second heart sound, 65, 87
Protein kinase A (PK-A), 46 Secondary hypertension, 138, 213t, 214–215
Proximal tubule, 171 Semilunar valve, 61
PS product, 181 Semipermeable membrane, 191
Pulmonary artery, 61, 174 Septic shock, 206, 210
Pulmonary blood flow, 173–175, 175t SERCA, 44, 45, 46, 48–49
Pulmonary capillary wedge pressure, 217 Severe hypotension, 210
Pulmonary circulation, 2, 3, 4 Shock
Pulmonary edema, 193 cardiogenic shock, 210, 212
Pulmonary stretch receptors, 135 hemorrhagic, 131, 210
Pulmonary stretch reflex, 135 irreversible, 211–212
Pulmonary vasoconstriction, 175 septic, 206, 210
Pulmonary veins, 61 Signal transduction, cardiac muscle, 47
Pulmonary vessels, 174 Signal transduction, vascular smooth muscle contraction, 53
Pulmonic regurgitation, 228 Sinoatrial node (SA node), 19–20, 20, 20t, 62, 125
Pulmonic stenosis, 225–226 Sinus bradycardia, 28
Pulmonic valve, 61, 61, 62–67 Sinus nerve, 130
Pulse pressure, 97 Sinus rhythm, 28
Purkinje fibers, 23 Sinus tachycardia, 28
Sinusoids, 170
SIRS (systemic inflammatory response syndrome), 206
Q Skeletal muscle circulation, 149t, 164–168, 167, 175t, 176
Skeletal muscle pump, 110, 110
Q-T interval, 26, 27t, 27 Skin
QRS complex, 26, 27t, 27, 29, 30, 31, 65 blood flow, 149t, 168, 168–169, 175t, 202
vasodilation, 202
Sleep apnea, 215
R Sliding filament theory, 44, 45
“Slow response” action potentials, 18
RA (right atrium), 2, 2, 4, 4, 60–61, 61 Small arteries, 94, 94t, 95
Rate of diffusion, 181 Smooth muscle cells, vascular, 51–54, 52–53, 153
Reactive hyperemia, organ blood flow, 156, 156 Sodium, cardiac ion channels and currents, 14t
Receptor-gated channels, 15 Sodium-calcium exchanger, 14
Reentry mechanism, 24–25, 24–25 Sodium-potassium ATPase, 13
Reflection coefficient, 191 Sodium channels, 15, 15, 16, 18, 21

Klabunde_Index.indd 241 6/10/2011 11:41:07 PM


242 INDEX

Sodium current, 16 Transcapillary fluid exchange, 186–192, 187–190, 192, 194


Sphygmomanometer, 97, 98 “Transient” calcium channels, 19
Splanchnic blood flow, 149t, 169–170, 202 Transmural pressure, 111
Spleen, blood flow, 149t, 170 Transverse tubules (T tubules), 43
ST segment, 26, 27, 27t Tricuspid regurgitation, 228
Standard limb leads, 32 Tricuspid valve, 61
Starling, Ernest, 74, 189 Tricuspid valve stenosis, 76, 226
Starling forces, 189 “Trigger” calcium, 44
Starling’s law of the heart, 74 Triple response, 169
Static exercise, 203 Tropomyosin, 43
Stenosis, 76, 105, 161 Troponin-C (TN-C), 43, 43, 46, 47–48
Stimulatory G-protein (Gs-protein), 46, 52 Troponin-I (TN-I), 43, 43
Stomach, blood flow, 149t Troponin-T (TN-T), 43, 43
Stroke volume, 4 Troponin-tropomyosin complex, 43, 44
afterload and, 77 Tubuloglomerular feedback, 173
exercise and, 202, 202t
Frank-Starling mechanism, 74–75, 74–75, 84, 87
heart rate, 67, 69 V
hypotension and, 207
inotropy, 80, 80 V1-V6 leads, 35, 35
pregnancy and, 205, 205–206 “v-wave”, 66
preload, 69 Vagal activation, 62
regulation, 69–84 Vagal stimulation, 19–20, 20, 126
units, 68 Vagal tone, 19, 125
Superior vena cava, 61, 61, 95 Vagus nerve, 61–62, 125, 131
Supraventricular tachycardia, 25 left vagus nerve, 62, 126
Sympathetic afferent nerve, 62 right vagus nerve, 61–62, 125–126
Sympathetic efferent fibers, 61 Valsalva maneuver, 112, 113, 203
Sympathetic escape, 167 Valve disease, 65, 84
Sympathetic innervation, 20, 61–62, 127, 127–128, 136, 142, Valve regurgitation, 226, 226–228, 228
143, 160, 163–164 Valve stenosis, 223–226, 224, 225
Sympathetic nerves, 200 Vanillylmandelic acid, 215
Sympathetic neurons, 127 Varicosities, 128
Syncytium, 41 Vasa recta, 172
Systemic circulation, 2, 94t, 96, 97–98 Vascular endothelial cells, 54, 54–55, 58
Systemic inflammatory response syndrome (SIRS), 206 Vascular function, 5
Systemic vascular function curve, 113–116, 114, 115 interdependence of circulatory and organ function, 5,
Systemic vascular resistance, 6, 118, 199t 104–105, 105, 118
calculation, 106 regulation, 5–6
hemodynamics, 100–105, 102–105 Vascular resistance, 4, 106, 118–119, 175
Systole, 62, 65, 159 Vascular smooth muscle, 50–54, 51
Systolic failure, 82, 217 cellular structure, 50–51, 51
Systolic murmur, 223 contraction, 51–54, 52–53
Systolic pressure, 97, 212 Vascular structure, 49–55
adventitia, 50, 50
endothelial cells, 54, 54–55
T intima, 49–50, 50
media, 50, 50
T tubules (transverse tubules), 43 smooth muscle cells, 50–54, 51–53
T-type calcium channels, 19 Vascular system, 5, 93–119 (see also Blood flow; Blood vessels;
T wave, 27t, 27, 65 Organ blood flow)
Tachycardia, 20 anatomy, 2–3, 93–95, 94, 94t
sinus, 28 arterial blood pressure, 97–100
supraventricular, 25 autonomic neural control, 124–135, 141–143
ventricular, 28, 29 cardiac function curves, 114, 116–118, 116–118
Temperature reflex, 135 exercise and, 198–205, 199t, 200, 201, 202t, 229
Terminal cisternae, 43 function, 93
Thebesian vessels, 158 hemodynamics, 100–105, 102–105
Thermoreceptors, 135, 202 humoral control, 135–141
Thick filaments, 42, 43, 43 lungs, 174
Thin filaments, 42, 43, 43 microcirculation, 180–182
Third-degree AV nodal block, 29 of organs, 5, 104–105, 105, 118
Third heart sound, 66 pregnancy and, 205, 205–206, 229
Thromboxanes, 152 pressures and volumes, 95–97, 96
Tissue hydrostatic pressure, 190 systemic circulation, 2, 94t, 96, 97–98
Tissue oncotic pressure, 191 systemic vascular function curve, 113–116, 114, 115
Titin, 42 systemic vascular resistance, 106
TN-C (troponin-C), 43, 43, 46, 47–48 varicosities, 128
TN-I (troponin-I), 43, 43 vascular tone, 107, 107
TN-T (troponin-T), 43, 43 venous blood pressure, 107–113
Tonic contraction, 51 venous return and cardiac output, 113–118
Total blood volume, central venous pressure, 112 Vascular tone, 107, 107
Total peripheral resistance (TPR) (see Systemic vascular Vasoactive intestinal polypeptide (VIP), 164
resistance) Vasoconstriction, 175, 176

Klabunde_Index.indd 242 6/10/2011 11:41:07 PM


INDEX 243

Vasodilation, 164, 176 Ventricular relaxation, 62, 70


active, 169 Ventricular stroke volume, 91, 110, 111, 201
cold-induced, 169 Ventricular stroke work, 67
cutaneous, 202 Ventricular tachycardia, 28, 29
flow-dependent, 152 Ventricular wall hypertrophy, 77
systemic vasodilators, 212 Venules, 94t, 95, 96
Vasodilator reserve, 149 Verapamil, 17, 216
Vasopressin, 133, 140, 140–141, 142t, 143, 208 Vertebral arteries, 161, 162
Vasovagal syncope, 129 Vesicular transport, 182
Veins, 93, 94t, 95, 108 VIP (vasoactive intestinal polypeptide), 164
Vena cava, 61, 94–96, 94t Voltage-gated channels, 15
Venous blood pressure, 107–113, 108–112
Venous blood volume, 96, 96, 107–109, 108
Venous compliance, 75, 108, 108, 109 W
Venous plexus, 168
Venous pressure, 75 Wall stress, 77
Venous return, cardiac output, 113–118
Ventilation-perfusion ratio, 175
Ventricular afterload (see Afterload) X
Ventricular arrhythmia, 25
“x-descent”, 64
Ventricular compliance, preload and, 69–71, 70
“x’-descent”, 65
Ventricular diastasis, 66
Ventricular dilation, 70, 77
Ventricular ejection, 94, 97
Ventricular ejection fraction, 87
Y
Ventricular fibrillation, 28, 29 “y-descent”, 66
Ventricular flutter, 29
Ventricular hypertrophy, 70
Ventricular inotropy, 76 Z
Ventricular preload (see Preload)
Ventricular pressure-volume relationship, 67, 68 Z-lines, 42

Klabunde_Index.indd 243 6/10/2011 11:41:07 PM

Vous aimerez peut-être aussi